Neurobiology of Addiction
Neurobiology of Addiction
An integrative review
Aviel Goodman*
Minnesota Institute of Psychiatry, 1347 Summit Avenue, St. Paul, MN 55105, USA
Addiction neuroscience may well be our eras most dynamic
eld  of  scientic  inquiry.   The  quality  as  well   as  quantity  of
information  that   it   generates   seems   almost   to  exceed  the
capacity of our theories to organize and make coherent sense
of  it.   This  article  offers  a  framework  that  may  be  helpful  in
organizing  and  integrating  the  wealth  of  information  that  is
the   neurobiology   of   addiction.   The   cornerstone   of   this
framework is the addictive process, an underlying biopsycho-
logical   process  that  addictive  disorders  are  hypothesized  to
share.  The  article  begins  by  introducing  the  hypothesis  that
psychoactive substance use disorders, bulimia nervosa (from
here on, bulimia), pathological gambling, and sexual addiction
share   an   underlying   biopsychological   process.   Research
ndings are demonstrated to accord with empirically testable
predictions that were generated from the hypothesis, thereby
conrming  it.  Denitions  are  then  offered  for  the  key  terms,
addiction and addictive process. The addictive process is brought
into focus as an interactionof impairments in three functional
systems: motivation-reward, affect regulation, and behavioral
inhibition.   The   review  itself   then   follows,   in   which   the
literature   that   addresses   the   neurobiology   of   addiction  is
selectively   organized   according   to   the   three   functional
systems  that  constitute  the  addictive  process.  The  review  is
directed  toward  identifying   candidate   neurochemical   sub-
strates   for   the   impairments   in   motivation-reward,   affect
regulation,   and  behavioral   inhibition  that   could  contribute
to an addictive process.
1.   Addictive disorders
1.1.   A shared underlying process
In the course of my work with individuals who suffered from
psychoactive  substance  use  disorders,   bulimia,   pathological
gambling, or sexual addiction, I noticed that these conditions
shared  a  number   of   characteristic   clinical   features.   These
included: (1) course of illness  the disorder typically begins in
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2
a r t i c l e   i n f o
Article history:
Received 8 May 2007
Accepted 23 July 2007
Keywords:
Addiction
Neurobiology
Neuroscience
Substance abuse
Bulimia
Gambling
a b s t r a c t
Evidence  that  psychoactive  substance  use  disorders,   bulimia  nervosa,   pathological   gam-
bling,  and  sexual  addiction  share  an  underlying  biopsychological  process  is  summarized.
Denitions  are  offered  for   addiction  and  addictive   process,   the  latter   being  the  proposed
designation   for   the   underlying   biopsychological   process   that   addictive   disorders   are
hypothesized to share. The addictive process is introduced as an interaction of impairments
in  three  functional  systems:   motivation-reward,   affect  regulation,  and  behavioral  inhibi-
tion. An integrative review of the literature that addresses the neurobiology of addiction is
then  presented,   organized  according  to  the  three  functional   systems  that  constitute  the
addictive  process.   The  review  is   directed  toward  identifying  candidate  neurochemical
substrates  for   the  impairments  in  motivation-reward,   affect   regulation,   and  behavioral
inhibition that could contribute to an addictive  process.
# 2007 Elsevier Inc. All rights reserved.
*   Tel.: +1 651 649 0847.
E-mail address: aviel@synergystudios.org.
avai l abl e  at   www. sci encedi r ect . com
j our nal   homepage:   www. el sevi er . com/ l ocat e/ bi ochemphar m
0006-2952/$  see front matter # 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.bcp.2007.07.030
adolescence or early adulthood and follows a chronic course
with  remissions  and  exacerbations;  (2)  behavioral  features  
narrowing   of   behavioral   repertoire,   continuation   of   the
behavior   despite   harmful   consequences;   (3)   individuals
subjective  experience  of   the  condition     sense  of   craving,
preoccupation, excitement during preparatory activity, mood-
altering  effects  of  the  behavior,   sense  of  loss  of  control;   (4)
progressive  development  of  the  condition    craving,   loss  of
control, narrowing of behavioral repertoire, and harmfulness
of consequences all tending to increase as the duration of the
condition   increases;   (5)   experience   of   tolerance      as   the
behavior is repeated, its potency to produce reinforcing effects
tends to diminish; (6) experience of withdrawal phenomena 
psychological   or   physical   discomfort   when  the  behavior   is
discontinued; (7) tendency to relapse  i.e., to returnto harmful
patterns of behavior after a period of abstinence or control has
been  achieved;   (8)   propensity  for   behavioral   substitution  
when  the  behavioral   symptoms  of   the  disorder  have  come
under  control,   tendency  for  addictive  engagement   in  other
behaviors to emerge or intensify; (9) relationship between the
condition and other aspects of affected individuals lives  for
example, neglect of other areas of life as the behavior assumes
priority;   and  (10)   recurrent   themes  in  the  ways  individuals
with  these  conditions  relate  to  others  and  to  themselves  
including low self-esteem, self-centeredness, denial, rationa-
lization,   and   conicts   over   dependency   and   control.   In
addition,   I   noticed  that   among   my   patients   who  suffered
from  psychoactive  substance  use  disorders,   bulimia,   patho-
logical   gambling,   or  sexual   addiction,   more  were  comorbid
with  at   least   one  of   the  other   three  conditions   and  more
reported  relatives  with  at  least  one  of  the  other  three  than
chance  would  have  predicted.   On  the  basis  of   the  clinical
features   that   these   four   conditions   shared,   as   well   as
similarities in  their  diagnostic  criteria,  I  provisionally  identi-
ed  them  as   addictive  disorders   (denition  to  follow).   My
observations   further   led   me   to   speculate   that   the   four
conditions  and, by extension, addictive disorders in general
 had something important in common. In 1990, I [1] proposed
the following hypothesis:
A  hypothesis  may  be  submitted,  the  gist  of  which  is  that
similar   patterns   in   behavioral   manifestations   of   the
various  addictive  disorders. . .reect   similarities  in  some
set of personality and/or biological variables, which may or
may not be measurable by instruments currently available.
In   other   words,   addictive   disorders   would   be   most
accurately  described,   not   as  a  variety  of   addictions,   but
as   a  basic   underlying  addictive  process,   which  may  be
expressed in one or more of various behavioral manifesta-
tions.
In brief, the hypothesis is that addictive disorders share an
underlying biopsychological process. From this hypothesis, a
number of empirically testable predictions can be generated:
(1)   A  person   who   has   been   diagnosed   with   an   addictive
disorder is at a signicantly higher risk than is the general
population to develop (or to have developed), at some point
in  his   or   her   life,   one   or   more   of   the   other   addictive
disorders.
(2)   Biological   relatives   of   an   individual   who   has   been
diagnosed  with  an  addictive  disorder  are  at  signicantly
higher risk than is the general population to develop (or to
have developed), at some point intheir lives, one or more of
the other addictive disorders.
(3)   Symptoms   of   the   various   addictive   disorders   respond
similarly   to   at   least   one   major   class   of   psychiatric
medications.
(4)   Results of at least one major class of empirically validated
psychological   tests  are  similar  for  individuals  who  have
been diagnosed with any one of the addictive disorders.
(5)   Addictive   disorders   have   in   common   one   or   more
identiable patterns of neurobiological activity, structure,
and development.
(6)   Individuals who have been or who will be diagnosed with
an  addictive  disorder  tend  to  exhibit  one  or  more  other
observable  manifestations  of  biopsychological  pathology,
such   as   symptoms   of   another   psychiatric   disorder   or
dysfunctional behavior patterns, prior to the onset of the
addictive disorder.
These   predictions   enable   scientic   assessment   of   the
validity  of  the  hypothesis  that  addictive  disorders  share  an
underlying  biopsychological   process.   To  the  extent  that  the
ndings of research accord with the predictions, the hypoth-
esis  is  conrmed  (factoring  in  that   the  predictions  vary  in
salience).   To  the  extent  that  they  do  not  accord  the  predic-
tions, the hypothesis is disconrmed. Of course, conrmation
or  disconrmation  is  relative  to  the  data  that  currently  are
available  particularly in a eld as dynamic as neuroscience,
where  potentially  signicant   data  emerge  with  astonishing
frequency.
A  wealth  of   published  research  ndings  are  relevant   to
assessing the accuracy of the six aforementioned predictions,
and hence the validity of the hypothesis fromwhichthey were
generated. Addictive disorders that have been the subjects of
enough  research  to  be  considered  in  the  validation  process
include   psychoactive   substance   use   disorders,   bulimia,
pathological   gambling,   and  sexual   addiction.   Findings  that
are   relevant   to  the   six  predictions      lifetime   comorbidity,
family  history,   response  to  medications,   psychometric  stu-
dies, neuroscience research, and temporal/predictive relation-
ships  will now be briey reviewed.
1.1.1.   Lifetime  comorbidity
Epidemiological and clinical research ndings indicate that a
person who has been diagnosed with psychoactive substance
dependence with respect to one type of substance (including
ethanol)   is  at  a  signicantly  higher  risk  than  is  the  general
population  for  dependence  on  (i.e.,   addictive  use  of)   one  or
more  other  psychoactive  substances  at  some  point  in  his  or
her  life  [29].  More  broadly,  research  ndings  indicate  that  a
person who has been diagnosed with psychoactive substance
dependence, bulimia, pathological gambling, or sexual addic-
tion   is   at   a   signicantly   higher   risk   than   is   the   general
population to develop (or to have developed), at some point in
his  or  her  life,  one  of  the  other  disorders.  A  person  who  has
been  diagnosed  with  bulimia  is  at  a  signicantly  higher  risk
than   is   the   general   population   to   develop   (or   to   have
developed),   at  some  point  in  his  or  her  life,   a  psychoactive
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   267
substance   dependence   [1032],   and   vice   versa   [3337].   A
person  who  has  been  diagnosed  with  pathological  gambling
is at a signicantly higher risk thanis the general populationto
develop (or to have developed), at some point in his or her life,
a psychoactive substance dependence [3,3557], and vice versa
[3,5867].   A  person  who   has   been  diagnosed   with  sexual
addiction  is  at  a  signicantly  higher  risk  than  is  the  general
population to develop (or to have developed), at some point in
his or her life, a psychoactive substance addiction [6876], and
vice  versa  [77,78].   A  person  who  has  been  diagnosed  with
pathological gambling is at a signicantly higher risk than is
the  general  population  to  develop  (or  to  have  developed),   at
some point in his or her life, sexual addiction [45,79], and vice
versa [71]. And a person who has been diagnosed with sexual
addiction  is  at  a  signicantly  higher  risk  than  is  the  general
population to develop (or to have developed), at some point in
his or her life, an eating disorder or pathological gambling or
both [68,73,75,76].
Studies have found that these four conditions  psychoac-
tive  substance  dependence,   bulimia,   pathological   gambling,
and   sexual   addiction     are   associated   also   with  affective
disorders,   anxiety  disorders,   attention  decit   disorder,   and
personality disorders at frequencies that are higher than  are
their   frequencies   in  the   general   population.   Affective   dis-
orders, primarily major depression, have a signicant degree
of  comorbidity  with  ethanol   dependence  [334,3544,8083],
other   psychoactive   substance   use   disorders   [2,4,5,8488],
bulimia  [15,16,32,8991],   pathological   gambling  [40,41,44,45,
51,9294,51,9597], and sexual addiction (paraphilic disorders
in most of these studies) [69,7176,98101]. Anxiety disorders
have   a   signicant   degree   of   comorbidity   with   ethanol
dependence  [2,79,80],   other   psychoactive  substance  depen-
dence   [3,4,85,86,88],   bulimia   [16,32,89,102,103],   pathological
gambling   [42,44,47,50,96],   and   sexual   addiction  (paraphilic
disorders  in  most   of   these  studies)   [29,71,7278,98,100,104].
Attention  decit  disorder  has  a  signicant  degree  of  comor-
bidity with ethanol dependence [105107], other psychoactive
substance   dependence   [108,109],   bulimia   [32],   pathological
gambling   [97,107,110,111],   and  sexual   addiction  (paraphilic
disorders in most of these studies) [74,112114]. And person-
ality  disorders  have  a  signicant  degree  of  comorbidity  with
ethanol   dependence  [2,13,115117],   other  psychoactive  sub-
stance dependence [3,85,115118], bulimia [32,119,120], patho-
logical   gambling   [47,97,111,121123],   and   sexual   addiction
[73,77,100,124126].
1.1.2.   Family history
Family history studies indicate that biological relatives of an
individual   who  has  been  diagnosed  with  psychoactive  sub-
stance dependence, bulimia, pathological gambling, or sexual
addiction  are  at  signicantly  higher  risk  than  is  the  general
population to develop (or to have developed), at some point in
their lives, one of the other disorders. First-degree relatives of
a  person  who  has  been  diagnosed  with  psychoactive  sub-
stance dependence are at a signicantly higher risk than is the
general   population   for   pathological   gambling   [41,43,44,
120,129].   First-degree   relatives   of   a   person  who   has   been
diagnosedwithbulimia are at a signicantly higher risk thanis
the   general   population  for   psychoactive   substance   depen-
dence   [12,1517,34,127,128].   First-degree   relatives   of   an
individual   who  has  been  diagnosed  with  pathological   gam-
bling  are  at   a  signicantly  higher   risk  than  is   the  general
population for compulsive overeating [129] and for psychoac-
tive  substance  dependence  [41,44,129,130].   And  rst-degree
relatives  of   a  person  who  has  been  diagnosed  with  sexual
addiction are at a signicantly higher risk than is the general
population  for   psychoactive  substance  dependence,   for   an
eating disorder, and for pathological gambling [69].
1.1.3.   Response  to medications
Research  indicates  that  symptoms  of  the  conditions  that  we
are considering respond similarly to a number of psychiatric
medications.   Antidepressants,   particularly  those  that   affect
the serotonin system, have been found to reduce craving and/
or  symptomatic  behavior  in  ethanol   dependence  [131141].
1
other  psychoactive  substance  dependence  [141149], bulimia
[150157], pathological gambling [154161], and sexual addic-
tion   [69,101,104,162170].   Opioid   antagonists,   most   often
naltrexone, have been found to be effective in treating ethanol
dependence   [171177],   other   nicotine/tobacco   dependence
[178,179],  pathological  gambling  [180183],  and  sexual  addic-
tion  [184,185].   Studies  have  supported  the  efcacy  of   stabi-
lizers,   primarily   topiramate,   in   the   treatment   of   ethanol
dependence   [186188],   cocaine   dependence   [189],   bulimia
[190193], pathological gambling [194197], and sexual addic-
tion [198200].
1.1.4.   Psychometric  studies
Similar patterns of results have been reported for alcoholics,
drug   abusers,   bulimics,   and  pathological   gamblers   on  the
Minnesota  Multiphasic  Personality  Inventory  (MMPI)  and  on
the  MacAndrew  Alcoholism  Scale.   Research  with  the  MMPI
has  demonstrated  similar  proles  for  alcoholics  and  heroin
addicts  [reviewed  in  201],   for  women  with  ethanol   or  other
drug abuse problems and women with bulimia (who have no
history   of   substance   abuse)   [202],   and   for   alcoholics   and
nonalcoholic   pathological   gamblers   [203,204].   Using   the
MacAndrew  Alcoholism  Scale,   researchers   have  found  the
same  range  of   scores  for  problem  drinkers,   heroin  addicts,
massively   obese   individuals,   and   smokers   [205207].   In   a
comprehensive  review,   Sutker   and  Archer   [208]   concluded
that   alcoholics,   opiate  addicts,   and  abusers   of   other   illicit
drugs share commonconstellations of MMPI features; but they
noted that alcoholics, as abusers of a socially sanctioned drug,
differ   from  abusers  of   illicit   drugs  on  dimensions  of   social
nonconformity   and  neurotic   symptomatology.   Since   MMPI
proles  for  these  groups  of   patients  are  not   homogeneous,
1
While Cornelius et al. [139] found that treatment of alcoholism
with antidepressants is effective only in the context of comorbid
depression or anxiety, the studies that are cited here specically
excluded subjects who had any (axis I) psychiatric diagnosis other
than alcohol abuse/dependence. Nonetheless, I agree with Corne-
lius   et   al.      with  the  caveat   that   the  comorbid  depression  or
anxiety  do  not  always  meet  the  criteria  for  DSM  diagnosis.   As  I
discuss  in  the  next  section,  I  believe  that  some  kind  of  affective
dysregulation  is  an  underlying  component  of  all  addictive  disor-
ders.  However,  the  affective  and/or  anxiety  symptoms  are  often
chronic, unconsciously excluded from the alcoholics awareness,
and  difcult  to  dissect  out  of  the  alcoholics  personality,  so  they
may  tend to y under the average clinicians  radar.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 268
some investigators have attempted to delineate homogeneous
MMPI prole subgroups with the help of multivariate  cluster
analysis.   Almost   all   studies   of   this   nature   have   been
conducted  with  alcoholics.   A  review  of   these  studies  [208]
found that they consistently delineated two major subtypes or
clusters, a neurotic subtype and a sociopathic subtype. Similar
delineations of two major subtypes or groups of subtypes were
found in one study of opiate addicts [209] and in two studies of
pathological gamblers [204,210].
Assessments   of   eld   dependence   have   yielded   similar
results of greater eld dependence (poorer performance onthe
Rod-and-Frame   Test)   for   alcoholics   [reviewed   in   201],   for
heroin  addicts  [211],   and  for   obese  individuals  [212,213].   A
related   condition,   overdependence   on   external   cues   and
impaired ability to recognize or correctly to interpret internal
cues,  has  been  found  to  be  associated  with  both  alcoholism
[213,214] and obesity [213,215].
1.1.5.   Neuroscience  research
Research  in  the   areas   of   clinical   phenomenology,   lifetime
comorbidity,   family   history,   response   to  medications,   and
psychometric studies has provided grounds for inferring that
psychoactive   substance   dependence,   bulimia,   pathological
gambling,  and  sexual  addiction  share  an  underlying  biopsy-
chological   process.   Neuroscience  research  has  accepted  the
challenge of this inferred underlying process by investigating
it directly.
To  set  the  stage,  studies  with  pairs  of  twins  have  yielded
evidence for a shared or common vulnerability that underlies
the abuse of psychoactive substances, regardless of the type of
substance   [216218].   These   studies   found  that   the   shared
vulnerability  comprised  both  genetically  determined  factors
and environmentally determined factors. Other studies either
did not examine the environmental component or found it to
be more substance-specic, while still concluding that most of
the  inherited  predisposition  to  abuse  different  psychoactive
substances  converges  in  a  shared  or   substance-nonspecic
liability [reviewed in 219221].
Neuroscience  research  has  led  beyond  demonstrating  a
shared vulnerability that underlies the abuse of psychoactive
substances  toward delineating the neurobiological  processes
that constitute this vulnerability. Among those mentioned are
dysregulated  mesolimbic  DA  circuits  [222224],   reduction  in
DA  D
2
  receptors  [224229],  abnormalities  in  the  orbitofrontal
cortex   and   the   anterior   cyngulate   gyrus   [228,230232],
abnormalities   in  the   ventromedial   prefrontal   cortex   [233
235],   genetic  variants  of   cannabinoid  receptor   1  (CB
1
/Cnr1)
[236,237],   up-regulation  of  brain-derived  neurotrophic  factor
(BDNF)   [238],   and   impaired   leptin   activity   [228,239].   This
research   also   has   expanded   the   realm   of   this   shared
vulnerability to include pathological gambling and pathologi-
cal   use   of   the   natural   rewards   food   and   sex,   as   well   as
psychoactive substance abuse [227,231,239248].
1.1.6.   Temporal/predictive relationships
Correlations  between  behavioral  syndromes  and  patterns  of
psychometric or neuroscience ndings raise questions about
their   causal   relationships.   Do  biological   and  social   conse-
quences of the behavioral syndromes cause the abnormalities
of   psychological   and   neurobiological   functioning   that   are
documented in the research ndings? Or do the abnormalities
of   psychological   and   neurobiological   functioning   that   are
documented  in  the  research  ndings  predispose  to  develop-
ment of the behavioral syndromes? While empirical research
does   not   provide   causal   information,   it   can   illuminate
temporal   and   predictive   relationships   from  which   causal
relationships may be inferred.
Archival   studies   found   elevations   in   the   MMPI   and
MacAndrew  scale   scores   of   young   individuals   who   later
became  abusers  of  psychoactive  substances  to  be  similar  to
the  score  elevations  of  psychoactive  substance  abusers.  The
results   for   subjects   who  were  tested  again  at   the  time  of
substance  abuse  treatment   correlated  well   with  their   pre-
morbid test results [249251]. In the case of eld dependence,
early  studies  determined  that   this  tendency  antedated  the
onset of drinking [252,253]. Anumber of archival, longitudinal,
and   prospective   studies   have   found   several   premorbid
personality traits to be associated with the later development
of   psychoactive  substance  abuse,   including:   unconvention-
ality or nonconformity, rejection of societal values, alienation,
social   anxiety,   pessimism,   depression,   sensation-seeking,
impulsivity, extraversion, aggressiveness, emphasis on inde-
pendence,   and   labile   or   erratic   mood   [105,249,254289].   A
recent   prospective   population-based  study  [55]   found  that
subjects   with  a   diagnosis   of   past-year   problem  gambling,
ethanol   dependence,   cannabis   dependence,   or   nicotine
dependence   at   age   21   years   were   more   characterized   by
anxiety, alienation, low stress tolerance, anger or aggressive-
ness,   impulsivity,   risk-taking,   and  nonconformity  measured
at age 18 years than were control subjects who did not have a
past-year   addictive   disorder   at   age   21   years.   Vanyukovs
review of research up to 2003 [220] concluded that variation in
the  liability  to  substance  use  disorder  is  shared  in  common
with   personality   phenotypic   variation   that   predates   the
initiation of substance use.
Similar  questions  about  causal   relationships  have  arisen
around  ndings  of   lifetime  comorbidity  between  the  beha-
viorally denedsyndromes that we have beenconsidering and
other psychiatric disorders. Retrospective epidemiologic  sur-
veys   have   consistently   found   that   in   respondents   with
comorbid   substance   use   disorders   and   other   psychiatric
disorders,   the   onset   of   the   other   psychiatric   disorders   is
typically 510 years earlier than is the onset of the substance
use   disorders   [290292,87,293,86].   The   WHO   International
Consortium  in   Psychiatric   Epidemiology   found   signicant
predictive  associations (odds ratios greater than  1.0  and  87%
statistically  signicant   at   the  .05  level)   between  temporally
primary  mental   disorders  and  the  subsequent  rst  onset  of
psychoactive   substance   use,   problems   among   users,   and
dependence  among  problem  users  [294,295].   The  results  of
prospective studies up to 2004 [reviewed in 220, 296] similarly
supported  the  temporal   and  predictive  primacy  of   anxiety,
mood,   and  attention  decit   disorders  when  comorbid  with
psychoactive  substance  abuse.  Subsequent  studies  [297299]
found  that   decits  in  affect  and  self-regulatory  functioning
usually  precede   and  increase   the   risk  for   development   of
substance use problems, though the data are more robust for
anxiety disorders than they are for depression.
Studies  of  temporal  and  predictive  relationships  between
bulimia, pathological gambling, or sexual addiction and other
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   269
psychiatric disorders are still scarce. A few studies found that
anxiety  disorders  that   were  comorbid  with  bulimia  usually
began  in  childhood  before  the  onset   of   the  eating  disorder
[108,300,301],  and  a  twin  study  identied  a  common  genetic
factor   that   inuences   liability   to   anxiety,   depression,   and
eating disorder symptoms [302]. One epidemiologic study with
pathological   gambling  found  that  among  problem  gamblers
with comorbid depression or anxiety, onset of the depression
or anxiety usually preceded onset of gambling [51].
1.1.7.   What to conclude?
The   preceding   blitz-review  indicates   that   the   ndings   of
scientic  research  accord  with  each  of   the  predictions  that
were generated from the hypothesis that addictive disorders
share an underlying biopsychological process. The hypothesis
is   thereby   conrmed.   In   accord   with   this   conrmation,
Krueger and colleagues [303,304] proposed that co-occurrence
of common psychiatric disorders at greater than chance rates
suggests that the disorders are indicators of latent factors or
hypothetical core psychopathological processes that underlie
putatively separate conditions. The foregoing reviewventured
beyond  comorbidity  data  to  include  a  range  of  research  that
provides substantial support for the hypothesis that addictive
disorders  have  in  common  an  underlying  biopsychological
process.  The  discussion  of  temporal  and  predictive  relation-
ships then indicated that, for the most part, the abnormalities
of   psychological   and   neurobiological   functioning   that   are
documented in the research ndings are temporally primary
to  and  predictive  of  the  behavioral  syndromes  that  we  have
been considering. From this we can infer that the underlying
biopsychological process that these conditions share precedes
their onset, and is not simply a consequence of the behavior or
life-style that characterizes them.
Our next step is to begin mining the neurobiology research
literature   for   ore   that   can   then   be   sifted,   rened,   and
eventually   fashioned   into   a   neurobiological   theory   of   the
process  that  underlies  addiction.  But  before  we  proceed  any
further, we need to dene our key terms.
1.2.   Denitions
1.2.1.   Addiction  (or  addictive disorder)
Presenting  a  theory  of  addiction  without  a  clear  and  mean-
ingful denition of the term is a recipe for misunderstanding.
The  denition  is  a  matter  of  controversy,   and  DSM-IV  [305]
does  not  employ  the  term  at  all.   However,   we  can  begin  to
formulate a denition by identifying the key features of drug
addiction  (in  DSM-IV,   psychoactive  substance  dependence),
the paradigm of addictive disorders.
We nowrecognize that neither tolerance nor withdrawal is
necessary or sufcient for a diagnosis of drug addiction [305].
These processes reect the natural adaptive responses of our
bodies cells to a changed chemical environment, regardless of
whether  the  chemicals  had  been  used  addictively.  Extensive
exploration  has  led  me  to  conclude  that  the  characteristics
that are both necessary and sufcient for identifying a pattern
of drug use as drug addictionare (1) recurrent failure to control
the use of one or more drugs, and (2) continuation of drug use
despite signicant harmful consequences. (Recurrent failure
to control means not that addicted individuals invariably lose
control  when  they  use  drugs,  but  that  their  predictions  that
they would remainincontrol of their drug use have repeatedly
proved to be unreliable.)
These  key  features  distinguish  drug  addiction  from  drug
use that does not constitute addiction. However, they do not
distinguish  addictive  behavior  from  compulsive  behavior  or
from impulsive behavior. These latter distinctions depend on
the  behaviors   motivational   functions.   Compulsive  behavior
functions  to  reduce  anxiety  or  other  painful   affects,   but  by
denition it does not produce pleasure or gratication [305]. It
is motivated by negative reinforcement (i.e., the alleviation of
aversive stimulus conditions). Impulsive behavior functions to
produce  pleasure  or   gratication  but   not   to  reduce  painful
affects.   It   is   motivated  by   positive   reinforcement.   Finally,
addictive behavior functions both to produce pleasure and to
reduce  painful   affects.   It   is  motivated  by  both  positive  and
negative reinforcement.
When   we   combine   this   distinctive   dual   motivational
function  of   addictive   behavior   with  the   key   features   that
distinguishdrug addictionfromordinary drug use, we arrive at
a  workable,   behaviorally  nonspecic  denition  of  addiction:
addiction is a condition in which a behavior that can function both to
produce   pleasure   and  to   reduce   painful   affects   is   employed  in  a
pattern that is characterized by two key features: (1) recurrent failure
to control the behavior, and (2) continuation of the behavior despite
signicant harmful consequences.
1.2.2.   Addictive  process
The  addictive  process  is  the  term  by  which  I  propose  that  we
designate the underlying biopsychological process that addic-
tive  disorders  are  hypothesized  to  share.   It   can  be  dened
operationally as an enduring, inordinately strong tendency to
engage  in  some  form  of   pleasure-producing  behavior   in  a
pattern   that   is   characterized   by   impaired   control   and
continuation  despite  signicant  harmful  consequences.   The
class  of  addictive  disorders  includes  psychoactive  substance
addiction, bulimia, pathological gambling, shopping or buying
addiction,
2
sexual addiction, and other enduring conditions in
which a behavior that can function both to produce pleasure
and to reduce painful affects is employed in a pattern that is
characterized  by  two  key  features:   (1)   recurrent   failure   to
control   the  behavior,   and  (2)   continuation  of   the  behavior
despite   signicant   harmful   consequences.   When   we   talk
about   addictive  disorders  as  a  group,   what   we  are  talking
about  is  not  a  collection  of  distinct  disorders,  but  an  under-
lying process that can be expressed in one or more of various
behavioral manifestations.
Thus,   we  can  recognize  that   two  sets   of   factors   shape
the development of an addictive disorder: those that concern
the underlying addictive process, and those that relate to the
selection of a particular substance or behavior as the one that
is preferredfor addictive use. The following discussionfocuses
on the former, which is the more important both theoretically
and practically.
2
Addiction or addictive disorder is a more suitable designation
for   this   condition  than  is   compulsion  or   compulsive  disorder,
since  the  symptomatic  behavior  usually  tends  to  be  associated
with  pleasure  or  gratication  as  well  as  alleviation  of  anxiety  or
other affective discomfort.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 270
2.   The addictive process
In   the   course   of   reviewing   evidence   that   psychoactive
substance   use   disorders,   bulimia,   pathological   gambling,
and  sexual   addiction  share  an  underlying  biopsychological
process,   we  noted  that   neuroscience  research  has  demon-
strated  that   a  shared  vulnerability  underlies   the   abuse   of
psychoactive  substances,   has  begun  to  delineate  the  neuro-
biological processes that constitute this vulnerability, and has
expanded  the  realm  of   this  shared  vulnerability  to  include
pathological gambling and pathological use of food and sex, as
well   as  psychoactive  substance  abuse.   These  developments
introduce us to the possibility of formulating a neurobiological
theory   of   the   addictive   process.   We   begin  the   project   of
actualizing this possibility with a comprehensive but selective
reviewof the neurobiology literature, insearchof rawmaterial
for   such  a  theory.   The  selection  process  is  guided  by  two
principles,   generality  and  specicity.   To  be  included  in  this
review, a researchnding or idea must be relevant to addictive
disorders   in  general,   not   just   to  a  particular   psychoactive
substance  or   behavior.   Findings   and  ideas   that   concern  a
particular   psychoactive   substance   or   behavior   and  do  not
generalize   to   the   rest   may   inuence   which  substance   or
behavior   a   person   who   is   predisposed   to   developing   an
addictive disorder  (by  virtue  of  an  addictive  process) is  most
inclined to use addictively, but they are unlikely to participate
signicantly in the genesis of that predisposition. Inclusion in
this reviewadditionally requires that a researchnding or idea
be   specic   to   addictive   patterns   of   using   a   substance   or
engaging ina behavioras distinct frombeing applicable to all
instances of a behavior, regardless of whether they instantiate
an addictive disorder. In other words, the object of our quest is
not the neurobiology of what makes cocaine or sexpleasurable
for people in general, but the neurobiology of what makes the
drive for cocaine or sex so much more inexorable for a person
who uses it addictively.
The   addictive   process   can   be   understood   to   involve
impairments in three functional systems: motivation-reward,
affect  regulation,  and  behavioral  inhibition.
3
Impaired  moti-
vation-reward exposes addicts to unsatised states of irritable
tension, emptiness, and restless anhedonia. In the context of
aberrant   motivation-reward   function,   behaviors   that   are
associated  with  activation  of   the  reward  system  are  more
strongly reinforced (via both positive and negative reinforce-
ment) than they otherwise would have been. Impaired affect
regulation  renders  addicts  chronically  vulnerable  to  painful
affects  and  emotional  instability.  In  the  context  of  impaired
affect  regulation,   behaviors  that  are  associated  with  escape
from  or   avoidance   of   painful   affects   are   more   strongly
reinforced  (via  negative  reinforcement)   than  they  otherwise
wouldhave been. Impairedbehavioral inhibitionincreases the
likelihood   that   urges   for   some   form   of   reinforcement
(negative,   positive,   or  both)   in  the  short   term  will   override
consideration of longer termconsequences, both negative and
positive.   When  motivation-reward  and  affect  regulation  are
impaired, impaired behavioral inhibition means that urges to
engage   in   behaviors   that   are   associated   with   both   (a)
activation  of   the   reward   system,   and   (b)   escape   from  or
avoidance  of   painful   affects,   are  extraordinarily  difcult   to
resist,   despite   the   harmful   consequences   that   they  might
entail.
We   now   embark   on   a   quest   to   identify   candidate
neurochemical   substrates   for   the   impairments   in  motiva-
tion-reward, affect regulation, and behavioral inhibition that
could  contribute  to  an addictive process. (Candidate  neuroa-
natomical substrates for these impairments are considered in
a   separate   publication,   as   are   genetic   and   environmental
factors that shape the development of an addictive  process.)
Our   initial   assumption   is   that   no   single   factor   is   either
necessary  or   sufcient,   and  that   an  addictive  process   can
result from any of a variety of multi-factor combinations. The
addictive  process  that  characterizes  a  person  is  the  unique
outcome of individual genetic and environmental inuences.
But  among  the  array  of  uniquenesses,  a  good  set  of  theories
will enable us to recognize some patterns.
2.1.   Aberrant motivation-reward
2.1.1.   Dopamine  (DA)
Administering  any  drug  of   abuse  [306323]   or   engaging  in
eating   (especially  sweets)   [324328],   gambling   [329,330],   or
sexual   behavior   [330334]   is   associated   with   increased
intrasynaptic levels of dopamine (DA) in the nucleus accum-
bens   (NAc).   Accumbal   DA  was   initially  thought   to  be   the
neurobiological   correlate   of   reward   or   pleasure.   However,
recent  research  has  claried  DAs  function  in  signaling  the
incentive  salience  of   events  (including  rewarding,   aversive,
novel, and unexpected stimuli), in driving motivated behavior,
in   predicting   reward   or   non-reward,   and   in   facilitating
consolidation of memory for salient events [335349].
Five DA receptors have been identied, all of which are G
protein-coupled.  They  can  be  classied  into  two  families:  D
1
and D
5
 receptors that stimulate adenylate cyclase to produce
cyclic   AMP;   and  D
2
,   D
3
,   and  D
4
  receptors   that   inhibit   the
production of cyclic AMP [350]. Most D
1
 and D
5
 receptors are
located postsynaptically, while most D
2
, D
3
, and D
4
 receptors
are located presynaptically [351353]. The functions of D
1
, D
2
,
and  D
3
  receptors  primarily  concern  motivation  and  reward,
while D
4
 and D
5
 receptors are more involved with behavioral
inhibition (and consequently will be discussed in the Section
2.3).
2.1.2.   DA D
1
 receptors
Activation of DA D
1
 receptors has been found to be associated
with  ethanol  reward  [354],  psychostimulant  reward  [355,356],
foodreward[357], cocaine-inducedlocomotor activity[355,358],
reinstatement   of   cue-induced   cocaine-seeking   behavior
[359,360],   reinstatement  of  extinguished  cocaine-conditioned
place  preference  [356],  and  enhancement  of  food  palatability
[361].   Activation  of   D
1
  receptors  has  been  found  also  to  be
critically  involved  in  enduring  cell-surface  and  intracellular
3
At this point, motivation-reward, affect regulation, and beha-
vioral   inhibition  are  heuristic  constructs  that   provide  an  intui-
tively   meaningful   framework   within   which   relevant   research
ndings  may  be  organized.   As  abstractions  from  the  nonlinear
system  of   an  organisms   neurobiology,   they   are   more   clearly
delineated from one another than are the processes to which they
refer.  The  organization  of  research  ndings  that  this  framework
enables can be expected to facilitate the eventual formulation of
operational denitions for these terms.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   271
changes   that   follow   administration   of   psychostimulants,
other  drugs  of  abuse,  and  palatable  food.  Changes  at  the  cell
surface  that   are  associated  with  activation  of   D
1
  receptors
include psychostimulant-induced externalization of a-amino-
3-hydroxy-5-methylisoxazole-4-propionic   acid   receptors
(AMPARs)   with  promotion  of   long-term  potentiation,   which
facilitates  reward-related  learning  [362],   and  dendritic  remo-
deling   of   medium  spiny   neurons   in  the   NAc,   an  adaptive
response to chronic cocaine exposure that is linked to addictive
patterns  of  behavior  [363,364].   Intracellular  changes  that  are
triggered by activation of D
1
 receptors include induction of c-
Fos, FosB, Fra-2 and JunB by acute cocaine exposure, induction
of  DFosB by repeated cocaine administration in both the  NAc
and  caudate-putamen  (CPu),  and  cocaine-induced expression
of olfactory-specic G protein a (Gaolf), b-catenin, and BDNF in
the NAc and CPu [364366]. The processes that are involved in
the   dendritic   remodeling   of   medium  spiny   neurons   are
integrated  by  the  c-Fos  that  has  been  induced  by  activation
of   D
1
  receptors   [364].   Zhang  et   al.   [366]   demonstrated  that
cocaine-induced  expression  of  Fos  family  genes,  including  c-
Fos, FosB, and Fra-2, is mediated by activation (via phosphor-
ylation) of extracellular signal-regulated kinase (ERK), which in
turn is mediated by D
1
 receptors. Valjent et al. [367] found that
activation  of   ERK  is  induced  not   only  by  administration  of
cocaine,  but  also  by  administration  of  morphine,  nicotine,  or
tetrahydrocannabinol   (THC).   Activation  of   D
1
  receptors   by
administration  of   ethanol   [368],   cocaine  [369],   and  palatable
food [370] also results in phosphorylation at threonine-34 (T35)
of the intracellular messenger DARPP-32 (dopamine and cyclic
3
0
, 5
0
  adenosine monophosphate-regulated phosphoprotein), a
process  that  Zachariou  et  al.   [369,371]   found  to  be  a  critical
mediator of cocaines rewarding effects.
The foregoing ndings provide valuable information about
the relationshipbetweenDAD
1
receptors andreward, but they
do  not  necessarily  enhance  our  understanding  of  addiction.
The latter depends less on knowing what makes a particular
substance  or  behavior  rewarding,   than  it   does  on  knowing
what makes some people more susceptible than are others to
developing addictive patterns of using a rewarding substance
or   engaging   in   a   rewarding   behavior.   So   how  might   D
1
receptors be involved in an addictive process? One possibility
is suggested by Haney et al.s [372] ndings that maintenance
administration  of   the  selective  D
1
  antagonist   ecopipam  to
human   subjects   enhanced   both   self-administration   and
subjective  effects  of   cocaine,   compared  to  maintenance  on
placebo.   These  ndings   are  consistent   with  the   results   of
preclinical   studies  in  which  doses  of  cocaine  that  had  been
maintaining   relatively   low   levels   of   self-administration
maintained  higher  levels  following  chronic  exposure  to  a  D
1
antagonist [373376]. These behavioral shifts were associated
with   an   increased   density   of   D
1
  receptors   [377379]   and
enhanced   D
1
  receptor   sensitivity   within   the   NAc   [380].
Considered  together,   these  data  suggest   that   maintenance
administration   of   a   D
1
  antagonist   results   in   D
1
  receptor
supersensitivity,   which  increases   the   reinforcing  and  sub-
jective  effects  of  cocaine.   Since  the  inuence  of  D
1
  receptor
supersensitivity  on  the  reward  effects  of   cocaine  is  almost
certainly  mediated  by  the  increase  in  intrasynaptic  DA  that
follows cocaine administration, D
1
receptor supersensitivity is
likely to have the same inuence onany substance or behavior
that   increases   the   intrasynaptic   availability   of   DAwhich
includes  every  substance  and  behavior   that   is  used  addic-
tively. Thus couldD
1
receptor supersensitivity, however it may
develop, contribute to an addictive process.
2.1.3.   DA D
2
 receptors
Preclinical  studies  in  which  D
2
  agonists  or  antagonists  were
administered   have   yielded   a   complex   array   of   results.
Administration  of  the  D
2
/D
3
  agonist  quinpirole  was  reported
to  eliminate rats preference for a  highly palatable  chocolate
food  [361],   to  block  expression  of   an  established  cocaine-
conditioned   place   preference   (CPP),   and   to   induce   place
aversions   to   the   cocaine-paired   side   of   the   conditioning
apparatus   following   extinction  of   the   established   cocaine
preference   [356].   However,   it   was   reported   also   to   evoke
reinstatement  of  self-administration  after  extinction  in  both
cocaine-trained   and   heroin-trained   rats   [381].   The   D
2
/D
3
partial   agonist  terguride  was  found  to  reduce  self-adminis-
tration  of   cocaine  and  of   food  [382].   Administration  of   D
2
receptor   antagonists   was   reported  to  reduce   ethanol   self-
administration [383], to inhibit reinstatement of cue-induced
cocaine-seeking behavior [384], and, in the case of raclopride,
to  potentiate  reinstatement  of  cue-induced  cocaine-seeking
behavior with a low dose and to attenuate it with a high dose
[359]. According to a model that was presented by Welter et al.
[385],   D
2
  receptors   tonically  inhibit   an  inhibitory  signaling
pathway   that   decreases   the   cocaine-induced   locomotion,
blocks   the   c-Fos   induction  that   cocaine   typically   triggers,
and  attenuates  cocaine-induced  CPP.   To  the  extent   that   D
2
receptor activity decreases, the inhibitory pathway is released
to block these characteristic responses to cocaine.
In  a  more  direct   and  readily  interpretable  vein,   clinical
studies have indicated an associationbetweenalcoholismand
decreased  D
2
  receptors   [386388].   Positron  emission  tomo-
graphy  (PET)   and  single  photon  emission  computed  tomo-
graphy (SPECT) studies of addictive users of ethanol, cocaine,
methamphetamine, andheroinhave revealedreductions of D
2
receptor density in the ventral striatum that persist long after
detoxication  [389392].   Studies   with  pathologically   obese
subjects  found  reductions  in  striatal   D
2
  receptors  that  were
similar  to  those  observed  in  studies  with  drug  addicts,   and
additionally   found   an   inverse   relationship   between   the
subjects   body   mass   index   and   their   D
2
  receptor   levels
[228,393]. An initial hypothesis that low levels of D
2
 receptors
predispose subjects to search for psychoactive substances as a
means to compensate for the consequent decrease in reward
circuit  activation  [224,394,395]  was  expanded,  in  response  to
the  studies  with  obese  subjects,   by  replacing  psychoactive
substances with the more general term, reinforcers [396].
This   hypothesis   has   been  supported  both  by  preclinical
research  and  by  research  with  nonclinical   populations.   Rats
that were high responders to novelty as measured by locomo-
tion  in  an  open  eld  and  thus  were  more  likely  to  acquire
amphetamine  self-administration  compared  to  low-respond-
ing rats [397] were found to have lower D
2
receptor levels in the
NAc [398], and an inbred ethanol-preferring strain of rats was
found  to  have  lower   D
2
  receptor  binding  than  did  ethanol-
nonpreferring rats [399402]. Virally mediated overexpression
of   D
2
  receptors   was   associated  with  marked  reductions   in
ethanol preference and intake in both ethanol-preferring and
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 272
ethanol-nonpreferring rats, which reverted to status quo ante
as theD
2
receptor densityreturnedtobaseline[402,403]. Astudy
with   rhesus   macaques   found   that   baseline   D
2
  receptor
availability  was   negatively  correlated  with  rates   of   cocaine
self-administration [404]. And mice that showed an increased
propensity to ethanol sensitization were found to have higher
levels of D
2
 receptor binding in localized brain areas than did
mice that showed less propensity to sensitization [405]. Mean-
while, Yoder et al. [406] reported a study in which a low (sub-
intoxicating) doseof ethanol was administeredtonon-addicted
human  subjects.   They  found  that  baseline  availability  of  D
2
receptors  in  the  left  NAc  was  correlated  with  peak  perceived
intoxication and marginally correlated with peak perceived
high. The moreD
2
receptors that were available for binding in
the   sober   state,   the   more   likely   was   the   subject   to   feel
intoxicated   and  high   from  a  low  dose  of   ethanol.   The
authors  speculated  that  individuals  with  fewer  D
2
  receptors
would  require  larger  quantities  of  ethanol   to  experience  the
same subjective high. Similarly, Volkowet al. [407,408] reported
that baseline measures of striatal D
2
 receptors in non-addicted
human  subjects  predicted  their   subjective  responses  to  the
reinforcing   effects   of   intravenous   methylphenidate   (MP).
Subjects who described MP as pleasant had signicantly lower
levels  of  D
2
  receptors  than  did  subjects  who  described  it  as
unpleasant.   The   authors   hypothesized  that   every   persons
brain  has  an  optimal   range  of  D
2
  stimulation  by  MP  that   is
experienced as pleasant, below which administration of MP is
perceived  as neutral  or insufcient,  and  above  which  admin-
istrationof thedrugisexperiencedas aversive. Theauthors also
noted that subjects who reported the effects of MP as pleasant,
as do most cocaine abusers [409], had D
2
receptor levels similar
to those that were previously reported to characterize cocaine
abusers [410,411]. They interpreted these results as suggesting
that   low  D
2
  receptor   levels   in  cocaine   abusers   may   have
antedated  their  use  of  cocaine  and  may  have  contributed  to
their shift from cocaine use to cocaine addiction.
While   consideration   of   genetic   factors   that   shape   the
development   of   an   addictive   process   is   in   general   being
deferred to another publication, no discussion of the relation-
ship   between   DA   D
2
  receptors   and   addiction   would   be
complete   without   at   least   mentioning   the   ndings   that
concern associations between the Al allele of the D
2
 receptor
gene Taq1A polymorphism and alcoholism [412417], cocaine
addiction   [418],   psychostimulant   addiction   [419],   cigarette
smoking   [420,421],   pathological   gambling   [422424],   and
exaggerated  reward  value  of   food  [425,426].   Interestingly,   a
number of studies have found that human subjects who carry
the Al allele of the D
2
receptor gene Taq1Apolymorphismhave
signicantly reduced D
2
 receptor density [427432].
2.1.4.   DA D
3
 receptors
Research   with   DA   D
3
  receptors   is   a   relatively   recent
phenomenon.   While  ndings  that  D
3
  receptors  are  located
primarily   in   limbic   regions   led   to   speculation   that   D
3
receptors   might   be  involved  in  addiction,   their   structural
similarity   to  D
2
  receptors   made   them  elusive   targets   for
molecular  sharpshooters.  Only  when  new  compounds  with
high selectivity for central D
3
receptors were synthesized and
characterized   was   research  with  these   receptors   able   to
proceed [433].
The   era   of   addiction   research   with   D
3
  receptors   was
launched by the publication in 1999 of Pilla et al.s [434] report
that   BP   897,   a   D
3
  antagonist   [435],   inhibited   cue-induced
reinstatement   of   drug-seeking  behavior   by  cocaine-trained
mice that had undergone response extinction. These ndings
were greeted by a urry of optimistic response [436438], and
were  later  extended  to  rats  and  rhesus  monkeys  [439441].
Other  studies  found  that  BP  897  blocked  the  expression  (but
not   the   acquisition)   of   amphetamine-conditioned   activity
[442,443], reduced both cue-induced ethanol-seeking behavior
and relapse-like drinking [444], and inhibited nicotine-condi-
tioned   locomotor   responses   [445].   Another   selective   D
3
antagonist,   SB-277011-A,   was  found  to  block  reinstatement
of   cocaine-seeking   behavior   that   had   been   triggered   by
cocaine-priming   [446],   to   attenuate   cue-induced  reinstate-
ment of cocaine-seeking [384,441,447], to block stress-induced
reinstatement   of   cocaine-seeking   [448],   and   to   lower   the
amount  of  work  that  rats  would  perform  for  a  given  dose  of
cocaine while raising the lower limit of the cocaine dose that
would  sustain  a  given  amount   of   work  [449].   SB-277011-A
similarly   was   found   to   reduce   oral   self-administration  of
ethanol [444,450,451],  cue-induced  reinstatement  of  ethanol-
seeking behavior [444,450], nicotine self-administration [452],
reinstatement   of   nicotine-seeking  behavior   [453],   nicotine-
induced  CPP   [454],   and  the   acquisition  and  expression  of
heroin-induced  CPP  [455].   The  D
3
  receptor   antagonist   NGB
2904 was found to inhibit cue-induced reinstatement of drug-
seeking  behavior  by  cocaine-trained  rats  [441].  Interestingly,
SB-277011-Awas found to potentiate the pharmacological MRI
response to D-amphetamine [456], and NGB 2904 was found to
enhance  amphetamine-stimulated  locomotion  in  wild-type
mice but to have no measurable effect in mice that had been
genetically  modied  so  as  not   to  develop  D
3
  receptors  (D
3
receptor knockout mice) [457], effects that intuitively seem to
contradict the other D
3
receptor antagonist ndings. However,
since NGB 2904 by  itself stimulated  spontaneous locomotion
in  wild-type  mice  while  having  no  measurable  effect   in  D
3
receptor knockout mice [457], the effects of SB-277011-A and
NGB  2904  on  response  to  amphetamine  may  have  pertained
more to amphetamines stimulant effect on locomotion than
to its reinforcing or addictive properties.
Addiction-related research with D
3
 receptors that does not
involve  antagonists  has  been  relatively  rare.   In  a  study  that
demonstratedthe role of DAD
1
receptors inactivating ERKand
inducing  c-Fos  in  response  to  acute  cocaine  administration,
Zhang et al. [366] also showed that D
3
 receptors have opposite
effects on the same intracellular systems. Meanwhile, Boyce-
Rustay  and  Risinger   [458]   found  no  difference  between  D
3
knockout   and  C57BL/6J   mice  in  ethanol   CPP,   in  two-bottle
drinking  preference,   or   in  operant   ethanol   self-administra-
tion, from which they inferred that elimination of D
3
 receptor
function has little inuence on ethanol reward or intake. The
apparent   discrepancy  between  this   nding  and  those  that
were  reviewed  in  the  preceding  paragraph  probably  reects
the  difference  between  acute  inactivation  of   a  functioning
receptor system and congenital absence of a receptor system,
for which the postnatal plasticity of the mammalian brain can
to some extent compensate.
The opposite effects of D
1
 and D
3
 receptors on ERK and c-
Fos, as well as ondynorphin, neogenin, andsynaptotagminVII
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   273
[366], may suggest that D
3
 receptors potential involvement in
an addictive process would similarly be opposite to that of D
1
receptorsi.e.,  that  an  addictive  process  could  be  facilitated
by   D
3
  hyposensitivity.   However,   D
3
  receptor   antagonists
inhibit   processes  that   are  associated  with  addiction,   which
suggests that an addictive process could be potentiated by D
3
receptor supersensitivity.
2.1.5.   Serotonin  (5-HT)
Serotonin (5-hydroxytryptamine, or 5-HT) activity is associated
with  behavioral   inhibition  [459,460],   emotional   stabilization
[461],   appetite  modulation  [462],   sensory  reactivity  [463,464],
pain   sensitivity   [464,465],   and   sleep,   sexual   behavior,   and
cognitive   function   [466,467].   At   least   14   subtypes   of   5-HT
receptors have been cloned and  identied. The  5-HT
1
 class is
inhibitory   both   pre-   and   post-synaptically,   and   reduces
adenylate  cyclase  activity  via  Gi  activation.  The  excitatory  5-
HT
2
  class  is predominantly  postsynaptic,  and  activates phos-
pholipase  C  via  Go.   The  5-HT
3
  receptor  exerts  its  excitatory
effects byacting as anionchannel. Andthe 5-HT
4
, 5-HT
5
, and5-
HT
6
 classes all activate adenylate cyclase via Gs [468].
Serotonin   does   not   directly   participate   in   motivation-
reward,   but   exerts  inuence  through  its  effects  on  the  DA
system. Application of 5-HT onto dopaminergic neurons from
the VTA increased their ring rate in vitro, an effect that was
attributed to action of 5-HT on 5-HT
2
receptors [469]. However,
increased release of DA in the NAc (presumably of VTA origin)
that was elicited by electronic stimulation of the dorsal raphe
nucleus (DRN) in vivo was counteracted by the selective 5-HT
3
antagonists ondansetron and (S)-zacopride [470].
In  a  study with  pathological  gamblers,  Pallanti  et  al.  [471]
found that direct postsynaptic serotonergic receptor stimula-
tion   with   meta-chlorophenylpiperazine   (m-CPP),   a   mixed
serotonergic agonist with highest afnity for 5-HT
2C
receptors,
elicited   an  enhanced   prolactin  response   that   the   authors
interpreted   as   a   hypersensitive   postsynaptic   serotonergic
function.   In   addition,   the   pathological   gambling   subjects
reported  that  the  high  sensation  that  they  experienced  in
response   to   m-CPP   was   similar   to   the   one   that   they
experienced while gambling, a result reminiscent of alcoholic
subjects   reports   that   their   m-CPP-induced  experience  was
comparable  to  their   experience  with  ethanol.   The  authors
speculated   that   increased  sensitivity   to   5-HT  stimulation,
shared by the other addictive diseases, could be a vulnerability
factor for addiction.
The most well researched and apparently most signicant
component   of   the   serotonergic   system   that   inuences
motivation-reward   is   the   5-HT
1B
  receptor,   a   G
i
-coupled
receptor  that  can  be  located  on  the  axon  terminals  of  many
types   of   neuron.   Axon   terminals   of   g-aminobutyric   acid
(GABA)  neurons  that  project  from  the  NAc  shell  to  the  VTA
contain  5-HT
1B
  receptors  that,  when  activated,  inhibit  GABA
release. Since GABA that is released in the VTA inhibits local
dopaminergic neurons, inhibition of GABA release disinhibits
the  mesolimbic  dopaminergic  neurons  and  thus  potentiates
the  DA-increasing  effects  of   cocaine  [472475]   and  of   other
rewarding  (or  salient)   substances  and  behaviors.   Up-regula-
tion  of  5-HT
1B
  receptors  on  the  axon  terminals  of  NAc  shell
GABAergic   neurons   could   then   contribute   to   a   persons
vulnerability to developing an addictive disorder.
A study that was designed to measure the effect of chronic
cocaine  injections  on  5-HT
1B
  mRNA  expression  in  the  NAc
shell [476] found that the latter was increased not only in rats
that   received  cocaine  but   also  in  control   rats  that   received
injections of vehicleand interestingly, only in those control
rats that were housed with cocaine-treated rats. The authors
interpreted this unexpected nding to have been mediated by
social   stress   that   the   control   rats   experienced   through
interaction with their cocaine-treated cagemates.
2.1.6.   Norepinephrine  (NE)
Norepinephrine   (NE)   is   both   a   neurotransmitter   that   is
produced  primarily  by  the  locus  coeruleus  (LC)   in  the  brain
stem, and a hormone that is produced by the adrenal medulla.
It   is   synthesized   from  DA  by   the   action   of   the   enzyme
dopamine beta hydroxylase (DBH). NE as well as epinephrine
(adrenalin)   is  a  ligand  for  adrenergic  receptors  (adrenocep-
tors),   G   protein-coupled   receptors   that   can   be   either   a-
adrenergic   or   b-adrenergic.   The   group  of   a-adrenoceptors
contains two subgoups, a
1
anda
2
, eachof whichhas three sub-
subgroups: a
1A
, a
1B
, and a
1D
; and a
2A
, a
2B
, and a
2C
. The group of
b-adrenoceptors contains three subgoups,  b
1
,  b
2
, and  b
3
. The
brain  noradrenergic  system  consists  of  two  main  ascending
projections:   the  dorsal   noradrenergic   bundle  (DNB),   which
originates   in   the   LC   and   projects   to   the   hippocampus,
cerebellum,   and   forebrain;   and   the   ventral   noradrenergic
bundle (VNB), which arises in a number of nuclei of the pons
and medulla and projects to the hypothalamus, midbrain, and
extended amygdala [477].
Evidence for the  involvement  of NE  in  motivation-reward
has   emerged   from   studies   of   self-administration,   CPP,
reinstatement, and locomotor activation. Ethanol self-admin-
istration was found to be attenuated by lofexidine, an agonist
at the  a
2A
 autoreceptor that reduces NE transmission, and to
be  enhanced  by  blockade  of  this  receptor  [478].   While  wild-
type mice readily self-administer cocaine or morphine orally,
a
1B
  knockout   mice   were   found   not   to   do   so   [479].   NE
transporter  (NET)  knockout  mice  lack  the  reuptake  function
that   terminates   the   action  of   NE   and   consequently   have
elevated   levels   of   extracellular   NE.   Such  mice   have   been
reported to self-administer cocaine at an average rate that is
four  times  that  of  wild-type  mice,  suggesting  that  a  chronic
NET deciency decreases the reinforcing properties of cocaine
[480].   Chronically   elevated   NE   levels   could   conceivably
dysregulate  the  dopaminergic  brain  reward  system,  perhaps
via  downregulation  of   presynaptic   DA  transmission  or   via
postsynaptic D
2
/D
3
 supersensitivity [481].
Establishment  of  CPP  for  opiates  seems  to  require  intact
noradrenergic   function.   Clonidine,   an   a
2A
  agonist   that
decreases  NE  release  by  activating  inhibitory  autoreceptors,
was  reported  to  disrupt  the  establishment  of  heroin  CPP  in
rats, presumably by inhibiting NE release [482]. Morphine CPP
in  mice  was  shown  to  be  attenuated  by  either  clonidine  or
prazosin (an a
1A
antagonist) and to be increased by yohimbine
(ana
2A
antagonist) [483,484]. Furthermore, bothDBHknockout
mice, which are incapable of producing NE, and a
1A
 knockout
mice  failed  to  express  a  CPP  over  a  wide  range  of  morphine
doses [479,485].
Noradrenergic function appears to be critical also in stress-
induced reinstatement of drug-seeking for multiple classes of
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 274
abused drugs. Administration of a
2A
 agonists has been found
to attenuate stress-induced reinstatement of ethanol-seeking
behavior  [486],   cocaine-seeking  behavior  [487,488],   and  her-
oin-seeking  behavior  [489,490].  DBH  inhibitors  that  block  NE
synthesis were shown to attenuate reinstatement of amphe-
tamine   self-administration   and   opiate   self-administration
[491].   Conversely,   blockade  of   a
2A
  autoreceptors  with  either
yohimbine  or   RS-79948  was   reported  to  reinstate  cocaine-
seeking in the absence of any stressors [492]. In addition, the
reinforcing  properties  of   morphine,   as  reected  in  the  CPP
paradigm,   seem  to  depend  on  NE.   Chronic  treatment   with
venlafaxine, a dual NE/5-HT reuptake inhibitor, was reported
to attenuate the reacquisition of morphine CPP by a priming
injection of morphine [493]. And selective depletion of medial
prefrontal  cortex  (PFC)  noradrenergic  afferents  was  found  to
abolish  the  reinstatement  of  an  extinguished  morphine  CPP
that  had  been  produced  by  a  priming  injection  of  morphine
[494],   as   well   as   the   reinstatement   of   an   extinguished
amphetamine   CPP   that   had   been  produced   by   a   priming
injection of amphetamine [495].
Finally,   locomotor  activation  that  is  induced  by  psychos-
timulants   or   by   opiates   seems   to   involve   and   in   some
instances to depend on NE. Administration of prazosin, either
systemically   or   directly   into   the   PFC,   has   been   found   to
attenuate  the  acute  locomotor   responses  and  sensitization
that   are  produced  by  psychostimulants  [479,496503]   or   by
morphine   [479,496,504].   The   locomotor   activation   that   is
induced  by  morphine  was   shown  to  be  decreased  also  by
the  nonspecic  a-adrenergic  antagonist,  phenoxybenzamine
[505,506]  and  by  pre-treatment  with  FLA-63,  a  DBH  inhibitor
[506].   LC   lesions   were   found   to   attenuate   amphetamine-
induced  locomotion  [507],   while  the  locomotor  response  to
psychostimulants was reported to be amplied by blockade of
a
2A
 inhibitory autoreceptors, which increases levels of extra-
cellular   NE   [508],   and   by   the   non-selective   b-adrenergic
antagonist   propranolol   [509].   Additional   evidence   for   NE
mediation  of   drug-induced  locomotion  comes  from  studies
in which NE function was modied genetically.  a
1B
 knockout
mice  were  found  to  be  refractory  to  both  psychostimulant-
induced and morphine-induced locomotor activity and sensi-
tization [479,496,500]. DBH knockout mice have been reported
not to develop morphine-induced locomotion, a decit that is
partially reversed by pharmacological restoration of NE or by
viral-mediated reexpression of DBH in the DNB or VNB [485].
Finally, genetic ablation of the NET was found to increase the
locomotor response to psychostimulants [481].
Weinshenker and colleagues [510] demonstrated that DBH
knockout   mice   were   hypersensitive   to   the   rewarding   and
locomotor  effects  of   amphetamine.   Further  work  with  DBH
knockout   mice   [511]   found   that   they   were   at   least   as
hypersensitive to the aversive effects of cocaine and amphe-
tamine  as  they  were  to  their  rewarding  effects.   They  even
developed   conditioned   place   aversion   to   cocaine,   which
control   mice  could  not   be  convinced  to  do.   Other   studies
have reported that DBH knockout mice are hypersensitive to
the   aversive   effects   of   ethanol   [512]   and   that   mice   that
specically lack NE in the PFC showa similar place aversion to
amphetamine [495]. These ndings can be understood in light
of  the  relationship  between  the  NE  and  DA  systems.  The  LC
noradrenergic system regulates the activity of the ascending
DA   pathways   [496,513].   They   meet   in   the   VTA,   where
noradrenergic  neurons  modulate  the  DA  cell   ring  pattern
via  excitatory  postsynaptic   a
1
-adrenoceptors   [514].   The   LC
noradrenergic   system   also   regulates   the   mesencephalic
dopaminergic  system  indirectly,   via  the  PFC.   DA  release  in
the  PFC  is  regulated  by  local   noradrenergic  nerve  terminals
[515],  and  electrical  stimulation  of  the  LC  neurons  increases
bothextracellular DAand NE inthe PFC[516]. WhenNE release
is blocked, DArelease is similarly attenuated. If the NE block is
chronic,   the   DA   system  gradually   compensates   by   up-
regulating  high-afnity  state  postsynaptic  DA  receptors  (i.e.,
increasing their density) by a factor of 36 [517]. This process
results  in  hypersensitivity  to  psychostimulants  and  to  any
other substance (or behavior) that increases intrasynaptic DA
levels.
In the course of a comprehensive reviewof the role of NE in
drug addiction, Weinshenker and Schroeder [517], reassessed
the operation of disulramfromthe perspective of its function
as a potent inhibitor of DBH. While the aversiveness of ethanol
after disulram administration had traditionally  been attrib-
uted  to  the  accumulation  of  acetaldehyde  that  results  from
disulrams inhibition of the enzyme acetaldehyde dehydro-
genase,   such   an   explanation   could   not   account   for   the
effectiveness of disulram in treating cocaine addiction when
ethanol   abuse  is   not   comorbid  [518520].   Amit   et   al.   [521]
compared  the  efcacy  of   disulram  in  decreasing   ethanol
intake with that of calcium carbamide and of FLA-63, and also
assessed  the  effect   that   each  of   these  compounds  had  on
acetaldehyde  levels  following  ethanol  injection.  Administra-
tion  of   disulram  and  FLA-63,   both  DBH  inhibitors,   signi-
cantly reduced ethanol intake. Calciumcarbamide, which had
the  greatest  effect  on  acetaldehyde  levels  following  ethanol
injection,  had  the  least  effect  on  ethanol  intake.  FLA-63  had
the  least   effect   on  acetaldehyde  levels,   but   was   the  most
potent suppressor of ethanol intake.
These research ndings suggest that potential contributors
to an addictive process could include blockade, hyposensitiv-
ity,   or   excessive  downregulation  of   a
2A
  autoreceptors,   and
chronic   deciency  or   malfunction  of   NE  transporters.   The
central   factor   in   a   potential   relationship   between   the
noradrenergic system and addiction seems to be an increased
level  of  extracellular  NE  and  its  effects  on  the  dopaminergic
system. The most frequent and signicant cause (or correlate)
of increased levels of extracellular NE is stress. The relation-
ship   between   the   noradrenergic   system,   stress,   and   the
addictive process will be addressed in Section 2.2.
2.1.7.   Endorphins  and opioid receptors
Administering  opiate  drugs  directly  stimulates  opioid  recep-
tors in the brain [522]. Administering any other drug of abuse
[519528]   or  engaging  in  eating  (especially  sweets)   [226,244,
529533],   gambling  [329],   or  sexual   behavior  [331,534,535]   is
associated  with  the  release  of   endogenous  opioids.   Studies
that measured  m opioid receptors in cocaine abusers showed
signicant  increases  in  receptor  availability  that  were  inter-
preted  to  reect  decreased  endogenous  opioid  release  [536].
CSF  b-endorphin  levels  in  bulimic  subjects  were found  to  be
lower   than   in   controls   [537].   Low  baseline   levels   of   b-
endorphin and consequent m opioid receptor hypersensitivity
would  constitute  a  vulnerability  to  addictive  engagement  in
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   275
any behavior that results in stimulation of m opioid receptors.
However,   these  cross-sectional   studies  do  not   address  the
question  of   whether   the  decreased  b-endorphin  levels  and
increased m opioid receptor availability preceded or developed
subsequent to the onset of the subjects disorders.
2.1.8.   Dynorphin
Dynorphin is an endogenous opioid peptide that functions as
an  agonist at  k  opioid  receptors.  Dopaminergic  VTA  neurons
that   project   to   the   NAc   contain   k   opioid   receptors   on
presynaptic axon terminals in the NAc and also on cell bodies
and  dendrites  in  the  VTA.   Action  of   dynorphin  on  these  k
opioid  receptors  inhibits  the  release  of  DA,  thus  attenuating
the reward-salience effects of substances and behaviors that
can  be  used  addictively,  and  moreover generating  dysphoria
[538540].   Dynorphin  expression  is  induced  in  the  NAc  and
related  striatal   regions  after  exposure  to  drugs  of  abuse,   an
effect  that  seems  to  be  mediated  by  the  gene  transcription
factor  CREB  (cAMP  response  element  binding  protein)   [540].
Drugs  of   abuse  also  induce  the  transcription  factor   DFosB,
which targets the gene that encodes dynorphin and decreases
its expression [541]. Variations in CREB, DFosB, or the balance
between them that result in decreased dynorphin expression
could contribute to an addictive process.
2.1.9.   g-Aminobutyric acid  (GABA)
g-Aminobutyric   acid  (GABA)   is   the   chief   inhibitory   neuro-
transmitter  in  the  central   nervous  system.   Three  classes  of
GABA  receptors   have   been  identied:   GABA
A
,   GABA
B
,   and
GABA
C
. GABA
A
 and GABA
C
 receptors are ionotropic receptors
(ligand-gated ion channels). The binding of a GABA molecule
to a GABA
A
or GABA
C
receptor directly triggers the opening of a
chloride   ion-selective   pore   that   allows   intracellular   and
extracellular  chloride  to  equilibrate,  thereby  hyperpolarizing
the neuron and inhibiting its ring [542,543]. In addition to an
active  binding  site  at   which  GABA  binds,   GABA
A
  receptors
have   specic   allosteric   sites   that   bind   benzodiazepines,
barbiturates,   ethanol,   picrotoxin,   neuroactive  steroids,   fur-
osemide,   and  inhalation  anesthetics  [544].   GABA
C
  receptors
seem to be variants of GABA
A
 receptors that are insensitive to
the  typical  allosteric  modulators  of  GABA
A
  receptors.  GABA
B
receptors are metabotropic (G protein-coupled) receptors that
can   open   transmembrane   potassium  channels,   suppress
calciumchannels, and reduce the activity of adenylate cyclase
[545547]. They too exert inhibitory effects when activated.
2.1.10.   GABA
A
 receptors
GABA
A
receptor antagonists that bind at or near the active site,
such as picrotoxin and bicuculline, have been found to reduce
self-administration  of   ethanol   [548552]   and  cocaine  [553].
Nowak et al. [550] reported that microinjections of picrotoxin
or of bicuculline into the VTA resulted in decreases in ethanol
consumption, but that microinjections in regions outside the
VTA failed to decrease ethanol intake. This neuroanatomical
specicity  could  reect   the  VTAs  role  in  the  dopaminergic
reward system. VTA dopaminergic neurons that project to the
NAc  are  under  tonic  inhibitory  control   mediated  by  GABA
A
receptors,   and  injections   of   picrotoxin  into  the   VTA  were
found  to  increase  DA  release  in  the  NAc  [554].   Nowak  et  al.
[550]   hypothesized   that   GABA
A
  antagonists,   by   producing
effects on the VTA DA system similar to those of ethanol, may
enable the animal to obtain the same rewarding effects while
consuming   less   ethanol.   Consistent   with   this   hypothesis,
administration of picrotoxin or of bicuculline during acquisi-
tion   of   ethanol-induced   CPP   was   found   to   increase   the
magnitude  of   ethanol-induced  CPP  relative  to  ndings   for
vehicle-treated  controls  [555],   and  simultaneous  microinfu-
sion  of   the  D
2
  antagonist   eticlopride  into  the  VTA  and  the
GABA
A
  receptor   antagonist   SR   95531   into   either   the   bed
nucleus  of  the  stria  terminalis  (BNST)   or  NAc  was  found  to
completely  attenuate  the  reduction  in  ethanol  self-adminis-
tration that was observed with eticlopride alone [383].
Partial inverse agonists that bindat the benzodiazepine site
on  the  GABA
A
  receptor   also  have  been  reported  to  reduce
ethanol self-administration [551,552,556566]. June et al. [561]
found   that   microinfusions   of   b-carboline-3-carboxylate-t-
butyl   ester   (bCCt)   into  the  ventral   pallidum  (VP)   produced
marked  reductions  in  ethanol-reinforced  behaviors,  but  that
no   effects   on  ethanol-reinforced  behaviors   were   observed
following infusion into the NAc or the CPu. The VP has been
found to play a role in regulating the rewarding properties of
both psychostimulant and opioid drugs [567572]. It has been
reported  to  code  the  normal   hedonic  impact   of   rewards  in
general   [573],   and  it  is  in  a  good  position  to  do  so,   since  it
receives efferent projections from the NAc [574,575], serves as
a centripetal nal common output path for mesocorticolimbic
circuits   [576578],   and   sends   projections   to   other   reward
structures  such  as  the  amygdala  [579,580],   orbitofrontal  and
insular   cortex   [578,581],   VTA,   and   parabrachial   nucleus
[575,582586].   Whether  the  VPs  role  with  respect  to  GABA
A
receptor  partial   inverse  agonists  is  analogous  to  that  of  the
VTAwith respect to GABA
A
receptor antagonists remains to be
hypothesized.
The effects of GABA
A
 agonists on ethanol self-administra-
tion  have  been  less   consistent   than  have  those  of   GABA
A
antagonists.   The   selective   GABA
A
  agonist   muscimol   was
found   to   decrease   operant   self-administration   of   ethanol
when  injected  intraperitoneally  [551]   or  into  the  NAc  [587].
When it was injected into the central nucleus of the amygdala,
it was found to decrease self-administration in dependent but
not nondependent rats [546]. Conversely, when it was injected
into the dorsal, but not the median, raphe nucleus it was found
to enhance ethanol self-administration[588]. Petry [563] found
that the effect of the benzodiazepine agonist chlordiazepoxide
dependedonthe dose administered: the lowest dose increased
ethanol   self-administration,   an  intermediate   dose   had   no
effect,  and  the  highest  dose decreased ethanol  self-adminis-
tration.   That   GABA
A
  agonists   produce   both  increases   and
decreases in ethanol self-administration could conceivably be
explained  by  a  combination  of   neuroanatomical   specicity
and the diversity in subunit composition of GABA
A
 receptors
[589].
These   ndings   with   GABA
A
  receptor   antagonists   and
agonists   could   lead   to   speculation   that   a   deciency   or
hyposensitivity  of  GABA
A
  receptors  in  the  VTA  or  in  the  VP,
or perhaps an excess or supersensitivity of GABA
A
receptors in
the  dorsal raphe, could contribute  to  an addictive  process. A
couple of other studies are more directly relevant to potential
relationships   between  GABA
A
  receptors   and   the   addictive
process.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 276
Tyndale and Tomkins [590] trained rats to self-administer
ethanol,   and  8   weeks   later   assessed  the   levels   of   GABA
A
receptor mRNAin various regions of their brains. Compared to
rats  that  were  in  the  lowest  15th  percentile  of  ethanol   self-
administration  (LES),   the  rats  that  were  in  the  highest  15th
percentile   (HES)   had   signicantly   higher   GABA
A
  receptor
mRNA  levels  in  the  dorsal  raphe,   medial  raphe,  cerebellum,
and hippocampus. The authors noted that the GABA
A
receptor
differences   between   the   two   groups   either   reected   the
groups  different  pre-existing  propensities  to  consume  etha-
nol or were caused by their differing ethanol exposure, adding
that they believed that the differences were part of the cause
or at least existed prior to exposure to ethanol. Their position
has  been  supported  by  more  recent   research  that   demon-
strated  that   long-term  ethanol   consumption  leads   to   sig-
nicant   decreases  in  expression  of   GABA
A
  receptor   mRNA
[591593].   So  to  whatever  extent  the  GABA
A
  receptor  mRNA
levels in Tyndale and Tomkinss [590] HES rats were affected
by  higher   levels  of   ethanol   consumption,   the  effect   would
have  been  in  the  direction  of   lowering  their   mRNA  levels
relative  to  LES  rats,   whereas  the  study  found  the  HES  rats
mRNA  levels  to  be  higher  than  those  of  the  LES  rats.   Thus,
elevated  GABA
A
  receptor  mRNA  levels  in  the  dorsal   raphe,
medial   raphe,   cerebellum,   or   hippocampus,   most   likely
associated  with  an  increased  density  of   GABA
A
  receptors,
may predispose to the development of an addictive process.
In a fascinating study, Laviolette et al. [594] proposed that a
discrete population of GABA
A
 receptors on non-dopaminergic
neurons  in  the  VTA  that  tonically  inhibit  VTA  dopaminergic
neurons serves as a potential addiction switching mechanism
by  gating  reward  transmission  through  one  of   two  neural
motivational systems, either a dopamine-independent system
or a dopaminergic system. And they demonstrated that in the
latter,   the  functional  conductance  properties  of  the  rat  VTA
GABA
A
  receptor  switch  from  an  inhibitory  to  an  excitatory
signaling  mode.   In  opiate-nave  animals,   animals  that  have
received  chronic  opiate  exposure  but   not   withdrawal,   and
animals   that   have   completed   and   recovered   from  opiate
withdrawal, opiates can produce their acute rewarding effects
through  a  DA-independent  system  that  is  mediated  through
brainstem  reward  circuits  [595597].   On  the  other  hand,   in
animals   that   are   in   a   state   of   opiate   withdrawal,   the
motivational   effects  of  opiates  are  dependent  on  the  meso-
limbic  DA  system  [596,598600].   Laviolette  et  al.   [594]   refer-
enced the suggestion by Robinson and Berridge [335,344] that
DA transmission mediates a drug wanting or craving signal,
independently  of  the  acute  rewarding  properties  of  opiates.
This suggestion makes particular sense in light of the opiate
withdrawal   states  mix  of  strongly  aversive  stimuli   that  are
relieved  by  administration  of  opiates.  Such  negative  reinfor-
cement motivation is incentively salient and thus suitable for
DA  signaling,   while  not   depending  at   all   on  reward.   While
Laviolette et al. [594] focused on opiate state  specically, the
difference between animals in opiate withdrawal and animals
not in opiate withdrawal  as the determinant of whether VTA
GABA
A
 receptors are in an inhibitory or an excitatory mode, I
believe that the scope of their ndings is deeper and more far-
reaching  than  opiate  states.   Neurobiologically,   the  state  of
opiate withdrawal is very similar to a state of acute stress. Both
involve   activation   of   the   hypothalamicpituitaryadrenal
(HPA)   axis  and  dysregulation  of   the  noradrenergic  system.
My hypothesis is that the component of the opiate withdrawal
state  that  determined  the  VTA  GABA
A
  receptor  switch  from
inhibitory to excitatory in Laviolette et al.s study [594] is one
that they share with the state of acute stress. The signicance
for addiction of the VTA GABA
A
 receptor inhibitory-excitatory
switch remains to be elucidated, but I believe that it also will
enhance our understanding of how recurrent acute stress can
potentiate an addictive process.
2.1.11.   GABA
B
 receptors
In  preclinical  research,   the  GABA
B
  receptor  agonist  baclofen
has  been  found  to  attenuate  self-administration  of   cocaine
[601604],   heroin  [605,606],   ethanol   [607],   nicotine  [608610],
and  d-amphetamine  [611].   It  has  been  found  also  to  reduce
reinstatement  of cocaine self-administration [612],  to  reduce
reinstatement   of   heroin   self-administration   [613],   and   to
decrease  stimulus-maintained  responding  for  either  cocaine
or  heroin  [614].   The  highly  selective  GABA
B
  receptor  agonist
CGP  44532  reduced  cocaine-induced  enhancement   of   brain
stimulation  reward  (BSR)   [615],   and  the   positive   allosteric
modulator of GABA
B
 receptors CGP 7930 reduced operant self-
administration of ethanol in ethanol-preferring rats [616].
Preliminary   clinical   studies   have   demonstrated   that
administration  of  baclofen  reduces  craving  for  both  cocaine
and   ethanol   in   addicted   patients   [617619].   Additionally,
baclofen  has  been  reported  to  attenuate  the  limbic  cortical
activation  induced  in  cocaine  addicts  by  conditioned  stimuli
that previously had been paired with cocaine use [602,620].
Baclofen  and   other   GABA
B
  agonists   are   understood   to
attenuate  the  reinforcing  effects  of   abusable  psychotropics
through modulation of DA transmission from the VTA to the
NAc,  and  perhaps  also  to  the  PFC  [606,621].  Their  targets are
inhibitory GABA
B
 receptors that are located on the cell bodies
of   VTA   dopaminergic   neurons   [622624]   and   that,   when
stimulated,   hyperpolarize   the   membrane   potential   and
decrease   the   ring   rate   of   these   neurons   [625,626].   Both
GABA
B
  agonists   and  GABA
B
  antagonists   can  increase   BSR
thresholds, whichsuggests a complex interactionbetweenthe
reward   system   and   GABA   function,   possibly   reecting
differential effects at pre- and post-synaptic receptors [627].
The  endogenous  ligand  for  the  GABA
B
  receptors  that  are
located on VTA DA neurons (i.e., GABA) is tonically produced
by local VTAGABAergic interneurons. Inadequate or abnormal
functioning   of   these   GABA   neurons   could   disinhibit   the
dopaminergic  neurons,   free  them  to  respond  more  enthu-
siastically when stimulated, and thus intensify the reinforcing
effects   of   substances   and   behaviors   that   can   be   used
addictively.
2.1.12.   Endocannabinoids  and cannabinoid  receptors
At   the   current   time,   there   are   two   known   cannabinoid
receptor subtypes: cannabinoid-type 1 (CB
1
), which are widely
expressed   throughout   the   peripheral   and   central   nervous
systems,   and   cannabinoid-type   2   (CB
2
),   which  show  high
levels of expression within the immune and enteric nervous
systems as well as in glial cells of the CNS. Both are coupled to
inhibitory   Gi/Go   proteins.   The   majority   of   neuronal   CB
1
receptors  appear  to  be  expressed  pre-synaptically.  Endogen-
ous   cannabinoids   or   endocannabinoids   (eCBs)   appear   to
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   277
function as retrograde neurotransmitters. Upon release from
postsynaptic   neurons   via   membrane   depolarization,   they
migrate   back   to   an   adjacent   presynaptic   membrane   and
activate   presynaptic   CB
1
  receptors,   which   then   inhibit
neurotransmitter release [223,724]. Two major classes of eCBs
have been identied thus far, exemplied by anandamide and
2-arachidonoyl  glycerol  [628631].   The  eCB  system  is  under-
stood   to   reinforce   both   the   motivation   and   the   reward
functions  of  the  mesolimbic  DA  system  in  its  regulation  of
eating behavior [632].
Dopaminergic terminals lack cannabinoid  receptors [633].
Nonetheless, genetic elimination of CB
1
 receptors (CB
1
 knock-
out   mice,   CB
1
1/)   abolished   DA   release   in   the   NAc   in
response   to   morphine   [634]   and   ethanol   [635],   and   the
cannabinoid  CB
1
  receptor   blocker   rimonabant   curtailed  DA
responses to administration of nicotine, ethanol, and cocaine
[636].   The  likely  anatomical  locus  for  the  CB
1
  receptors  that
made the critical difference in these studies is the presynaptic
terminals of VTA GABAergic neurons that synapse onto VTA
dopaminergic   neurons   and  modulate   their   activity.   Under
ordinary circumstances, eCBs that had been launched by VTA
dopaminergic   neurons   would   drift   over   to   activate   the
presynaptic CB
1
 receptors that mediate the inhibition of GABA
release, thereby decreasing the GABA-mediated inhibition of
DA  release.   Inactivation  or  elimination  of  the  CB
1
  receptors
interrupts this process and thus attenuates the DA responses
to administration of psychoactive substances and to any other
behavior that is associated withDArelease inthe NAc. Genetic
variants of cannabinoid CB
1
 receptors have been identied as
probable  factors   in  a  persons   vulnerability   to  develop  an
addictive disorder, especially one that involves consumption
of food or of psychoactive substances [236,237].
2.1.13.   Cyclic AMP response element  binding protein (CREB)
Cyclic   AMP   response   element   binding   protein  (CREB)   is   a
transcription factor that mediates effects of the cAMP second
messenger pathway on gene expression. Once CREB has been
phosphorylated by proteinkinase A(a proteinkinase activated
by cAMP) or another protein kinase, it forms dimers that bind
to specic CRE (cAMP response element) sites on target genes
andinteract withthe basal transcriptional complex to regulate
gene transcription [637].
Administration   of   psychostimulant   and   opiate   drugs
induces the phosphorylation and activation of CREB in several
reward-related   regions   [638643].   The   induction   of   CREB
activity appears to become greater and more persistent with
repeated   drug   exposures   [644].   In  the   NAc,   the   ability   of
psychostimulants to induce CREB is mediated via activation of
the  DA  D
1
  receptor   [638,639].   Cocaine  increases  cAMP-PKA
signaling in the NAc, which directly decreases medium spiny
neuron  (MSN)   excitability,   while  also  activating  CREB.   CREB
increases   MSN  excitability   and   thus   counterbalances   the
magnitude  of  the  cocaine-induced  decrease  [645].   Increased
CREB function in the NAc decreases an animals sensitivity to
the rewarding effects of cocaine, morphine, or sucrose, while
reduction in CREB activity produces opposite effects [646,647].
Chronic   administration   of   cocaine   or   other   stimulants
induces dynorphin expression in the NAc, and this induction
is dependent on CREB [639,646]. Dynorphin activates k opioid
receptors   on   VTA  dopaminergic   neurons   to   decrease   DA
release in the NAc and thus to dampen the reward-reinforce-
ment process [648]. At least some of the CREB-related decrease
in   the   rewarding   properties   of   drugs   is   mediated   by   the
induction of prodynorphin mRNA, which encodes dynorphin
[639].
Exposure of an animal to aversive stimuli activates CREB in
the NAc in muchthe same way as do drugs of abuse. Following
the parallel, researchers foundthat increasedCREBfunctionin
the NAc decreased an animals responsiveness to a variety of
aversive  or   negative  emotional   stimuli,   including  stressful,
anxiogenic, and nociceptive events; and that decreased CREB
function  in  this  region  increased  the  animals  sensitivity  to
these   conditions   [647,649].   Activation  of   CREB  in  the   NAc
appears  to  result  from  exposure  to  stimuli  of  high  hedonic-
emotional   charge,   whether  they  are  rewarding  or  aversive.
And   it   appears   to   mediate   a   behavioral   state   that   is
characterized  by   reduced  sensitivity   to   hedonic-emotional
stimuli in general, again regardless of their valence [637]. This
behavioral state resembles  the syndromes  of anhedonia and
emotional   numbing  that  can  characterize  depression,   post-
traumatic  stress   disorder   (PTSD),   and  some  forms   of   drug
withdrawal [650].
2.1.14.   DFosB
DFosB is a member of the Fos family of transcription factors.
These  proteins  dimerize  with  a  Jun  family  member  to  form
activator   protein-1   (AP-1)   transcription   factor   complexes,
which  bind  to  AP-1  sites   within  the  regulatory  regions   of
certain genes [637].
DFosB is induced by virtually all drugs of abuse, including
psychostimulants,   opiates,   ethanol,   and   nicotine,   among
others   [651655].   DFosB   also   is   induced   by   repetition   of
naturally   rewarding   behavior,   such   as   wheel   running   or
sucrose  drinking  [541,656],   and  by  several   forms  of   chronic
stress  [657,658].   Interestingly,   levels  of   stress-related  DFosB
induction  negatively  correlate  with  the  degree  to  which  the
animals develop learned helplessness, suggesting that induc-
tion of  DFosB represents an adaptive, active coping mechan-
ism  that   opposes  the  development   of   learned  helplessness
[657]. And  DFosB is induced by the chronic administration of
pharmaceutical antidepressants [651].
Inducible transgenic mice that overexpressed DFosB within
the NAc and dorsal striatum showed increased sensitivity to
the behavioral effects of cocaine, enhanced incentive motiva-
tion for cocaine, increased sensitivity to the behavioral effects
of morphine, and greater responsiveness to naturally reinfor-
cing   behaviors,   such  as   running   and   eating   [539,540,637].
Nestler, the author of these studies, interpreted their ndings
to suggest that  DFosB could be part of a sustained molecular
switch  that  functions  rst  to  induce  and  later  to  maintain  a
state  of   heightened  incentive  motivation  toward  reinforced
behaviors.
DFosB  appears  to  be  the  antithesis  of   CREB.   While  CREB
activation  mediates  a  state  of  reduced  reward  and  reduced
emotional reactivity, DFosB accumulation mediates a state of
heightened drug sensitivity and increased drive for rewarding
behavior.   Drug-induced   activation   of   CREB   in   the   NAc
dissipates  within  a  few  days  of   coming  off   the  drug,   while
DFosB increases over time and persists in the brain for up to 2
months [659].
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 278
At   one  time,   DFosB  may  have  seemed  to  epitomize  the
argument that drug addiction is caused by drug use. DFosB is
induced  by  drug  use,  it  increases  sensitivity  to  and  drive  for
drugs,  and  its  extended  duration  of  action  could  account  for
abstinent addicts vulnerability to relapse. However, as Nestler
noted  [539],  DFosB  does  not  last  long  enough  to  underlie  the
near-permanent   predispositions   that   are   seen   in   many
addicted   individuals.   He   suggested   that   variations   in   the
genes encoding DFosB could contribute to the genetic risk for
addiction:   for   example,   an   individual   with   a   gene   that
expresses   DFosB   at   high   levels   might   be   more   prone   to
addiction [660]. Another means by which levels of DFosB could
increase andthus contribute to anaddictive process is through
its induction by chronic stress [658,657].
2.1.15.   Dopamine and  cAMP-regulated  phosphoprotein
(DARPP-32)
The  dopamine and  cAMP-regulated  phosphoprotein  (DARPP-
32)   is   a   key   regulator   of   kinase-phosphatase   signaling
cascades  that  is  found  in  dopaminergically  innervated  areas
of the brain. cAMP that has been generated by activation of D
1
receptors  activates  protein  kinase  A  (PKA),  which  phosphor-
ylates DARPP-32 at the threonine-34 site (T34) [661,662]. When
DARPP-32 is phosphorylated at this site, it acts as an inhibitor
of protein phosphatase-1 (PP-1) [663], thereby maintaining the
phosphorylation state of various neuronal proteins. DARPP-32
can  be  phosphorylated  also  at  the  threonine-75  site  (T75)  by
cyclin-dependent   kinase  5.   DARPP-32  that   has   been  phos-
phorylated   at   this   site   inhibits   PKA  activity,   resulting   in
reduced efcacy of DA signaling [664]. In this way, DARPP-32
acts as a bidirectional signaling protein that regulates protein
phosphorylation  and  dephosphorylation  via  PKA  and  PP-1.
The  phosphorylation  state of  DARPP-32 is inuenced also  by
the   phosphatases,   calcineurin  (dephosphorylates   T34)   and
protein phosphatase 2A (dephosphorylates T75) [665].
DARPP-32  knockout  mice  were  found  to  have  heightened
substance   P-like   immunoreactivity   and   not   to   show  the
characteristic   increase   in   DFosB   after   repeated   cocaine
administration [666]. They also were reported to have reduced
cocaine   CPP   [371],   suggesting   that   a   DARPP-32   decit   can
decrease the rewarding properties of cocaine. Zachariou et al.
[369]  used  a  mutation  of  T34  to  demonstrate  that  phosphor-
ylation at T34 of DARPP-32 is a necessary mediator of cocaine-
induced place conditioning, locomotor activity, and sensitiza-
tion. The T34 mutationalso diminished the induction of DFosB
in the ventral striatum by chronic cocaine administration.
Donohue  et   al.   [667]   reported  that   transgenic  mice  that
overexpress an ethanol-sensitive isoform of adenylate cyclase
(AC7) had higher basal levels of T34 DARPP-32 in the striatum
and amygdala than did wild-type mice, whereas basal levels of
T75 DARPP-32 did not differ between wild-type and transgenic
mice. Ethanol administrationwas foundtoincreaseT34DARPP-
32 in the NAc and amygdala (but not in the striatum) of wild-
type and transgenic mice, with a greater effect in the amygdala
of transgenic mice. It was found also to increase T75 DARPP-32
in the amygdala of the wild-type mice only, and in the NAc and
striatumof boththetransgenicandwild-typemice. Theauthors
concludedthat the effect of ethanol onthe balance of DARPP-32
phosphorylation, especially inthe amygdala, may contribute to
differential motivational effects of ethanol.
2.1.16.   Neuropeptide Y (NPY)
Neuropeptide Y(NPY) is a36-aminoacidneuromodulator that
is  expressed  throughout   the  central   nervous  system  [668].
Most of it derives from neurons in the arcuate nucleus (ARC)
of   the  hypothalamus,   which  project   dorsally  to  the  para-
ventricular  nucleus  (PVN)  as  well  as  to  other  hypothalamic
and  extrahypothalamic  nuclei  [669].  NPY  is  involved  with  a
diverse set of biological functions that include integration of
emotional behavior [670,671], control of food intake [672,673],
neuronal   development   [674,675],   circadian  rhythms   [676
679],  pain  modulation  [680,681],  and  reproduction  [682,683].
NPY acts through at least ve receptor subtypes  the Y1, Y2,
Y4,  Y5, and  Y6  receptors  all  of which  couple  to G  proteins
that   inhibit   the  production  of   cyclic  adenosine  monopho-
sphate [684]. Y5 receptors are present at signicant levels in
the PVN, ARC, thalamus, and amygdala, which suggests the
presence  of  functional  hypothalamiclimbic  neural  circuits
[685].
A  number   of   studies  found  that   administration  of   NPY
[686,687] reduces ethanol consumptioninethanol-preferring P
rats and high ethanol-drinking HAD rats, but not in ethanol-
nonpreferring   NP   rats,   low-ethanol-drinking   LAD  rats,   or
nonselected  rats.  However,   other  studies  found  that  admin-
istration  of   NPY  increases  [688,689]   and  of   NPY  antagonist
decreases   [690693]   ethanol   self-administration.   Schroeder
et  al.  [694]  suggested  that  these  discrepant  ndings  could  be
reconciled   with  the   recognition  that   the   nature   of   NPYs
inuence   on   ethanol   intake   is   brain   region   dependent.
Research indicating that NPY increases ethanol intake infused
the  peptide  directly  into  the  PVN,   where  it  functions  as  an
orexigenic   agent   [688,689].   Meanwhile,   research  indicating
that   NPY   decreases   ethanol   intake   administered   it   via
intracerebroventricular infusion [686,687].
Other   research  speaks  with  a  more  unied  voice  about
NPYs   role  in  modulating   ethanol   consumption.   Mice  that
overexpress   NPY   consume   less   ethanol   than   wild-type
controls, while transgenic mice that lack NPY (NPY knockout
mice)   consume  more  ethanol   than  wild-type  controls  [695].
Ethanol consumption is suppressed by blockade of either NPY
Y1 receptors [693] or NPY Y2 receptors [696,697], but elevated
in  Y1  receptor   null   mutant   mice  [698].   And  in  humans,   a
polymorphism  in  NPY  (Leu7Pro)  was  signicantly  associated
with addiction in European American and Finnish alcoholics,
both  exhibiting   an  increased  frequency   of   the   Pro7   allele
compared with controls [699,700].
NPY is a highly potent activator of feeding behavior. When
administered   into   the   PVN,   it   induces   feeding   in  satiated
animals   and   it   seems   to   selectively   stimulate   prodigious
carbohydrate  intake  [222].   Studies  indicate  that   NPY  and  5-
HTplay antagonistic roles inthe regulationof feeding [701,702],
and  that   NPYs   stimulation  of   feeding  is   mediated  via  the
internal opioidsystem, sinceNPY-inducedfeedingisblockedby
the opioid antagonists naloxone and naltrexone [703]. NPYalso
hasbeenobservedtobindtonon-selectiveopiatereceptorswith
modest  activity similar  to that  of the  endogenous  opioid  leu-
enkephalin   [704].   NPYs   induction   of   feeding   in   satiated
animals,  its  involvement  with  the  internal  opioid  system,  its
antagonistic   relationship   with   5-HT,   and   its   presence   in
functional hypothalamiclimbic neural circuits suggest that it
could contribute to an addictive process.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   279
2.1.17.   Galanin
Galanin is a 29-amino acid neuropeptide that activates at least
three receptor subtypes coupled to Gi, Gq, or Go [705,706]. It is
synthesized inmany types of neuron, including brainstemNE-
producing cells of the LC and 5-HT-producing neurons of the
DRN.   Cells  that  express  galanin  are  concentrated  in  several
hypothalamic  areas:   the  dorsomedial   nucleus,   PVN,   perifor-
nical   lateral   hypothalamus   (PLH)   and  ARC.   They  send  out
dense projections throughout the hypothalamus as well as to
other   parts   of   the  limbic   system,   including  the  amygdala,
BNST, andhippocampus [707711]. Galaninserves a number of
disparate functions. It inhibits the ring of NE, 5-HT, and DA
neurons  and  reduces  release  of   these  neurotransmitters  in
forebrain   target   regions   [712].   It   inhibits   glucose-induced
insulin release and reduces levels of 5-HT, NE, and acetylcho-
line  through  the  inhibition  of   adenylate  cyclase  and  phos-
phatidyl   inositol   hydrolysis   [713].   Galanin  also  is   a  potent
endogenous   modulator   of   ring   pattern   in   hypothalamic
neuroendocrine cells [714], a hypothalamic-hypophysiotropic
hormone that modulates the secretion and action of luteiniz-
ing hormone releasing hormone (LHRH) [715], and a regulator
of food intake [716,717].
Injection of galanin in the PVN at a dose known to induce
feeding  has  been  found  to  potentiate  intake  of  4%  ethanol,
even  when  food   and   water   were   available   as   sources   of
calories and uid [718]. In rats that have been trained to drink
ethanol,   galanin   seems   to   stimulate   the   appetite   for   it
[718,719].   We  can  wonder  whether  a  similar  process  would
occur   with  rats   that   have   been  trained  to   drink   avored
solutions  that  have  been  paired  with  infusions  of  cocaine  or
heroin.
Galanin has interesting relationships with the endogenous
opioid  and  dopamine  systems   that   could  contribute  to  its
involvement in an addictive process. Galanin has been found
to potentiate morphine analgesia [720]. Meanwhile, naloxone
has been found to block galanin-induced feeding [721,722] and
to decrease PVNgalanin mRNAin ethanol-drinking rats, while
having little or no effect on galanin mRNA in water-drinking
rats [723]. Another study found that injection of galanin into
the PVN of rats releases DA and inhibits acetylcholine release
in  the NAc,  an effect  that occurs  only in  rats  that previously
had   demonstrated   signicant   increases   in   their   feeding
behavior   in  response  to  galanin  [724].   These  ndings  were
interpreted   in   one   later   study   as   indicating   that   ethanol
consumption  induced  by  PVN  injections  of   galanin  may  be
mediated  through  DA  in  the  NAc  [718],   and  in  another   as
indicating that galanin-induced overeating is associated with
DA release [723].
Studies   have   demonstrated   that   hypothalamic   galanin
increases  the  consumption  of   a  fat-rich  diet   [725,726]   and,
conversely,   that   the  consumption  of   fats  can  increase  the
expression   of   galanin   in   hypothalamic   nuclei   [727,728].
Leibowitz  et  al.  [723]  commented  that  these ndings suggest
the operation of a positive feedback loop that could contribute
to overeating until the cycle is interrupted by postingestional
satiety  signals  [717,729].  Interestingly,  Thiele  et  al.  [730]  and
Rada  et   al.   [718]   identied  a  similar  positive  feedback  loop
between galanin and ethanol intake, since ethanol increases
galaninexpressionin the PVNand injection of galanin into the
PVN  potentiates  consumption  of   ethanol.   A  key  difference
between the two positive feedback loops is that in the latter,
ethanol produces no satiety signal. (Actually, some sufferers of
bulimia  and  binge-eating  disorder   report   that   they  do  not
experience satiety signals after eating.).
2.1.18.   Orexin
Orexins  (or  hypocretins)   are  neuropeptides  that  are  synthe-
sized  in  neurons  of  the  posterior  and  lateral   hypothalamus
(LH).   Neurons  that   synthesize  the  excitatory  peptide  hypo-
cretin  also  synthesize  dynorphin,   a  peptide  that   usually  is
inhibitory [731].  Orexin neurons that  are located  in  posterior
hypothalamic  areas  are  involved  in  regulating  wakefulness,
thermogenesis,  and  energy  expenditure,  whereas  those  that
are located in the LH are involved in stimulation of appetitive
behaviors and in reward processing. Orexin A and orexin B are
produced by cleavage of a single precursor protein, and their
actions   are   mediated  by  two  G  protein-coupled  receptors,
orexin  receptor   type   1   (OXR1)   and  orexin  receptor   type   2
(OXR2). OXR1 shows higher afnity for orexin A, while OXR2
shows  equal   afnity  for  both  ligands  [732].   Orexin  neurons
receive input from the amygdala,  basal forebrain cholinergic
neurons,  GABAergic  neurons  in  the  preoptic  area  (POA),  and
serotonergic neurons in the median/paramedian raphe nuclei.
During  periods  of   wakefulness,   emotional   stimuli   from  the
limbic   system  and   cholinergic   inuences   from  the   basal
forebrain stimulate orexin neurons to maintain the activity of
the monoaminergic system, while sleep-active neurons in the
POAinhibit themduring periods of sleep [733]. Orexinneurons
of the LH-perifornical area send widespread axonal efferents
to  the  LC,   VTA,   NAc,   cortex,   and  midline  thalamus,   and  to
other  regions  of  the  lateral  and  medial  hypothalamus.  They
also maintain local axonal collaterals that terminate on other
cells   in   the   LH,   including   orexin   cells   and   neurons   that
synthesize  melanin  concentrating  hormone  (MCH),   though
orexin neurons show little direct response to orexin [731].
Orexin A has been demonstrated to increase the ring rate
and  in  some  cases  to  cause  burst   ring  of   VTA  dopamine
neurons   in   rat   brain   slices   [734].   Orexin   neurons   send
excitatory  projections  to  the  VTA  and  substantia  nigra  pars
compacta.  Intra-VTA  infusions  of  orexin  A  in  vivo  have  been
found to increase extracellular DA levels in the PFC and in the
shell   region  of   the  NAc,   but   not   in  the  NAc  core  [735,736].
Through   their   enhancement   of   midbrain   dopaminergic
system  activity,  orexins  can  potentiate  an  increase  in  motor
activity and thereby reduce the threshold for emitting specic
(previously rewarded) behaviors [737].
Orexin  neurons  in  the  LH  become  activated  by  cues  that
have  been  associated  with  consummatory  rewards  such  as
food and drugs [738]. Several studies demonstrated orexin to
have a critical role in activating reward-seeking or appetitive
behavior in response to conditioned or discriminative stimuli.
Intracerebroventricular  (icv)  infusion  of  orexin  was  found  to
evoke  a  dose-related  reinstatement  of  extinguished  cocaine
self-administration  in  rats,   an  effect  that  was  prevented  by
antagonists  of  receptors  for  NE  or  for  corticotropin  releasing
factor   (CRF)   [739].   The  inference  that   orexins  activation  of
cocaine self-administration reinstatement is mediated via NE
and CRF is consistent with ndings that icv administration of
orexin A activates CRF-expressing neurons in the PVN and the
central   nucleus  of  the  amygdala  (CeA)   [740].   Antagonism  of
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 280
orexin OXR1 receptors by the selective orexin Aantagonist SB-
334867  was  found  to  reduce  operant  responding  for  ethanol
and  also  to  abolish  cue-induced  reinstatement   of   ethanol-
seeking  behavior   in  ethanol-preferring  iP  rats   [741].   Harris
et  al.  [738]  reported  that  the  amount  of  Fos  activation  in  LH
orexin neurons of animals that had been conditioned via CCP
protocol for morphine, cocaine, or food reward was correlated
with the intensity of their reward-seeking. They reported also
that microinjection of orexin into the VTAcaused a signicant
reinstatement  response  for  morphine  reward;   that  adminis-
tration of the orexin antagonist SB-334867 after morphine CPP
training   produced   a   signicant   reduction   in   preference
compared to animals that were given a vehicle injection; that
reinstatement of extinguished CPP by microinfusionof rPP (rat
pancreatic  polypeptide,   an  agonist   at   NPY  Y4  receptors  on
orexin  neurons)   in  the  LH  was  similar  to  the  reinstatement
produced by systemic morphine; and that reinstatement was
completely  blocked  by  prior  systemic  administration  of  SB-
334867. Harris and Aston-Jones [742] concluded that both the
orexin  neurons   that   originate  in  the  perifornical   and  dor-
somedial   hypothalamic   areas   (PFADMH)   and   those   that
originate in the LHcan participate in drug relapse, but through
different   processes.   The  PFADMH  orexin  system,   which  is
involved  in  regulating  wakefulness  and  energy  expenditure,
drives  relapse  through  activation  of  stress  systems  (perhaps
involving CRF or NE), whereas the LH orexin system, which is
involved in reward processing, drives relapse through activa-
tion of brain circuits that are associated with reward learning
and reward-seeking behavior [742].
Neuroplasticity   at   VTA   glutamatergic   synapses   that   is
induced  by  drugs   of   abuse  has   been  suggested  to  play  an
important role in the behavioral consequences of in vivo drug
exposure [343,659,743]. A recent study by Borgland et al. [744]
demonstratedthat orexinAisacritical substrateinthisprocess.
Glutamatergic  N-methyl-D-aspartate  receptors  (NMDARs)   on
VTA DA neurons perform two major functions: they promote
burst ring[743,745], andtheyarenecessaryfor theinductionof
long-term potentiation [746,747]. Burst ring of VTA dopamine
neurons,   which  increases  extracellular  DA  in  the  projection
areas  more  efciently  than  does  a  regularly  spaced  train  of
action  potentials  [745,748],   signals  the  occurrence  of   salient
stimuli   and  facilitates  consolidation  of  the  relevant  memory
traces  [749].   Long-term  potentiation  is  an  enduring  decrease
in   depolarization   threshold   that   functionally   strengthens
synapses   and   contributes   to   synaptic   plasticity.   Borgland
et   al.   [744]   reported   that   in   vitro   application   of   orexin   A
potentiates NMDAR responses in VTA dopamine neurons; that
the  OXR1  receptor  antagonist  SB  334867  blocks  induction  of
cocaine-induced  potentiation  of   excitatory  inputs  onto  VTA
neurons; that orexin A causes late-phase increases in AMPAR-
mediated synaptic transmission; and that microinjection of SB
334867directlyintotheVTAblocks thedevelopment of cocaine-
induced locomotor sensitization. These data provide evidence
that  orexin  signaling  pathways  play  an  important  role  in  the
drug-induced   neural   plasticity   that   contributes   to   cocaine
addictionand by inference, to other addictions as well.
2.1.19.   Substance P  (SP)
Substance   P   (SP)   is   the   most   common  of   the   ve   known
mammalian neurokinins (or tachykinins), the others of which
are neurokinin A (NKA), neurokinin B (NKB), neuropeptide K,
and   neuropeptide   a.   Three   G   protein-coupled   neurokinin
receptors  have  been  identied    NK-1,   NK-2,   and  NK-3    for
which  SP,   NKA,   and  NKB  have  the  highest  binding  afnity,
respectively,   but   all   neurokinins   bind   to   all   three   NK-Rs
[750,751].  SP  is colocalized with other  neurotransmitters and
has  important  neuromodulatory  effects.   Examples  are  colo-
calizations  with  5-HT  in  the  nuclei   raphes,   with  DA  in  the
midbrain  and  striatum,  with  GABA  and  acetylcholine  in  the
cortex, and with CRH in the hypothalamus [752]. Examples for
direct   neuromodulatory  effects  of   SP  are  the  regulation  of
acetylcholine   release   in   the   human   cortex   [753]   and   the
modulation  of   noradrenergic   neurotransmission  in  the   LC
[754].
SP  and  the  NK-1  receptor  play  a  role  in  maintaining  the
activity  of  mesocorticolimbic  DA  neurons,   under  both  basal
and  drug-induced  conditions.  Blockade  of  NK-1  receptors  by
systemic  injection  of  the  NK-1  receptor  antagonist  CP-96345
decreased the number of spontaneously active DA neurons in
the  VTA  [755],   and  injection  of   a  SP  antibody  into  the  NAc
produced increases in concentrations of DA and metabolites,
an   effect   consistent   with   intracellular   accumulation   and
metabolism  of   DA  following  decreases  in  DA  release  [756].
SP   afferents   have   synaptic   contacts   with   dopaminergic
neurons   in   the   VTA   [757],   and   SP   is   present   in   high
concentration  in  terminals  close  to  the  VTA  dopaminergic
cell bodies [758,759]. Injection of SP directly into the VTA has
been found to increase the levels of DA and/or its metabolites
in  the  PFC  and  the  NAc,   suggesting  that   SP  stimulates  the
release of DA from mesocortical and mesolimbic DA neurons
[760762].   SP   has   been   shown   to   preferentially   activate
mesocortical   DA   neurons   in   a   manner   similar   to   acute
stressors  such  as  mild  footshock  or  restraint  [763].   Further-
more, evidence suggests that the increased turnover of DA in
the PFC and the NAc in response to stress may be mediated by
SP.   Increases  in  DA  metabolism  in  the  PFC  in  response  to
footshock-stress can be blocked by a SP antibody in the VTA
[764]. These ndings suggest that not only does SP have a tonic
facilitatory inuence on mesocorticolimbic DA activity, it also
may contribute to the DA-dependent behavioral responses to
drugs.   They  also  raise  the  possibility  that   the  activation  of
these  DA  neurons  in  response  to  stress  may  be  mediated  by
the endogenous SP system [765,766].
Injection  of  SP  into  the  VTA  has  been  shown  to  enhance
responding for conditioned reward, but not selectively for the
reward-paired lever [767]. Similarly, on a test of xed interval
responding, intra-VTA injection of SP was shown to increase
responding  on  the  non-reinforced  lever  [768].  These  ndings
suggest that activation of VTA cell bodies by SP or its analogue
produces increases in reward, but may also disrupt discrimi-
nation   processes   and   thereby   result   in   some   degree   of
response generalization [765].
The  experimental  literature consistently  afrms  that SP  is
involvedinopiaterewardprocesses. Reward-relatedbehavioral
effects   of   morphine   or   heroin  are   substantially  attenuated
when SPs favorite receptor, NK-1, is blocked by the antagonist
GR82334, ablated, or geneticallydeleted[766,769771]. However,
when  we  turn  to  consider  the  relationship  between  SP  and
psychostimulants, the research results are mixed and perplex-
ing.   Placenza   et   al.   [766]   reported   a   study   in   which   icv
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   281
administration   of   the   selective   NK-1   receptor   antagonist
GR82334hadnoeffect oncocaine-inducedlocomotor activation
or   cocaine  self-administration,   though  it   had  produced  sig-
nicant  increases  in  heroin  self-administration  and  attenua-
tion   of   morphine-induced   locomotion.   Their   results   are
consistent   with  studies  that   found  that   genetic  deletion  of
the  NK-1  receptor   did  not   impair   the  reinforcing  effects  of
cocaine   [769,770];   that   ablation   of   NK-1   receptors   in   the
amygdala  did  not  block  cocaine-induced  CPP  [771];   and  that
icv injections of NK-1 receptor antagonists had no effect on the
reinstatement   of   cocaine   seeking   induced   by   a   priming
injection  of  cocaine  [772].   However,  other  studies  found  that
infusion of the high-afnity nonpeptide NK-1 receptor antago-
nist   L-733,060   prior   to   a   systemic   injection   of   cocaine
signicantly  attenuated  the  cocaine-evoked  release  of  DA  in
the   striatum  [773,774];   that   infusion  of   the   NK-1   receptor
antagonist   WIN-51,708   prevented   the   massive   release   of
acetylcholine   in  the   striatum  that   cocaine   injection  would
ordinarily evoke [774]; that intrastriatal NK-1 receptor blockade
by  the  specic  NK-1  receptor  antagonist  LY306740  decreased
amphetamine-induced behavior [775]; and that pre-treatment
with  the  NK-1  receptor  antagonist  WIN-51,708  30 min  before
injections  of   methamphetamine  prevented  both  the  loss  of
dopamine  transporters  (DAT)   in  the  striatum  and  metham-
phetamine-inducedcell death[774]. Anattempt toreconcilethe
conicting  studies   by  questioning  whether   different   NK-1R
antagonists are functionally equivalent would miss the larger
point of perplexity: that a disruption of the mesocorticolimbic
DA system that blocks incentive-reward for opiates but not for
psychostimulants seems to be inconsistent withthe consensus
that   the  mesocorticolimbic   DA  system  where  SP  exerts   its
inuence is the primary conduit of incentive-reward-reinforce-
ment for both opiates and psychostimulants. Whether SP is to
be considered as a potential contributor to anaddictive process
hingesonhowtheseapparent contradictionsareresolved. If the
SP functions that are  most relevant to addiction  are  found  to
characterize opiates but not psychostimulants, then variations
of the SP-NK1 systemcould be factors that inuence whether a
person who is predisposed to developing an addictive disorder
is more likely to addictively  use opiates or psychostimulants,
but they would not be factors in an addictive process.
2.1.20.   Melanocortins (MCs)  and melanocortin  receptors
(MCRs)
Adrenocorticotropin  (ACTH)   and h-,   1-,   and  #-melanocyte-
stimulating  hormones  (h-,   1-,   and  #-MSH)   are  derived  by
enzymatic  processing  from  proopiomelanocortin  (POMC),   a
propeptide  that   is   produced  in  the  arcuate  nucleus  of   the
hypothalamus.   Collectively,   they  are  called  melanocortins.
The   hypothalamic   arcuate   nucleus   (ARC)   contains   two
discrete cell groups. One group co-expresses the anorexigenic
peptides   a-MSH  and  cocaine-   and  amphetamine-regulated
transcript peptide (CART). The second group coexpresses the
orexigenic peptides agouti-related protein (AGRP) and neuro-
peptide Y. Melanocortins act via ve receptor subtypes (MC1R
MC5R), all of which belong to the G protein-coupled receptor
superfamily andare positively coupledto adenylate cyclase. In
the   brain,   MC3R   and   melanocortin-4   receptor   (MC4R)   are
mainly  expressed,   with  little  expression  of   MC5R.   They  are
expressed   in   brain  regions   that   modulate   the   reinforcing
properties of drugs of abuse and natural reinforcers (e.g., food
and sex), including the NAc, the VTA, and the hypothalamus
[776,777].  All  melanocortin  receptors  are  activated  by  ACTH,
whereas all melanocortin receptors except MC2R are activated
by MSH. While a-MSH and AGRP bind as high-afnity agonist
and  antagonist,   respectively,   at  the  melanocortin  MC3R  and
MC4R receptors, their opponent effects on feeding seem to be
mediated by MC4R [778780].
Studies indicate that melanocortins operate inrelationships
of mutual inuence with the mesolimbic DA and endogenous
opioidreward-relatedsystems. Melanocortins have beenfound
to enhance dopaminergic neurotransmission, and DAhas been
found  to increase melanocortin function [781]. MCR  signaling
has   been   reported   to   regulate   ethanol   consumption   by
modulating   endogenous   opioid   activity   within   mesolimbic
DA  pathways  [782].   Repeated  administration  of   cocaine  has
been  found  to  increase  the  expression  of  MC4R  mRNA,   and
administrationof a lowdoseof morphine has beenfoundtoup-
regulate  the expression  of MC4R mRNA in the striatum  [783].
Meanwhile,   chronic   administration  of   morphine   has   been
found to result in down-regulation of MC4R mRNA expression
inthe striatumandperiaqueductal gray[784]. This MC4Rdown-
regulation and the associated decrease in melanocortin func-
tion  have  been  hypothesized  to  promote  the  development  of
opiate addiction [784].
At   rst   glance,   the   results   of   research   that   concerns
melanocortin  and  drugs  of   abuse  may  seem  to  be  contra-
dictory.   The   selective  MC4R  agonist   MTII   was   reported  to
reduce   ethanol   self-administration,   and   MCR   antagonists
were  reported  to  increase  it   [785].   Similarly,   melanocortins
were reported to reduce opiate self-administration [786], and
mutations  in  MC4R  were  found  to  be  associated  with  binge-
eating   disorder   [787].   On  the   other   hand,   infusion  of   the
melanocortin  MC4R  antagonist   SHU-9119  into  the  NAc  was
reported  to  block  the  rewarding  effects  of  cocaine:   cocaine-
induced  CPP,   cocaine-enhanced  responding  for  conditioned
reinforcement,   and  the  reinforcing  effectiveness  of   stimuli
that were conditioned by being paired with cocaine [781]. And
treatment with the MC4R agonist MTII was found to produce a
robust augmentation of amphetamine reward, as measured in
terms   of   its   ability   to   lower   the   threshold   for   lateral
hypothalamic   self-stimulation   (LHSS)   [788].   Whether   the
contradiction  is  more  apparent  than  real  is  hard  to  discern,
since   the   outcome   measures   are   not   really   comparable.
Measures  of  self-administration  of  a  substance  are  unques-
tionably related to its reward value, but not necessarily in the
same way as is CPP or the lowering of the LHSS threshold. In
fact,   the   correlation  is   not   always   in  the   same   direction:
increased reward is associated sometimes with increased self-
administration  and  sometimes  with  decreased  self-adminis-
tration.
2.1.21.   Leptin
Leptin  is  a  protein  hormone  that  is  generated  in  adipocytes
and  interacts   with  six  types   of   receptor   (LepRaLepRf).   Its
primary  physiological   function  is  the  regulation  of  appetite.
When  leptin  binds  to  LepRb  receptors  in  the  ventral  medial
hypothalamus   (VMH),   a   satiety   signal   is   generated   that
instructs the brain to direct the body to stop eating. The cells
of the ARC, located in the VMH, receive leptin signals and then
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 282
communicate to other hypothalamic  and extrahypothalamic
structures  via  neuropeptide  transmission  [reviewed  in  789].
Leptinreceptors are expressedalso by some neurons inthe LH,
including orexin and MCH neurons [790], which suggests that
these   neurons   may   respond   directly   to   peripheral   leptin
signals.   In  addition,   the  VMH  has  extensive  connections  to
the LH.
Leptin  works   by  inhibiting  the  activity  of   neurons   that
contain   NPY   and   agouti-related   peptide   (AgRP),   and   by
increasing the activity of neurons that express a-MSH. It also
down-regulates  the  expression  of   endocannabinoids  in  the
hypothalamus [632].
Leptin  not  only  suppresses  food  intake,  but  also  reverses
the  effects  of   food  restriction  on  brain  stimulation  reward
thresholds  [791]   and  on  the  reinstatement   of   drug-seeking
[792].   Food   and   drugs   work   through   common   molecular
substrates  within  the  brain  [793],   and  connections  between
the  hypothalamus  and  the  NAc  may  underlie  some  of   the
behavioral   observations  of   cross-sensitization  between  nat-
ural   rewards  and  drugs  of  abuse  [243].   Most  drugs  are  self-
administered to higher levels after food deprivation [794], and
food deprivation increases cocaine-conditioned place prefer-
ence  [795].   Leptin  acts  to  reduce  heroin  self-administration
[792],   and  has  been  shown  to  modulate  LHSS  [796].   Investi-
gators are beginning to consider impaired leptin activity as a
potential   factor   in  a   shared   vulnerability   to   psychoactive
substance addiction and bulimia [228,239].
2.1.22.   Glutamate
Burst   ring  in  mesencephalic  DA  neurons  is  dependent   on
excitatory   afferents   that   activate   ionotropic   glutamatergic
receptors  on  DA  cells  [498,797,798]   The  pause  after  a  burst
can   be   mediated   by   glutamate   acting   on   metabotropic
glutamatergic  receptors  (mGluRs),   which  induces  inhibitory
postsynaptic   potentials   (IPSPs)   [799,800].   Psychostimulants
selectively  reduce  the  mGluR-induced  IPSPs  in  DA  neurons
through  cross-desensitization  that  is  mediated  by  a-adreno-
ceptors [801]. As a result, acute exposure to a psychostimulant
increases DA neuron bursting via a-adrenoceptors [802]. This
suggests that a-adrenergic receptors play an important role in
mediating   drug   reinforcement   [803].   Indeed,   Drouin  et   al.
found that mice that lacked a-adrenoceptors exhibited neither
psychostimulant-induced  locomotor   hyperactivity  [804]   nor
rewarding   effects   [479].   Therefore,   an  interaction  between
noradrenergic and  glutamatergic systems may  modulate  the
ring pattern of DA neurons, which in turn may underlie the
reinforcing value of drugs and the establishment of addictive
behavior [805].
In a study by Tremblay et al. [806], a group of patients with
major depressive  disorder who were administered d-amphe-
tamine demonstrated enhanced rewarding effects, compared
with  the  control   group.   The  degree  of  enhancement  that  d-
amphetamine   elicited   correlated   with   the   severity   of   the
depressed patients anhedonic symptoms. The hypersensitive
response  to  d-amphetamine   was   associated  with  negative
blood  oxygen  level-dependent  (BOLD)  signals  in  most  of  the
regions   of   interaction.   The   physiological   counterparts   of
negative BOLD  signals  are  thought  to  be  induced  by  reduced
blood   ow  (i.e.,   active   neuronal   inhibition  and   decreased
cortical  excitability)  [807809].  The  relative  decrease  in  brain
activity  among  depressed  subjects  could  reect  exaggerated
deactivation of glutamatemediated transmission by amphe-
tamine (as per the preceding paragraph).
We  are  considering  a  distinction  between  two  groups  of
people that renders one of them more susceptible to develop
an addictive disorder, and three possible constituents of this
distinction  that   emerge   out   of   the   preceding   discussion.
First,  Tremblay  et  al.  [806]  referenced  a  study  that  reported
evidence   of   abnormal   glutamate   transmission   in   major
depression   [810],   by   way   of   suggesting   that   abnormal
glutamate transmission could have generated the depressed
groups  enhanced  sensitivity  to  the  effects  of   d-ampheta-
mine. Second, the converse of Drouin et al.s ndings that a-
adrenoceptor hypoactivity reduces psychostimulant-induced
locomotor  hyperactivity  and  reward  is  that  a-adrenoceptor
hyperactivity could intensify these psychostimulant-induced
effects.   Such   adrenergic   hyperactivity   is   not   unusual   in
states  of  chronic  stress.  Finally,  Tremblay  et  al.  [806]  noted
that   the   degree   of   enhancement   that   d-amphetamine
elicited   correlated   with   the   severity   of   the   depressed
patients   anhedonic  symptoms.   Thus,   anhedonia  emerges
as a risk factor or marker for the development of an addictive
disorder.
2.1.23.   Glucocorticoids
Both  in  rodents  and  in  primates,   positive  correlations  have
been  observed  between  circulating  glucocorticoid  levels  and
psychostimulant  self-administration  [811813].   Most  studies
on   self-administration   have   reported   increases   in   drug
responding   following   repeated   or   prolonged   exposure   to
stress   levels   of   glucocorticoids   [812815].   Glucocorticoids,
probably via glucocorticoid receptors, facilitate DA transmis-
sion in the NAc shell [816]. During chronic stress, the repeated
increase  in  glucocorticoid  hormones  and  DA  could  result  in
sensitization  of   the   reward   system.   This   sensitized  state,
which can persist after the end of the stress, would render the
subject more responsive to drugs of abuse and consequently
more vulnerable to the development of addiction.
Glucocorticoids, the hypothalamicpituitaryadrenal (HPA)
axis,   and  the   stress   response   system  are   discussed  more
extensively  in  the  following  section,   where  we  consider  the
impaired   affect   regulation   component   of   the   addictive
process.
2.2.   Impaired affect regulation
Affect regulation comprises the neurobiological processes that
maintain  emotional   states   within  ranges   of   intensity   and
stability   that   do   not   impair   functioning   or   lead   to   over-
whelming distress. The addictive disorders literature employs
the  term impaired affect  regulation  less often than  it uses
terms  that  refer  to  its  more  clearly  dened  and  experience-
near manifestations, such as chronic stress, stress hypersen-
sitivity,   depression,   and  anxiety.   A  term  as  important   and
apparently  vague  as   stress   also  merits   denition.   While
diverse and elaborate denitions abound, the words etymol-
ogy   might   be   an   expeditious   beginning:   Middle   English
stresse,   short   for   destresse   (from  Old   French),   distress.
Stress/distress  seems  to  be  a  synonym  of  pain  or  suffering,
with a connotation of tension or strain.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   283
2.2.1.   Corticotropin  releasing factor  (CRF)
Corticotropin releasing factor (CRF) is released from two brain
regions  to  participate  in  two  separate  (but  connected)  stress
systems  [817].   In  the  hypothalamicpituitaryadrenal   (HPA)
stress system, CRF neurons in the parvocellular region of the
paraventricular nucleus of the hypothalamus (PVNh) regulate
the  release  of  pituitary  adrenocorticotropic  hormone  (ACTH)
and  adrenal   glucocorticoids  (GC).   In  the  extrahypothalamic
(EH) stress system, CRF neurons in the central nucleus of the
amygdala   (CeA)   project   to   the   locus   coeruleus   (LC)   and
increase  the  ring  rate  of  LC  neurons,  resulting  in  increased
NE  release  in  the  terminal   elds  of  this  ascending  noradre-
nergic  system  [818821].   One  of  the  principal   noradrenergic
targets of this systemis actually the CRF neurons of the PVNh.
NE is the major known source of drive over CRF release from
PVNh  neurons  during  stress  [822,823].   The  activation  of  the
CRF neurons of the PVNh is associated with increased activity
in   the   nucleus   tractus   solitarius   (NTS)   and   the   dorsal
medullary nucleus, as well as the LC.
Either stress system can contribute  to  both a  pre-existing
vulnerability to use drugs addictively and a later vulnerability
to  relapse.   The  HPA  stress  system  seems  to  have  the  more
important   role  in  the  initiation  of   drug-seeking  and  in  the
maintenance  of   drug-taking  behavior,   while  the  EH  stress
system  seems   to   have   the   more   important   role   in   the
motivational   effects   of   both   protracted   abstinence   and
stress-induced reinstatement [824827].
In  the  stress-induced  reinstatement  procedure,   rats  that
previously   had  been  trained  to  self-administer   cocaine   or
heroinand thenextinguished will reinstate their responding if
a  mild  footshock  is   administered  immediately  prior   to  the
testing session[828832]. This reinstatement was reportedtobe
blocked  by  pre-treatment  with  CRF  antagonists  administered
directly  into  the  brain,  but  not  by  removal  of  glucocorticoids
[833836].   Research   results   indicated   that   the   key   neural
pathway  for   this   process   originates   in  CRF  neurons   of   the
CeA and ends at CRF
1
 receptors on the ventral BNST [835838].
Stress  stimuli  that  activate  CRF  circuits  have  been  found
also to potentiate mesolimbic dopaminergic reward pathways
in   laboratory   animals   [839].   Similarly,   human   laboratory
studies have shown that emotional stress and negative affect
states   increase   drug   craving   in   drug-addicted   individuals
[841,842]. Preclinical studies have demonstrated that early life
stress  and  chronic  stress  can  result  in  enduring  changes  in
stress  responses  [817,839850].   Such  changes  can  alter   the
sensitivity   of   the   DA  system  to   stress   and   can   increase
susceptibility  to  self-administration  of   substances  of   abuse
[840,851,852].
A suggestion by some investigators [853855] that anxiety
and  affective  disorders   be  considered  to  be  chronic  stress
states  makes  immediate  sense  of   the  data  on  comorbidity
between  these  disorders  and  addictive  disorders.   The  high
levels  of  serum  cortisol   that  issue  from  a  hyper-responsive
HPA axis during chronic stress enhance the sensitivity of the
mesolimbic   DA   system  to   the   reinforcing   properties   of
psychoactive  substances  and  rewarding  behaviors,   thereby
increasing  the  risk  that   affected  individuals   will   use  such
substances  and  behaviors  addictively  [824,856].   Accordingly,
Tremblay   et   al.   [857]   found  a   strong   positive   relationship
between  the  severity  of  subjects   depressive  symptoms  and
the degree of reward effect that they experienced from a dose
of d-amphetamine. Compared to healthy controls, individuals
with PTSD [858,859] and with depression [860862] were found
to have higher levels of CRF in their CSF.
Finally, a series of stressful episodes were reported to have
led to a marked and prolonged increase in ethanol consump-
tion in CRF
1
 receptor-decient mice but not in wild-type mice,
even  though  CRF
1
  receptor-decient  mice  and  the  wild-type
mice  had  not   differed  in  ethanol   consumption  prior  to  the
stressful   episodes   [863].   These  data  suggest   that   the   CRF
1
receptor is involved in adaptive responses to stress, the lack of
which  leads   the   CRF
1
  receptor-decient   mice   to   resort   to
ethanol to manage their stress [864].
Research  results  seem  to  concur  that  a  hyper-responsive
HPAaxis and chronic stress conditions  including anxiety and
affective disorders  are likely to be signicant risk factors for
the development of addictive disorders.
2.2.2.   Cortisol
Receptors  for  cortisol   (or  glucocorticoid  receptors  [GRs])   are
located  in  the  hippocampus,  the  limbic  system,  and  the  PFC
[865,866].   During  periods  of  psychological   distress,   cortisols
diurnal pattern is overridden by signals to the hypothalamus
that originate in the amygdala and the BNST, structures that
are  activated  by  conditioned  and  unconditioned  stimuli  and
that convey information having survival value [867869]. The
BNST   also   provides   the   primary   inputs   to   the   PVN  that
generate   an   HPA  response   to   psychological   stress.   These
inuences   are   augmented  during   periods   of   psychological
stress  by  NE  inputs  that  ascend  from  the  LC  to  activate  the
cerebral cortex and limbic system [870,871].
Acute cortisol administration has been found to precipitate
cocaine  craving  in  human  addicts  [872],   as  has  stress  [873].
Stress   also   has   been  demonstrated  to   increase   drug   self-
administration  in  animal   models  [874].   The  stress-charged
drive  to  self-administer   drugs  of   abuse  has  been  linked  to
increased  activation  of  the  mesolimbic  DA  system,  which  is
mediated by glucocorticoid release [874877]. Repeated expo-
sure   to   stress   has   been   shown   to   induce   a   long-lasting
enhancement   of   the   mesolimbic   DA  response  to  drugs   of
abuse [878].
Interestingly, a hypo-responsive HPAaxis withlowlevels of
cortisol   also  has  been  associated  with  enhanced  drug  self-
administration  [879,880].   These  ndings  are  consistent  with
human studies that demonstrated lower cortisol response to
stress  in  individuals  who  had  behavioral  conduct  problems,
externalizing symptoms, and antisocial personality [881885].
In  adolescent  boys,   lower  stress-related  cortisol   levels  were
found to be associated with subsequent increased frequency
of drug use [886].
Wei   et   al.   [887]   found  that   transgenic   mice   with  over-
expressed  GRs  manifested  a  signicant  increase  in  anxiety-
like  and  depression-like  behaviors  relative  to  wild  type,  and
also  to  show  enhanced  sensitization  to  cocaine  [887].  Mean-
while,   decreasing  the  production  of  central   nervous  system
(CNS)   GRs  reduced  cocaine  self-administration  [888],   which
suggests that self-administrationdepends onfeedback signals
from cortisol receptors to the CNS [889].
The  cortisol   variant  that  seems  to  be  the  more  likely  to
contribute  to  an  addictive  process  is  enhanced  secretion  or
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 284
increased  receptor  sensitivity,   which  would  increase  activa-
tion  of   the   mesolimbic   DA  system  in  the   short   run  and
chronically would  induce  a  long-lasting enhancement  of  the
mesolimbic DA response to drugs of abuse.
2.2.3.   Norepinephrine (NE)
When the LC is ring at normal rates, NE increases the signal
to  noise  ratio  of   responses  evoked  by  other  afferents,   both
excitatory  and  inhibitory,   and  enhances  synaptic  transmis-
sion  in  target  circuits  [890892].  In  an  acute  stress  situation,
the  LC  ring  rate  increases,  enhancement  of  signal  to  noise
ratio decreases, and the LC becomes the brains alarmsystem.
Another  important  role  of  the  LC-NE  system  during  stress  is
inhibition   of   the   PFC,   thereby   favoring   rapid   instinctual
responses over more complex ones in the service of surviving
acute life-threatening situations [890].
Early   studies   suggested   that   LC   neuronal   activity   was
driven   primarily   by   aversive   stimuli   and   that   NE   was
essentially   a   stress   neurotransmitter.   These   observations
led  to  a  number  of  hypotheses  that  the  function  of  the  LC-
NE  system  was  alarm-  or  anxiety-related.   However,   electro-
physiological studies demonstrated that phasic responses are
elicited by appetitive as well as aversive stimuli, provided that
a stimulus is perceived as salient [893,894]. And microdialysis
studies   demonstrated   elevated   extracellular   NE   levels   in
response  to  appetitively  conditioned  stimuli  [895897].  Com-
bined, these observations suggest that both phasic and tonic
LC discharge activity is related to the overall salience and/or
arousing nature of a given stimulus more closely than it is to
the affective valence of the stimulus [898]. In this regard, our
understanding   of   NE   seems   to   be   following   the   same
developmental   path  as   has   our   understanding   of   DA  and
CREB, from affective-hedonic valence to salience.
Noradrenergic   antagonists   were   found   to   block   stress-
induced reinstatement [487,489,899], much as did CRF antago-
nists. The brain sites for these effects appear to have been the
ventral noradrenergic bundle projections to the BNST. Neuro-
toxin-specic lesions of the ventral noradrenergic bundle were
found  to  attenuate   stress-induced  reinstatement   of   heroin
responding [489], and local injection of a b-adrenergic receptor
antagonist intothe BNSTalsoblockedstress-inducedreinstate-
ment in cocaine-trained rats [488]. The conditioned release of
NE in the  BNST in response to stressors may elevate anxiety,
which  then  augments   the   reward   value   of   drugs   through
negative reinforcement processes [827,900].
2.2.4.   Norepinephrine (NE) and  serotonin (5-HT)
The   College   de   France   group  [496]   rst   demonstrated  that
psychostimulant- or opiate-induced locomotor activation and
behavioral sensitizationare entirely dependent onthe stimula-
tion of two non-dopaminergic monoaminergic receptors,  a
1b
-
adrenergic  and  5-HT
2A
.   They  then  followed  up  with  another
study [901], in which they found that repeated treatments with
d-amphetamine increased the reactivity of both noradrenergic
and serotonergic neurons, and that this hyperreactivity could
be blocked by pre-treatment withana
1b
-adrenergic or a 5-HT
2A
receptor antagonist. Theypostulatedthat innaveanimals both
types   of   neuron   regulate   one   another   through   these   two
receptors.   For  example,   the  activation  of  noradrenergic  cells
by   external   stimuli   would   be   immediately   attenuated   by
serotonergic  cells,   the  activation  of   which  is  itself   triggered
by noradrenergic neurons. They hypothesized that this closely
coupled  control   vanishes  after  repeated  administration  of  d-
amphetamine. And they proposed that this long-term uncou-
pling  between  noradrenergic  and  serotonergic  neurons  may
explain   the   extreme   sensitivity   to   emotions   described   by
human addicts during withdrawal. After noting that stressful
situations cross-sensitize with the effects of psychostimulants
or opiates on behavioral sensitization, they concluded with the
statement  that  chronic  stress  could  therefore  also  induce  an
uncoupling  between  noradrenergic  and  serotonergic  systems
and  thus  be  one  source  of   mental   illnesses  such  as  bipolar
disorder.   They  might  just  as  easily  have  concluded  that  this
noradrenergic-serotonergic  uncoupling  due  to  chronic  stress
could be a source of addictive disorders.
2.2.5.   Serotonin  (5-HT)
Serotonin (5-HT), 5-HT receptors, and the 5-HT transporter (5-
HTT) have been the subjects of considerable interest in recent
years.   Genetic   research  has   focused   on  the   5-HTT-linked
polymorphic  region  (5-HTTLPR),   the  promoter  region  of  the
gene  that  encodes  5-HTT,   which  contains  a  common  func-
tional   polymorphism  with   a   variable   number   of   tandem
repeats (the short allele has 14 repeat elements, the long allele
has   16)   [226,680,902910].   Research   that   investigates   the
effects   of   the   postnatal   environment   on   neurobiological
development   has  focused  on  the  5-HT  receptors,   primarily
the  5-HT
1A
  and  to  a  lesser  extent  the  5-HT
1B
  and  the  5-HT
2A
[911914]. And research that explores the interaction between
genetic   and   environmental   factors   has   thus   far   focused
primarily  on  the  5-HTTLPR  [915918],   and  to  a  lesser  extent
on  the  5-HT
2A
  receptor   [919].   While  discussion  of   develop-
mental research has been deferred to another publication, its
mention  here  may  serve  to  indicate  that  the  5-HT  system  is
receiving a lot more scientic attention than might be inferred
from the following review.
The   5-HTT  is   an  integral   membrane   glycoprotein  that
occurs in pre-synaptic neuronal membranes. Its job is to take
up  5-HT  into  the  pre-synaptic  neurons  after   its   release  in
synaptic spaces, with the function of terminating the synaptic
action of 5-HT and recycling it [689].
A study reported that alcoholic subjects had a lower 5-HTT
density   in   perigenual   anterior   cingulate   cortex   than   did
control subjects [920]. The difference was not explained by a
nonspecic  ethanol-induced  down-regulation  of   5-HTT,   nor
by a general neuronal loss inthe frontal cortex [921,922]. These
results   also   indicated   that   lower   5-HTT   density   in   the
perigenual  anterior  cingulate  cortex  was  not  correlated  with
age at time of death (which was presumed roughly to reect
the  duration  of   ethanol   abuse).   Another  study  found  lower
midbrain and amygdala 5-HTT radioligand [11C]McN 5652 BP2
(binding potential) in subjects with major depressive disorder
than  in  controls  [850].  BP2  did  not  correlate  with  depression
severity. The authors interpreted the lower [11C]McN5652 BP2
in the subjects with major depressive disorder to reect lower
Bmax, or lower total number of available 5-HTT binding sites.
Less 5-HTinput to the amygdala, as suggestedby the nding of
lower  5-HTT  BP2,   may  result  in  increased  amygdala  activity
[923], as 5-HT enhances inhibition in the amygdala, presum-
ably through activation of GABA interneurons [924].
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   285
2.2.6.   Dopamine  (DA)
The chronic mild stress model has been suggested to have the
best face  validity  of any  animal model of  depression,  in  that
repeated  mild  stresses  over  time  gradually  induce  a  state  of
decreased responsiveness to rewards and reduced sexual and
aggressive  behaviors   [925].   Rodents   exposed  to  this   model
demonstrate  decreased  D
2
/D
3
  receptor   binding  in  the  NAc,
which is reversed by chronic antidepressant treatment (TCAs,
SSRIs, or mianserin) [926].
A   recent   study   used   functional   magnetic   resonance
imaging  to  assess  the  activity  of  brain  reward  systems  after
d-amphetamine challenge in 12 drug-free depressed patients
and 12 matched controls [806]. The depressed subjects had a
markedly greater behavioral response to the rewarding effects
of   the  psychostimulant   and  altered  brain  activation  of   the
ventrolateral PFC, orbitofrontal cortex, caudate, and putamen.
These   ndings   suggest   that   major   depressive   disorder
involves dysfunction of the dopaminergic system. Two earlier
sets of research results  round out  the picture:  (1) the  nding
that glucocorticoids selectively facilitated DA transmission in
the  NAc  [816];   and  (2)   the  nding  that   when  subjects  who
reported  poor   early   life   maternal   care   were   exposed  to  a
psychosocial stressor, their ventral striatal DA concentrations
increased  more  than  did  those  of   subjects  who  did  not   so
report, and the DAincrease was correlated with an increase in
salivary cortisol concentrations [927].
The  high  incidence  of   hypercortisolemia  in  depression,
particularly   in   severe   depression,   raises   speculation   that
elevated  cortisol   concentrations  alter   dopaminergic  reward
systems,  thereby  altering  hedonic  responsiveness  [928].  One
proposed model posits that over time, frequent bouts of stress
associated with intermittent increased exposure to glucocor-
ticoids   sensitizes   the   mesolimbic   DA   system  [929].   Such
sensitization of the mesolimbic DA system would predispose
a person to develop an addictive disorder.
2.2.7.   Endocannabinoids
The endocannabinoid (eCB) system appears to be involved in
modulation  of  depression  [930],   stress  [931,932]  and  anxiety
[933935], while its absence results in a greater vulnerability to
stress [936]. Chronic stress was reported to down-regulate CB
1
receptor  expression  and  signicantly  reduce  the  content  of
the   endocannabinoid   2-arachidonoyl   glycerol   within   the
hippocampus   [937].   CB
1
  knockout   mice   were   found   to
show   increased   aggressiveness,   anxiety-like   responses,
depressive-like  responses  in  the  chronic  unpredictable  mild
stress  procedure  [938],   and  HPA  axis  changes  that  included
reduced basal corticosterone secretion and hypersensitivity to
restraint  stress  [939].  Evidence  for  an  endogenous  anxiolytic
cannabinoid  tone  also  comes  from  the  anxiogenic  effects  of
the CB
1
 receptor antagonist rimonabant [940]. The eCB system
might  be  activated  in  response  to  anxiogenic  situations  and
might   regulate   emotional   states   by   modulating   amygdala
outputs,   as  part   of   a  negative  feedback  system  that   limits
anxiety [932,941].
The   primary   function  of   the   eCB   system  seems   to   be
regulation or containment of chronic stress. Disruption of the
eCB  system  would  be  likely  to  increase  the  level   of  chronic
stress,   which  in  turn  would  increase   the   likelihood  of   an
addictive disorder developing.
2.2.8.   Cyclic AMP response element  binding protein  (CREB)
A review article [942] proposed that cAMP response element-
binding  protein  (CREB)   has   a  role  in  anxiety  and  ethanol-
drinking  behaviors.   Wistar  rats  with  high  levels  of   anxiety
were reported to consume more ethanol than did those with
lowlevels of anxiety [943], and strains of rats that were bred to
consume large quantities of ethanol were reported to exhibit
more  anxiety-like  behaviors  than  did  their  ethanol-nonpre-
ferring counterparts  [944946]. The  transcription  factor CREB
regulates the expression of the gene that encodes neuropep-
tide Y(NPY), which is involved in the regulation of anxiety and
the  modulation  of  ethanol  consumption.  Intracerebroventri-
cular (icv) infusionof NPYwas found to signicantly attenuate
the  ethanol   intake  of  P  rats  but  not  of  NP  rats  [686]   and  to
produce   electrophysiological   effects   similar   to   those   that
ethanol   produced   in   P   rats   [947].   Transgenic   mice   that
overexpress NPY also were found to have a lower preference
for   ethanol   [695].   Conversely,   NPY-null   mutant   mice  were
observed to consume greater quantities of ethanol, and mice
that lacked the RII1 regulatory subunit of the PKA gene and
thus  were  unable  to  phosphorylate  CREB  were  reported  to
consume greater amounts of ethanol than did wild-type mice
[948]. CREB has been proposed to regulate anxiety and ethanol
abuse behaviors via NPY [949]. Noting that CREB is associated
also  with  the  molecular  effects  of  other  addictive  drugs,  the
author suggested that changes in synaptic plasticity that are
mediated by CREB might be a common factor in ethanol and
drug addiction.
Another  review  [950]  reported  that  CREB  is  stimulated  in
the  NAc  by  exposure  to  several   types  of   drugs  of   abuse  or
stress,   and   that   CREB   function   in   the   NAc   normally   is
regulated  by  glutamatergic  and  dopaminergic   inputs   [951].
Numerous studies have established that CREB activity in this
region has a profound effect on an animals responsiveness to
emotional   stimuli   [952,953].   The  authors   understood  these
ndings to suggest that  by determining the set point of NAc
neurons   [954]      CREB   represents   an   emotional   gate   for
behavioral   responsivity.   Viral   vector-mediated  elevations  of
CREB  within  the  rat  NAc  were  found  to  produce  anhedonia-
like signs and a generalized numbing of behavioral responses
to   both   aversive   and   pleasurable   emotional   stimuli
[646,647,649].   Similarly,   overexpression  of   CREB  in  the  NAc
of   inducible   transgenic   mice   was   found   to   produce   a
depression-like phenotype [955] and to reduce the rewarding
effects of cocaine [659]. The authors observed that short-term
increases   in  CREB  activity  in  the  NAc,   induced  by  normal
rewarding or aversive stimuli, can serve to dampen responses
to subsequent stimuli and facilitate the ability to actively deal
with the situation at hand. However, they noted, under more
pathological conditions, larger and more sustained increases
in CREB activity, induced by drugs of abuse or excessive stress,
can  lead  to  an  excessive  dampening  of  emotional   reactivity
and to the depression-like phenotype outlined above.
These two reviews  the rst by Pandey in 2003, the second
by Nestler andCarlezonin2006  present perspectives onCREB
that are thought-provokingly divergent, yet mutually compa-
tible.   For   immediate   purposes,   we   can   note   that   affect
regulation  is  a  non-linear  dynamic  balance,   and  that  anhe-
donia is probably just as conducive to addictive behavior as is
anxiety.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 286
2.2.9.   Brain-derived  neurotrophic factor  (BDNF)
Brain-derived neurotrophic factor (BDNF) is a member of the
neurotrophin  family.   It  is  highly  expressed  in  various  brain
regions   and   is   involved   in   neuronal   survival,   functional
differentiation,  and  synaptic  strength  [956961].   Its  action  is
mediated by binding to tyrosine kinase B (TrkB), a high-afnity
receptor that is localized on cell bodies and dendrites as well
as  postsynaptically  [962,963].   BDNF  is  involved  in  synaptic
plasticity and cellular processes of learning, such as long-term
potentiation   (LTP)   or   memory   consolidation   [956].   BDNF
application  facilitates   LTP,   while  reduction  of   BDNF  levels
attenuates   LTP   [467].   BDNF   is   synthesized   in   mesolimbic
dopaminergic  neurons  [964].   It  is  responsible  for  the  devel-
opmental appearance of the D
3
 receptor, which is selective for
the NAc, andfor maintaining expressionof the receptor during
adulthood [965,966]. BDNF has been strongly connected to the
effects of serotonergic agents in animal studies [967,968], and
augmentation  of   serotonergic  activity  within  various  brain
areas   after   infusion  of   BDNF   into   the   midbrain  has   been
reported [969].
Infusion  of   BDNF  into  the  VTA  promotes  the  behavioral
actions  of  drugs  of  abuse  [540].   When  administered  directly
into the VTAor NAc, it causes a signicant increase incocaine-
induced  locomotor  activity  and  in  cocaine  reward  [970972].
Meanwhile, a deciency inBDNF has beenreportedto promote
ethanol intake in mice [973,974]. Application of any of several
abusable   drugs      including   amphetamine   [975],   morphine
[976],   cocaine   [977],   D9-tetrahydrocannabinol   [978],   and
nicotine  [979]     was  found  to  up-regulate  the  expression  of
BDNF.   Intermittent,   exogenous   application  of   BDNF   either
centrally or into the VTA was reported to decrease food intake
and body weight and to increase behavioral activity [980983],
a  set  of  effects  that  is  reminiscent  of  the  psychostimulants.
Patients   with  eating   disorders   were   reported  to  have   low
serum levels of BDNF, which rose to the normal range when
the  patients  were  treated  with  antidepressant   medications
[984].
Pandey  et   al.   [985]   reported  nding  that   CREB-decient
mice   that   had  been  displaying   anxiety-like   behaviors   and
increased   ethanol   consumption   turned   out   also   to   have
decient   expression   of   BDNF   in   several   brain   regions,
including  the  amygdala  [986].   These  results  suggested  that
the effects of decreased CREB levels on anxiety-like behaviors
and  ethanol   consumption  may  have  been  mediated  by  the
reduction  of   BDNF  levels  in  amygdaloid  structures.   Pandey
et al. proposed that a deciency of BDNF in the CeA and MeA
(medial amygdala) results in decreased CREB phosphorylation
inthe CeAand MeAand inlowlevels of NPYinthe CeA, bothof
which promote anxiety-like behaviors and ethanol consump-
tion.   A  similar  association  between  low  BDNF  level   and  the
combination of anxiety-like behaviors and increased ethanol
consumption was reported by Yan et al. [987] to occur in P rats,
though  the  low  BDNF  levels  (as  compared  to  NP  rats)   were
observed in the NAc, not the amygdala.
Most of the clinically relevant researchabout BDNF that has
been published concerns the relationship between BDNF and
depression.   Serum  levels   of   BDNF   in  patients   with  major
depressive   disorder   prior   to   initiation   of   antidepressant
treatment were found to be lower than were those of healthy
subjects [988992], and moreover were found to be negatively
correlated with the severity of depression [988,990,992]. After
several   weeks  of  treatment  with  antidepressant  medication
(presumably   effective),   patients   BDNF   levels   increased   to
levels similar to those of the control subjects [991,993995]. In
studies  that   used  animal   models  of   depression,   depressive
states were shown to be associated with reduced BDNF levels
in the brain, and central administration of BDNF was shownto
reverse such depressive states [9961000].
What  about  the  relationship  between  BDNF  and  depres-
sion?   One  hypothesis   would  be  that   reduced  BDNF  might
reect a genetic vulnerability to develop depression. Another
possible   explanation   would   be   that   stress-induced   BDNF
reductions  might   cause  neuronal   damage,   which  would  in
turn lead to acquired biological vulnerability. Stress decreases
levels of BDNF expression in the dentate gyrus and pyramidal
cell layer of hippocampus [999,1001]. This reductionappears to
be   mediated  partly   via   stress-induced  glucocorticoids   and
partly via other processes, such as stress-induced increases in
serotonergic activity [9991003]. Stress, which can precipitate
and   exacerbate   depression,   causes   neuronal   atrophy   and
death, especially in the hippocampus [10041008].
From the viewpoint of stress-induced BDNF reduction, low
BDNF   levels   in   our   antidepressant-na ve   patients   with
depression  may  reect  the  collapse  of  the  stress-adaptation
systemand its failure to protect the brain fromstress-induced
neuronal   degeneration   [990].   Addictive   behavior   can   be
understood  as  the  last  resort  of  a  stress-adaptation  system
that is failing to maintain allostasis.
2.2.10.   Neuropeptide Y (NPY)
Inrecent years, NPYhas emergedas asignicant factor inaffect
regulation. In rodent models, central NPY is released following
stress  and  attenuates  the  behavioral   consequences  of  stress
[10091012]. WhileNPYknockout micewere foundtodisplayan
anxiety-like  phenotype  [684,1013],  transgenic  rats  with  selec-
tive NPY overexpression were shown to be resistant to stress-
induced increases in anxiety-like behavior [1014,1015]. The Y1
subtype of NPY receptor has been most strongly implicated in
mediating   anxiolytic   behaviors   [10161021].   Meanwhile,   Y2
receptors seem to be anxiogenic [10221026], with the possible
exception of Y2 receptors  in the  locus  coeruleus, which  were
reported to be involved in mediating decreases in anxiety-like
behavior [1008].
The anxiolytic effects of NPY appear to be mediated by Y1
receptors in the amygdala, particularly in its central nucleus
[670,1025].   The  CeA  receives  NPYergic  innervation  from  the
nucleus   of   the   solitary   tract,   arcuate   nucleus,   and  lateral
septum  [1009,1027].  NPY  neurons  in  the  amygdala  project  to
the  BNST  [1028],   which  also  contains  NPY  Y1  receptors  and
NPY Y1 and Y2 receptor mRNA[10291031]. The BNST projects
to   the   dorsal   vagal   complex   and  consequently   may   have
effects on the autonomic nervous system [1028,1032].
The Y2 receptor is believedto be a presynaptic autoreceptor
that limits the transmission of NPY [1033,1034]. Therefore, Y2
agonists could produce an anxiogenic-like effect by inhibiting
NPY  release.  NPY  is  found  within  GABA  interneurons  in  the
basolateral  nucleus  of  the  amygdala  (BLA) [1035],  and  also  is
colocalized with GABA in the suprachiasmatic nucleus. In the
latter region, NPY can decrease the inhibitory effects of tonic
GABA release via presynaptic Y2 receptors [10361038]. When
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   287
the inhibitory actions of GABA are reduced, both inhibition of
and opposition  to stimulatory  glutamatergic function dimin-
ish, and neuronal activity increases signicantly. Activation of
Y2 receptors in the BLAcould suppress the release of both NPY
and   GABA,   thereby   producing   an   excitatory   state   in   the
nucleus that results inthe expression of anxiety-like behavior.
A number of studies have reported that ethanol-preferring
P   rats   have   lower   levels   of   NPY   in   the   CeA   than   do
nonpreferring  NP  rats  [10391041],  and  that  P  rats  also  have
been  found  to  display  more  anxiety-like  behavior  and  to  be
more sensitive to the anxiolytic effects of ethanol [944]. Some
investigators have taken the next step and hypothesized that
the   higher   consumption   of   ethanol   by   P   rats   could   be
motivated  by  higher anxiety  levels  [10411043].  Pandey et  al.
[1041]  reported  that  both  anxiety  and  ethanol  drinking  were
reduced  in P  rats  when NPY  activity in  the central or  medial
amygdala was increased, either directly by infusion of NPY, or
indirectly by increasing CREBfunction. They reportedalso that
anxiety and ethanol drinking were increased in NP rats when
NPY activity was reduced by decreasing CREB function in the
same brain area [1041]. Primeaux et al. [1044] found that rats
that  had  been  identied  as  anxious   on  the  basis  of   their
performance   in   an   elevated   plus   maze   consumed   more
ethanol   solution  than  did  the  nonanxious   rats.   They  then
found that treatment of anxious rats  with a viral vector that
mediated  an  increase  in  CeA  NPY  decreased  their   ethanol
preference more than did treatment of other anxious rats with
an  antisense  NPY  vector.   Combined  with  previous  research
ndings that virally mediated increases in CeA NPY decreased
anxiety-related behaviors [1045], these data canbe understood
to suggest that treatment that increased CeA NPY activity in
anxious rats led to a reduction in their anxiety, and thus to a
reduction in their preference for ethanol. The results of these
studies support Valdez and Koobs [1046] revisionist tension
reduction  hypothesis   that   consumption  of   ethanol   (espe-
cially to the point that it becomes self-damaging) subserves a
motivation to alleviate negative affect and stress.
2.2.11.   Galanin
In rodent models, expression of galanin in the brain is altered
by   various   stressors,   while   administration  of   galanin  can
modulate  anxiety-like  responses  to  stress.  A  recent  study  of
the central  amygdala  showed that,  while mild stress did not
alter galanin levels, a model of high stress did increase galanin
release [1047]. Other studies of centrally administered galanin
in  rats  [1048]  and  of  galanin  overexpressing  transgenic  mice
[1049,1050]   appear   to   support   the   view  that   galanin  may
modulate behavioral responses to signicant stress (i.e., high
levels of noradrenergic activationinthe central amygdala), but
may  remain  dormant  under  conditions  of  mild  stress  [1051].
Emerging  evidence  further  supports  a  role  for  galanin  in  the
mediation of depression-related behaviors in rodents [1052].
Stress can evoke a variety of potential modulatory interac-
tions involving NE andgalanininthe CeAand BNST, depending
on  the  nature  of  the  stressor  and  the  response  elicited,   the
subset  of  noradrenergic neurons  activated,  and  the degree to
whichthesesystems are activated. Dysregulationof the normal
interaction  between  NE  and  galanin  may  contribute  to  the
development of stress-related behavioral disorders, including,
for   example,   stress-induced  reinstatement   of   drug-seeking
behavior, aprocessthat hasbeenassociatedwithnoradrenergic
mechanisms   in   the   CeA   and   BNST   [488]   More   generally,
dysregulation of the interaction between NE and galanin may
be involved in stress-related neuropsychiatric illnesses such as
depression, PTSD, or other anxiety disorders.
Ethanol intake increases galanin mRNA expression in the
rat   hypothalamus  [1053],   whereas  galanin  injected  into  the
third  ventricle  or  the  hypothalamus  increases  ethanol   con-
sumption  in  rats  that   have  learned  to  consume  ethanol   at
moderate  levels  [1054,781].  This  suggests  the  possibility  of  a
positive  feedback  loop  between  galanin  and  ethanol   intake
[1055].
A  signicant  association  between galanin  haplotypes  and
alcoholism has been demonstrated in both Finnish Caucasian
and Plains American Indian men [1056]. In both populations,
the  two  haplotypes  A  and  B,  differing  by  only  one  allele  and
therefore  originating  from  a  common  ancestral   haplotype,
were risk factors for alcoholism. The other two haplotypes, C
and D, also differing by only one allele and also derived from
a  shared  ancestral   haplotype,   were  protective  against   alco-
holism.   These  ndings   from  two  independent   populations
suggest   that   galanin   may   contribute   to   vulnerability   to
alcoholism, perhaps mediated by dimensional anxiety.
2.2.12.   Substance  P (SP)
Anatomical and functional studies suggest that SP is a central
stress neurotransmitter. SP and its preferred tachykinin NK-1
receptor  are  expressed  throughout  the  fear-processing  path-
ways   of   the   brain,   including   the   amygdala,   hippocampus,
hypothalamus  and  periaqueductal   grey  [10571059].   Central
injection   of   SP   agonists   produces   a   range   of   defensive
behavioral and cardiovascular reactions in animals, including
conditioned place aversion [1060].
Observations  from  basic  research  had  suggested  that  SP
might   be  involved  in  the  etiology  of   affective  and  anxiety
disorders [10611064]. Inanimal models of depression(chronic
mild  stress)   and  anxiety  (social   interaction  test),   the  NK-1
receptor  antagonist  NKP608  was  shown  to  exert  antidepres-
sant and anxiolytic activity [1061,1064]. A randomized double-
blind placebo-controlled study was conducted to evaluate the
safety  and  efcacy  of  the  NK-1  receptor  antagonist  MK-869,
and the results were encouraging [1065]. Rupniak and Kramer
[1061] suggested that SP might be the neurobiological correlate
of  the  subjective  experience  that  has  been  called  emotional
pain  a state in which the type of affect caused by trauma is
expressed,   but   devoid   of   the   sensation   of   pain.   They
speculated  that  autonomous  hyperactivity  in  SP  neurotrans-
mission might contribute to the anxiety, fear, and emotional
pain that accompany affective and anxiety disorders.
2.2.13.   Dynorphin
Shirayama et al. [1066] reported that levels of dynorphin A and
dynorphin B immunoreactivity in rats hippocampus and NAc
increased when the rats were exposed to learned helplessness
(LH)   and  immobilization  stress,   and  that  exposure  to  forced
swim stress increased dynorphin A levels in the hippocampus.
Additionally,   they   found   that   infusions   of   the   k-opioid
antagonist nor-binaltorphimine dihydrochloride into the den-
tate gyrus or CA3 regions of the hippocampus and into the shell
or core regions of the NAc produced antidepressant-like effects
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 288
intheLHparadigm. Earlier studies hadreportedthat infusionof
an antagonist of the k-opioid receptor, the primary receptor for
dynorphin, producedanantidepressant effect intwobehavioral
models of depression, the forced swim test [649,1067] and the
learned helplessness paradigm [955].
Dynorphin  is   co-localized  with  glutamate,   the   primary
neurotransmitter   in  granule   cells,   and  synaptic   release   of
dynorphin has been reported to cause pre-synaptic inhibition
of   glutamate   release   from  the   mossy   ber   and   perforant
pathway terminals [1068].
Shirayama  et  al.  [997]  found  that  microinfusions  of  BDNF
into the hippocampus produced an effect similar to blockade
of k-opioid receptors. Previous studies had provided evidence
of   a  link  between  BDNF  and  dynorphin.   Infusions  of   BDNF
were  reported to  decrease  levels  of  dynorphin  [1069],  raising
the possibility that the actions of BDNF could be accounted for
by down-regulation of dynorphin. This also is consistent with
reports   that   stress   decreases   BDNF,   which  could  result   in
increased dynorphin,  and  that  antidepressant  treatment  up-
regulates  the  expression  of   this  neurotrophic  factor   in  the
hippocampus [996,999; reviewed in 1070].
GABAergic  projection  neurons  in  the  NAc  receive  inputs
from  VTA  DA  neurons  that   express  dynorphin.   Dynorphin
serves a negative feedback process by acting on presynaptic k-
opioid receptors to inhibit DA neuronal function.
2.2.14.   DFosB
The expression of DFosB can be induced by either chronic drug
exposure  or  chronic  stress.  Chronic  drug  exposure  has  been
reported to induce DFosB expression primarily in the NAc and
dorsal striatum, with lower levels of induction observed in the
frontal   cortex  and  amygdala  [654,10711075].   Chronic  stress
has been reported to induce DFosB expression predominantly
in the frontal cortex, NAc, and amygdala. Perrotti et al. [1075]
suggested that stress induction of DFosB within dynorphin + -
NAc and dorsal striatal neurons would increase the drive for
drugs,   and  could  thereby  mediate  in  part   the  tendency  of
stress to increase vulnerability for drug addiction and relapse.
2.2.15.   GABA
Ethanol has been found to produce anti-conict (anti-anxiety)
actions in the social interaction test, elevated plus maze, and
in operant procedures [1076]. These anti-anxiety effects were
shown to be blocked by administration of the GABA
A
 receptor
antagonist picrotoxin [1077] and by isopropylcyclophosphate,
a   compound   that   binds   near   or   at   the   GABA
A
  chloride
ionophore   [1078].   Low   doses   of   benzodiazepine   inverse
agonists  also  were  found  to  block  the  anti-anxiety  effects  of
ethanol,   but  to  have  anxiogenic-like  effects  on  their  own  at
these   doses   [1079,1080].   Administration  of   picrotoxin  was
reported   to   decrease   ethanol   intake   [1081],   as   was   pre-
treatment   with  the   picrotoxin  ligand  isopropylbicyclopho-
sphate or with the benzodiazepine inverse agonist RO 15-4513
[552]. GABA
A
 receptors seem to mediate a critical component
of ethanols anti-anxiety effect, and to affect ethanol intake in
ways  that  suggest  the  signicant  contribution  that  negative
reinforcement (alleviation of anxiety) makes to the motivation
to consume ethanol.
A study that was reported by Castelli et al. [1082] demon-
strated  that  GABA
B
  receptors  in  limbic  areas  and  to  a  lesser
extent in the cortex functioned about half as well in ethanol-
na ve ethanol-preferring sP  rats  as they  did in ethanol-na ve
ethanol-nonpreferring  sNP  rats.   The  sP  rats  required  more
than twice the concentration of the GABA
B
agonist baclofen as
did  the  sNP  rats  to  achieve  the  same  result.  The  diminished
responsivity   of   the   GABA
B
  receptors   was   attributed   to
genetically  determined  differences   in  G-protein  activation.
Previous  studies  were  reported  to  have  found  that  ethanol-
na ve  sP  rats   displayed  a  higher   degree  of   anxiety-related
behaviors than did ethanol-na ve sNP rats [945,1083,1084], and
that   GABA
B
  receptors  are  involved  in  the  neural   substrate
mediating   anxiety-related   behaviors   [1085].   The   authors
proposed  that  the  lower  GABA
B
  receptor  function  in  sP  than
in sNP rats that they observed could have contributed to the
development of the higher degree of anxiety-related behaviors
that  sP  rats  had  been  found  to  display.   They  suggested  that
baclofen-induced suppression of ethanol-drinking behavior in
sP  rats  might  have  been  secondary  to  the  substitution  of  its
anxiolytic  effect   for   that   of   voluntarily  consumed  ethanol.
Their   hypothesis   seems   to  be  that   genetically  determined
alterations of their GABA
B
 receptors left sP rats susceptible to
higher  degrees  of  anxiety  (chronic  stress),   the  alleviation  of
whichprovided themwith substantial negative reinforcement
for   consuming  ethanol.   The  rats   predilection  for   imbibing
thus resulted not from their preference for ethanol, as their sP
designation  implies,   but   from  their  preference  for  freedom
from  anxiety.   This   hypothetical   vignette   exemplies   the
impaired affect regulation aspect of the addictive process.
2.3.   Impaired behavioral inhibition
Impulsivity   has   increasingly   come   to   be   understood   as   a
heterogeneous  phenomenon  that   includes  impaired  inhibi-
tory  control  of  behavior,  intolerance  to  delay  of  reward,  and
premature decision-making [1086,1087]. However, these three
forms   of   impulsivity   are   not   on   the   same   level,   neither
theoretically  nor   practically.   Theoretically,   impaired  inhibi-
tory  control  of  behavior  is  the  nal  common  pathway  of  all
forms of impulsivity. If an organism can refrain from acting in
response  to  an  instance  of  delay  intolerance  or  a  premature
decision,   impulsive   behavior   does   not   occur.   Practically,
unless we are dealing with organisms that can communicate
their   subjective  experiences   of   delay-intolerance,   decision-
making,   and  intent  to  inhibit  behavior,   we  must  infer  these
processes   from  behavior   that   we   observe.   Compared   to
intolerance   to   delay   of   reward   and   premature   decision-
making,   impaired   inhibitory   control   of   behavior   remains
closer  to  the  observational   data  and  depends  on  a  simpler,
more direct form of inference. These considerations informed
the identicationof this component of the addictive process as
impaired behavioral inhibition.
2.3.1.   Serotonin  (5-HT)
Serotonin (5-HT) has come to be recognized as being the main
player   in  behavioral   inhibition  [1088].   Studies   have   deter-
mined  that   low  levels  of   cerebrospinal   uid  (CSF)   5-hydro-
xylindolacetic  acid (5-HIAA), a major metabolite of 5-HT and
an  indicator  of   5-HT  activity,   are  associated  primarily  with
impulsivity   and   to   a   lesser   but   still   signicant   extent
with   aggression,   depression,   and   early-onset   alcoholism
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   289
[1089,1090;   reviewed  in  1091,   1092].   An  association  between
low  5-HT  activity  and  impulsive  behavior  has  been  demon-
strated also by prolactin release after fenuramine challenge
[1093]. Low levels of 5-HIAA were found in the CSF of bulimic
subjects [1094], and a preclinical study found decreased levels
of 5-HT and 5-HIAAin the brains of rats that tended to acquire
self-administration of amphetamine [1095].
The  5-HT receptors  that have been  most often associated
withaddictive disorders are 5-HT
1A
, 5-HT
1B
, and5-HT
2A
. The 5-
HT
2C
  receptor   is   the   only   receptor   that   when  stimulated
inhibits many addiction-related behaviors [468]. Interestingly,
most  of  the  recent research  on  5-HT  receptors  and  impaired
behavioral   inhibition  has   focused   on  bulimia   (other   than
genetic  and  developmental  studies,  which  are  reviewed  in  a
separate publication).
PET studies have demonstrated increased 5-HT
1A
 receptor
binding inseveral cortical areas of patients withactive bulimia
[1096],   and  in  subjects  who  had  recovered  from  bulimia  or
from  bulimic-type   anorexia   [1097,1098].   A   wide   cortical
distribution of increased 5-HT
1A
receptor binding might reect
a   diffuse   dysregulation   of   5-HT   activity   that   could   be
associated   with   impaired   impulse   control   [1096,1099].
Increased  receptor  binding  (i.e.,   binding  of  labeled  ligand  to
receptor)   can  indicate  either   increased  receptor   density  or
decreased   intrasynaptic   neurotransmitter/neuromodulator
concentration.   In  the  absence  of  evidence  that  a  functional
5-HT
1A
  receptor   gene   polymorphism  is   associated   with
bulimia [1096], increased 5-HT
1A
 receptor binding most likely
reects low levels of intrasynaptic 5-HT.
A study with mice revealed a specic association between
the  5-HT
1B
  receptor  and  impulsivity  [1100].  5-HT
1B
  receptors
are expressed on the terminals of GABAergic striatal neurons
that   project   to   the   substantia   nigra   and   VTA  [1101,1102].
Activation of these receptors may inhibit the release of GABA
onto  dopaminergic  neurons,   thereby  disinhibiting  the  dopa-
minergic neurons [1103].
5-HT
2A
  receptor  binding  was  found  to  be  reduced  in  the
orbitofrontal   cortex   of   subjects   who   had   recovered   from
bulimia   [1104],   and   in  the   cingulate,   parietal,   and   mesial
temporal cortices of subjects who hadrecoveredfrombulimic-
type anorexia [1105]. Decreased 5-HT activity at orbitofrontal,
cingulate, parietal, or mesial temporal 5-HT
2A
 receptors could
reect impulsiveness and altered emotional processing [1106].
5-HTtransporter (5-HTT) binding was found to be decreased
in the midbrain of obese binge-eating women [1107]. A follow-
up  of   treated  patients   whose  binge-eating  disorder   was   in
remission  showed  signicantly  increased  5-HTT  binding,   as
compared to unchanged results in controls [1108].
While decreased 5-HTT binding in association with active
binge-eating disorder that increases when the disorder remits
indicates a transitory state condition, reduced 5-HT activity
(or   increased  5-HT  receptor   binding)   in  bulimic  syndromes
that  persists  long  after  the  disorder  has  gone  into  remission
implies   a   chronic   trait   condition.   The   latter   is   likely   to
represent a primary vulnerability that arose independently of
the   eating   disorder   and   contributed   to   its   pathogenesis
[1104,1105,1109].   Collateral   information  about  the  5-HT  sys-
tem  suggests   that   this   vulnerability   relates   not   to   food
ingestion  or   gustatory-gastric   sensations,   but   to   impaired
behavioral inhibition or impulsivity. Thus it is not specic to
bulimia,   but   could   contribute   to   the   development   of   any
addictive disorder.
2.3.2.   Dopamine (DA)
Jentsch  and  Taylor   [1110]   hypothesized  that   the  inhibitory
modulation of reward-seeking behavior may depend critically
upon  the  corticostriatal   projections  from  the  medial   frontal
cortex  to  the  caudate  nucleus  and  NAc.  Studies  had  demon-
strated that lesions to the frontal cortex [1111] or DA depletion
within   the   PFC   [1112,1113]   can   augment   the   locomotive
and  reinforcing   effects   of   psychostimulants.   A  hypothesis
emerged, according to which the cortical DA system tonically
inhibits  subcortical   DA  systems  [1114].   Under  such  circum-
stances,   dopaminergic   hypofunction   in   the   frontal   cortex
can  result   in  the   disinhibition  of   mesolimbic   DA  systems
[1115,1116; review1117]. Thus, a loss of DAfunction in the PFC
can  result   in  an  increased  vulnerability  to  self-administer
psychostimulant drugs [1118,1119] or to engage addictively in
another  behavior  that  activates  the  mesolimbic  DA  system.
This  hypothesis  is  supported  by  the  preclinical   nding  that
animals that  were more  vulnerable  to  acquiring  intravenous
drug self-administration showed reduced dopaminergic activ-
ity in the PFC [1117].
Amphetamine   has   been   shown   to   increase   premature
responding in the 5-choice serial reaction time task (5-CSRTT),
which  is  generally  considered  to  represent  behavioral  disin-
hibition[11201122]. This premature responding was shownto
be   attenuated  by   DA  depletion  of   the   NAc   [1121]   and  by
systemic   administration  of   the   non-selective   DA  receptor
antagonist   a-upentixol   [1120].   Van   Gaalen   et   al.   [1087]
investigated  the  effects  of   a  number  of   substances  on  rats
that   had   been   well-trained   in   the   5-CSRTT.   Premature
responding   was   found   to   be   increased   by   amphetamine,
cocaine, nicotine, and the DA reuptake inhibitor GBR 12909. It
was   found   to   be   decreased   by   the   NE   reuptake   inhibitor
desipramine and by the DA D
1
 receptor antagonist SCH 23390,
but  not  by  the  DA  D
2
  receptor  antagonist  eticlopride.   Mean-
while, the increments in premature responding that had been
evoked by amphetamine, cocaine, and nicotine were found to
be attenuated by eticlopride, whereas SCH 23390 reduced the
drug-induced  behavioral  disinhibition  only  at  a  dose  that  by
itself   decreased   premature   responding.   The   authors   con-
cluded  that  premature  responding  in  general  is  regulated  by
DA D
1
, while behavioral disinhibition that is induced by drugs
of   abuse  depends  on  activation  of   DA  D
2
  receptors.   Super-
sensitivity   of   either   D
1
  or   D
2
  receptors   could  conceivably
result,  directly  or  indirectly  (respectively),  in  impaired  beha-
vioral inhibitionand thus could contribute to the development
of an addictive process.
An  elegant   study   by   Dalley   et   al.   [1123]   demonstrated
relationships between trait impulsivity, reduced D
2
/D
3
 recep-
tor  availability,  and  tendency  to  escalate  self-administration
of   cocaine.   Rats   performance  on  the  5-CSRTT  was  used  to
identify  a  group  of  high-impulsive  rats  and  a  group  of  non-
impulsive  rats.   Positron  emission  tomography  then  demon-
strated   that   D
2
/D
3
4
receptor   availability   was   signicantly
4
Dalley  et   al   (2007)   used  the  high-afnity  DA  D
2
/D
3
  receptor
radiotracer  [
18
F]   fallypride,   which  does  not  distinguish  between
D
2
 and D
3
 receptors.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 290
reduced  in  the  ventral   striatum  but   not   in  the  dorsolateral
striatumof high-impulsive rats as compared to non-impulsive
rats,   and   that   D
2
/D
3
  receptor   availability   in   the   ventral
striatum  was   inversely   correlated  with  impulsivity.   When
the   heretofore   cocaine-nave   rats   were   trained   to   self-
administer   cocaine,   rats  that   exhibited  trait   impulsivity  on
the   5-CSRTT  showed  a   greater   tendency  for   escalation  of
intravenous  cocaine  self-administration  than  did  their  non-
impulsive   counterparts.   The   authors   concluded   that   their
ndings demonstrated that trait impulsivity predicts cocaine
reinforcement  and  that  decreased  D
2
  receptor  availability  in
the striatum may be a predisposing neurobiological trait and
not   only  a  consequence  of   chronic  cocaine  exposure.   This
study echoes and afrms the ndings related to D
2
 receptors
that  we  reviewed  in  the  Motivation-Reward  section.  Further
investigation   of   the   relationships   between   D
2
  receptors,
motivation-reward, behavioral inhibition, and addictive beha-
vior patterns is likely to be both fascinating and productive.
Research on the relationship between addiction and DA D
4
and  D
5
  receptors  has  been  concerned  primarily  with  genetic
factors. A variable number tandem repeat (VNTR) polymorph-
ism located in the third exon of the DA D
4
 receptor gene has
been found to be associated with impulsive personality traits
[1124]   and  to  be  a  risk  factor  for  adolescent   ethanol   abuse
[1125],   adolescent  hard  drug  use  [1126],   heroin  abuse  [1127],
cue-elicited  heroin  craving   [1128],   severity   of   ethanol   and
opiate  addiction  [1129],   pathological   gambling  [1124],   binge-
eating [1130], and cue-elicited craving for food [1131]. On the
other  hand,  a  few  studies  found  no  or  negligible  association
between   D
4
  receptor   gene   exon   III   polymorphism   and
alcoholism  [1132,1133]   or  heroin  addiction  [1134].   The  com-
mon 148 bp allele of a microsatellite polymorphism at the D
5
receptor gene was foundto be correlatedwithsubstance abuse
and novelty seeking in females [220]. It also  was found to be
associated   with   attention-decit/hyperactivity   disorder
[1135,1136], which is only weakly suggestive of a relationship
to  addiction  but  indicates  a  potentially  fruitful  direction  for
further research.
3.   Conclusion
The  present  article  represents  the  owering  of  an  idea  that
was planted in 1990:
A  hypothesis  may  be  submitted,  the  gist  of  which  is  that
similar   patterns   in   behavioral   manifestations   of   the
various  addictive  disorders. . .reect   similarities  in  some
set of personality and/or biological variables, which may or
may not be measurable by instruments currently available.
In   other   words,   addictive   disorders   would   be   most
accurately  described,   not   as  a  variety  of   addictions,   but
as   a  basic   underlying  addictive  process,   which  may  be
expressed in one or more of various behavioral manifesta-
tions. [1]
At the time that the preceding sentences were published,
addiction neuroscience was young, and much of the research
that could evaluate the hypothesis of an underlying addictive
process had not yet been conducted. In the intervening years,
addiction neuroscience has advanced so considerably that the
hypothesis   is   no   longer   radical.   Perhaps   the   continuing
trajectory  of  scientic  progress  soon  will  render  it  no  longer
necessary.
r e f e r e n c e s
[1]   Goodman A. Addiction: denition and implications. Br J
Addict 1990;85:14038.
[2]   Hesselbrock MN, Meyer RE, Keener JJ. Psychopathology in
hospitalized alcoholics.  Arch  Gen Psychiatry
1985;42:10505.
[3]   Ross  HE, Glaser FB, Germanson T. The prevalence of
psychiatric disorders in patients  with alcohol and other
drug problems. Arch Gen Psychiatry  1988;45:102331.
[4]   Regier DA, Farmer ME, Rae DS, Locke BZ, Keith SJ, Judd LL,
et al. Comorbidity of mental disorders with alcohol and
other drug abuse: results from the epidemiological
catchement area (ECA) study. J Am Med Assoc
1990;264:25118.
[5]   Kosten  TR, Kosten TA, Rounsaville  BJ. Alcoholism and
depressive disorders in opioid addicts and their family
members. Compr Psychiatry 1991;32:5217.
[6]   Kandel D, Yamaguchi K. From beer to crack:
developmental patterns of drug involvement. Am J Public
Health 1993;83:8515.
[7]   Kendler KS, Prescott  CA. Cannabis use, abuse, and
dependence in a population-based  sample of female
twins.  Am J Psychiatry  1996;155:101622.
[8]   Degenhardt L, Hall W, Lynskey M. Alcohol, cannabis and
tobacco use among  Australians: a comparison  of their
associations with  other drug use and use disorders,
affective and anxiety disorders, and psychosis. Addiction
2001;96:160314.
[9]   Wagner FA, Anthony JC. Into the world of illegal drug use:
exposure opportunity and other mechanisms linking the
use of alcohol, tobacco, marijuana, and cocaine. Am J
Epidemiol 2002;155:91825.
[10]   Mitchell  JE, Goff G. Bulimia in male patients.  Psychosom
1984;25:90913.
[11]   Jones  DA, Cheshire N, Moorhouse H. Anorexia nervosa,
bulimia, and alcoholism: Association of eating disorder
and alcohol. J Psychiatry  Res 1985;19:37780.
[12]   Mitchell  JE, Hatsukami D, Eckert ED, Pyle RL.
Characteristics of 275 patients  with bulimia. Am J
Psychiatry 1985;142:4825.
[13]   Beary MD, Lacey JH, Merry J. Alcoholism and eating
disorders in women  of fertile age. Br J Addict  1986;81:
6859.
[14]   Robinson PH, Holden NL. Bulimia nervosa  in the male: a
report of nine cases. Psychol Med 1986;16:795803.
[15]   Bulik CM. Drug and alcohol abuse by bulimic women and
their families. Am J Psychiatry  1987;144:16046.
[16]   Hudson JI, Pope HG, Jonas JM, Yurgelun-Todd D. A
controlled family history study of bulimia. Psychol  Med
1987;17:88390.
[17]   Hudson JI, Pope HG, Yurgelun-Todd  D, Jonas JM,
Frankenburg FR. A controlled study of lifetime prevalence
of affective  and other psychiatric disorders in bulimic
outpatients. Am J Psychiatry 1987;144:12837.
[18]   Johnson  C, Connors ME. The etiology and treatment  of
bulimia nervosa:  a biopsychosocial perspective. New
York: Basic Books;  1987.
[19]   Schneider JA, Agras WS. Bulimia in males:  a matched
comparison with  females. Int J Eat Disord  1987;6:23542.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   291
[20]   Powers PS, Coovert DL, Brightwell DR, Stevens BA. Other
psychiatric disorders among bulimic  patients.  Campr
Psychiatry 1988;29:5038.
[21]   Mitchell JE, Specker  SM, de Zwaan  M. Comorbidity and
medical complications of bulimia  nervosa.  J Clin
Psychiatry 1991;52(suppl lO):1320.
[22]   Lacey  JH. Self-damaging  and addictive  behaviour in
bulimia nervosa: a catchment area study. Br J Psychiatry
1993;163:1904.
[23]   Braun DL, Sunday SR, Halmi KA. Psychiatric co-morbidity
in patients  with eating  disorders. Psychol Med
1994;24:85967.
[24]   Fichter  MM, Quadieg N, Rief W. Course of multi-
impulsive  bulimia.  Psychol Med 1994;24:591604.
[25]   Holderness CC, Brooks Gunn J, Warren MP. Co-morbidity
of eating disorders and substance abuse review of the
literature. Int J Eating Disord 1994;16:134.
[26]   Epik A, Arikan Z, Boratav C, et al. Bulimia in a male
alcoholic: a symptom substitution  in alcoholism. J Eat
Disord  1995;17(2):2014.
[27]   Bulik  CM, Sullivan PF, Carter FA, Joyce PR. Lifetime
comorbidity  of alcohol dependence in women with
bulimia  nervosa. Addict Behav 1997;22:43746.
[28]   Lilenfeld LR, Kaye  WH, Greeno CG, et al. Psychiatric
disorders in women with bulimia nervosa and their rst-
degree relatives: effects of comorbid substance
dependence. Int J Eat Disord 1997;22:25364.
[29]   Nagata  T, Kawarada  Y, Kiriike N, Iketani  T. Multi-
impulsivity  of Japanese  patients with  eating disorders:
primary  and secondary impulsivity. Psychiatry  Res
2000;94:23950.
[30]   Wolfe WL, Maisto SA. The relationship  between eating
disorders and substance use: moving  beyond  co-
prevalence research. Clin Psychol Rev 2000;20:61731.
[31]   Anzengruber D, Klump KL, Thornton L, Brandt  H,
Crawford  S, Fichter  MM, et al. Smoking  in eating
disorders. Eat Behav 2006;7(4):2919.
[32]   Hudson JI, Hiripi E, Pope  HG, Kessler RC. The prevalence
and correlates  of eating disorders in the national
comorbidity  survey replication.  Biol Psychiatry
2007;61:34858.
[33]   Jonas J, Gold M. Cocaine abuse and eating disorders.
Lancet  1986;14:3901.
[34]   Jonas D, Gold MS, Sweeney  D, Pottash ALC. Eating
disorders and cocaine abuse: a survey of 259 cocaine
abusers. J Clin Psychiatry  1987;48:4750.
[35]   Peveler R, Fairburn C. Eating disorders in women who
abuse alcohol. Br J Addict  1990;85:16338.
[36]   Timmerman MG, Wells LA, Chen S. Bulimia nervosa and
associated alcohol abuse among secondary school
students. J Am Acad Child Adol Psychiatry 1990;29:11822.
[37]   Pyle RL, Mitchell JE, Eckert ED, Halvorson PA, Neuman PA,
Goff GM. The incidence  of bulimia in freshman college
students. Int J Eat Disord  1983;2:7585.
[38]   Haberman  PW. Drinking and other self-indulgences:
Complements or counter-attractions? Int J Addict
1969;4:15767.
[39]   Miller WR, Hedrick KE, Taylor CA. Addictive  behaviors
and life problems before and after behavioral  treatment
of problem drinkers. Addict Behav 1983;8:40312.
[40]   McCormick RA, Russo AM, Ramirez LF, Taber JI. Affective
disorders among  pathological  gamblers seeking
treatment. Am J Psychiatry  1984;141:2158.
[41]   Ramirez LF, McCormick RA, Russo AM, Taber JI. Patterns
of substance abuse in pathological gamblers seeking
treatment. Addict Behav 1984;8:4258.
[42]   Linden RD, Pope HG, Jonas JM. Pathological gambling and
major  affective disorders: preliminary  ndings. J Clin
Psychiatry 1986;47:2013.
[43]   Lesieur HR, Heineman M. Pathological gambling among
youthful  multiple substance abusers in a therapeutic
community. Br J Addict 1988;83:76571.
[44]   Roy A, Adinoff B, Roerich L, Lamparski D, Custer R,
Lorenz V, et al. Pathological gambling: a psychobiological
study. Arch  Gen Psychiatry 1988;45:36973.
[45]   Steinberg MA. Sexual addiction  and compulsive
gambling.  Am J Prevent Psychiatry Neurol 1990;2:
3941.
[46]   Lynch WJ, Maciejewski PK, Potenza  MN. Psychiatric
correlates  of gambling  in adolescents and young adults
grouped by age at gambling  onset. Arch Gen Psychiatry
2006;61:111622.
[47]   Petry NM, Stinson FS,  Grant BF. Comorbidity of DSM-IV
pathological gambling  and other psychiatric disorders:
results from the national  epidemiologic survey  on
alcohol and related conditions.  J Clin Psychiatry
2005;66:56474.
[48]   Gerstein DR, Volberg RA, Toce MT, et al. Gambling impact
and behavior study: report to the National  Gambling
Impact Study  Commission. Chicago: National  Opinion
Research Center; 1999.
[49]   Welte J, Barnes G, Wieczorek W, et al. Alcohol and
gambling  pathology  among  US adults: prevalence,
demographic patterns and comorbidity. J Stud Alcohol
2001;62:70612.
[50]   Bland RC, Newman SC, Orn H, et al. Epidemiology  of
pathological gambling  in Edmonton. Can J Psychiatry
1993;38:10812.
[51]   Cunningham-Williams RM, Cottler LB, Compton III WM,
Spitznagel EL. Taking  chances:  problem gamblers and
mental health  disorders: results from the St. Louis
Epidemiologic Catchment Area (ECA) Study.  Am J Public
Health 1998;88:10936.
[52]   Shaffer HJ, Hall MN, Vander Bilt J. Estimating  the
prevalence of disordered gambling behavior in the United
States and Canada: a research synthesis. Am J Public
Health 1999;89:136976.
[53]   Crockford  DN, el-Guebaly  N. Psychiatric comorbidity  in
pathological gambling: a critical review. Can J Psychiatry
1998;43:4350.
[54]   Black DW, Moyer T. Clinical  features and psychiatric
comorbidity  of subjects with pathological gambling
behavior. Psychiatr Serv 1998;49:14349.
[55]   Slutske  WS, Caspi A, Moftt TE, Poulton R. Personality
and problem gambling:  a prospective study of a birth
cohort of young adults. Arch  Gen Psychiatry 2005;62:
76975.
[56]   Vitaro F, Ferland F, Jacques C, Ladouceur R. Gambling,
substance abuse, and impulsivity  during adolescence.
Psych  Addict Behav 1998;12(3):18594.
[57]   Maccallum F, Blaszczynski A. Pathological  gambling  and
comorbid  substance use. Aust N Z J Psychiatry
2002;36:4115.
[58]   Lesieur HR, Blume SB, Zoppa PM. Alcoholism, drug abuse,
and gambling.  Alc Clin Exp  Res 1986;10:338.
[59]   Steinberg MA, Kosten  TA, Rounsaville BJ. Cocaine  abuse
and pathological gambling.  Am J Addict 1992;1:12132.
[60]   Daghestani  AN, Elenz E, Crayton JW. Pathological
gambling  in hospitalized substance abusing veterans.  J
Clin psychiatry 1996;57(8):3603.
[61]   Toneatto  T, Brennan J. Pathological gambling  in
treatment-seeking substance abusers. Addict Behav
2002;27(3):4659.
[62]   Petry NM. Substance  abuse, pathological  gambling,  and
impulsiveness. Drug Alcohol Depend 2001;63(1):
2938.
[63]   Hall GW, Carriero  NJ, Takushi  RY, Montoya ID, Preston
KL, Gorelick  DA. Pathological gambling among
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 292
cocaine-dependent  outpatients.  Am J Psychiatry
2000;157:112733.
[64]   Welte J, Barnes G, Wieczorek W, Tidwell MC, Parker J.
Alcohol and gambling  pathology  among US adults:
prevalence, demographic patterns and comorbidity. J
Stud Alcohol  2001;62:70612.
[65]   Spunt B, Lesieur H, Hunt D, Cahill L. Gambling  among
methadone patients.  Int J Addict 1995;30:92962.
[66]   Feigelman W, Wallisch LS, Lesieur HR. Problem gamblers,
problem substance users, and dual problem individuals:
an epidemiological study. Am J Public Health 1998;88:467
70.
[67]   Shaffer HJ, Korn DA. Gambling  and related  mental
disorders: a public health analysis. Annu Rev Public
Health 2002;23:171212.
[68]   Schneider JP, Schneider BH. Couple recovery from sexual
addiction/coaddiction: results of a survey  of 88
marriages. Sex Addict Compulsivity 1996;3:111226.
[69]   Kruesi MJP, Fine S, Valladares L, Phillips RA, Rapoport JL.
Paraphilias: a double-blind crossover comparison of
clomipramine versus desipramine. Arch Sex Behav
1992;21:58793.
[70]   Irons RR, Schneider JP. Sexual addiction: signicant
factor in sexual exploitation by health care professionals.
Sex Addict Compulsivity 1994;1:20814.
[71]   Kafka MP, Prentky R. Preliminary observations of DSM-
III-R axis I comorbidity  in men with  paraphilias and
paraphilia-related  disorders. J Clin Psychiatry
1994;55:4817.
[72]   Kafka MP, Hennen J. A DSM-IV Axis I comorbidity study
of males (n = 120) with paraphilias and paraphilia-related
disorders. Sex Abuse 2002;14:34966.
[73]   Black DW, Kehrberg LLD, Flumerfelt  DL, Schlosser  SS.
Characteristics  of 36 subjects reporting compulsive
sexual behavior. Am J Psychiatry 1997;154:2439.
[74]   Raymond  NC, Coleman E, Miner MH. Psychiatric
comorbidity  and compulsive/impulsive traits in
compulsive sexual behavior. Compr Psychiatry
2003;44:37080.
[75]   Carnes P. Dont  call it love: recovery from sexual
addiction. New York: Bantam; 1991.
[76]   Carnes PJ, Delmonico DL. Childhood abuse and multiple
addictions: research  ndings in a sample of self-
identied sexual addicts. Sexual  Addict Compulsivity
1996;3:25868.
[77]   Coleman E. Sexual compulsivity: denition, etiology and
treatment considerations.   In: Coleman  E, editor.
Chemical dependency  and intimacy  dysfunction. New
York: Haworth  Press; 1987.
[78]   Washton AM. Cocaine may trigger sexual compulsivity.
US J Drug Alcohol Depend 1989;13:8.
[79]   Lesieur HR. Report on pathological gambling in New
Jersey. In: Report and Recommendations of the
Governors Advisory Commission on Gambling. Trenton,
NJ, Governors Advisory  Commission on  Gambling,
1988:124.
[80]   Powell BJ, Penick EC,  Othmer E, Bingham SF,  Rice AS.
Prevalence of additional  psychiatric syndromes among
male alcoholics.  J Clin Psychiatry  1982;43:4047.
[81]   Weiss KJ, Rosenberg DJ. Prevalence of anxiety disorders
among alcoholics.  J Clin Psychiatry  1985;46:35.
[82]   Grant B, Harford T. Comorbidity between DSM-IV alcohol
use disorders and major depression: results of a national
survey. Drug Alcohol Depend 1995;39:197206.
[83]   Kessler RC, Crum RM, Warner LA, Nelson  CB,
Schulenberg J, Anthony JC. Lifetime co-occurrence  of
DSM-III-R alcohol abuse and dependence with other
psychiaric disorders in the National  Comorbidity Study.
Arch Gen Psychiatry  1997;54:31321.
[84]   Khantzian EJ, Treece  C. DSM-III  psychiatric diagnosis of
narcotic  addicts: recent  ndings. Arch Gen Psychiatry
1985;42:106771.
[85]   Merikangas KR, Mehta RL, Molnar BE, Walters EE,
Swendsen  JD, Aguilar-Gaziola  S, et al. Comorbidity of
substance use disorders with  mood and anxiety
disorders: results  of the International Consortium in
Psychiatric Epidemiology. Addict Behav 1998;23:
893907.
[86]   Couwenbergh C, van den Brink W, Zwart K, Vreugdenhil
C, van Wijngaarden-Cremers P, van der Gaag RJ.
Comorbid psychopathology in adolescents and young
adults treated for substance use disorders. Eur Child
Adolesc Psychiatry 2006;15:31928.
[87]   Escobedo LG, Reddy M, Giovino GA. The relationship
between depressive  symptoms and cigarette smoking in
US adolescents. Addiction 1998;93:43340.
[88]   Patton GC, Carlin JB, Coffey C, Wolfe R, Hibbert M, Bowes
G. Depression, anxiety, and smoking initiative: a
prospective study over 3 years. Am J Public Health
1998;88:151822.
[89]   Hudson JI, Pope HG, Jonas JM, Yurgelun-Todd D.
Phenomenologic relationship of eating disorders to major
affective disorder. Psychiatry  Res 1983;9:34554.
[90]   Perugi G, Toni  C, Passino MCS, Akiskal KK, Kaprinis S,
Akiskal  HS. Bulimia  nervosa in atypical  depression: The
mediating role of cyclothymic temperament. J Affect
Disord 2006;92(1):917.
[91]   Ramacciotti  CE, Paoli RA, Marcacci  G, Piccinni A,
Burgalassi A, DellOsso  L, et al. Relationship  between
bipolar illness and binge-eating disorders. Psychiatry Res
2005;135(2):16570.
[92]   Dell LJ, Ruzicka  MF, Palisi AT. Personality  and other
factors associated with gambling addiction. Int J Addict
1981;16:14956.
[93]   Hollander E, Buchalter  AJ, DeCaria CM. Pathological
gambling. Psychiatr Clin North Am 2000;22:62942.
[94]   Lynch Wj, Maciejewski PK, Potenza  MN. Psychiatric
correlates of gambling  in adolescents  and young adults
grouped by age at gambling  onset. Arch Gen Psychiatry
2006;61:111622.
[95]   Potenza MN, Xian It, Shah K, Scherrer JF, Eisen SA. Shared
genetic contributions to pathological gambling and major
depression in man. Arch Gen Psychiatry 2005;62:101521.
[96]   National Research Council. Pathological gambling:  a
critical  review. Washington,  DC: National Academy
Press; 1999.
[97]   Ibanez A, Blanco C, Donahue E, Lesieur HR, Perez dCI,
Fernandez-Piqueras J, et al. Psychiatric comorbidity  in
pathological gamblers seeking treatment.  Am J
Psychiatry 2001;158:17335.
[98]   Wise TN. Fetishism  and transvestism. In: Karasu TB,
editor.   Treatment of psychiatric disorders a task force of
the american psychiatric association V 1, vol. 1.
Washington,  DC: American Psychiatric Press; 1989.  p.
63346.
[99]   Chalkley AJ, Powell GE. The clinical  description of forty-
eight cases of sexual fetishism. Br J Psychiatry
1983;142:2925.
[100]   Fagan PJ, Wise TN, Schmidt CW, Ponticas Y, Marshall RD.
A comparison  of ve-factor personality  dimensions in
males  with sexual dysfunction and males with
paraphilia. J Pers Assess 1991;57:43448.
[101]   Kafka  MP, Prentky R. Fluoxetine  treatment  of
nonparaphilic sexual addictions and paraphilias in men.
J Clin Psychiatry 1992;53:3518.
[102]   Bulik  CM, Sullivan PF, Fear JL, Joyce  PR. Eating disorders
and antecedent  anxiety disorders: a controlled  study.
Acta Psychiatrica  Scandinavica 1997;96:1017.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   293
[103]   Kaye WH, Bulik CM, Thornton L, Barbarich N, Masters K.
Price foundation  collaborative group comorbidity of
anxiety disorders with anorexia and bulimia nervosa. Am
J Psychiatry 2004;161(12):221521.
[104]   Kafka MP. Successful antidepressant  treatment  of
nonparaphilic sexual addictions and paraphilias in men.
J Clin Psychiatry 1991;52:605.
[105]   Goodwin  DW, Schulsinger  F, Hermansen L, Guze S,
Winokur G. Alcoholism and the hyperactive child
syndrome. J Nerv Ment Dis 1975;160:34953.
[106]   Wood D, Wender PH, Reimherr FW. The prevalence of
attention decit disorder, residual type, or minimal brain
dysfunction, in a population of male alcoholic patients.
Am J Psychiatry  1983;140:958.
[107]   Carlton PL, Manowitz P. Behavioral  restraint  and
symptoms of attention decit  disorder in  alcoholics
and pathological gamblers. Neuropsychobiology
1992;25:448.
[108]   Eyre SL,  Rounsaville  BJ, Kleber HD. History of childhood
hyperactivity in a clinic population of opiate addicts. J
Nerv Ment Dis 1982;170:5229.
[109]   Kessler RC, Adler R, Barkley R, Biederman J, Conners CK,
Demler O, et al. The prevalence and correlates of adult
ADHD in the United  States: results from the National
Comorbidity Survey Replication. Am J Psychiatry
2006;163:71623.
[110]   Carlton PL, Manowitz P, McBride H, Nora R, Swartzburg
M, Goldstein L. Attention decit  disorder and
pathological gambling.  J Clin Psychiatry 1987;48:4878.
[111]   Specker  SM, Carlson GA, Christenson  GA, Marcotte M.
Impulse  control disorders and attention decit  disorder
in pathological gamblers. Ann Clin Psychiatry
1995;7(4):1759.
[112]   Kavoussi RJ, Kaplan M, Becker  JV. Psychiatric diagnoses
in adolescent sex offenders.  J Am Acad Child Adolesc
Psychiatry 1988;27:2413.
[113]   Hunter JA, Goodwin  DW. The clinical  utility of satiation
therapy  with juvenile offenders:  variations and efcacy.
Ann Sex Res 1992;5:7180.
[114]   Kafka MP, Prentky  RA. Attention-decit/hyperactivity
disorder in males with paraphilias and paraphilia-related
disorders. J Clin Psychiatry 1998;59:38896.
[115]   Koenigsberg  HW, Kaplan RD, Gilmore  MM, Cooper AM.
The relationship  between syndrome and personality  in
DSM-III: experience with 2462 patients. Am J Psychiatry
1985;142:20712.
[116]   Mezzich  JE, Fabrega H, Coffman  GA, Haley R. DSM-III
disorders in a large sample of psychiatric patients:
frequency and specicity of diagnoses. Am J Psychiatry
1989;146:2129.
[117]   DeJong AJ, van den  Brink W, Harteveld  FM, van der
Wielen EGM. Personality disorders in alcoholics and drug
addicts. Compr Psychiatry  1993;34:8794.
[118]   Nace  EP, Davis CW, Gaspari JP. Axis II comorbidity  in
substance abusers. Am J Psychiatry 1991;148:11820.
[119]   Gartner AF, Marcus RN, Halmi K, Loranger AW. DSM-III-R
personality disorders in patients  with eating  disorders.
Am J Psychiatry  1987;144:12837.
[120]   Herzog DB, Keller MB, Lavori PW, Kenny GM, Sacks NA.
The prevalence of personality  disorders in 210 women
with eating  disorders. J Clin Psychiatry  1992;53:
14752.
[121]   Slutske  WS, Eisen  RM, Xian H, True  WR, Lyons  MJ,
Goldberg J, et al. A twin study of the association between
pathological gambling  and antisocial personality
disorder, J. Abnorm Psychol 2001;110:297308.
[122]   Steel Z, Blaszczynski A. Impulsivity,  personality
disorders and pathological  gambling  severity. Addiction
1998;93:895905;
Blaszczynski AP, McConaghy  N, Frankova A. Crime,
antisocial personality, and pathological  gambling.  J
Gambling  Behav 1989;5:13752.
[123]   Fernandez-Montalvo J, Echeburua  E. Pathological
gambling and personality disorders: an exploratory study
with the IPDE. J Personal Disord  2004;18:5005 (also:
Bellaire,  W., Caspari, D., Diagnosis and therapy of male
gamblers in a university psychiatric hospital. J Gambl
Stud 1992;8:143150).
[124]   Blair CD, Lanyon RI. Exhibitionism:  etiology and
treatment.  Psychol Bull 1981;89:43963.
[125]   Schmidt CW, Meyer JK, Lucas J. Paraphilias  and
personality  disorders.   In: Lion JR, editor. Personality
disorders: diagnosis and management. 2nd ed.,
Baltimore:  Williams and Wilkins; 1981. p. 26995.
[126]   Irons RP, Schneider JP. Addictive sexual disorders.   In:
Miller N, editor. Principles and practice  of addictions in
psychiatry.  Philadelphia: Saunders; 1997. p. 44157.
[127]   Hudson JI, Laffer PS, Pope HG. Bulimia related to affective
disorder by family history and response to
dexamethasone suppression test. Am J Psychiatry
1982;139:6857.
[128]   Kassett JA, Gershon ES, Maxwell ME, Guroff  JJ, Kazuba
DM, Smith AL, et al. Psychiatric disorders in the rst-
degree relatives of probands with  bulimia nervosa. Am J
Psychiatry  1989;146:146871.
[129]   Jacobs DF. Children  of problem gamblers. J Gambling
Behav 1989;5:2618.
[130]   Shah KR, Potenza  MN, Eisen SA. Biological  basis for
pathological gambling.   In: Grant JE, Potenza MN, editors.
Pathological gambling:  a clinical  guide to treatment.
Washington,  DC: American Psychiatric  Publishing Inc.;
2004. p. 12742.
[131]   Gorelick DA. Effect of uoxetine on alcohol consumption
in male alcoholics. Alcohol Clin Exp Res 1986;10:13.
[132]   Gorelick  DA. Serotonin  uptake blockers and the
treatment  of alcoholism. Rec Dev Alcohol 1989;7:26781.
[133]   Naranjo CA,  Sellars EM, Lawrin MO. Modulation of
ethanol  intake by serotonin  uptake inhibitors. J Clin
Psychiatry  1986;47:1622.
[134]   Naranjo CA, Sellars EM, Sullivan JT, Woodley DV, Kadlec
K, Sykora K. The serotonin reuptake inhibitor citalopram
attenuates ethanol intake.  Clin Pharmacol  Ther
1987;41:26674.
[135]   Naranjo CA,  Sellars EM. Serotonin  uptake inhibitors
attenuate ethanol intake  in problem drinkers. Rec Dev
Alcohol 1989;7:25566.
[136]   Slywka S, Hart LL. Fluoxetine  in alcoholism. Ann
Pharmacother 1993;27:10667.
[137]   Janiri L, Gobbi G, Mannelli P, Pozzi G. Effects of uoxetine
and antidepressant  doses on short-term  outcome of
detoxied  alcoholics.  Int Clin Psychopharmacol
1996;11:10917.
[138]   Janiri L, Hadjichristos A, Buonanno A, Rago R, Mannelli P,
de Riso S. Adjuvant trazodone in the treatment of
alcoholism: an open study. Alcohol Alcohol 1998;34:
3625.
[139]   Cornelius JR, Bukstein O, Salloum I, Clark D. Alcohol and
psychiatric comorbidity. Recent Dev Alcohol 2003;16:
36174.
[140]   Pettinati HM, Volpicelli JR, Kranzler HR, Luck G, Rukstalis
MR, Cnaan A. Sertraline treatment for alcohol
dependence: interactive effects of medication  and
alcoholic  subtype. Alcohol Clin Exp Res 2000;24:10419.
[141]   Gawin FH, Kleber HD, Byck R, Rounsaville BJ, Kosten  TR,
Jatlow PI, et al. Desipramine facilitation of initial cocaine
abstinence.  Arch  Gen Psychiatry 1989;46:11721.
[142]   Batki SL, Manfredi LB, Sorensen JL, Jacob  P, Dumontet R,
Jones RT. Fluoxetine  for cocaine abuse in methadone
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 294
patients: preliminary  ndings. NIDA Res Monogr Ser
1991;105:5167.
[143]   Pollack MH, Rosenbaum  JF. Fluoxetine  treatment  of
cocaine  abuse in heroin addicts. J Clin Psychiatry
1991;52:313.
[144]   Jorenby DE, Leischow SJ, Nides MA, Rennard SI, Johnston
JA, Hughes AR, et al. A controlled  trial of sustained-
release bupropion, a nicotine patch, or both for smoking
cessation. N Engl J Med 1999;340:68591.
[145]   Hays JT, Hurt RD, Rigotti NA, Niaura  R, Gonzales D,
Durcan MJ, et al. Sustained-release bupropion  for
pharmacologic  relapse prevention after smoking
cessation: a randomized, controlled trial. Ann Intern Med
2001;135:42333.
[146]   Ahluwalia JS, Harris KJ, Catley D, Okuyemi KS, Mayo MS.
Sustained-release bupropion  for smoking cessation in
African  Americans:  a randomized controlled trial. J Am
Med Assoc 2002;288:46874.
[147]   Tonnesen P, Tonstad S, Hjalmarson  A, Lebargy F, Van
Spiegel PI, Hider A, et al. A multicentre,  randomized,
double-blind,  placebo-controlled, 1-year study of
bupropion SR for  smoking cessation. J Intern Med
2003;254:18492.
[148]   Dalsgareth OJ, Hansen NC, Soes-Petersen  U, Evald  T,
Hoegholm A, Barber J, et al. A multicenter,  randomized,
double-blind,  placebo-controlled, 6-month trial  of
bupropion hydrochloride sustained-release tablets as an
aid to smoking cessation in hospital employees. Nicotine
Tob Res 2004;6:5561.
[149]   Collins BN, Wileyto EP, Patterson F, Rukstalis M, Audrain-
McGovern J, Kaufmann V, et al. Gender differences in
smoking cessation in a placebo-controlled trial of
bupropion with behavioral counseling. Nicotine Tob Res
2004;6:2737.
[150]   Pope  HG, Hudson JI. Antidepressant drug therapy  for
bulimia: current status. J Clin Psychiatry  1986;47:
33945.
[151]   Edelstein CK, Yager J, Gitlin M, Landsverk J. A clinical
study of anti-depressant medications in the treatment of
bulimia. Psychiatric Med 1989;7:11121.
[152]   Walsh BT, Hadigan CM, Devlin MJ, Gladis M, Roose  SP.
Long-term outcome of antidepressant treatment for
bulimia nervosa.  Am J Psychiatry 1991;148:120612.
[153]   Steven  J, Romano MD, Katherine  A, Halmi MD, Neena  P,
Sarkar PhD, et al. Placebo-controlled study of uoxetine
in  continued treatment  of bulimia nervosa  after
successful acute uoxetine treatment. Am J Psychiatry
2002;159:96102.
[154]   Hollander E, DeCaria CM, Mari E, Wong CM, Mosovich S,
Grossman R, et al. Short-term single-blind uvoxamine
treatment of pathological gambling. Am J Psychiatry
1998;155:17813.
[155]   Hollander E, DeCaria  CM, Finkell JN, Begaz T, Wong CM,
Cartwright C. A randomized double-blind uvoxamine/
placebo crossover trial in pathologic gambling.  Biol
Psychiatry 2000;47(9):8137.
[156]   Blanco C, Petkova E, Ibanez A, Saiz-Ruiz J. A pilot placebo-
controlled study of uvoxamine for  pathological
gambling. Ann  Clin Psychiatry  2002;14:915.
[157]   Kim SW, Grant  JE, Adson DE, Shin YC, Zaninelli R. A
double-blind placebo-controlled study of the efcacy and
safety  of paroxetine  in the treatment  of pathological
gambling. J Clin Psychiatry  2002;63:5017.
[158]   Grant  JE, Kim SW, Potenza MN, Blanco C, Ibanez A,
Stevens L, et al. Paroxetine  treatment  of pathological
gambling: a multi-centre randomized controlled trial. Int
Clin Psychopharmacol  2003;18:2439.
[159]   Pallanti S, Baldini-Rossi  N, Sood E, Hollander  E.
Nefazodone treatment  of pathological  gambling:  a
prospective open-label controlled  trial. J Clin Psychiatry
2002;63:10349.
[160]   Zimmerman M, Breen  RB, Posternak MA. An open-label
study of citalopram  in the treatment  of pathological
gambling.  J Clin Psychiatry 2002;63:448.
[161]   Black DW. An open-label trial of bupropion in the
treatment of pathological gambling.  J Clin
Psychopharmacol 2004;24(1):10810.
[162]   Snaith RP. Collins SA: ve exhibitionists and a method of
treatment. Br J Psychiatry 1981;138:12630.
[163]   Perilstein  RD, Lipper S, Friedman LJ. Three cases of
paraphilias responsive to uoxetine  treatment.  J Clin
Psychiatry 1991;52:16970.
[164]   Stein DJ, Hollander  E, Anthony DT, Schneier FR, Fallon
BA, Liebowitz MR, et al. Serotonergic  medications  for
sexual obsessions, sexual addictions, and paraphilias. J
Clin Psychiatry 1992;53:26771.
[165]   Kafka  MP. Sertraline pharmacotherapy for paraphilias
and paraphilia-related disorders: an open trial. Ann Clin
Psychiatry 1994;6:18995.
[166]   Zohar J, Kaplan Z, Benjamin J. Compulsive exhibitionism
successfully treated with uvoxamine: a controlled  case
study. J Clin Psychiatry  1994;55:868.
[167]   Greenberg DM, Bradford JMW, Curry S, ORourke A. A
comparison of treatment  of paraphilias with three
serotonin reuptake inhibitors: a retrospective study. Bull
Am Acad Psychiatry  Law 1996;24:52532.
[168]   Abouesh A, Clayton A. Compulsive voyeurism  and
exhibitionism: a clinical response to paroxetine. Arch Sex
Behav  1999;28:2330.
[169]   Coleman  E, Gratzer  T, Nesvacil L, Raymond  NC.
Nefazodone  and the treatment of nonparaphilic
compulsive sexual behavior: a retrospective study. J Clin
Psychiatry 2000;61:66470.
[170]   Wainberg ML, Muench F, Mogenstern J, Hollander  E,
Irwin TW, Parsons  JT, et al. A double-blind study of
citalopram versus placebo  in the treatment of
compulsive sexual behaviors  in gay and bisexual men. J
Clin Psychiatry 2006;67(12):196873.
[171]   Volpicelli JR, Alterman AI, Hayashida  M, OBrien CP.
Naltrexone in the treatment of alcohol dependence. Arch
Gen Psychiatry  1992;49:87680.
[172]   OMalley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE,
Rounsaville B. Naltrexone  and coping skills therapy for
alcohol  dependence: a controlled study. Arch Gen
Psychiatry 1992;49:8817.
[173]   Swift RM, Whelihan  W, Kuznetsov  O, Buongiorno G,
Hsuing H.  Naltrexone-induced  alterations in human
ethanol  intoxication. Am J Psychiatry 1994;151:
14637.
[174]   Mason BJ, Ritvo EC, Morgan RO, Salvato FR, Goldberg G,
Welch B, et al. A double-blind,  placebo-controlled pilot
study to evaluate the efcacy and safety of oral
nalmefene HCl for alcohol dependence. Alcohol Clin Exp
Resv 1994;18:11627.
[175]   Monti PM, Rohsenow DJ, Swift RM, Abrams DB, Colby SM,
Mueller TI. Effect of naltrexone  on urge to drink  during
alcohol  cue exposure: preliminary results. Alcohol Clin
Exp  Res 1996;20:92A.
[176]   King  AC, Volpicelli JR, Frazer A, OBrien CP. Effect of
naltrexone on  subjective alcohol  response in subjects at
high and low risk for future alcohol dependence.
Psychopharmacol 1997;129:1522.
[177]   Davidson D, Palfai T, Bird C, Swift R. Effects of naltrexone
on alcohol self-administration in heavy drinkers. Alcohol
Clin Exp  Res 1999;23:195203.
[178]   Wewers ME, Dhatt R, Tejwani GA. Naltrexone
administration affects ad libitum smoking behavior.
Psychopharmacology  (Berl) 1998;140:18590.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   295
[179]   Hutchison  KE, Monti  PM, Rohsenow  DJ, Swift  RM, Colby
SM, Gnys M, et al. Effects of naltrexone with nicotine
replacement on smoking cue reactivity:  preliminary
results. Psychopharmacology (Berl) 1999;142:13943.
[180]   Kim SW. Opioid antagonists in the treatment of impulse-
control  disorders. J Clin Psychiatry  1998;59(4):15964.
[181]   Kim SW, Grant  JE, Adson DE, Shin YC. Double-blind
naltrexone and placebo  comparison study in the
treatment  of pathological  gambling.  Biol Psychiatry
2001;49:91421.
[182]   Kim SW, Grant JE. An open naltrexone treatment study in
pathological gambling  disorder. Int Clin
Psychopharmacol 2001;16:2859.
[183]   Grant  JE, Potenza  MN, Hollander  E, Cunningham-
Williams R, Nurminen  T, Smits G, et al. Multicenter
investigation of the opioid antagonist  nalmefene in the
treatment  of pathological  gambling.  Am J Psychiatry
2006;163:30312.
[184]   Grant JE, Kim SW. A case of kleptomania and compulsive
sexual behavior treated with naltrexone. Ann Clin
Psychiatry 2001;113:22931.
[185]   Raymond NC, Grant JE, Kim SW, Coleman E. Treatment of
compulsive sexual behaviour with naltrexone and
serotonin  reuptake inhibitors: two case studies.  Int Clin
Psychopharmacol 2002;17:2015.
[186]   Myrick H, Anton R. Recent  advances in the
pharmacotherapy of alcoholism. Curr Psychiatry Rep
2004;6:3328.
[187]   Ma JZ, Alt-Daoud N, Johnson BA. Topiramate reduces the
harm of excessive drinking: implications for public
health and primary  care. Addiction  2006;101(11):
15618.
[188]   Johnson  BA, Ait-Daoud  N, Bowden CL, DiClemente CC,
Roache  JD, Lawson K. Oral topiramate  for treatment of
alcohol dependence: a randomized controlled  trial.
Lancer 2003;361(9370):167785.
[189]   Kampman  KM, Pettinati  H, Lynch KG, Dackis C,
Sparkman T, Weigley C, et al. A pilot trial of topiramate
for the treatment  of cocaine  dependence. Drug Alcohol
Depend 2004;75:23340.
[190]   Shapira NA,  Goldsmith TD,  McElroy SL. Treatment  of
hinge eating disorder with topiramate: a clinical  case
series. J Clin Psychiatry 2000;61:35872.
[191]   Barbee JG. Topiramate in  the Treatment  of Severe
Bulimia Nervosa with Comorbid Mood Disorders: A Case
Series.  Int J Eat Disord 2003;33(4):46872.
[192]   Hoopes SP, Reimherr  FW, Hedges DW, Rosenthal  NR,
Kamin M, Karim  R, et al. Treatment  of bulimia nervosa
with topiramate in a randomized, double-blind, placebo-
controlled  trial.  Part 1. Improvement  in binge and purge
measures.  J Clin Psychiatry  2003;64:133541.
[193]   Nickel  C, Tritt  K, Muehlbacher M, Pedrosa Gil GF,
Mitterlehner FO, Kaplan P, et al. Topiramate treatment in
bulimia  nervosa patients:  a randomized, double-blind,
placebo-controlled trial. Int J Eat Disord 2005;38(4):
295300.
[194]   Moskowitz JA. Lithium and lady luck: use of lithium
carbonate  in compulsive gambling.  NY State  J Med
1980;80:7858.
[195]   Haller R, Hinterhuber H. Treatment of pathological
gambling  with carbamazepine. Pharmacopsychiatry
1994;27(3):129.
[196]   Pallanti S, Quercioli L, Sood E, Hollander  E. Lithium and
valproate  treatment  of pathological  gambling:  a
randomized  single-blind study. J Clin Psychiatry
2002;63:55964.
[197]   Hollander  E, Pallanti S, Allen A, Sood E, Baldini Rossi N.
Does sustained-release  lithium reduce impulsive
gambling  and affective instability versus placebo  in
pathological  gamblers with  bipolar spectrum disorders?
Am J Psychiatry 2005;162(1):13745.
[198]   Cesnik JA, Coleman E. Use of lithium carbonate  in the
treatment  of autoerotic  asphyxia.  Am J Psychother
1989;63:27786.
[199]   Fong TW, De La Garza  R, Newton  TF. A case report of
topiramate  in the treatment of nonparaphilic sexual
addiction.  J Clin Psychopharmacol 2005;25:5124 (also:
Khazaal, Y., Zullino, D.F., Topiramate in the treatment of
compulsive sexual behavior: case report. BMC Psychiatry
2006;6:22.
[200]   Shiah I-S, Chao C-Y, Mao  W-C, Chuang Y-J. Topiramate
for adolescent/young adult fetishism. Brown University
Psychopharmacology Update 2006;17:8.
[201]   Barnes GE. Clinical and prealcoholic personality
characteristics.   In: Kissin B, Begleiter H, editors. The
biology  of alcoholism. vol. 6, The pathogenesis of
alcoholism: psychosocial factors. New York: Plenum
Press; 1983. p. 11395.
[202]   Hatsukami D, Owen P, Pyle R, Mitchell JE. Similarities and
differences on the MMPI between women with  bulimia
and women with alcohol or drug abuse problems. Addict
Behav 1982;7:4359.
[203]   Ciarrocchi  JW, Kirschner NM, Fallik F. Personality
dimensions  of male pathological  gamblers, alcoholics,
and dually addicted  gamblers. J Gambling  Stud
1991;7:13341.
[204]   Lowenfeld BH. Personality  dimensions of the
pathological  gambler. Diss Abstr Int 1979;40(1-B):456.
[205]   Kranitz  L. Alcoholics,  heroin addicts and nonaddicts.
Comparisons on the MacAndrew Alcoholism Scale of the
MMPI. Quart J Stud Alcohol  1972;33:8079.
[206]   Burke HR, Marcus R. MacAndrew MMPI alcoholism scale:
alcoholism and drug addictiveness. J Psychol
1977;96:1418.
[207]   Leon G, Kolotkin R, Korgeski G. MacAndrew  Addiction
Scale  and other MMPI characteristics  associated with
obesity, anorexia, and smoking  behavior. Addict Behav
1979;4:4017.
[208]   Sutker PB, Archer RP. MMPI characteristics of opiate
addicts, alcoholics, and other drug abusers.   In: Newmark
CS, editor. MMPI: clinical and research trends. New York:
Prager; 1979. p. 10548.
[209]   Berzins JI, Ross  WF, English GE, Haley JV. Subgroups
among opiate  addicts: a typological investigation.  J Abn
Psychol 1974;83:6573.
[210]   Glen AM. Personality research on pathological gamblers.
Paper presented at the 87th Annual Convention of the
American Psychological Association, New York, 1979.
Cited in McCormick RA, Taber JI. The pathological
gambler: Salient personality  variables. In: Galski T,
editor. The Handbook of Pathological  Gambling.
Springeld, Il: Charles C. Thomas, 1979:939.
[211]   Arnon D, Kleinman MH, Kissin B. Psychological
differentiation in  heroin addicts. Int J Addict 1974;9:
1519.
[212]   Karp SA, Pardes H. Psychological  differentiation  (eld
dependence) in obese  women. Psychosom Med
1965;27:23844.
[213]   Brown R, Williams R. Internal and external cues relating
to uid intake  in obese and alcoholic  persons. J Abnorm
Psychol 1975;84:6605.
[214]   Lansky D, Nathan  PE, Lawson DM. Blood alcohol level
discrimination by alcoholics:  the role of internal  and
external  cues. J Consult Clin Psychol 1978;46:95360.
[215]   Nisbett RE, Storms MD. Cognitive and social
determinants of food intake.   In: London H, Nisbett  RE,
editors. Thought  and feeling: cognitive  alteration  of
feeling states. Chicago: Aldine; 1974.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 296
[216]   Tsuang MT, Lyons  MJ, Meyer JM, Doyle T, Eisen SA,
Goldberg J, et al. Co-occurrence of abuse of different
drugs in men: the role of drug-specic and shared
vulnerabilities. Arch  Gen Psychiatry 1998;55:96772.
[217]   Karkowski LM,  Prescott CA, Kendler KS. Multivariate
assessment of factors inuencing illicit substance use in
twins  from femalefemale pairs. Am J Med Genet
2000;96:66570.
[218]   Kendler KS, Jacobson KC, Prescott CA,  Neale MC.
Specicity  of genetic and environmental  risk factors for
use and abuse/dependence of cannabis, cocaine,
hallucinogens, sedatives, stimulants, and opiates in male
twins.  Am J Psychiatry 2003;160:68795.
[219]   Uhl GR, Liu Q-R, Naiman  D. Substance abuse
vulnerability loci: converging  genome scanning data.
Trends Genet 2002;18(8):4205.
[220]   Michael M, Vanyukov RE, Tarter LK, Galina PK, Brion SM,
Duncan B. Clark Liability  to substance use disorders: 1.
Common mechanisms and manifestations. Neurosci
Biobehav Rev 2003;27:50715.
[221]   Susan EY, Soo HR, Michael CS, Robin PC, John KH. Genetic
and environmental vulnerabilities underlying adolescent
substance use and problem use: general  or specic?
Behav Genet 2006;36(4):60315.
[222]   Dackis C, OBrien C. Neurobiology of addiction: treatment
and public policy ramications.  Nat Neurosci
2005;8(11):14316.
[223]   Gardner E. Brain  reward mechanisms.   In: Lowinson  JH,
Ruiz P, Millman RB, Langrod JG, editors. Substance abuse:
a comprehensive textbook. 4th ed., Lippincott Williams &
Wilkins; 2005. p. 4896.
[224]   Gold MS, Starr J. Eating disorders.   In: Lowinson  JH, Ruiz
P, Millman RB, Langrod JG, editors. Substance abuse: a
comprehensive textbook. 4th ed., Lippincott Williams  &
Wilkins; 2005. p. 46987.
[225]   Blum K, Cull JG, Braverman  FR, Comings DE. Reward
deciency syndrome. Am Sci 1996;84:13245.
[226]   Goudriaan AE, Oosterlaan J, de Beurs E, Van den Brink W.
Pathological gambling:  a comprehensive review of
biobehavioral ndings. Neurosci  Biobehav Rev
2004;28:12341.
[227]   Spangler R, Wittkowski  KM, Goddard NL, Avena  NM,
Hoebel BG, Leibowitz SF. Opiate-like effects of sugar on
gene expression in reward areas of the rat brain,
Molecular. Brain  Res 2004;124:13442.
[228]   Volkow ND, Wise RA. How can drug addiction  help us
understand obesity? Nat Neurosci  2005;8(5):55560.
[229]   Wang G-J, Volkow ND, Telang F, Jayne M, Ma J, Rao M,
et al. Exposure  to appetitive  food stimuli markedly
activates the human  brain. Neuroimage 2004;21:
17907.
[230]   Koob GF, LeMoal M. Neurobiology  of addiction. London:
Academic Press; 2006.
[231]   Rudolf U, Tara M, Michael JB, Tim D, Mary LP, Virginia
WNg, et al. Medial  prefrontal cortex  activity associated
with  symptom provocation in  eating disorders. Am J
Psychiatry 2004;161:123846.
[232]   Volkow ND, Wang FG.-J., Ma Y, Fowler JS, Wong C, Ding
Y-S, et al. Activation of orbital  and medial prefrontal
cortex by methylphenidate  in cocaine-addicted subjects
but  not in controls:  relevance to  addiction. J Neurosci
2005;25(15):39329.
[233]   Hommer D, Andreasen P, Rio D, Williams W, Ruttiman U,
Momenan R, et al. Effects of m-clorophenylpiperazine on
regional brain glucose utilization:  a positron emission
tomographic comparison  of alcoholic and control
subjects. J Neurosci  1997;17:2796806.
[234]   Volkow ND, Fowler JS, Wolf AP, Hitzemann R, Dewey S,
Bendriem B, et al. Changes in brain  glucose metabolism
in cocaine dependence and withdrawal.  Am J Psychiatry
1991;148:6216.
[235]   Grant  JE, Brewer JA, Potenza MN. The neurobiology  of
substance and behavioral addictions. CNS  Spectr
2006;3(12):92430.
[236]   Zhang  P-W, Ishiguro  H, Ohtsuki  T, Hess J, Carillo F,
Walther D, et al. Human  cannabinoid  receptor 1: 5
0
exons,  candidate regulatory regions, polymorphisms,
haplotypes and association with polysubstance abuse.
Mol Psychiatry  2004;9:91631.
[237]   Fattore L, Spano MS, Deiana S, Melis V, Cossu G, Fadda P,
et al. An endocannabinoid mechanism in relapse to drug
seeking: A review of animal studies  and clinical
perspectives.. Brain  Res Rev 2007;53:116.
[238]   Undine EL, Thomas S, Falk WL, Rainer H, Malek B, Georg
W, et al. Association of the met66 allele of brain-derived
neurotrophic factor (BDNF) with  smoking.
Psychopharmacology  2007;190:4339.
[239]   Simerly R. Feeding signals and drugs meet in  the
midbrain. Nat Med 2006;12(11):12446.
[240]   Adinoff  B. Neurobiologic processes in drug reward and
addiction. Harv  Rev Psychiatry 2004;12:30520.
[241]   Avena  NM, Rada P, Moise N, Hoebel BG. Sucrose sham
feeding  on a binge  schedule releases accumbens
dopamine  repeatedly  and eliminates the acetylcholine
satiety response.  Neuroscience  2006;139:81320.
[242]   Cheer JF, Wassum KM, Sombers LA, Heien MLAV,
Ariansen JL, Aragona BJ, et al. Phasic dopamine  release
evoked  by abused substances requires cannabinoid
receptor activation.  J Neurosci  2007;27(4):7915.
[243]   DiLeone RJ, Georgescu D, Nestlet EJ. Lateral hypothalamic
neuropeptides in reward and drug addiction. Life Sci
2003;73:75968.
[244]   Fletcher PJ, Korth KM, Chambers JW. Selective destruction
of brain serotonin neurons by 5,7-dihydroxytryptamine
increases responding for a conditioned reward.
Psychopharmacology 1999;147:2919.
[245]   Kelley AE.  Ventral striatal  control of appetitive
motivation: role in ingestive behavior and reward-related
learning.  Neurosci  Biobehav Rev 2004;27:76576.
[246]   Pelchat ML. Of human bondage: food craving, obsession,
compulsion, and addiction. Physiol Behav  2002;76:
34752.
[247]   Shaffer HJ, LaPlante DA, LaBrie RA, Kidman RC, Donato
AN, Stanton MV. Toward a syndrome model of addiction:
multiple expressions, common etiology. Harv Rev
Psychiatry 2004;12:36774.
[248]   Wong T, Tomasi D, Thanos PK, Fowler JS, Wang G-J, Yang
J, et al. Gastric stimulation in obese subjects activates the
hippocampus and other regions involved in brain reward
circuitry. PNAS 2006;103:156415.
[249]   Kammeier ML, Hoffman H, Loper RG. Personality
characteristics  of alcoholics  as college freshman and at
time of treatment.  Quart J Stud Alc 1973;34:3909.
[250]   Loper R, Kammeier M, Hoffman H. MMPI characteristics
of college freshman males who later became alcoholic.  J
Abnorm  Psychol 1973;82:15962.
[251]   Hoffman H, Loper R, Kammeier ML. Identifying future
alcoholics with MMPI alcoholism scales. Quart J Stud Alc
1974;35:4908.
[252]   Witkin H, Karp S, Goodenough  D. Dependence in
alcoholics. Quart J Stud Alcohol 1959;20:493504.
[253]   Karp SA, Kronstadt  NL. Alcoholism and psychological
differentiation: long-range effect of heavy  drinking on
eld dependence. J Nerv Ment Dis 1965;140:4126.
[254]   McCord W, McCord J. Origins of alcoholism. Stanford, CA:
Stanford University Press; 1960.
[255]   McCord W, McCord J. A longitudinal  study of the
personality of alcoholics.   In: Pittman  DJ, Snyder CR,
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   297
editors. Society,  culture, and drinking patterns. New
York: Wiley; 1962. p. 41330.
[256]   Robins LN. Deviant children grown up: a sociological and
psychiatric study of sociopathic personality. Baltimore:
Williams and Wilkins; 1966.
[257]   Robins LN, Bates WM, ONeal P. Adult drinking patterns
of former problem children.   In: Pittman  DJ, Snyder CR,
editors. Society,  culture, and drinking patterns. New
York: Wiley; 1962. p. 395412.
[258]   Jones MC. Personality  correlates  and antecedents of
drinking patterns  in adult males.  J Consult Clin Psychol
1968;32:212.
[259]   Jones MC. Personality  correlates  and antecedents of
drinking patterns  in women. J Consult Clin Psychol
1971;36:619.
[260]   Jessor R, Jessor S. Problem behavior and psychosocial
development: a longitudinal  study of youth. New York:
Academic  Press; 1977.
[261]   Jessor R, Jessor SL.  Theory testing in longitudinal
research  on marijuana research.   In: Kandel D, editor.
Longitudinal research on drug use: empirical ndings
and methodological issues. Washington,  DC:
Hemisphere; 1978. p. 4171.
[262]   Kandel DB. Convergences in prospective longitudinal
surveys of drug use in normal populations.   In: Kandel D,
editor. Longitudinal research on drug use: empirical
ndings and methodological  issues. Washington,  DC:
Hemisphere; 1978. p. 338.
[263]   Kandel DB. Drug and drinking behavior among  youth.
Ann Rev Sociol 1980;6:23585.
[264]   Zucker  RA. Developmental aspects of drinking through
the young adult years.   In: Blane HT, Chafetz ME, editors.
Youth, alcohol, and social policy. New York: Plenum
Press; 1979. p. 91146.
[265]   Kellam SG, Ensminger ME, Simon MB. Mental health in
rst grade and teenage drug, alcohol, and cigarette use.
Drug Alcohol Depend 1980;5:273304.
[266]   Wingard JA, Huba GJ, Bentler PM. A longitudinal analysis
of personality  structure and adolescent substance use.
Person Ind Diff 1980;1:25972.
[267]   Jessor R, Chase JA, Donovan JE. Psychosocial correlates of
marijuana use and problem drinking in a national sample
of adolescents. Am J Public Health 1980;70:60413.
[268]   Zucker  RA, Noll  RB. Precursors and developmental
inuences  on drinking and alcoholism: etiology from a
longitudinal  perspective. In: Alcohol and health
monograph. 1. Alcohol consumption and related
problems. DHHS Publication No. (ADM)  82-1190.
Rockville,  MD: National Institute on Alcohol Abuse and
Alcoholism, 1982.
[269]   Vicary JR, Lerner JV. Longitudinal  perspectives  on drug
use: analyses  from the New York longitudinal  study. J
Drug Ed 1983;13:27585.
[270]   Labouvie  EW, McGee CR. Relation of personality  to
alcohol and drug use in adolescence. J Consult Clin
Psychol 1986;54:28993.
[271]   Sadava SW. Initiation  to cannabis use: a longitudinal
social psychological study of college freshmen. Can J
Behav Sci  1973;5:37184.
[272]   Gulas I, King FW. On the question  of pre-existing
personality differences between users  and nonusers of
drugs. J Psychol 1976;92:659.
[273]   Goldstein JW, Sappington  JT. Personality  characteristics
of students who became heavy drug users: an MMPI
study of an avant-garde. Am J Drug Alcohol Abuse
1977;4:40112.
[274]   Sadava SW, Forsyth R. Person-environment interaction
and college student drug use: a multivariate longitudinal
study. Genet Psychol Monogr 1977;96:21145.
[275]   Smith GM. Correlates of personality  and drug use. I.
RAUS cluster review  no. 3. Rockville, MD: National
Institute  of Drug Abuse; 1977.
[276]   Ginsberg IJ, Greenley JR. Competing theories of marijuana
use: a longitudinal study. J Health Soc Behav 1978;19:
2234.
[277]   Smith GM, Fogg CP. Psychological predictors of early use,
late use, and nonuse of marijuana  among teenage
students.   In: Kandel D, editor. Longitudinal research on
drug use: empirical ndings and methodological  issues.
Washington,  DC: Hemisphere; 1978. p. 10113.
[278]   Brook JS, Lukoff  IF, Whiteman M. Initiation into
adolescent  marijuana use. J Genet  Psychol 1980;137:
13342.
[279]   Huba GJ, Bentler PM. A developmental theory of drug use:
Derivation  and assessment  of a causal modeling
approach.   In: Baltes PB, Brim OG, editors. Life-span
development  and behavior. New York: Academic  Press;
1982.
[280]   Kellam SG, Brown  CH, Rubin BR, Ensminger ME. Mental
health and going to school: the woodlawn program of
assessment, early intervention, and evaluation. Chicago:
University  of Chicago Press; 1983.
[281]   Kellam SG, Brown  CH, Rubin BR, Ensminger ME. Paths
leading to teenage  psychiatric symptoms and substance
abuse: developmental epidemiological studies  in
Woodlawn.   In: Guze SB, Earls FJ, Barrett JE, editors. Child
psychopathology and development.  New York: Raven
Press; 1983. p. 1747.
[282]   Brook JS, Whiteman M, Gordon AS, Cohen P. Dynamics of
childhood and adolescent personality traits and
adolescent  drug use. Dev Psychol 1986;22:40314.
[283]   Block J, Block JH, Keyes S. Longitudinally foretelling drug
usage in adolescence: early childhood personality  and
environmental precursors. Child Dev 1988;59:33655.
[284]   Shedler J, Block J. Adolescent drug use and psychological
health: a longitudinal  inquiry. Am Psychol  1990;45:
61230.
[285]   Kellam SG. Developmental epidemiological and
prevention research  on early risk behaviors.   In: Lipsitt
SP, Mitnick LL, editors. Self-Regulatory behaviors and risk
taking:  causes and consequences. Norwood, NJ: Ablex;
1991. p. 5170.
[286]   Martin CS, Clifford PR, Clapper  RL. Patterns  and
predictors of simultaneous  and concurrent use of
alcohol, tobacco,  marijuana, and hallucinogens  in rst-
year college students. J Subst  Abuse 1992;4(3):31926.
[287]   Brook, JS, Whiteman,  M, Flinch, S, Cohen, P. Mutual
attachment,  personality, and drug use: pathways from
childhood to  young adulthood. Genetic, social & general
psychology  monographs, Nov. 98, vol. 124 Issue 4, p.
492510.
[288]   Kosten TA, Ball SA, Rounsaville BJ, A sibling study of
sensation seeking and opiate  addiction. J Nerv Ment Dis
l994;182(5):2849.
[289]   Brook JS, Whiteman  M, Finch  S, Cohen P. Longitudinally
foretelling  drug use in the late twenties: adolescent
personality  and socialenvironmental  antecedents.  J
Genet Psychol 2000;161(1):3751.
[290]   Shaffer HJ, Eber GB. Temporal progression of cocaine
dependence symptoms in the US National Comorbidity
Survey.  Addiction  2002;97:54354.
[291]   Nelson  CB, Heath AC, Kessler RC. Temporal progression
of alcohol dependence symptoms in the U.S. household
population: results from the National Comorbidity Study.
J Consult Clin Psychol  1998;66:47483.
[292]   Kessler RC, Nelson  CB, McGonagle KA, Edlund MJ, Frank
RG, Leaf PJ. The epidemiology of co-occurring addictive
and mental disorders: implications for prevention and
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 298
service utilization.  Am J Orthopsychiatry  1996;66:
1731.
[293]   Patton GC, Carlin JB, Co
 !
ey C, et al. Depression, anxiety,
and smoking initiative: a prospective study over 3 years.
Am J Public Health  1998;88:151822.
[294]   Kessler RC, Aguilar-Gaxiola S, Andrade L, Bijl R, Borges G,
Caraveo-Anduaga  JJ, et al. Mental-substance
comorbidities in the ICPE surveys. Psychiatria Fennica
2001;32:6280.
[295]   Kessler RC, Aguilar-Gaxiola S, Andrade L, Bijl R, Borges G,
Caraveo-Anduaga JJ, et al. Cross-national comparisons of
comorbities between substance use disorders and
mental disorders: results from the International
Consortium in Psychiatric Epidemiology.   In: Bukoski WJ,
Sloboda Z, editors. Handbook for drug abuse prevention
theory, science, and practice. New York: Plenum
Publishers; 2003. p. 44871.
[296]   Kessler RC. The epidemiology  of dual diagnosis. Biol
Psychiatry 2004;56:7307.
[297]   de Graaf R, Bijl RV, ten Have M, Beekman ATF, Vollebergh
WAM. Pathways to comorbidity:  the transition  of pure
mood, anxiety and substance use disorders into
comorbid conditions  in a longitudinal  population-based
study. J Affect Disord 2004;82:4617.
[298]   Marquenie LA, Schade   A, van B, Anton  JLM, Comijs HC,
de Graaf R, et al. Origin of the comorbidity  of anxiety
disorders and alcohol dependence: ndings of a
general population  study. Eur Addict Res 2007;13(1):
p3949.
[299]   Simons  JS, Carey KB, Gaher RM. Lability and impulsivity
synergistically increase risk for alcohol-related problems.
Am J Drug Alc Abuse 2004;30(3):68594.
[300]   Bulik CM, Sullivan PF, Fear JL, Joyce  PR. Eating disorders
and antecedent anxiety disorders: a controlled  study.
Acta Psychiatr Scand 1997;96:1017.
[301]   Godart NT, Flament MF, Lecrubier Y, Jeammet P. Anxiety
disorders in anorexia nervosa and bulimia  nervosa: co-
morbidity and chronology of appearance. Eur Psychiatry
2000;15:3845.
[302]   Silberg JL, Bulik CM. The developmental association
between eating  disorders symptoms and symptoms of
depression and anxiety in juvenile twin girls. J Child
Psychol Psychiatry  2005;46(12):131726.
[303]   Krueger RF, Caspi A, Moftt TE, Silva PA. The structure
and stability of common mental disorders (DSM-III-R): a
longitudinal-epidemiological study. J Abnorm Psychology
1998;107:21627;
Krueger RF. The structure of common mental disorders.
Arch  Gen Psychiatry 1999;56:9216.
[304]   Krueger RF. Psychometric perspectives on comorbidity.
In: Helzer JE, Hudziak JJ, editors. Dening
psychopathology in the 21st century: DSM-V and beyond.
Washington,  DC: American Psychiatric Publishing; 2002.
p. 4154.
[305]   American Psychiatric Association. Diagnostic  and
statistical manual of mental disorders, (DSM-IV),   4th ed.,
Washington,  DC: American Psychiatric Press; 1994.
[306]   Yoshimoto K, McBride WJ, Lumeng  L, Li T-K. Alcohol
stimulates the release of dopamine and serotonin in the
nucleus accumbens. Alcohol 1991;9:1722.
[307]   Di Chiara G. Alcohol and dopamine. Alcohol Health
ResWorld  1997;21:10814.
[308]   Weiss F, Lorang MT, Bloom FE, Koob GF. Oral alcohol self-
administration  stimulates dopamine release  in the rat
nucleus accumbens: genetic and motivational
determinants. J Pharm  Exp Ther 1993;267:2508.
[309]   Johnson SW, North RA. Opioids excite dopamine neurons
by hyperpolarization of local interneurons. J Neurosci
1992;12:4838.
[310]   Devine  DP, Leone P, Pocock  D, Wise RA. Differential
involvement of ventral  tegmental mu,  delta and kappa
opioid receptors in modulation  of basal mesolimbic
dopamine  release:  in vivo microdialysis studies.  J
Pharmacol Exp  Ther 1993;266:123644.
[311]   Pettit HO, Justice Jr JB. Effect of dose on cocaine
selfadministration behavior and dopamine  levels in the
nucleus accumbens. Brain Res 1991;539:94102.
[312]   Bergman J, Kamien JB, Spealman  RD. Antagonism of
cocaine  self-administration  by selective dopamine  D(1)
and D(2) antagonists.  Behav Pharmacol 1990;1:
35563.
[313]   Caine SB, Koob  GF. Effects of mesolimbic  dopamine
depletion on  responding maintained by cocaine  and
food. J Exp Anal  Behav 1994;61:21321.
[314]   Breiter HC, Gollub RL, Weisskoff RM, et al. Acute effects
of cocaine on human brain activity and emotion. Neuron
1997;19:591611.
[315]   Wise RA. The role of reward pathways in  the
development of drug dependence. Pharmacol Ther
1987;35:22762.
[316]   Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ. Cocaine
receptors on dopamine  transporters  are related to self-
administration of cocaine. Science 1987;237:121923.
[317]   Di Chiara G, Imperato A. Drugs abused  by humans
preferentially increase synaptic dopamine
concentrations in the mesolimbic system of freely
moving  rats. Proc Natl Acad Sci USA 1988;85:52748.
[318]   Brody AL, Olmstead RE, London ED, Farahi J, Meyer JH,
Grossman P, et al. Smoking-induced ventral striatum
dopamine  release.  Am J Psychiatry 2004;161:12118.
[319]   Carlezon Jr WA, Wise RA. Rewarding actions of
phencyclidine and related drugs in nucleus accumbens
shell and frontal  cortex. J Neurosci  1996;16:311222.
[320]   Mifsud JC, Hernandez L, Hoebel BG. Nicotine infused into
the nucleus accumbens  increases synaptic dopamine as
measured by in vivo microdialysis. Brain Res
1989;478:3657.
[321]   Cheer JF, Wassum KM, Heien ML, Phillips PE, Wightman
RM. Cannabinoids enhance subsecond dopamine release
in the nucleus accumbens  of awake rats. J Neurosci
2004;24:4393400.
[322]   French  ED, Dillon K, Wu X. Cannabinoids excite
dopamine  neurons in  the ventral tegmentum and
substantia nigra. NeuroReport  1997;8:64952.
[323]   Gessa GL, Melis M, Muntoni  AL, Diana  M. Cannabinoids
activate mesolimbic dopamine  neurons by an action on
cannabinoid CB1 receptors. Eur J Pharmacol 1998;341:
3944.
[324]   Hernandez  L, Hoebel BG. Feeding and hypothalamic
stimulation increase dopamine  turnover in the
accumbens. Physiol Behav 1988;44:599606.
[325]   Avena  NM, Rada P, Moise N, Hoebel BG. Sucrose sham
feeding  on a binge  schedule releases accumbens
dopamine  repeatedly  and eliminates the acetylcholine
satiety response.  Neuroscience  2006;139(3):81320.
[326]   Hajnal R, Norgren R. Accumbens dopamine mechanisms
in sucrose intake. Brain  Res 2001;904:7684.
[327]   Hernandez L, Hoebel BG. Feeding can enhance dopamine
turnover in the prefrontal  cortex. Brain  Res Bull
1990;25:9759.
[328]   Yoshida M, Yokoo H, Mizoguchi K, Tsuda A, Nishikawa T,
Tanaka M. Eating and drinking cause increased
dopamine release in the nucleus accumbens and ventral
tegmental area in the rat: meaurement by in vivo
microdialysis. Neurosci  Lett 1992;139:736.
[329]   Blaszczynski A, Wilson A, McConaghy  N. Sensation
seeking and pathological gambling.  Br J Addict
1986;81:1137.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   299
[330]   Bergh C, Eklund  T, Soderstcn  P, Nordin  C. Altered
dopamine  function in  pathological  gambling.  Psycho/
Med 1997;27:4735.
[331]   Agmo A, Berenfeld  R. Reinforcing  properties of
ejaculation in the male rat: role of opioids and dopamine.
Behav Neurosci 1990;104:17782.
[332]   Pfaus JG, Damsma G, Nomikos GG, Wenkstern DG, Blaha
CD, Phillips  AG, et al. Sexual behavior enhances central
dopamine  transmission  in the male rat. Brain Res
1990;530:3458.
[333]   Mas M. Neurobiological correlates of masculine  sexual
behavior. Neurosci  Biobehav  Rev 1995;19:26177.
[334]   Balfour  ME, Yu L, Coolen  LM. Sexual behavior and sex-
associated environmental cues activate the mesolimbic
system in male rats. Neuropsychopharmacology
2004;29:71830.
[335]   Robinson TE, Berridge KC. The psychology and
neurobiology of addiction: an incentive-sensitization
view. Addiction  2000;95(2):S91117.
[336]   Hyman SE, Malenka  RC. Addiction and the brain: the
neurobiology of compulsion and its persistence. Nat Rev
Neurosci  2001;2:695703.
[337]   Schultz  W. Predictive reward signal of dopamine
neurons.  J Neurophysiol 1998;80:127.
[338]   Schultz  W, Dayan P, Montague PR. A neural substrate  of
prediction and reward. Science  1997;275:15939.
[339]   Fenu S, Di Chiara G. Facilitation  of conditioned  taste
aversion  learning  by systemic amphetamine:  role of
nucleus accumbens shell dopamine D
1
 receptors. Eur J
Neurosci  2003;18:202530.
[340]   Schultz  W. Dopamine neurons  and their role in
reward mechanisms.  Curr Opin Neurobiol 1997;7(2):
1917.
[341]   Gurden  H, Tassin JP, Jay TM.  Integrity of mesocortical
dopaminergic  system is necessary  for complete
expression of in vivo hippocampal-prefrontal  cortex
long-term  potentiation.  Neurosci 1999;94:101927.
[342]   Mulder AB, Arts MPM, Lopes da Silva FH. Short- and long-
term plasticity  of the hippocampus to nucleus
accumbens and prefrontal cortex pathways in the rat, in
vivo. Eur J Neurosci  1997;9:160311.
[343]   Robinson T, Kolb  B. Alterations in the morphology of
dendrites and dendritic spines in the nucleus accumbens
and prefrontal cortex following  repeated treatment with
amphetamine  or cocaine. Eur J Neurosci 1999;11:
1598604.
[344]   Robinson TE, Berridge KC. The neural basis of drug
craving: an incentive-sensitization theory of addiction.
Brain  Res Rev 1993;18:24791.
[345]   Deroche V, Le Moal M, Piazza PV. Cocaine self-
administration increases the incentive motivational
properties  of the drug in rats. Eur J Neurosci
1999;11:27316.
[346]   Berridge KC, Robinson TE. What is the role of dopamine
in reward: hedonic impact, reward learning, or incentive
salience? Brain  Res Rev 1998;28:30969.
[347]   Garris PA, Kilpatrick M, Bunin MA, Michael D, Walker QD,
Wightman RM. Dissociation  of dopamine release  in the
nucleus accumbens from intracranial self-stimulation.
Nature 1999;398:679.
[348]   Montague PR, Berns GS. Neural economics and biological
substrates  of valuation. Neuron 2002;36:26584.
[349]   McClure SM, Daw ND, Montague  PR. A computational
substrate  for incentive  salience. Trends Neurosci
2003;25:4238.
[350]   Comings DE, Gade R, Wu S, Dietz CCG, Muhleman D,
Saucier G, et al. Studies of the potential role of the
dopamine  D1 receptor gene in addictive  behaviors.  Mol
Psychiatry 1997;2:4456.
[351]   Centonze  D, Grande C, Usiello A, Gubellini P, Erbs E,
Martn AB, et al. Receptor subtypes involved in the
presynaptic  and postsynaptic  actions of dopamine  on
striatal interneurons. J Neuroscience 2003;23:624554.
[352]   Momiyama T, Sim JA, Brown DA. Dopamine D1-like
receptor-mediated presynaptic inhibition of excitatory
transmission  onto  rat magnocellular basal  forebrain
neurons.  J Physiol 1996;495:97106.
[353]   Civelli O. Molecular biology  of the dopamine  receptor
subtypes.   In: Bloom FE, Kupfer DJ, editors.
Psychopharmacology: the fourth generation of progress.
New York: Raven Press; 1995. p. 1557.
[354]   DSouza MS, Ikegami A, Olsen CM, Duvauchelle CL.
Chronic D1 agonist  and ethanol coadministration
facilitate ethanol-mediated  behaviors..  Pharmacol
Biochem Behav 2003;76(2):33542.
[355]   Karasinska JM, George SR, Cheng R, ODowd BF. Deletion
of dopamine D1 and D3 receptors differentially affects
spontaneous  behaviour and cocaine-induced  locomotor
activity,  reward and CREB phosphorylation.  Eur J
Neurosci  2005;22(7):174150.
[356]   Graham DL, Hoppenot R, Hendryx A, Self DW.
Differential ability of D1 and D2 dopamine  receptor
agonists  to induce and modulate expression and
reinstatement  of cocaine place preference in rats.
Psychopharmacology 2007;191(3):71930.
[357]   Sharf R, Lee DY, Ranaldi R. Microinjections of SCH 23390
in the ventral tegmental area reduce operant responding
under a progressive ratio schedule of food reinforcement
in rats. Brain Res 2005;1033(2):17985.
[358]   Sorg BA, Li N, Wu W, Bailie TM. Activation of dopamine
D1 receptors  in the medial prefrontal cortex produces
bidirectional  effects on cocaine-induced locomotor
activity in  rats: effects of repeated stress. Neuroscience
2004;127(1):18796.
[359]   Berglind WJ, Case JM, Parker MP, Fuchs  RA, See RE.
Dopamine D1  or D2 receptor antagonism  within  the
basolateral amygdala differentially alters the acquisition
of cocaine-cue associations necessary  for cue-induced
reinstatement  of cocaine-seeking. Neuroscience
2006;137(2):699706.
[360]   Alleweireldt AT, Kirschner KF, Blake CB, Neisewander JL.
D1-receptor drugs and cocaine-seeking behavior:
investigation  of receptor  mediation and behavioral
disruption  in rats. Psychopharmacology 2003;168(1/2):
10917.
[361]   Cooper SJ, Al-Naser HA. Dopaminergic control  of food
choice:  contrasting effects of SKF 38393 and quinpirole
on high-palatability food preference in  the rat.
Neuropharmacology 2006;50(8):95363.
[362]   Wolf ME, Sun X, Mangiavacchi  S, Chao SZ. Psychomotor
stimulants  and neuronal plasticity.  Neuropharmacology
2004;47(1):6179.
[363]   Lee K-W, Kim Y, Kim AM, Helmin K, Nairn AC, Greengard
P. Cocaine-induced  dendritic spine  formation  in D1 and
D2 dopamine  receptor-containing medium spiny
neurons in  nucleus accumbens.  In: Proceedings  of the
National  Academy  of Sciences  of the United  States of
America, 2/28/2006, Vol. 103 Issue 9; 2006. p. 3399404.
[364]   Zang J, Zang L, Jiao H, Zang Q, Zang D, Lou D, et al. c-Fos
facilitates the acquisition  and extinction  of cocaine-
induced  persistent changes.  J Neurosci  2006;26(51):
1328796.
[365]   Zhang D, Zhang L, Lou DW, Nakabeppu Y, Zhang J, Xu M.
The dopamine D1 receptor is a critical mediator for
cocaine-induced gene expression. J Neurochem
2002;82(6):145364.
[366]   Zang L, Lou  D, Jiao H, Zang  D, Wang X, Xia Y, et al.
Cocaine-induced intracellular  signaling  and gene
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 300
expression are oppositely regulated by the dopamine D1
and D3 receptors. J Neurosci  2004;24(13):334454.
[367]   Valjent  E, Page` s C, Herve   D, Girault J-A, Caboche  J.
Addictive and non-addictive drugs induce distinct and
specic patterns of ERK activation in mouse  brain. Eur J
Neurosci 2004;19(7):182636.
[368]   Donohue T, Hoffman PL, Tabakoff B. Effect of ethanol on
DARPP-32 phosphorylation  in transgenic mice that
express human  type VII adenylyl cyclase in brain.
Alcohol Clin Exp Res 2005;29(3):3106.
[369]   Zachariou V, Sgambato-Faure V, Sasaki T, Svenningsson
P, Berton O, Fienberg AA, et al. Phosphorylation of
DARPP-32 at threonine-34 is required for  cocaine action.
Neuropsychopharmacology 2006;31(3):55562.
[370]   Rauggi R, Scheggi S, Cassanelli A, Graziella De Montis M,
Tagliamonte A, Gambarana C. The mesolimbic
dopaminergic response  to novel palatable food
consumption increases dopamine-D1 receptor-mediated
signalling with  complex  modications of the DARPP-32
phosphorylation pattern. J Neurochem 2005;92(4):86777.
[371]   Zachariou V, Benoit-Marand  M, Allen PB, Ingrassia P,
Fienberg AA, Gonon F, et al. Reduction of cocaine place
preference in mice lacking  the protein phosphatase 1
inhibitors DARPP-32 or Inhibitor 1. Biol Psychiatry
2002;51(8):61220.
[372]   Haney M, Ward AS, Foltin RW, Fischman MW. Effects of
ecopipam, a selective dopamine D1 antagonist, on
smoked cocaine self-administration  by humans.
Psychopharmacology  2001;155(4):3307.
[373]   Barone  P, Tucci I, Parashos SA, Chase TN.
Supersensitivity to a D-1 dopamine receptor agonist and
subsensitivity to a D-2 receptor agonist following chronic
D-1 receptor  blockade.  Eur J Pharmacol  1988;149:22532.
[374]   Braun  AR, Laruelle M, Mouradian MM. Interactions
between D1 and D2 dopamine receptor family agonists
and antagonists: the effects of chronic exposure on
behavior and receptor binding in rats and their clinical
implications. J Neural  Transm  1997;104:34162.
[375]   DallOlio R, Gandol O, Vaccheri A, Roncada  P,
Mantanaro N. Changes in behavioural responses  to the
combined administration of D1 and D2 dopamine
agonists  in normosensitive and D1 supersensitive rats.
Psychopharmacology  1988;95:3815.
[376]   Vaccheri A, DallOlio  R, Gandol  O, Roncada  P,
Montanaro N. Enhanced  stereotyped response  to
apomorphine after chronic D-1 blockade with SCH 23390.
Psychopharmacology  1987;91:3946.
[377]   Creese  I, Chen A. Selective D-1 dopamine  receptor
increase following  chronic treatment  with SCH. Eur  J
Pharmacol 1985;23390(109):1278.
[378]   Gui-Hua C, Perry BD, Woolverton  WL. Effects of chronic
SCH 23390 or acute EEDQ on the discriminative stimulus
effects of SKF 38393. Pharmacol  Biochem  Behav
1992;41:3217.
[379]   Hess EJ, Albers LF, Le H, Creese I. Effects of chronic SCH
23390 treatment on  the biochemical and behavioral
properties of D1 and D2 dopamine receptors: potentiated
behavioral responses  to a D2 dopamine agonist  after
selective D1 dopamine receptor up-regulation. J
Pharmacol Exp Ther 1986;238:84654.
[380]   White FJ, Joshi A, Koeltzow  TW, Hu X-T. Dopamine
receptor antagonists  fail to prevent induction of cocaine
sensitization. Neuropsychopharmacology  1998;18:2640.
[381]   De Vries TJ, Schoffelmeer  ANM, Binnekade R, Raas  H,
Vanderschuren LJMJ. Relapse to cocaine-  and heroin-
seeking behavior mediated by dopamine d2 receptors is
time-dependent and associated with behavioral
sensitization. Neuropsychopharmacology  2002;26(1):
1826.
[382]   Platt DM, Rodefer JS, Rowlett  JK, Spealman RD.
Suppression of cocaine-  and food-maintained behavior
by the D2-like receptor  partial agonist terguride in
squirrel monkeys.  Psychopharmacology 2003;166(3):
298305.
[383]   Eiler WJA, June HL. Blockade of GABA
A
 receptors within
the extended amygdala attenuates D
2
 regulation of
alcohol-motivated  behaviors  in the ventral tegmental
area  of alcohol-preferring (P) rats. Neuropharmacology
2007;52(8):15709.
[384]   Ga l K, Gyertya n I. Dopamine D3 as well as D2 receptor
ligands attenuate the cue-induced cocaine-seeking  in  a
relapse model in rats. Drug Alcohol Depend
2006;81(1):6370.
[385]   Welter M, Vallone  D, Samad TA, Meziane H, Usiellot A,
Borrelli E. Absence of dopamine D2 receptors unmasks
an inhibitory  control over the brain circuitries activated
by cocaine. In: Proceedings of the National  Academy of
Sciences  of the United  States of America, vol. 104, 16;
2007. p. 68405.
[386]   Matsumoto  I, Wilce PA, Buckley T, Dodd P, Puzke J,
Spanagel R, et al. Ethanol and gene expression in brain.
Alcohol  Clin Exp Res 2001;25:82S6S. Suppl ISBRA.
[387]   Lu RB, Lee JF, Ko HC, Lin WW. Dopamine  D2 receptor
gene (DRD2) is associated with  alcoholism with conduct
disorder. Alcohol  Clin Exp Res 2001;25(2):17784.
[388]   Guardia J, Catafau  AM, Batlle F, Martin JC, Segura L,
Gonzalvo B, et al. Striatal dopaminergic D(2) receptor
density measured by [(123)I]iodobenzamide SPECT in the
prediction of treatment outcome of alcohol-dependent
patients.  Am J Psychiatry  2000;157(1):1279.
[389]   Martin-Soelch C, Chevalley  AF, Kunig  G, Missimer J,
Magyar S, Mino A, et al. Changes in reward-induced brain
activation in opiate addicts. Eur J Neurosci  2001;14:
13608.
[390]   Volkow  ND, Chang L, Wang G-J, Fowler JS, Ding Y-S,
Sedler M, et al. Low level of brain dopamine D2 receptors
in methamphetamine abusers: association with
metabolism in the orbitofrontal cortex. Am J Psychiatry
2001;158:201521.
[391]   Volkow  ND, Fowler JS, Wang G-J. The addicted human
brain  viewed in the light of imaging studies: brain
circuits and treatment strategies. Neuropharmacology
2004;47(1):313.
[392]   Volkow ND, Fowler JS, Wang G-J, Swanson JM. Dopamine
in drug abuse and addiction:  results from imaging
studies  and treatment  implications. Mol Psychiatry
2004;9:55769.
[393]   Wang G-J, Volkow ND, Logan J, Pappas NR, Wong CT, Zhu
W, et al. Brain dopamine and obesity. Lancet  2001;3:
3547.
[394]   Volkow ND, Wang GJ, Fowler JS, Logan J, Gatley SJ, Gifford
A, et al. Prediction of reinforcing responses to
psychostimulants in humans  by brain  dopamine D2
receptor levels. Am J Psychiatry 1999;156:14403.
[395]   Volkow ND, Wang GJ, Fowler JS, Logan J, Gatley SJ, Wong
C, et al. Reinforcing effects of psychostimulants in
humans are associated with increases in brain dopamine
and occupancy  of D(2) receptors. J Pharmacol Exp Ther
1999;291:40915.
[396]   Wang G-J, Volkow  ND, Thanos PK, Fowler JS. Similarity
between obesity and drug addiction  as assessed by
neurofunctional imaging: a concept review. J Addict Dis
2004;23(3):3953.
[397]   Piazza PV, Deminiere J-M, Le Moal M, Simon H. Factors
that predict individual vulnerability to  amphetamine
self-administration. Science 1989;245:15113.
[398]   Piazza PV, Deroche-Gamonent  V, Rouge-Pont  F, Le Moal
M. Vertical shifts in  self-administration  dose-response
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   301
functions  predict a drug-vulnerable phenotype
predisposed to addiction. J Neurosci 2000;20:422632.
[399]   McBride WJ, Chernet E, Dyr W, Lumeng L, Li TK. Densities
of dopamine D2 receptors are reduced in CNS regions of
alcohol-preferring P rats. Alcohol 1993;10:38790.
[400]   McBride WJ, Murphy JM, Gatto GJ, Levy AD, Yoshimoto K,
Lumeng  L, et al. CNS mechanisms  of alcohol self-
administration. Alcohol Alcohol 1993;2:4617.
[401]   Stefanini E, Frau M, Garau  MG, Garau B, Fadda F, Gessa
GL. Alcohol-Preferring rats have fewer dopamine  D2
receptors  in the limbic  system. Alcohol Alcohol
1992;27(2):12730.
[402]   Thanos  PK, Taintor NB, Rivera  SN, Umegaki H,  Ikari H,
Roth G, et al. DRD2 Gene  transfer into the nucleus
accumbens of the alcohol preferring (P) and non
preferring  (NP) rats attenuates alcohol drinking. Alcohol
Clin Exper  Res 2004;28:7208.
[403]   Thanos  PK, Volkow ND, Freimuth P, Umegaki H, Ikari H,
Roth G, et al. Overexpression  of dopamine D2 receptors
reduces alcohol  self-administration.  J Neurochem
2001;78(5):1094103.
[404]   Nader MA, Morgan D, Gage HD, Nader SH, Calhoun  TL,
Buchheimer N, et al. PET imaging of dopamine D2
receptors  during chronic cocaine self-administration in
monkeys.  Nat Neurosci  2006;9(8):10506.
[405]   Souza-Formigoni  MLO, De Lucca EM, Hipo lide DC, Enns
SC, Oliveira MGM, Nobrega JN. Sensitization to ethanols
stimulant  effect is associated with  region-specic
increases in  brain D2 receptor  binding.
Psychopharmacology  1999;146(3):p2627.
[406]   Yoder KK, Kareken DA, Seyoum RA, OConnor SJ, Wang C,
Zheng Q-H, et al. Dopamine  D2 receptor  availability  is
associated with  subjective  responses to alcohol. Alcohol
Clin Exp Res 2005;29(6):96570.
[407]   Volkow ND, Wang GJ, Fowler JS, Logan J, Gatley SJ, Gifford
A, et al. Prediction of reinforcing responses to
psychostimulants in humans by brain  dopamine D2
receptor  levels. Am J Psychiatry  1999;156:14403.
[408]   Volkow ND, Wang GJ, Fowler JS, Thanos  PP, Logan  J,
Gatley SJ, et al. Brain DA D2 receptors predict reinforcing
effects of stimulants  in humans: replication study.
Synapse  2002;46:7982.
[409]   Davidson ES, Finch JF, Schenk S. Variability in subjective
responses  to cocaine: initial experiences of college
students. Addict Behav 1993;18:44553.
[410]   Volkow ND, Wang GJ, Fowler JS, Logan  J, Gatley SJ,
Hitzemann  R, et al. Decreased striataldopaminergic
responsivity  in detoxied cocaine abusers. Nature
1997;386:8303.
[411]   Volkow ND, Wang G-J, Fowler JS, Logan J, Franceschi  D,
Maynard  L, et al. Relationship  between blockade  of
dopamine transporters by oral methylphenidate and the
increases in  extracellular  dopamine:  therapeutic
implications. Synapse 2002;43:1817.
[412]   Munafo`   MR, Surtees PG, Wainwright  NWJ, Brixey RD,
Flint J. The serotonin  transporter  length polymorphism,
neuroticism, and depression: a comprehensive
assessment  of association. Biol  Psychiatry  2005;58(6):
4516.
[413]   Huang S-Y, Lin W-W, Wan F-J, Chang A-J, Ko H-C, Wang
T-J, et al. Monoamine oxidase-A  polymorphisms might
modify the association between the dopamine  D
2
receptor  gene and alcohol dependence. J Psychiatry
Neurosci  2007;32(3):18592.
[414]   Konishi T, Calvillo  M, Leng A-S, Lin K-M, Wan Y-JY.
Polymorphisms of the dopamine  D2 receptor,  serotonin
transporter, and GABA
A
receptor  b
3
 subunit  genes and
alcoholism in  Mexican-Americans.  Alcohol  2004;32(1):
4552.
[415]   Limosin F, Corwood P, Loze JY, Fedeli LP, Hamon M,
Rouillon  F, et al. Male limited association of the
dopamine  receptor D2 gene TaqI a polymorphism and
alcohol dependence. Am J Med Genet 2002;2(4):
3436. 11.
[416]   Connor JP, Young RM, Lawford BR, Ritchie TL, Noble EP.
D(2) dopamine receptor  (DRD2) polymorphism is
associated with  severity of alcohol dependence. Eur
Psychiatry  2002;17:1723.
[417]   Noble EP, Zhang  X, Ritchie TL, Sparkes  S. Haplotypes at
the DRD2 locus and severe alcoholism. Am J Med Genet
2000;96(5):62231.
[418]   Noble EP, Blum K, Khalsa ME, Ritchie T, Montgomery A,
Wood RC, et al. Allelic association of the D2 dopamine
receptor  gene with cocaine  dependence. Drug Alcohol
Depend 1993;33:27185.
[419]   Persico AM, Bird G, Gabbay FH, Uhl GR. D2 dopamine
receptor gene TaqI A1 and B1 restriction fragment length
polymorphisms: enhanced frequencies  in
psychostimulant-preferring polysubstance  abusers. Biol
Psychiatry  1996;40:77684.
[420]   Lerman C, Caporaso NE, Audrain J, Main D, Bowman ED,
Lockshin B, et al. Evidence suggesting the role of specic
genetic factors in cigarette smoking. Health Psychol
1999;18:1420.
[421]   Sabol SZ, Nelson ML, Fisher C, Gunzerath L, Brody CL, Hu
S, et al. A genetic association for cigarette smoking
behavior. Health  Psychol 1999;18 (t):L37.
[422]   Comings DE, Rosenthal  RJ, Lesieur HR, Rugle LJ,
Muhleman  D, Chiu C, et al. A study of the dopamine  D2
receptor  gene in pathological gambling.
Pharmacogenetics 1996;6:22334.
[423]   Comings DE, Gade R, Wu S, Chiu C, Dietz G, Muhleman D,
et al. Studies of the potential role of the dopamine D1
receptor  gene in addictive  behaviors.  Mol Psychiatry
1997;2:4456.
[424]   Comings DE, Gade-Andavolu  R, Gonzalez  N, Wu S,
Muhleman  D, Chen  C, et al. The additive effect of
neurotransmitter genes  in pathological  gambling.  Clin
Genet 2001;60:10716.
[425]   Lerman C, Berrettini  W, Pinto A, Patterson F, Crystal-
Mansour  S, Wileyto EP, et al. Changes in food reward
following  smoking cessation: a pharmacogenetic
investigation.  Psychopharmacology  2004;174(4):5717.
[426]   Epstein LH, Leddy JJ. Food reinforcement. Appetite
2006;46(1):225.
[427]   Pohjalainen  T, Rinne JO, Nagren K, Lehikoinen P, Anttila
K, Sylvalahti EK,  et al. The Al allele of the human D2
dopamine  receptor gene predicts low D2 receptor
availability  in healthy volunteers.  Mol Psychiatry
1998;3:25660.
[428]   Noble EP, Blum K, Ritchie T, Montgomery A, Sheridan PJ.
Allelic association  of the D2 dopamine  receptor gene
with receptor-binding characteristics in alcoholism. Arch
Gen Psychiatry 1991;48:64854.
[429]   Noble EP, Gottschalk LA, Fallon JH, Ritchie TL, Wu JC. D2
dopamine  receptor polymorphism and brain regional
glucose metabolism.  Am J Med Genet  1997;74:1626.
[430]   Noble EP. The D2 dopamine  receptor gene: a review of
association  studies in alcoholism and phenotypes.
Alcohol 1998;16:3345.
[431]   Noble EP. D2 dopamine receptor  gene in psychiatric and
neurologic disorders and its phenotypes. Am J Med Genet
B Neuropsychiatr Genet 2003;116:10325.
[432]   Jonsson EG, Nothen MM, Grunhage F, Farde L, Nakashima
Y, Propping P, et al. Polymorphisms in the dopamine D2
receptor  gene and their relationships  to striatal
dopamine  receptor density of healthy  volunteers.  Mol
Psychiatry  1999;4:2901290.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 302
[433]   Heidbreder CA, Gardner EL, Xi ZX, Thanos  PK, Mugnaini
M, Hagan JJ, et al. The role of central dopamine  D3
receptors in drug addiction: a review of pharmacological
evidence. Brain Res Brain  Res Rev 2005;49(1):77105.
[434]   Pilla M, Perachon S, Sautel F, Garrido F, Mann A,
Wermuth  CG, et al. Selective inhibition  of cocaine-
seeking behaviour by a partial dopamine  D3 receptor
agonist.  Nature 1999;400(6742):3715.
[435]   Wicke  K, Garcia-Ladona  J. The dopamine D3 receptor
partial agonist, BP 897, is an antagonist at human
dopamine D3 receptors and at rat somatodendritic
dopamine D3 receptors. Eur J Pharmacol 2001;424(2):
8590.
[436]   Aston-Jones G, Druhan J. Breaking the chain of addiction.
Nature  1999;400(6742):3178.
[437]   Koob GF, Caine  SB. Cocaine addiction  therapy? Are we
partially there? Nat Med 1999;5(9):9935.
[438]   Childress AR, OBrien CP. Dopamine  receptor  partial
agonists  could address the duality of cocaine craving.
Trends Pharmacol  Sci 2000;21(1):69.
[439]   Duarte C, Lefebvre C, Chaperon2 F, Hamon M, Thie bot M-
H. Effects of a dopamine D3 receptor ligand, BP 897, on
acquisition and expression of food-, morphine-, and
cocaine-induced conditioned place preference, and food-
seeking behavior in  rats. Neuropsychopharmacology
2003;28:190315.
[440]   Garcia-Ladona FJ, Cox BF. BP 897, a selective dopamine D
3
receptor ligand with  therapeutic potential for the
treatment of cocaine-addiction. CNS Drug Rev
2003;9(2):14158.
[441]   Gilbert  JG, Newman AH, Gardner EL, Ashby CR,
Heidbreder CA, Pak AC, et al. Acute administration of SB-
277011A, NGB 2904, or BP 897 inhibits  cocaine cue-
induced reinstatement of drug-seeking behavior in rats:
role of dopamine  D3 receptors. Synapse  2005;57(1):
1728.
[442]   Aujla H, Beninger RJ. Intra-BLA or intra-NAc infusions of
the dopamine  D
3
 receptor partial agonist,  BP 897, block
intra-NAc amphetamine  conditioned  activity.  Behav
Neurosci 2004;118(6):132430.
[443]   Aujla H, Beninger RJ. The dopamine D
3
 receptor-
preferring partial agonist  BP 897 dose-dependently
attenuates the expression of amphetamine-conditioned
place preference  in rats. Behav Pharmacol  2005;16(3):
1816.
[444]   Vengeliene V, Leonardi-Essmann  F, Perreau-Lenz S,
Gebicke-Haerter P, Drescher K, Gross  G, et al. The
dopamine D
3
 receptor plays an essential role in alcohol-
seeking and relapse.  FASEB J 2006;20(13):222333.
[445]   Le Foll B, Schwartz  J-C, SokoloffB P, Foll L, Schwartz J-C,
Sokoloff P. Disruption  of nicotine conditioning by
dopamine D
3
 receptor ligands. Mol Psychiatry 2003;8:
22530.
[446]   Vorel SR, Ashby  Jr CR, Paul  M, Liu X, Hayes R, Hagan JJ,
et al. Dopamine D3  receptor antagonism  inhibits
cocaine-seeking and cocaine-enhanced brain  reward in
rats. J Neurosci  2002;22:9595603.
[447]   Cervo L, Cocco  A, Petrella  C, Heidbreder CA. Selective
antagonism at dopamine  D
3
 receptors attenuates
cocaine-seeking behaviour in the rat. Int J
Neuropsychopharmacol  2007;10(2):16781.
[448]   Xi ZX, Gilbert J, Campos AC, Kline N, Ashby CR, Hagan JJ,
et al. Blockade  of mesolimbic dopamine  D
3
 receptors
inhibits stress-induced reinstatement of cocaine-seeking
in  rats. Psychopharmacology  (Berl) 2004;176(1):5765.
[449]   Xi ZX, Gilbert JG, Pak  AC, Ashby CR, Heidbreder CA,
Gardner EL. Selective dopamine D3 receptor antagonism
by SB-277011A attenuates cocaine reinforcement as
assessed by progressive-ratio and variable-cost-variable-
payoff xed-ratio cocaine self-administration in rats. Eur
J Neurosci 2005;21(12):342738.
[450]   Heidbreder CA, Andreoli M, Marcon C, Hutcheson  DM,
Gardner EL, Ashby CR. Evidence for the role of dopamine
D3 receptors in oral operant  alcohol self-administration
and reinstatement of alcohol-seeking  behavior in mice.
Addict Biol  2007;12(1):3550.
[451]   Thanos PK, Katana JM, Ashby CR, Michaelides M, Gardner
EL, Heidbreder CA,  et al. The selective dopamine D3
receptor antagonist  SB-277011-A attenuates ethanol
consumption in ethanol preferring (P) and non-preferring
(NP)  rats. Pharmacol Biochem Behav 2005;81(1):
1907.
[452]   Ross JT, Corrigall WA, Heidbreder CA, LeSage MG. Effects
of the selective dopamine D3 receptor antagonist  SB-
277011A  on the reinforcing  effects of nicotine  as
measured by a progressive-ratio  schedule in rats. Eur J
Pharmacol 2007;559(2/3):1739.
[453]   Andreoli M, Tessari M, Pilla M, Valerio E, Hagan JJ,
Heidbreder CA. Selective  antagonism  at dopamine  D3
receptors prevents nicotine-triggered relapse to nicotine-
seeking behavior. Neuropsychopharmacology
2003;28(7):127280.
[454]   Pak AC, Ashby CR, Heidbreder CA, Pilla M, Gilbert J, Xi ZX,
et al. The selective dopamine D3 receptor antagonist SB-
277011A  reduces nicotine-enhanced brain reward and
nicotine-paired environmental cue functions.  Int J
Neuropsychopharmacol 2006;9(5):585602.
[455]   Ashby  CR, Paul M, Gardner EL, Heidbreder CA, Hagan JJ.
Acute administration  of the selective D3 receptor
antagonist SB-277011A blocks the acquisition and
expression of the conditioned place preference response
to heroin  in male rats. Synapse  2003;48(3):1546.
[456]   Schwarz A, Gozzi A, Reese T, Bertani S, Crestan V, Hagan
J, et al. Selective dopamine D(3) receptor antagonist SB-
277011-A potentiates phMRI response to  acute
amphetamine challenge in  the rat brain. Synapse
2004;54(1):110.
[457]   Pritchard  LM, Newman AH, McNamara RK, Logue AD,
Taylor B, Welge JA, et al. The dopamine D3 receptor
antagonist NGB 2904 increases spontaneous  and
amphetamine-stimulated  locomotion.  Biochem  Behav
2007;86(4):71826.
[458]   Boyce-Rustay JM, Risinger FO. Dopamine  D3 receptor
knockout  mice and the motivational effects of ethanol,
pharmacology. Biochem  Behav 2003;75(2):3739.
[459]   Coccaro EF, Siever LJ, Klar HM, et al. Serotonergic studies
in patients with affective and personality disorders. Arch
Gen Psychiatry  1989;46:58799.
[460]   Stein DJ, Hollander  E, Liebowitz MR. Neurobiology  of
impulsivity  and the impulse control  disorders. J
Neuropsychiatry Clin Neurosci  1993;5:917.
[461]   Mandell AJ, Knapp S. Asymmetry and mood, emergent
properties  of serotonin regulation. Arch Gen Psychiatry
1979;36:90916.
[462]   Blundell JE. Serotonin  and appetite. Neuropharmacology
1984;23:153752.
[463]   Sheard  MH, Aghajanian GK.  Stimulation of midbrain
raphe neurons:  Behavioral  effects of serotonin  release.
Life Sci 1968;7:1925.
[464]   Harvey JA, Yunger LM. Relationship  between
telencephalic content of serotonin and pain  sensitivity.
In: Barchas  J, Usdin E, editors. Serotonin and behavior.
New York: Academic  Press; 1973. p. 17989.
[465]   Akil  H, Liebeskind  JC. Monoaminergic  mechanisms  of
stimulation produced analgesia.  Brain  Res 1975;94:
27996.
[466]   Ressler  KJ, Nemeroff CB. Role of serotonergic and
noradrenergic systems in the pathophysiology  of
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   303
depression and anxiety  disorders. Depress Anxiety
2000;12(suppl. 1):219.
[467]   Patterson SL, Abel T, Deuel TA, Martin KC, Rose JC,
Kandel ER. Recombinant BDNF rescues decits in basal
synaptic transmission and hippocampal LTP  in BDNF
knockout  mice. Neuron 1996;16:113745.
[468]   Muller CP, Huston JP. Determining the region-specic
contributions  of 5-HT receptors to the psychostimulant
effects of cocaine. Trends  Pharmacol  Sci 2006;27:10512.
[469]   Pessia M, Jiang Z-G, Alan North R, Johnson SW. Actions of
5-hydroxytryptamine on ventral tegmental area neurons
of the rat in vitro.  Brain Res 1994;654:32430.
[470]   De Deurwaerdere P, Stinus  L, Spampinato U. Opposite
change  of in vivo dopamine release  in the rat nucleus
accumbens and striatum  that follows electrical
stimulation  of dorsal raphe nucleus: role of 5-HT
3
receptors.  J Neurosci  1998;18(16):652838.
[471]   Pallanti S, Bernardi S, Quercioli L, DeCaria C, Hollander E.
Serotonin  dysfunction in pathological  gamblers:
increased prolactin  response to oral  m-CPP versus
placebo. CNS Spectr 2006;11:95664.
[472]   David V, Segu L, Buhot MC, Ichaye M, Cazala P.
Rewarding  effects elicited by cocaine microinjections
into the ventral tegmental area of C57BL/6 mice:
involvement of dopamine  D(1) and serotonin(1B)
receptors.  Psychopharmacology (Berl) 2004;174:36775.
[473]   ODell LE, Parsons  LH. Serotonin1B receptors in the
ventral tegmental area modulate cocaine-induced
increases in  nucleus accumbens  dopamine levels. J
Pharmacol  Exp  Ther 2004;311:7119.
[474]   Yan QS, Zheng SZ, Yan  SE. Involvement  of 5-HT1B
receptors within the ventral tegmental area in regulation
of mesolimbic dopaminergic neuronal  activity via  GABA
mechanisms:  a study with dual-probe microdialysis.
Brain  Res 2004;1021:8291.
[475]   Guan  XM, McBride  WJ. Serotonin  microinfusion into the
ventral tegmental area increases accumbens  dopamine
release.  Brain Res Bull 1989;23:5417.
[476]   Hoplight  BJ, Vincow ES, Neumaier JF. Cocaine  increases
5-HT
1B
 mRNA in  rat nucleus accumbens shell  neurons.
Neuropharmacology  2007;52(2):4449.
[477]   Moore RY, Bloom FE. Central catecholamine neuron
systems: anatomy and physiology of the norepinephrine
and epinephrine  systems. Annu  Rev Neurosci  1979;2:
11368.
[478]   Le AD, Harding S, Juzytsch W, Funk D, Shaham Y. Role of
alpha-2 adrenoceptors in stress-induced reinstatement
of alcohol seeking and alcohol self-administration  in
rats. Psychopharmacology (Berl) 2005;179:36673.
[479]   Drouin C, Darracq L, Trovero F, Blanc G, Glowinski J,
Cotecchia  S, et al. Alpha1b-adrenergic receptors control
locomotor and rewarding effects of psychostimulants
and opiates. J Neurosci 2002;22:287384.
[480]   Rocha BA. Stimulant and reinforcing effects of cocaine in
monoamine transporter knockout mice. Eur J Pharmacol
2003;479:10715.
[481]   Xu F, Gainetdinov RR, Wetsel WC. Mice lacking  the
norepinephrine transporter are supersensitive to
psychostimulants. Nat Neurosci  2000;3(5):46571.
[482]   Hand TH, Stinus  L, Le Moal M. Differential mechanisms
in the acquisition and expression of heroin-induced
place preference. Psychopharmacology  (Berl) 1989;98:
617.
[483]   Zarrindast MR, Bahreini T, Adl M. Effect of imipramine on
the expression and acquisition of morphine-induced
conditioned  place preference  in mice. Pharmacol
Biochem Behav 2002;73:9419.
[484]   Sahraei  H, Ghazzaghi  H, Zarrindast MR, Ghoshooni H,
Sepehri H, Haeri-Rohan A. The role of alpha-
adrenoceptor mechanism(s)  in morphine-induced
conditioned place preference in female mice. Pharmacol
Biochem Behav 2004;78:13541.
[485]   Olson VG, Heusner CL, Bland RJ, During MJ, Weinshenker
D, Palmiter RD. Role of noradrenergic signaling  by the
nucleus tractus solitarius in mediating opiate reward.
Science 2006;311:101720.
[486]   Le AD, Harding S, Juzytsch W, Funk D, Shaham Y. Role of
alpha-2 adrenoceptors in stress-induced  reinstatement
of alcohol seeking and alcohol self-administration in
rats. Psychopharmacology (Berl) 2005;179:36673.
[487]   Erb S, Hitchcott PK, Rajabi H, Mueller D, Shaham Y,
Stewart J. Alpha-2 adrenergic receptor agonists  block
stress-induced  reinstatement of cocaine seeking.
Neuropsychopharmacology  2000;23:13850.
[488]   Leri F, Flores J, Rodaros D, Stewart J. Blockade of stress-
induced  but not cocaine-induced reinstatement  by
infusion of noradrenergic antagonists  into the bed
nucleus of the stria terminalis  or the central nucleus of
the amygdala. J Neurosci 2002;22:57138.
[489]   Shaham Y, Higheld D, Delfs J, Leung S, Stewart J.
Clonidine  blocks stress-induced  reinstatement  of heroin
seeking in rats: an effect independent of locus coeruleus
noradrenergic neurons. Eur J Neurosci 2000;12:292302.
[490]   Wang X, Cen X, Lu L. Noradrenaline in the bed nucleus of
the stria terminalis is critical for stress-induced
reactivation  of morphine-conditioned  place preference
in rats. Eur J Pharmacol  2001;432:15361.
[491]   Davis WM, Smith SG, Khalsa JH. Noradrenergic role in the
self-administration of morphine or amphetamine.
Pharmacol  Biochem Behav 1975;3:47784.
[492]   Lee B, Tiefenbacher S, Platt DM, Spealman  RD.
Pharmacological blockade  of alpha2-adrenoceptors
induces reinstatement of cocaine-seeking  behavior in
squirrel monkeys. Neuropsychopharmacology
2004;29:68693.
[493]   Lu L, Su WJ, Yue W, Ge X, Su F, Pei G, et al. Attenuation of
morphine dependence and withdrawal  in rats by
venlafaxine,  a serotonin  and noradrenaline  reuptake
inhibitor. Life Sci 2001;69:3746.
[494]   Ventura R, Alcaro A, Puglisi-Allegra S. Prefrontal cortical
norepinephrine release is critical  for morphine-induced
reward, reinstatement  and dopamine release in the
nucleus accumbens.  Cereb Cortex 2005;15:187786.
[495]   Ventura R, Cabib S, Alcaro  A, Orsini C, Puglisi-Allegra S.
Norepinephrine  in the prefrontal cortex is critical for
amphetamine-induced reward and mesoaccumbens
dopamine  release. J Neurosci 2003;23:187985.
[496]   Auclair A, Drouin C, Cotecchia S, Glowinski J, Tassin JP. 5-
HT2A and alpha1b-adrenergic receptors entirely mediate
dopamine  release, locomotor  response and behavioural
sensitization  to opiates and psychostimulants. Eur J
Neurosci  2004;20:307384.
[497]   Blanc G, Trovero F, Vezina  P, Herve D, Godeheu AM,
Glowinski J, et al. Blockade of prefronto-cortical alpha 1-
adrenergic  receptors  prevents locomotor  hyperactivity
induced  by subcortical D-amphetamine  injection. Eur J
Neurosci  1994;6:2938.
[498]   Darracq L, Blanc G, Glowinski J, Tassin JP. Importance of
the noradrenaline-dopamine coupling in the locomotor
activating  effects of D-amphetamine. J Neurosci
1998;18:272939.
[499]   Dickinson  SL, Gadie B, Tulloch IF. Alpha 1- and alpha 2-
adrenoreceptor antagonists  differentially inuence
locomotor and stereotyped behaviour induced  by D-
amphetamine  and apomorphine  in the rat
ATPsychopharmacology (Berl)  1988;96:5217.
[500]   Salomon  L, Lanteri C, Glowinski J, Tassin  JP. Behavioral
sensitization  to amphetamine results from an
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 304
uncoupling between noradrenergic and serotonergic
neurons.  Proc Natl Acad Sci USA  2006;103:747681.
[501]   Snoddy AM, Tessel RE. Prazosin: effect on psychomotor-
stimulant cues and locomotor activity  in mice. Eur J
Pharmacol 1985;116:2218.
[502]   Weinshenker D, Miller NS, Blizinsky K, Laughlin  ML,
Palmiter RD. Mice with  chronic norepinephrine
deciency resemble amphetamine-sensitized  animals.
Proc Natl Acad  Sci USA 2002;99:138737.
[503]   Wellman P, Ho D, Cepeda-Benito A, Bellinger L, Nation J.
Cocaine-induced hypophagia and hyperlocomotion in
rats are attenuated  by prazosin. Eur J Pharmacol
2002;455:11726.
[504]   Drouin C, Blanc G, Trovero F, Glowinski J, Tassin  JP.
Cortical alpha 1-adrenergic regulation of acute and
sensitized morphine locomotor effects. NeuroReport
2001;12:34836.
[505]   Estler CJ. Effect of - and -adrenergic  blocking  agents and
para-chlorophenylalanine  on morphine- and caffeine-
stimulated locomotor activity of mice.
Psychopharmacologia  1973;28:2618.
[506]   Ayhan IH, Randrup A. Behavioural  and pharmacological
studies on morphine-induced excitation of rats. Possible
relation to brain catecholamines. Psychopharmacologia
1973;29:31728.
[507]   Mohammed AK, Danysz  W, Ogren SO, Archer T. Central
noradrenaline depletion attenuates amphetamine-
induced locomotor  behavior. Neurosci  Lett 1986;64:
13944.
[508]   Ville gier AS, Drouin C, Bizot JC, Marien M, Glowinski J,
Colpaert F, et al. Stimulation  of postsynaptic alpha1b-
and alpha2-adrenergic  receptors amplies  dopamine-
mediated locomotor activity in both  rats and mice.
Synapse  2003;50:27784.
[509]   Vanderschuren LJ, Beemster P, SchoffelmeerF  A.N.. On
the role of noradrenaline in psychostimulant-induced
psychomotor activity  and sensitization.
Psychopharmacology  (Berl)  2003;169:17685.
[510]   Weinshenker D, Miller NS, Blizinsky K, Laughlin  ML,
Palmiter RD. Mice with  chronic norepinephrine
deciency resemble amphetamine-sensitized  animals.
Proc Natl Acad  Sci USA 2002;99:138737.
[511]   Schank JR, Ventura R, Puglisi-Allegra S, Alcaro A, Cole CD,
Liles LC, et al. Dopamine b-hydroxylase knockout  mice
have alterations in dopamine  signaling  and are
hypersensitive to cocaine. Neuropsychopharmacology
2006;31:222130.
[512]   Weinshenker D, Rust NC, Miller NS, Palmiter RD.
Ethanol-associated  behaviors  of mice lacking
norepinephrine. J Neurosci  2000;20:315764.
[513]   Auclair A, Cotecchia  S, Glowinski J, Tassin JP. D-
Amphetamine fails to increase extracellular dopamine
levels in mice lacking  alpha 1b-adrenergic  receptors:
relationship between functional and nonfunctional
dopamine release.  J Neurosci  2002;22:91504.
[514]   Grenhoff J, Svensson  TH. Prazosin modulates the ring
pattern of dopamine  neurons in rat ventral tegmental
area.  Eur J Pharmacol  1993;233:7984.
[515]   Gresch  PJ, Sved AF, Zigmond MJ, Finlay JM. Local
inuence of endogenous norepinephrine on extracellular
dopamine in rat medial prefrontal cortex. J Neurochem
1995;65:1116.
[516]   Devoto P, Flore G, Saba P, Fa M, Gessa GL. Stimulation of
the locus coeruleus elicits noradrenaline and dopamine
release in the medial prefrontal and parietal cortex. J
Neurochem 2005;92:36874.
[517]   Weinshenker D, Schroeder JP. There and back again: a
tale of norepinephrine and drug addiction.
Neuropsychopharmacology 2007;32:143351.
[518]   George TP, Chawarski MC, Pakes J, Carroll KM, Kosten TR,
Schottenfeld RS. Disulram versus placebo for cocaine
dependence in  buprenorphine-maintained subjects: a
preliminary  trial. Biol Psychiatry 2000;47:10806.
[519]   Petrakis IL, Carroll KM, Nich C, Gordon LT, McCance-Katz
EF, Frankforter T, et al. Disulram treatment for cocaine
dependence in  methadone-maintained opioid addicts.
Addiction 2000;95:21928.
[520]   Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi
J, et al. Efcacy of disulram and cognitive behavior
therapy in cocaine-dependent outpatients: a randomized
placebo-controlled  trial. Arch Gen Psychiatry
2004;61:26472.
[521]   Amit Z, Levitan  DE, Lindros KO. Suppression  of ethanol
intake  following  administration  of dopamine-beta-
hydroxylase inhibitors in rats. Arch Int Pharmacodyn
Ther 1976;223:1149.
[522]   Kreek MJ. Effects of opiates, opioid antagonists and
cocaine  on the endogenous opioid system: clinical  and
laboratory studies. NIDA Res Monogr Ser 1992;119:
448.
[523]   Schulz  R, Wuster  M, Duka T, Herz A. Acute  and chronic
ethanol treatment changes endorphin levels in brain and
pituitary. Psychopharmacology 1980;68:2217.
[524]   Seizinger BR, Bovermann K, Maysinger D, Hollt V, Herz A.
Differential effects of acute and chronic ethanol
treatment on particular opioid peptide systems in
discrete regions of rat brain  and pituitary. Pharmacol
Biochem  Behav 1983;18(suppl):3619.
[525]   Koob  GF, Bloom  FE. Cellular and molecular mechanisms
of drug dependence. Science  1988;242:71523.
[526]   Houdi AA, Bardo MT, Van Loon  GR. Opioid mediation of
cocaine-induced hyperactivity and reinforcement. Brain
Res 1989;497:1958.
[527]   Hollt V, Horn G. Nicotine and opioid peptides, Prog. Brain
Res 1989;79:18793.
[528]   Walters CL, Cleck JN, Kuo Y-c, Blendy  JA. m-Opioid
Receptor  and CREB Activation Are Required  for Nicotine
Reward. Neuron 2005;46:93343.
[529]   Kelley AE,  Bakshi VP, Haber SN, Steininger TL, Will MJ,
Zhang M. Opioid modulation of taste hedonics within the
ventral  striatum.  Physiol Behav 2002;76:36577.
[530]   Dum J, Gramsch C, Herz A. Activation of hypothalamic b-
endorphin pools by reward induced by highly palatable
food. Pharmacol  Biochem Behav 1983;18:4437.
[531]   Kirkham TC, Cooper SJ. Attenuation  of sham feeding
by naloxone  is stereospecic: evidence  for opioid
mediation of orosensory reward. Physiol Behav
1988;43:8457.
[532]   Colantuoni  C, Schwenker J, McCarthy J, Rada P,
Ladenheim B, Cadet  J-L, et al. Excessive  sugar intake
alters binding to dopamine  and mu-opioid receptors in
the brain. Neuroreport 2001;12(16):354952.
[533]   Colantuoni  C, Rada P, McCarthy  J, Patten C, Avena  NM,
Chadeayne A, et al. Evidence that intermittent, excessive
sugar intake  causes endogenous opioid dependence.
Obes  Res 2002;10:47888.
[534]   Szechtman H, Hershkowitz  M, Simantov  R. Sexual
behavior decreases pain sensitivity  and stimulates
endogenous opioids in male rats. Eur J Pharmacol
1981;70:27985.
[535]   Balfour ME, Yu L, Coolen LM. Sexual behavior and sex-
associated environmental  cues activate the mesolimbic
system in male rats. Neuropsychopharmacology
2004;29:71830.
[536]   Zubieta JK, Gorelick  DA, Stauffer R, Ravert HT, Dannals
RF, Frost  JJ. Increased  mu opioid receptor binding
detected by PET in cocaine-dependent men is associated
with  cocaine craving. Nat Med 1996;2:12259.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   305
[537]   Brewerton TD, Lydiard RB, Laraia MT, Shook JE, Ballenger
JC. CSF beta-endorphin and dynorphin in bulimia
nervosa.  Am J Psychiatry 1992;149:108690.
[538]   Nestler EJ, Barrot M, Self DW. Delta FosB: a sustained
molecular switch for  addiction. NAS  2001;98:110426.
[539]   Nestler EJ. Molecular neurobiology of addiction. Am J
Addict 2001;10:20117.
[540]   Nestler EJ. Molecular basis of long-term  plasticity
underlying  addiction. Nat Rev Neurosci  2001;2:11928.
[541]   McClung CA, Ulery PG, Perrotti LI, Zachariou V, Berton O,
Nestler EJ.  DFosB: a molecular switch for long-term
adaptation in the brain. Mol Brain Res 2004;132(2):14654.
[542]   Macdonald  RL, Olsen RW. GABA
A
 receptor channels.
Annu  Rev Neurosci  1994;17:569602.
[543]   Sieghart  W. Structure  and pharmacology of gamma-
aminobutyric acid
A
 receptor  subtypes. Pharmacol  Rev
1995;47:181234.
[544]   Johnston  GAR. GABA
A
 receptor pharmacology.
Pharmacol  Ther 1996;69(3):17398.
[545]   Chen K, Li H-Z, Ye N, Zhang J, Wang J-J. Role of GABAB
receptors  in GABA and baclofen-induced inhibition  of
adult rat cerebellar interpositus nucleus neurons in vitro.
Brain  Res Bull  2005;67:10318.
[546]   Koob GF. A role for GABA  mechanisms  in the
motivational effects of alcohol. Biochem Pharmacol
2004;68(8):151525.
[547]   Kuriyama  K, Hirouchi M, Kimura H. Neurochemical and
molecular pharmacological aspects of the GABA
B
receptor.  Neurochem Res 2000;25:12339.
[548]   Buczek Y, Le   AD, Sellers EM, Tomkins  DM. Effect of
pentylenetetrazole on ethanol  intake,  ethanol kinetics,
and social behavior in male Wistar rats. Alcohol Clin Exp
Res 1998;22:42836.
[549]   Hyytia   P, Koob GF. GABA
A
 receptor antagonism in the
extended  amygdala decreases ethanol self-
administration in  rats. Eur J Pharmacol  1995;283:1519.
[550]   Nowak KL, McBride  WJ, Lumeng  L, Li T-K, Murphy JM.
Blocking  GABA
A
 receptors in the anterior ventral
tegmental area attenuates ethanol intake of the alcohol-
preferring  P rat. Psychopharmacology 1998;139(1/2):
10816.
[551]   Petry NM. Benzodiazepine-GABA modulation  of
concurrent ethanol and sucrose reinforcement in the rat.
Exp Clin Psychopharmacol  1997;5:18394.
[552]   Rassnick S, DAmico E, Riley E, Koob GF. GABA antagonist
and benzodiazepine partial inverse  agonist reduce
motivated  responding for ethanol.  Alcohol Clin Exp Res
1993;17:12430.
[553]   Valles  R, Rocha  A, Cardon  A, Bratton GR, Nation JR. The
effects of the GABA
A
 antagonist  bicuculline on  cocaine
self-administration in rats exposed to  lead during
gestation/lactation, Pharmacology. Biochem  Behav
2005;80(4):6119.
[554]   Ikemoto  S, Kohl RR, McBride  WJ. GABA
A
 receptor
blockade in the anterior ventral tegmental area increases
extracellular  levels of dopamine in the nucleus
accumbens of rats. J Neurochem 1997;69:13743.
[555]   Chester JA, Cunningham CL. GABA
A
 receptors modulate
ethanol-induced conditioned  place preference  and taste
aversion  in mice. Psychopharmacology (Berl)
1999;144:36372.
[556]   Buczek Y, Tomkins DM, Le   AD, Sellers EM. Opposite
effects of Ro 15-4513 on acquisition and maintenance  of
ethanol  drinking behavior in male Wistar rats. Alcohol
Clin Exp Res 1997;21:166775.
[557]   June HL, Greene  TL, Murphy JM, Hite ML, Williams JA,
Cason  CR, et al. Effects of the benzodiazepine inverse
agonist  RO19-4603 alone and in combination  with the
benzodiazepine receptor  antagonists  umazenil, ZK
93426 and CGS 8216, on ethanol  intake  in alcohol-
preferring (P) rats. Brain  Res 1996;734:1934.
[558]   June HL, Eggers MW, Warren-Reese C, DeLong J, Ricks-
Cord A, Durr L, et al. The effects of the novel
benzodiazepine receptor inverse  agonist  Ru 34000 on
ethanol-maintained behaviors.  Eur J Pharmacol
1998;350:1518.
[559]   June HL, Torres L, Cason CR, Hwang BH, Braun  MR,
Murphy  JM. The novel benzodiazepine  inverse agonist
RO19-4603 antagonizes ethanol motivated  behaviors:
neuropharmacological studies. Brain  Res 1998;784:
25675.
[560]   June HL, Zuccarelli D, Torres L, Craig KS, DeLong J, Allen
A, et al. High-afnity benzodiazepine antagonists reduce
responding maintained by ethanol presentation in
ethanol-preferring rats. J Pharmacol  Exp Ther
1998;284:100614.
[561]   June HL, Foster KL, McKay  PF, Seyoum R, Woods II JE,
Harvey SC, et al. The reinforcing properties of alcohol are
mediated by GABA
A1
 receptors in the ventral pallidum.
Neuropsychopharmacology  2003;28(12):212437.
[562]   McBride WJ, Murphy JM, Lumeng L, Li T-K. Effects of Ro
15-4513, uoxetine and desipramine  on the intake of
ethanol, water and food by the alcohol-preferring (P) and
-nonpreferring (NP)  lines of rats. Pharmacol Biochem
Behav 1988;30:104550.
[563]   Petry NM. Ro 15-4513 selectively  attenuates ethanol,  but
not sucrose, reinforced responding in a concurrent
access procedure; comparison to other drugs.
Psychopharmacology (Berl) 1995;121:192203.
[564]   Samson HH, Tolliver GA, Pfeffer AO, Sadeghi KG, Mills FG.
Oral ethanol reinforcement in the rat: effect of the partial
inverse  benzodiazepine agonist  RO15-4513. Pharmacol
Biochem Behav 1987;27:5179.
[565]   Samson HH, Haraguchi  M, Tolliver  GA, Sadeghi KG.
Antagonism  of ethanol-reinforced behavior by the
benzodiazepine inverse agonists  Ro15-4513 and FG7142:
relation to sucrose reinforcement. Pharmacol Biochem
Behav 1989;33:6018.
[566]   Wegelius K, Honkanen A, Korpi ER. Benzodiazepine
receptor  ligands modulate ethanol  drinking in alcohol-
preferring rats. Eur J Pharmacol 1994;263:1417.
[567]   Austin  MC, Kalivas PW. Enkephalinergic  and GABAergic
modulation  of motor activity in the ventral pallidum. J
Pharmacol  Exp Ther 1990;252:13707.
[568]   Hubner CB, Koob GF. The ventral pallidum plays a role in
mediating  cocaine and heroin  self-administration  in the
rat. Brain Res 1990;508:209.
[569]   Hiroi N, White NM. The ventral pallidum area is involved
in the acquisition but  not the expression of the
amphetamine  conditioned  place preference. Neurosci
Lett 1993;156:912.
[570]   Gong W, Neill D, Justice JB. Conditioned place preference
and locomotor activation produced  by injection of
psychomotor  stimulants in the ventral pallidum. Brain
Res 1996;707:6474.
[571]   Gong W, Justice JB, Neill D. Dissociation of locomotor and
conditioned  place preference responses following
manipulation  of GABA-A  and AMPA receptors in ventral
pallidum. Prog Neuropsychopharmacol Biol Psychiatry
1997;21:83952.
[572]   Johnson  PI, Napier TC. Morphine modulation  of GABA
and glutamate-induced changes  of ventral pallidal
neuronal activity.  Neuroscience  1997;77:18797.
[573]   Tindell AJ, Smith KS, Pecin a S, Berridge KC, Aldridge JW.
Ventral pallidum ring codes  hedonic reward: when a
bad taste turns good. J Neurophysiol 2006;96:2399409.
[574]   Gulley JM, Kosobud AEK, Rebec GV. Amphetamine
inhibits  behavior-related  neuronal responses in
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 306
substantia nigra pars reticulata of rats working for
sucrose reinforcement. Neurosci Lett  2002;322:1658.
[575]   Peoples LL, Lynch KG, Lesnock J, Gangadhar N. Accumbal
neural responses during the initiation and maintenance
of intravenous cocaine self-administration. J
Neurophysiol 2004;91:31423.
[576]   Groenewegen HJ, Wright CI, Beijer AV, Voorn  P.
Convergence and segregation of ventral striatal inputs
and outputs.  Ann  NY Acad Sci 1999;877:4963.
[577]   Usuda  I, Tanaka  K, Chiba  T. Efferent projections of the
nucleus accumbens in the rat with special reference to
subdivision of the nucleus: biotinylated dextran amine
study. Brain  Res 1998;797:7393.
[578]   Zahm  DS. The evolving  theory of basal  forebrain
functional-anatomical  macrosystems.,.  Neurosci
Biobehav Rev 2006;30:14872.
[579]   Galaverna OG, Seeley RJ, Berridge KC, Grill HJ, Epstein AN,
Schulkin J. Lesions of the central nucleus of the
amygdala. I. Effects on taste reactivity, taste aversion
learning and sodium appetite. Behav Brain Res
1993;59:117.
[580]   Napier TC. Contribution  of the amygdala and nucleus
accumbens to ventral pallidal  responses to  dopamine
agonists.  Synapse  1992;10:1109.
[581]   Reep  RL, Winans SS. Efferent connections of dorsal and
ventral agranular insular  cortex in the hamster,
Mesocricetus auratus. Neuroscience 1982;7:260935.
[582]   Grove EA. Efferent  connections of the substantia
innominata in the rat. J Comp Neurol 1988;277:34764.
[583]   Grove EA. Neural  associations of the substantia
innominata in the rat: afferent  connections.  J Comp
Neurol 1988;277:31546.
[584]   Kalivas  PW, Churchill L, Romanides A. Involvement  of
the pallidal-thalamocortical circuit in adaptive behavior.
Ann  NY Acad Sci 1999;877:6470.
[585]   Mitrovic  I, Napier TC. Substance P attenuates and
DAMGO potentiates amygdala glutamatergic
neurotransmission within  the ventral pallidum.. Brain
Res 1998;792:193206.
[586]   Napier TC, Mitrovic I. Opioid modulation of ventral
pallidal inputs.  Ann NY  Acad Sci 1999;877:176201.
[587]   Hodge  CW, Cox AA. The discriminative stimulus effects
of ethanol  are mediated by NMDA and GABA
A
 receptors
in  specic limbic brain  regions. Psychopharmacology
1998;139(1/2):95107.
[588]   Tomkins DM, Sellers EM, Fletcher PJ. Median and dorsal
raphe injections  of the 5-HT1A agonist, 8-OH-DPAT, and
the GABAA agonist,  muscimol, increase  voluntary
ethanol intake  in Wistar rats. Neuropharmacology
1994;33:34958.
[589]   Chester, Julia A, Cunningham,  Christopher L. GABA
A
receptor modulation  of the rewarding and aversive
effects of ethanol.  Alcohol 2002;26(3):13143.
[590]   Tyndale  RF, Tomkins  DM. Differences in propensity for
drinking alcohol are reected in subunit- and region-
specic GABA. Addict Biol  1999;4(3):30916.
[591]   Anderson NJ, Daunais JB, Friedman DP, Grant KA, McCool
BA. Long-term ethanol  self-administration  by the
nonhuman primate, macaca fascicularis,  decreases the
benzodiazepine sensitivity  of amygdala GABA
A
receptors. Alcoh  Clin Exp Res 2006;30(12):197885.
[592]   Hemby SE, OConnor  JA, Acosta G, Floyd D, Anderson N,
McCool BA, et al. Ethanol-induced regulation of GABA
A
subunit  mRNAs in prefrontal  elds of cynomolgus
monkeys. Alcohol Clin Exp Res 2006;30(12):197885.
[593]   Sarviharju M, Hyytia  P, Hervonen A, Jaatinen P, Kiianmaa
K, Korpi ER. Lifelong ethanol  consumption  and brain
regional GABA
A
 receptor subunit  mRNA expression in
alcohol-preferring rats. Alcohol 2006;40(3):15966.
[594]   Laviolette SR, Gallegos  RA, Steven  JH, Roger  A, Van Der
Kooy D. Opiate state controls bi-directional reward
signaling  via GABA
A
 receptors in the ventral tegmental
area.  Nat Neurosci 2004;7(2):p1609.
[595]   Bechara  A, Nader K, van der Kooy. A two-separate
motivational systems hypothesis  of opioid addiction.
Pharmacol. Biochem Behav 1998;59:117.
[596]   Nader K, van der Kooy D. Deprivation state switches the
neurobiological substrates  mediating  opiate reward in
the ventral tegmental  area. J Neurosci 1997;17:38390.
[597]   Olmstead MC, Munn EM, Franklin KB, Wise RA. Effects of
pedunculopontine tegmental  nucleus lesions on
responding for intravenous heroin under different
schedules of reinforcement. J Neurosci  1999;18:
503544.
[598]   Dockstader CL, Rubinstein M, Grandy DK, Low MJ, van
der Kooy D. The D2  receptor is critical  in mediating
opiate motivation only in opiate-dependent and
withdrawn mice. Eur J Neurosci 2001;13:9951001.
[599]   Gerrits M, Ramsey NF, Wolternink G, van Ree JM. Lack of
evidence  for an involvement of the nucleus accumbens
dopamine  D1 receptors  in the initiation of heroin self-
administration in the rat. Psychopharmacology
1994;114:48694.
[600]   Laviolette SR, Nader K, van der Kooy D. Motivational state
determines the role of the mesolimbic dopamine system
in the mediation of opiate reward processes. Behav Brain
Res 2002;129:1729.
[601]   Roberts  DCS, Andrews MM, Vickers GJ. Baclofen
attenuates the reinforcing effects of cocaine  in rats.
Neuropsychopharmacology  1996;15:41723.
[602]   Brebner K, Froestl W, Andrews M, Phelan R, Roberts  DC.
The GABA(B) agonist  CGP 44532 decreases cocaine
selfadministration in rats: demonstration using a
progressive ratio and a discrete trials procedure.
Neuropharmacology  1999;38:1797804.
[603]   Brebner K, Phelan R, Roberts  DC. Effect of baclofen on
cocaine  self-administration  in rats reinforced under
xed-ratio 1 and progressive-ratio schedules.
Psychopharmacology  (Berl) 2000;148:31421.
[604]   Brebner K, Phelan R, Roberts  DC. Intra-VTA baclofen
attenuates cocaine self-administration on  a progressive
ratio schedule of reinforcement. Pharmacol  Biochem
Behav  2000;66:85762.
[605]   Xi ZX, Stein EA. Baclofen inhibits  heroin  self-
administration behavior and mesolimbic dopamine
release.  J Pharmacol  Exp  Ther 1999;290:136974.
[606]   Di Ciano P, Everitt BJ. The GABA(B) receptor  agonist
baclofen  attenuates cocaine-  and heroin-seeking
behavior by rats. Neuropsychopharmacology
2003;28:5108.
[607]   Colombo G, Serra S, Brunetti G, Atzori G, Pani M, Vacca G,
et al. The GABA(B)  receptor agonists  baclofen and CGP
44532 prevent acquisition of alcohol drinking behaviour
in alcohol-preferring rats. Alcohol Alcohol 2002;37:
499503.
[608]   Fattore  L, Cossu G, Martellotta  MC, Fratta W. Baclofen
antagonizes intravenous  self-administration  of nicotine
in mice and rats. Alcohol Alcohol 2002;37:4958.
[609]   Paterson  NE, Froestl W, Markou A. The GABA
B
 receptor
agonists baclofen and CGP44532 decreased nicotine self-
administration in the rat. Psychopharmacology  (Berl)
2004;172:17986.
[610]   Paterson  NE, Froestl W, Markou A. Repeated
administration of the GABA
B
receptor  agonist CGP44532
decreased nicotine  self-administration, and acute
administration decreased cue-induced  reinstatement  of
nicotine-seeking in rats. Neuropsychopharmacology
2004;3:11928.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   307
[611]   Brebner K, Ahn S, Phillips AG. Attenuation  of d-
amphetamine self-administration by baclofen in the rat:
behavioral  and neurochemical correlates.
Psychopharmacology  (Berl) 2005;177:40917.
[612]   Campbell UC, Lac ST,  Carroll ME. Effects of baclofen on
maintenance and reinstatement of intravenous  cocaine
selfadministration in rats. Psychopharmacology (Berl)
1999;143:20914.
[613]   Spano  MS, Fattore  L, Fratta W, Fadda P. The GABA
B
receptor  agonist baclofen prevents  heroin-induced
reinstatement  of heroin-seeking  behavior in rats.
Neuropharmacology  2007;52(7):155562.
[614]   Di Ciano P, Everitt BJ. The GABA
B
 receptor agonist
baclofen attenuates cocaine-and  heroin-seeking
behavior by rats. Neuropsychopharmacology
2003;28(3):5108.
[615]   Dobrovitsky V, Pimentel P, Duarte A, Froestl W, Stellar JR,
Trzcin ska M. CGP 44532 a GABA
B
 receptor agonist,  is
hedonically neutral and reduces cocaine-induced
enhancement of reward. Neuropharmacology
2002;42(5):62632.
[616]   Liang J-H, Chen  F, Krstew E, Cowen MS, Carroll  FY,
Crawford  D, et al. The GABA
B
 receptor  allosteric
modulator CGP7930  like baclofen, reduces operant self-
administration of ethanol  in alcohol-preferring rats.
Neuropharmacology  2006;50(5):6329.
[617]   Ling W, Shoptaw  S, Majewska D. Baclofen as a cocaine
anticraving  medication:  a preliminary clinical  study.
Neuropsychopharmacology  1998;18:4034.
[618]   Addolorato G, Caputo F, Capristo E, Domenicali M,
Bernard M, Janiri  L, et al. Baclofen  efcacy in reducing
alcohol craving and intake:  a preliminary double blind
randomized  controlled  study. Alcohol Alcohol
2002;37:5048.
[619]   Shoptaw  S, Yang X, Rotheram-Fuller EJ, Hsieh YC,
Kintaudi PC, Charuvastra VC, et al. Randomized placebo-
controlled  trial of baclofen  for cocaine dependence:
preliminary effects for individuals with chronic patterns
of cocaine use. J Clin Psychiatry 2003;64:14408.
[620]   Childress AR. Subjective and brain responses during cue-
induced craving: evidence for an affective memory state.
Marseilles: EBPS/EBBS; 2001.
[621]   Cousins CS, Roberts DCS. Harriet de Wit GABAB receptor
agonists for the treatment of drug addiction: a review of
recent  ndings. Drug Alcohol Depend 2002;65:20920.
[622]   Bardo MT. Neuropharmacological  mechanisms  of drug
reward: beyond  dopamine in the nucleus accumbens.
Crit Rev Neurobiol  1998;12(12):3767.
[623]   Bartholini  G. GABA receptor agonists:  pharmacological
spectrum and therapeutic  actions.  Med Res Rev
1985;5(1):5575.
[624]   Gong W, Neill DB, Justice JBJ. GABAergic modulation  of
ventral pallidal dopamine release  studied by in vivo
microdialysis in the freely  moving rat. Synapse
1998;29(4):40612.
[625]   Pinnock  RD. Hyperpolarizing  action of baclofen on
neurons  in the rat substantia nigra slice. Brain  Res
1984;332:33740.
[626]   Lacey  MG, Mercuri NB, North RA. On the potassium
conductance increase activated by GABA-B  and
dopamine D-2 receptors in rat substantia nigra neurones.
J Physiol 1988;401:40135.
[627]   Macey DJ, Froestl W, Koob GF, Markou A. Both GABA-B
receptor  agonist and antagonists decreased brain
stimulation  reward in the rat. Neuropharmacology
2001;40:67685.
[628]   Chhatwal JP, Ressler KJ. Modulation of fear and anxiety
by the endogenous cannabinoid system. CNS Spectr
2007;12:21120.
[629]   Mackie K. Mechanisms of CB1 receptor signaling:
endocannabinoid modulation  of synaptic strength. Int J
Obes (Lond) 2006;30(suppl. 1):S1923.
[630]   Iversen L. Pharmacology. Endogenous cannabinoids.
Nature 1994;372:619.
[631]   Iversen L. Cannabis and the brain. Brain  2003;126:
125270.
[632]   Di Marzo V, Matias  I. Endocannabinoid control  of food
intake  and energy balance. Nat Neurosci  2005;8:5859.
[633]   Lupica CR, Riegel AC. Endocannabinoid release from
midbrain  dopamine  neurons: a potential substrate  for
cannabinoid receptor antagonist  treatment  of addiction.
Neuropharmacology 2005;48:110516.
[634]   Mascia MS, Obinu MC, Ledent C, Parmentier M, Bohme
GA, Imperato A, et al. Lack of morphine-induced
dopamine  release in the nucleus accumbens  of
cannabinoid CB(1)  receptor knockout  mice. Eur J
Pharmacol  1999;383:R12.
[635]   Hungund BL, Szakall I, Adam A, Basavarajappa BS,
Vadasz  C. Cannabinoid  CB1 receptor knockout  mice
exhibit  markedly reduced voluntary alcohol
consumption and lack alcohol-induced dopamine release
in the nucleus accumbens. J Neurochem 2003;84:698704.
[636]   Cheer JF, Wassum KM, Sombers LA, Heien MLAV,
Ariansen JL, Aragona BJ, et al. Phasic dopamine release
evoked by abused substances requires cannabinoid
receptor  activation. J Neurosci  2007;27(4):7915. January
24.
[637]   Nestler EJ. Molecular mechanisms  of drug addiction.
Neuropharmacology 2004;47(suppl. 1):2432.
[638]   Konradi C, Cole RL, Heckers S, Hyman SE. Amphetamine
regulates gene expression in rat striatum  via
transcription  factor CREB. J Neurosci  1994;14:562334.
[639]   Cole RL, Konradi C, Douglass J, Hyman SE. Neuronal
adaptation  to amphetamine  and dopamine:  molecular
mechanisms  of prodynorphin  gene regulation in rat
striatum.  Neuron 1995;14:81323.
[640]   Olson VG, Zabetian CP, Bolanos CA, Edwards S, Barrot M,
et al. Regulation of drug reward by CREB: evidence for
two functionally distinct subregions  of the ventral
tegmental  area. J Neurosci  2005;25:555362.
[641]   Shaw-Lutchman TZ, Barrot M, Wallace T, Gilden  L,
Zachariou V, et al. Regional and cellular mapping of CRE-
mediated transcription during naltrexone-precipitated
morphine withdrawal.  J Neurosci  2002;22:366372.
[642]   Shaw-Lutchman TZ, Impey S, Storm D, Nestler  EJ.
Regulation of CRE-mediated transcription in mouse brain
by amphetamine.  Synapse  2003;48:107.
[643]   Walters CL, Kuo YC, Blendy JA. Differential distribution of
CREB in the mesolimbic dopamine  reward pathway. J
Neurochem 2003;87:123744.
[644]   Hyman SE, Malenka  RC, Nestler EJ. Neural mechanisms
of addiction: the role of reward-related learning  and
memory. Annu Rev Neurosci  2006;29:56598.
[645]   Dong Y, Green T, Saal D, Marie H, Neve R, Nestler EJ, et al.
CREB modulates excitability of nucleus accumbens
neurons.  Nat Neurosci  2006;9:4757.
[646]   Carlezon Jr WA, Thome J, Olson VG, Lane-Ladd SB,
Brodkin ES, et al. Regulation of cocaine  reward by CREB.
Science 1998;282:22725.
[647]   Barrot M, Olivier  JDA, Perrotti LI, Ralph JD, Olivier  B,
Amelia JE, et al. CREB activity in the nucleus accumbens
shell controls  gating of behavioral  responses to
emotional  stimuli.  Proc Natl Acad Sci USA 2002;99:
1143540.
[648]   Shippenberg TS,  Rea W. Sensitization to the behavioral
effects of cocaine: modulation  by dynorphin and
kappaopioid  receptor agonists.  Pharmacol  Biochem
Behav 1997;57:44955.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 308
[649]   Pliakas  AM, Carlson RR, Neve RL, Konradi C, Nestler EJ,
Carlezon Jr WA. Altered responsiveness to cocaine  and
increased immobility in the forced swim test associated
with  elevated CREB expression in the nucleus
accumbens. J Neurosci  2001;21:7397403.
[650]   Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ,
Monteggia LM. Neurobiology of depression. Neuron
2002;34:1325.
[651]   Hope BT, Nye HE, Kelz MB, Self DW, Iadarola  MJ,
Nakabeppu Y, et al. Induction of a long-lasting AP-1
complex  composed of altered Fos-like proteins in brain
by chronic cocaine and other chronic treatments. Neuron
1994;13:123544.
[652]   Moratalla R, Elibol B, Vallejo M, Graybiel AM. Network-
level changes in expression of inducible Fos-Jun proteins
in  the striatum  during chronic cocaine treatment and
withdrawal. Neuron 1996;17:14756.
[653]   Nye HE, Nestler EJ. Induction of chronic Fras (Fos-related
antigens) in rat brain by chronic morphine
administration. Mol Pharmacol  1996;49:63645.
[654]   Nye H, Hope BT, Kelz M, Iadarola  M, Nestler EJ.
Pharmacological  studies of the regulation by cocaine  of
chronic Fra (Fos-related antigen) induction  in the
striatum and nucleus accumbens. J Pharmacol  Exp Ther
1995;275:167180.
[655]   Pich EM, Pagliusi SR, Tessari M, Talabot-Ayer D, hooft van
Huijsduijnen R, Chiamulera C. Common neural
substrates for the addictive  properties of nicotine  and
cocaine. Science 1997;275:836.
[656]   Werme M, Messer C, Olson L, Gilden L, Thore n P, Nestler
EJ, et al.  DFosB regulates  wheel running.  J Neurosci
2002;22:81338.
[657]   Berton O, Sears R, Carle T, Ulery  P, Dileone  R, Barrot M,
et al.  DFosB in the dorsal raphe mediates  stress-induced
transcriptional changes  in substance P neurons, and
modulates behavioral  proles in the learned
helplessness model of depression. Abstr-Soc Neurosci
2003;29:5218.
[658]   Perrotti LI, Hadeishi Y, Barrot M, Duman RS, Nestler EJ.
Induction of  DFosB in reward-related brain structures
after chronic stress. Abstr-Soc  Neurosci  2003;29. p.
112.15.
[659]   McClung CA,  Nestler EJ. Regulation  of gene expression
and cocaine  reward by CREB and  DFosB. Nat Neurosci
2003;6:120815.
[660]   Nestler EJ. The neurobiology of cocaine addiction.  Sci
Pract Perspect 2005;3:410.
[661]   Hemmings  Jr HC, Williams  KR, Konigsberg WH,
Greengard P. DARPP-32, a dopamine- and adenosine 3:5-
monophosphate-regulated neuronal phosphoprotein. I.
Amino acid sequence around  the phosphorylated
threonine. J Biol Chem 1984;259:1448690.
[662]   Nishi A, Snyder GL, Greengard P. Bidirectional regulation
of DARPP-32  phosphorylation  by dopamine.  J Neurosci
1997;17:814755.
[663]   Hemmings  Jr HC, Greengard P, Tung HY, Cohen P.
DARPP-32, a dopamine-regulated neuronal
phosphoprotein, is a potent inhibitor of protein
phosphatase-1. Nature 1984;310:5035.
[664]   ibb JA, Snyder GL, Nishi A, Yan Z, Meijer L, Fienberg AA,
et al. Phosphorylation of DARPP-32  by Cdk5 modulates
dopamine signalling in neurons. Nature 1999;402:66971.
[665]   Nishi A, Bibb JA, Matsuyama S, Hamada M, Higashi  H,
Nairn  AC, et al. Regulation  of DARPP-32
dephosphorylation at PKA- and Cdk5-sites by NMDA and
AMPA receptors: distinct roles of calcineurin and protein
phosphatase-2A. J Neurochem 2002;81:83241.
[666]   Hiroi  N, Fienberg AA, Haile CN, Alburges M, Hanson  GR,
Greengard P, et al. Neuronal and behavioural
abnormalities in striatal function  in DARPP-32-mutant
mice. Eur J Neurosci  1999;11(3):11148.
[667]   Donohue T, Hoffman PL, Tabakoff B. Effect of ethanol on
DARPP-32  phosphorylation  in transgenic mice that
express human type VII adenylyl cyclase  in brain.
Alcohol  Clin Exp Res 2005;29(3):3106.
[668]   Gray TS,  Morley JE. Neuropeptide Y: anatomical
distribution and possible function in mammalian
nervous system. Life Sci 1986;38:389401.
[669]   Schroeder JP, Iller KA, Clyde CW. Neuropeptide-Y Y5
receptors modulate the onset and maintenance  of
operant  ethanol self-administration. Alcohol Clin Exp
Res 2003;27(12):191220.
[670]   Heilig  M, McLeod S, Brot M, Heinrichs SC, Menzaghi F,
Koob  GF, et al. Anxiolytic-like  action  of neuropeptide Y:
mediation by Y1 receptors in amygdala, and dissociation
from food intake effects. Neuropsychopharmacology
1993;8:35763.
[671]   Heilig M, Widerlov E. Neurobiology and clinical aspects of
neuropeptide Y. Crit Rev Neurobiol 1995;9:11536.
[672]   Clark JT, Kalra PS, Crowley WR, Kalra SP. Neuropeptide Y
and human  pancreatic  polypeptide  stimulate feeding
behavior in rats. Endocrinology 1984;115:4279.
[673]   Levine AS, Morley JE. Neuropeptide  Y: a potent  inducer
of consummatory behavior in rats. Peptides 1984;5:
10259.
[674]   Hansel DE, Eipper BA, Ronnett  GV. Neuropeptide Y
functions  as a neuroproliferative factor. Nature
2001;410:9404.
[675]   Hansel DE, Eipper BA, Ronnett GV. Regulation of olfactory
neurogenesis by amidated neuropeptides. J Neurosci Res
2001;66:17.
[676]   Biello SM, Golombek DA, Harrington ME. Neuropeptide Y
and glutamate  block each others phase shifts in the
suprachiasmatic nucleus in vitro. Neuroscience
1997;77:104957.
[677]   Golombek  DA, Biello SM, Rendon  RA, Harrington ME.
Neuropeptide Y phase shifts the circadian  clock in vitro
via  a Y2 receptor. Neuroreport 1996;7:13159.
[678]   Gribkoff VK, Pieschl RL, Wisialowski TA, van den Pol AN,
Yocca FD.  Phase shifting of circadian rhythms  and
depression of neuronal  activity in the rat
suprachiasmatic nucleus by neuropeptide Y: mediation
by different receptor subtypes. J Neurosci 1998;8:301422.
[679]   Harrington ME, Schak KM. Neuropeptide Y phase
advances the in vitro hamster circadian clock during the
subjective day with  no effect on phase during the
subjective night. Can J Physiol  Pharmacol  2000;78:
8792.
[680]   Shi TJ, Cui JG, Meyerson BA, Linderoth  B, Hokfelt T.
Regulation of galanin and neuropeptide Y in dorsal root
ganglia and dorsal horn in rat mononeuropathic models:
possible relation to tactile hypersensitivity. Neuroscience
1999;93:74157.
[681]   Shi TJ, Tandrup T, Bergman E, Xu ZQ, Ulfhake B, Hokfelt
T. Effect of peripheral nerve injury  on dorsal root
ganglion  neurons  in the C57 BL/6J mouse: marked
changes  both in  cell numbers and neuropeptide
expression. Neuroscience  2001;105:24963.
[682]   Kalra SP, Xu B, Dube MG, Moldawer LL, Martin D, Kalra
PS. Leptin and ciliary neurotropic factor (CNTF) inhibit
fasting-induced suppression of luteinizing  hormone
release  in rats: role of neuropeptide Y. Neurosci  Lett
1998;240:459.
[683]   Kasuya E, Mizuno M, Watanabe G, Terasawa E. Effects of
an antisense oligodeoxynucleotide for neuropeptide  Y
mRNA on  in vivo luteinizing hormone-releasing
hormone  release in  ovariectomized  female rhesus
monkeys. Regul Pept 1998;75/76:31925.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   309
[684]   Palmiter  RD, Erickson  JC, Hollopeter G, Baraban  SC,
Schwartz  MW. Life without  neuropeptide Y. Recent Prog
Horm Res 1998;53:16399.
[685]   Gerald C, Walker MW, Criscione L, Gustafson  EL, Batzl-
Hartmann C, Smith KE, et al. A receptor subtype involved
in neuropeptide-Y-induced  food intake.  Nature
1996;382:16871 (6587).
[686]   Badia-Elder NE, Stewart RB, Powrozek TA, Roy KF,
Murphy JM, Li TK. Effect of neuropeptide Y (NPY) on oral
ethanol  intake in  Wistar, alcohol-preferring (P), and -
nonpreferring (NP) rats. Alcohol Clin Exp  Res
2001;25(3):38690.
[687]   Gilpin NW, Stewart RB, Murphy JM, Li TK, Badia-Elder NE.
Neuropeptide Y reduces oral  ethanol intake  in alcohol-
preferring  (P) rats following a period of imposed ethanol
abstinence.  Alcohol  Clin Exp Res 2003;27(5):78794.
[688]   Gilpin NW, Stewart RB, Murphy JM, Badia-Elder NE.
Neuropeptide Y in the paraventricular  nucleus of the
hypothalamus increases ethanol intake in high- and low-
alcohol-drinking rats. Alcohol Clin Exp  Res
2004;28(10):14928.
[689]   Kelley SP, Nannini MA, Bratt AM, Hodge  CW.
Neuropeptide-Y in the paraventricular nucleus increases
ethanol  self-administration.  Peptides 2001;22(3):
51522.
[690]   Ehlers CL, Somes C, Cloutier D. Are some of the effects of
ethanol  mediated through NPY? Psychopharmacology
(Berl) 1998;139(12):13644.
[691]   Schroeder JP, Iller KA, Hodge CW. Neuropeptide-Y Y5
receptors  modulate the onset and maintenance of
operant ethanol self-administration. Alcohol Clin Exp
Res 2003;27(12):191220.
[692]   Schroeder JP, Olive F, Koenig  H, Hodge CW. Intra-
amygdala  infusion of the NPY Y1 receptor antagonist
BIBP 3226 attenuates operant ethanol self-
administration. Alcohol Clin Exp Res 2003;27(12):188491.
[693]   Sparta DR, Fee JR, Hayes DM, Knapp DJ, MacNeil DJ,
Thiele TE. Peripheral central administration of a selective
neuropeptide Y Y1 receptor  antagonist  suppresses
ethanol  intake by C57BL/6J mice. Alcohol Clin Exp  Res
2004;28(9):132430.
[694]   Schroeder JP, Overstreet  DH, Hodge  CW. The
neuropeptide-Y Y5 receptor antagonist L-152,804
decreases alcohol self-administration  in inbred  alcohol-
preferring  (iP) rats. Alcohol 2005;36:17986.
[695]   Thiele TE, Marsh DJ, Ste Marie L, Bernstein IL, Palmiter
RD. Ethanol consumption and resistance are inversely
related to neuropeptide Y levels. Nature
1998;396(6709):3669.
[696]   Rimondini R, Thorsell A, Heilig M. Suppression of ethanol
self-administration by the neuropeptide  Y (NPY) Y2
receptor  antagonist  BIIE0246: evidence for  sensitization
in rats with a history of dependence. Neurosci  Lett
2005;375(2):12933.
[697]   Thorsell  A, Rimondini R, Heilig M. Blockade  of central
neuropeptide Y (NPY) Y2 receptors reduces ethanol  self-
administration in  rats. Neurosci Lett  2002;332(1):1.
[698]   Thiele TE, Koh MT,  Pedrazzini  T. Voluntary alcohol
consumption is controlled  via  the neuropeptide Y Y1
receptor.  J Neurosci  2002;22:RC208.
[699]   Lappalainen J, Kranzler HR, Malison R, Price LH, Van Dyck
C, Rosenheck  RA, et al. A functional neuropeptide Y
Leu7Pro polymorphism associated with alcohol
dependence in a large population sample from the
United  States. Arch  Gen Psychiatry 2002;59:82531.
[700]   Zhu G, Pollak L, Mottagui-Tabar S, Wahlestedt  C,
Taubman J, Virkkunen M, et al. NPY Leu7Pro and alcohol
dependence in Finnish and Swedish populations. Alcohol
Clin Exp Res 2003;27:1924.
[701]   Sahu A, Kalra SP. Neuropeptide  regulation of feeding
behavior: Neuropeptide Y. Trends Endocrinol Metab TEM
1993;4:21724.
[702]   Pu S, Jam MR, Honath  TL, et al. Interactions between
neuropeptide  y and gammaaminobutyric acid in
stimulation  of feeding:  morphological  and
pharmacological analysis. Endocrinology
1999;140(2):93340.
[703]   Kotz CM, Grace MK, Briggs J, Levine AS, Billington CJ.
Effects of opioid antagonists naloxone and naltrexone on
neuropeptide  Y-induced feeding and brown  fat
thermogenesis in the rat. J Clin Invest 1995;96:16370.
[704]   Kiddle JJ, McCreery  HJ, Soles S. Synthesis and binding
afnity of neuropeptide Y at opiate receptors. Bioorg Med
Chem Lett  2003;13:102931.
[705]   Bedecs K, Langel U, Bartfai  T, Wiesenfeld-Hallin Z.
Galanin receptors and their second messengers in  the
lumbar dorsal spinal  cord. Acta Physiol  Scand
1992;144:21320.
[706]   Branchek TA, Smith KE, Gerald C, Walker MW. Trends
Pharmacol  Sci 2000;21:10917.
[707]   Melander T, Hokfelt T, Rokaeus A. Distribution of
galanin-like immunoreactivity in the rat central nervous
system. J Comp  Neurol 1986;248:475517.
[708]   Bartfai T, Langel U, Bedecs K, Andell S, Land T, Gregersen
S, et al. Galanin-receptor  ligand  m40 peptide
distinguishes between putative galanin-receptor
subtypes. Proc Natl Acad Sci  USA 1993;90:1128791.
[709]   Merchenthaler  I, Lopez FJ, Negro-Vilar A. Anatomy and
physiology of central galanin-containing  pathways. Prog
Neurobiol  1993;40:71169.
[710]   Branchek TA, Smith KE, Gerald C, Walker MW. Galanin
receptor subtypes. Trends Pharmacol Sci 2000;21:10917.
[711]   Perez SE, Wynick D, Steiner RA, Mufson EJ. Distribution of
galaninergic immunoreactivity in the brain of the mouse.
J Comp Neurol 2001;434:15885.
[712]   Holmes A, Picciotto  MR. Galanin: a novel therapeutic
target for depression, anxiety disorders and drug
addiction?  CNS Neurol Disord   Drug Targets
2006;5(2):22532.
[713]   Wrenn CC, Crawley JN. Pharmacological evidence
supporting a role for galanin in cognition and affect. Prog
Neuropsychopharmacol Biol Psychiatry 2001;25:28399.
[714]   Papas S, Bourque  CW. Galanin inhibits  continuous  and
phasic ring in  rat hypothalamic  magnocellular
neurosecretory cells. J Neurosci 1997;17:604856.
[715]   Lo pez FJ, Merchenthaler I, Ching  M, Wisniewski MG,
Negro-Vilar A. Galanin: a hypothalamic-
hypophysiotropic hormone modulating  reproductive
functions.  Proc Natl Acad Sci USA 1991;88(10):450812.
[716]   Crawley  JN. The role of galanin in feeding behavior.
Neuropeptides  1999;33:36975.
[717]   Leibowitz SF,  Hoebel BG. Behavioral neuroscience of
obesity.   In: Bray GA, Bouchard C, editors. The handbook
of obesity. 2nd ed., New York: Marcel Dekker Inc.; 2004. p.
30171.
[718]   Rada P, Avena  NM, Leibowitz SF, Hoebel BG. Ethanol
intake  is increased  by injection of galanin in the
paraventricular nucleus and reduced by a galanin
antagonist.  Alcohol 2004;33:917.
[719]   Lewis MJ, Johnson DF, Waldman D, Leibowitz SF, Hoebel
BG. Galanin microinjection in the third ventricle
increases voluntary ethanol intake. Alcohol Clin Exp Res
2004;28(12):18228.
[720]   Przewlocka B, Machelska H, Rekowski P, Kupryszewski G,
Przewlocki R. Intracerebroventricular galanin and N-
terminal  galanin fragment  enhance  the morphine-
induced  analgesia in the rat. J Neural  Transm Gen Sect
1995;102:22935.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 310
[721]   Barton C, York DA, Bray GA. Opioid receptor subtype
control of galanin-induced feeding. Peptides 1996;17:
23740.
[722]   Dube MG, Horvath  TL, Leranth C, Kalra PS, Kalra SP.
Naloxone reduces the feeding  evoked by
intracerebroventricular  galanin injection. Physiol Behav
1994;56:8113.
[723]   Leibowitz SF, Avena NM, Chang G-Q, Karatayeva O, Chau
DT, Hoebel BG. Ethanol intake increases galanin mRNA in
the hypothalamus and withdrawal decreases it. Physiol
Behav 2003;79:10311.
[724]   Rada P, Mark GP, Hoebel BG. Galanin in the
hypothalamus raises dopamine and lowers acetylcholine
release in the nucleus accumbens: a possible mechanism
for hypothalamic initiation of feeding behavior. Brain Res
1998;798:16.
[725]   Leibowitz SF. Hypothalamic galanin in relation to feeding
behavior and endocrine systems.   In: Ho kfelt T, Barfai T,
Jacobowitz D, Ottoson D, editors. Galan: a new
multifunctional peptide in the neuro-endocrine system.
New York: Macmillan;  1991. p. 393406.
[726]   Tempel DL, Leibowitz KJ, Leibowitz SF.  Effects of PVN
galanin on macronutrient  selection.  Peptides 1988;9:
30914.
[727]   Akabayashi A, Zaia CT, Koenig JI, Gabriel SM, Silva I,
Leibowitz SF. Diurnal  rhythm of galanin-like
immunoreactivity  in the paraventricular and
suprachiasmatic  nuclei and other hypothalamic areas.
Peptides 1994;15:143744.
[728]   Leibowitz SF, Akabayashi A, Wang J. Obesity on a
high-fat  diet: role of hypothalamic  galanin in neurons
of the anterior paraventricular nucleus projecting
to  the median eminence. J Neurosci 1998;18:
270919.
[729]   Leibowitz SF. Hypothalamic galanin, dietary fat, and
body fat.   In: Bray GA, Ryan DH, editors. Nutrition,
genetics and obesity. BatonRouge: Louisiana State
University Press; 1999. p. 33881.
[730]   ThieleF T.E., Stewart RB, Badia-Elder NE, Geary N, Massi
M, LeibowitzF S.F.. et al. Overlapping  peptide control of
alcohol self-administration and feeding alcohol. Clin Exp
Res 2004;28(2):28894.
[731]   Li Y, van den Pol  AN. Differential target-dependent
actions of coexpressed  inhibitory  dynorphin and
excitatory hypocretin/orexin neuropeptides. J Neurosci
2006;26(50):130374.
[732]   Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM,
Tanaka H, et al. Orexin and orexin receptors: a family of
hypothalamic  neuropeptides and G protein-coupled
receptors that regulate  feeding behavior. Cell
1998;92:57385.
[733]   Sukurai T, Nagata R, Yamanaka A, Kawamuro H, Tsujino
N, Muraki Y, et al. Input of orexin/hypocretin neurons
revealed by a genetically encoded tracer in mice. Neuron
2005;46:297308.
[734]   Korotkova TM,  Sergeeva OA, Eriksson KS, Haas HL,
Brown RE. Excitation of ventral tegmental area
dopaminergic and nondopaminergic neurons  by orexin/
hypocretins. J Neurosci  2003;23:711.
[735]   Narita M, Naguma Y, Hashimoto  S, Narita M, Khotib J,
Miyataka M, et al. Direct involvement of orexinergic
systems in  the activation  of the mesolimbic dopamine
pathway and related behaviors  induced  by morphine. J
Neurosci 2006;26:398405.
[736]   Zahm  DS. Functional-anatomical implications of the
nucleus accumbens core and shell subterritories. Ann N
Y Acad Sci  1999;877:11328.
[737]   Scammell TE, Saper Orexin CB. drugs and motivated
behaviors. Nat Neurosci 2005;8(10):12868.
[738]   Harris  GC, Wimmer M, Aston-Jones G. A role for  lateral
hypothalamic  orexin neurons in reward seeking. Nature
2005;437:5569.
[739]   Boutrel B, Kenny PJ, Specio SE, Martin-Fardon R, Markou
A, Koob GF, et al. Role for hypocretin in mediating stress-
induced reinstatement  of cocaine-seeking  behavior.
PNAS 2005;102:1916873.
[740]   Sakamoto  F, Yamada S, Ueta Y. Centrally administered
orexin-A activates corticotropin-releasing factor-
containing neurons in the hypothalamic paraventricular
nucleus and central amygdaloid nucleus of rats: possible
involvement of central  orexins on stress-activated
central CRF neurons.  Regul Pept 2004;118:18391.
[741]   Lawrence  AJ, Cowen MS, Yang  H-J, Feng C, Brian O. The
orexin system regulates  alcohol-seeking  in rats. Br J
Pharmacol 2006;148:7529.
[742]   Harris  GC, Aston-Jones G. Arousal and reward: a
dichotomy  in orexin function. Trends  in Neurosci
2006;28(10):5717.
[743]   Overton PG, Clark D. Burst ring in midbrain
dopaminergic  neurons.  Brain Res Brain  Res Rev
1997;25:31234.
[744]   Borgland SL, Taha SA, Sarti F, Fields HL, Bonci A. Orexin A
in the VTA is critical  for the induction  of synaptic
plasticity and behavioral  sensitization  to cocaine.
Neuron 2006;49:589601.
[745]   Komendantov AO, Komendantova OG, Johnson  SW,
Canavier C. A modeling  study suggests complementary
roles for GABAA and NMDA receptors and the SK channel
in regulating the ring pattern in midbrain dopamine
neurons.  J Neurophysiol 2004;91:34657.
[746]   Bonci  A, Malenka RC. Properties  and plasticity of
excitatory synapses on dopaminergic and GABAergic
cells in the ventral tegmental  area. J Neurosci
1999;19:372330.
[747]   Overton PG, Richards CD, Berry MS, Clark  D. Long-term
potentiation at excitatory  amino acid synapses on
midbrain dopamine neurons. Neuroreport 1999;10:2216.
[748]   Gonon FG. Non-linear relationship between impulse ow
and dopamine  released by rat midbrain dopaminergic
neurons  as studied by in vivo electrochemistry.
Neuroscience 1988;24:1928.
[749]   Schultz W. Getting formal with  dopamine reward.
Neuron 2002;36:24163.
[750]   Massi M, Panocka I, De Caro G. The psychopharmacology
of tachykinin  NK-3  receptors in laboratory animals.
Peptides 2000;21:1597609.
[751]   Otsuka M, Yoshioka K. Neurotransmitter functions of
mammalian tachykinins. Physiol Rev 1993;73:229308.
[752]   Sergeyev V, Hokfelt T, Hurd Y. Serotonin and substance P
co-exist  in dorsal raphe neurons  of the human brain.
Neuroreport 1999;10:396770.
[753]   Feuerstein  TJ, Seeger W. Modulation of acetylcholine
release in human cortical slices: possible implications for
Alzheimers disease. Pharmacol  Ther 1997;74:33347.
[754]   Hahn MK, Bannon MJ. Stress-induced c-Fos expression in
the rat locus  coeruleus is dependent on neurokinin 1
receptor activation.  Neuroscience  1999;94:11838.
[755]   Minabe Y, Emori  K, Toor A, Stutzmann  GE, Ashby Jr CR.
The effect of the acute and chronic administration of CP-
96345, a selective neurokinin1  receptor  antagonist,  on
midbrain dopamine  neurons in  the rat: a single unit,
extracellular recording study. Synapse 1996;22(1):
3545.
[756]   Elliott PJ, Nemeroff CB, Kilts CD. Evidence for a tonic
facilitatory inuence of substance P on dopamine release
in the nucleus accumbens.  Brain  Res 1986;385(2):37982.
[757]   Tamiya R, Hanada M, Kawai Y, Inagaki S, Takagi H.
Substance P afferents have synaptic contacts  with
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   311
dopaminergic  neurons in  the ventral tegmental area of
the rat. Neurosci Lett  1990;110:115.
[758]   Brownstein MJ, Mroz EA, Kizer S, Palkovits M, Leeman SE.
Regional distribution of substance P of the rat. Brain Res
1976;116:299305.
[759]   Kanazawa I, Jessel T. Post-mortem changes and regional
distribution of substance P in the rat and mouse nervous
systems. Brain  Res 1976;117:3627.
[760]   Deutch  AY, Maggio JE, Bannon MJ, Kalivas PW, Tam  SE,
Goldstein M, et al. Substance K and substance P
differentially modulate  mesolimbic and mesocortical
systems. Peptides 1985;6:11322.
[761]   Cador M, Rivet JM, Kelley AE, Le Moal M, Stinus L.
Substance P, neurotensin and enkaphalin injections into
the ventral tegmental  area: comparative study on
dopamine turnover in several forebrain structures. Brain
Res 1989;486:35763.
[762]   Barnes JM, Barnes NM, Costall B, Cox AJ, Domeney  AM,
Kelly ME, et al. Neurochemical  consequences following
injection of the substance P analogue, DiMe-C7, into the
rat ventral tegmental  area. Pharmacol Biochem  Behav
1990;37:83941.
[763]   Elliott PJ, Alpert JE, Bannon MJ, Iversen SD. Selective
activation  of mesolimbic and mesocortical dopamine
metabolism in rat brain by infusion of a stable substance
P analogue into the ventral tegmental area. Brain  Res
1986;363:1457.
[764]   Bannon MJ, Elliot PJ, Alpert JE, Goedert M, Iversen SD,
Iversen LL. Role of endogenous substance P in stress-
induced  activation  of mesocortical dopamine neurons.
Nature 1983;306:7912.
[765]   Placenza  FM, Fletcher  PJ, Rotzinger S, Vaccarino  FJ.
Infusion of the substance P analogue, DiMe-C7, into the
ventral-tegmental area induces reinstatement of
cocaine-seeking behaviour in rats. Psychopharmacology
2004;177:11120.
[766]   Placenza  FM, Fletcher  PJ, Vaccarino FJ. Suzanne Erb
Effects of central neurokinin-1  receptor antagonism on
cocaine- and opiate-induced locomotor activity and self-
administration behaviour in rats. Pharmacol  Biochem
Behav 2006;84(1):94101.
[767]   Kelley AE, Delfs JM. Dopamine and conditioned
reinforcement II. Contrasting the effects of amphetamine
microinjection into the nucleus accumbens with peptide
microinjection into the ventral tegmental  area.
Psychopharmacology  1991;103:197203.
[768]   Kelley AE, Cador  M, Stinus L, Le Moal M. Neurotensin,
substance P, neurokinin-alpha, and enkephalin: injection
into ventral tegmental area  in the rat produces
differential effects on operant responding.
Psychopharmacology  1989;97:24352.
[769]   Murtra  P, Sheasby  AM, Hunt SP, De Felipe C. Rewarding
effects of opiates are absent in mice lacking the receptor
for substance P. Nature 2000;405(6783):1803.
[770]   Ripley  L, Gadd  CA, De Felipe C, Hunt SP, Stephens DN.
Lack of self-administration and behavioural sensitisation
to morphine, but  not cocaine, in mice lacking  NK1
receptors.  Neuropharmacology 2002;43(8):125868.
[771]   Gadd CA,  Murtra P, De Felipe C, Hunt SP. Neurokinin-1
receptor-expressing neurons  in the amygdala modulate
morphine reward and anxiety behaviors  in the mouse. J
Neurosci  2003;23(23):827180.
[772]   Placenza FM, Vaccarino FJ, Fletcher PJ, Erb S. Activation of
central neurokinin-1 receptors induces reinstatement of
cocaine-seeking behavior. Neurosci  Lett 2005;390(1):
427.
[773]   Noailles  And P-AH, Angulo JA. Neurokinin receptors
modulate the neurochemical actions of cocaine. Ann NY
Acad Sci  2002;965:26773.
[774]   Loonam TM, Noailles  PAH, Yu J, Zhu  JPQ, Angulo JA.
Angulo substance P and cholecystokinin  regulate
neurochemical responses to cocaine  and
methamphetamine in the striatum.  Life Sci 2003;73:
72739.
[775]   Gonzalez-Nicolini V, McGinty  JF. NK-1 receptor  blockade
decreases amphetamine-induced  behavior and
neuropeptide  mRNA expression in the striatum.  Brain
Res 2002;931(1):419.
[776]   Hadley ME, Haskell-Luevano C. The proopiomelanocortin
system. Ann NY  Acad Sci 1999;885:121.
[777]   Lindblom J, Wikberg JES, Bergstrom L. Alcohol-preferring
AA rats show a derangement in their central
melanocortin  signalling  system. Pharm  Biochem Behav
2002;72:4916.
[778]   Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH,
Fang Q, Berkemeier LR, et al. Targeted disruption of the
melanocortin-4 receptor results in  obesity in mice. Cell
1997;88:13141.
[779]   Kask A, Ra go L, Mutilis F, Pa hkla R, Wikberg JES, Schio th
HB. Selective antagonist  for the melanocortin  4 receptor
(HS014) increases food intake  in free-feeding  rats.
Biochem Biophys Res Commun 1998;245:903.
[780]   Kask A, Ra go L, Wikberg JES, Schio th HB. Differential
effects of melanocortin  peptides on ingestive behaviour
in rats: evidence against the involvement  of MC3
receptor  in the regulation of food intake.  Neurosci  Lett
2000;283:14.
[781]   Hsu R, Taylor JR, Newton SS, Alvaro JD, Haile C, Han G,
et al. Blockade  of melanocortin  transmission inhibits
cocaine reward. Eur J Neurosci 2005;21:223342.
[782]   Ploj K, Roman E, Kask A, Hyytia P, Schioth HB, Wikberg J,
et al. Effects of melanocortin receptor ligands on ethanol
intake  and opioid levels in alcohol-preferring AA rats.
Brain Res Bull 2002;59:97104.
[783]   Alvaro JD, Hsu R, Duman RS. Chronic cocaine
administration  increases the expression of MC4-R  in rat
neostriatum.  J Pharmacol Exp  Ther 2003;304:3919.
[784]   Alvaro J, Tatro JB, Quillan JM, Fogliano M, Eisenhard M,
Lerner MR, et al. Morphine down-regulates
melanocortin-4 receptor expression in brain regions that
mediate opiate addiction. Mol Pharmacol 1996;50:58391.
[785]   Navarro M, Cubero I, chen AS, Chen HY, Knap DJ, Breese
GR, et al. Effects of Melanocortin Receptor Activation and
Blockade  on Ethanol Intake:  A Possible Role for the
Melanocortin-4 Receptor.  Alcohol Clin Exp Res
2005;29(6):94957.
[786]   VanRee JM, Bohus B, Csontos KM, Gispen WH, Greven
HM, Nijkamp FP, et al. Behavioral prole of
n.melanocyte-stimulating  hormone:  relationship  with
ACTH and f3-endorphin action. Life Sci  1981;28:287588.
[787]   Branson R, Potoczna N, Kral JG, Lents  K-U, Hoehe MR,
Horber FF. Binge eating as a major  phenotype of
melanocortin  4 receptor  gene mutations.  N Engl J Med
2003;348:1096103.
[788]   de Vaca SC, Kim G-Y, Carr KD. The melanocortin receptor
agonist  MTII augments the rewarding effect of
amphetamine in ad-libitum-fed and food-restricted rats.
Psychopharmacology 2002;161:7785.
[789]   Cone RD, Cowley MA, Butler AA, Fan W, Marks DL, Low
MJ. The arcuate nucleus as a conduit  for diverse signals
relevant to energy homeostasis. Int J Obesity Relat Metab
Disord 2001;25(Suppl.  5):S637.
[790]   Hakansson ML, Brown H, Ghilardi N, Skoda RC, Meister B.
Leptin receptor  immunoreactivity in chemically  dened
target neurons  of the hypothalamus. J Neurosci
1998;18(1):55972.
[791]   Fulton S, Woodside B, Shizgal P. Modulation of brain
reward ciorcuitry by leptin.  Science 2000;287:1281128.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 312
[792]   Shalev U, Yap J, Shaham Y. Leptin attenuates acute food
deprivation-induced relapse to heroin seeking. J Neurosci
2001;21. RC129-1RC129-5.
[793]   Carr KD. Augmentation of drug reward by chronic food
restriction: behavioral  evidence and underlying
mechanisms. Physiol Behav 2002;76(3):35364.
[794]   Carroll ME, France CP,  Meisch RA. Food deprivation
increases oral and intravenous  drug intake in rats.
Science 1979;205(4403):31921.
[795]   Bell SM, Stewart RB, Thompson SC, Meisch RA. Food-
deprivation increases cocaine-induced conditioned place
preference and locomotor activity  in rats.
Psychopharmacology  1997;131(1):18.
[796]   Fulton S, Woodside B, Shizgal P. Modulation of brain
reward circuitry by leptin.  Science 2000;287:1258.
[797]   Kitai ST, Shepard PD, Callaway  JC, Scroggs R. Afferent
modulation of dopamine  neuron ring patterns.  Curr
Opin Neurobiol  1999;9:6907.
[798]   Jones  DL, Mobley CC. Treatment  of nicotine  addiction.
Tex Dent J 2000;117:2632.
[799]   Fiorillo CD, Williams  JT. Glutamate  mediates an
inhibitory postsynaptic potential in  dopamine neurons.
Nature  1998;394:7882.
[800]   Marino MJ, Wittmann M, Bradley  SR, Hubert  GW, Smith
Y, Conn PJ. Activation of group I metabotropic glutamate
receptors produces a direct excitation and disinhibition
of GABAergic projection neurons in the substantia nigra
pars  reticulata. J Neurosci  2001;21:700112.
[801]   Paladini  CA, Fiorillo CD, Morikawa H, Williams  JT.
Amphetamine selectively blocks inhibitory glutamate
transmission in dopamine neurons.  Nat Neurosci
2001;4:27581.
[802]   Shi WX, Pun CL, Zhang XX, Jones MD, Bunney BS. Dual
effects of D-amphetamine on dopamine neurons
mediated by dopamine and nondopamine  receptors.  J
Neurosci 2000;20:350411.
[803]   Carlos AP, Jennifer MM, John  TW, Gregory PM. Cocaine
self-administration  selectively  decreases noradrenergic
regulation of metabotropic glutamate receptor-mediated
inhibition in dopamine neurons.  J Neurosci
2004;24(22):520915.
[804]   Drouin C, Blanc G, Villegier AS, Glowinski J, Tassin  JP.
Critical role of alpha1-adrenergic  receptors  in acute and
sensitized locomotor effects of D-amphetamine, cocaine
and GBR 12783: inuence  of preexposure conditions and
pharmacological characteristics. Synapse  2002;43:5161.
[805]   Bonci  A, Bernardi G, Grillner P, Mercuri NB. The
dopaminecontaining  neuron: maestro or simple
musician in the orchestra  of addiction? Trends
Pharmacol Sci 2003;24:1727.
[806]   Tremblay LK, Naranjo CA, Graham SJ, Hermann N,
Mayberg  HS, Hevenor S, et al. Functional
neuroanatomical substrates of altered reward processing
in major depressive disorder revealed by a dopaminergic
probe. Arch Gen Psychiatry  2005;62:122836.
[807]   Shmuel A, Yacoub E, Pfeuffer J, Van  de Moortele PF,
Adriany G, Hu X, et al. Sustained negative BOLD, blood
ow and oxygen consumption response and its coupling
to  the positive response in the human brain.  Neuron
2002;36:1195210.
[808]   Hamzei F, Dettmers C, Rzanny  R, Liepert J, Buchel C,
Weiller C. Reduction  of excitability (inhibition) in the
ipsilateral primary motor cortex is mirrored by f MRI
signal decreases. Neuroimage 2002;17:4906.
[809]   Stefanovic B, Warnking JM, Pike GB. Hemodynamic  and
metabolic responses to neuronal inhibition. Neuroimage
2004;22:7718.
[810]   McCullumsmith RE, Meador-Woodruff  JH. Striatal
excitatory amino acid transporter  transcript expression
in schizophrenia, bipolar disorder, and major depressive
disorder. Neuropsychopharmacology  2002;26:36875.
[811]   Goeders NE, Guerin GF. Effects of surgical and
pharmacological adrenalectomy on  the initiation and
maintenance of intravenous cocaine self-administration
in rats. Brain  Res 1996;722:14552.
[812]   Goeders NE, Guerin GF. Role of corticosterone in
intravenous cocaine self-administration  in rats.
Neuroendocrinology 1996;64:33748.
[813]   Deroche V, Marinelli M, Le Moal  M, Piazza  PV.
Glucocorticoids  and behavioral  effects of
psychostimulants. II. cocaine intravenous self-
administration and reinstatement  depend on
glucocorticoid levels. J Pharmacol  Exp Ther
1997;281:14017.
[814]   Piazza PV, Maccari S, Deminiere JM, Le Moal M, Mormede
P, Simon H. Corticosterone  levels determine individual
vulnerability to amphetamine self-administration.  Proc
Natl Acad  Sci USA 1991;88:208892.
[815]   Mantsch JR, Saphier  D, Goeders NE. Corticosterone
facilitates the acquisition of cocaine self-administration
in rats: opposite effects of the type II glucocorticoid
receptor agonist  dexamethasone.  J Pharmacol Exp  Ther
1998;287:7280.
[816]   Marinella M, Piazza  PV. Interaction  between
glucocorticoid hormones, stress and psychostimulant
drugs. Eur J Neurosci  2002;16:38794.
[817]   Francis  DD, Caldji C, Champagne F, Plotsky  PM, Meaney
MJ. The role of corticotropin-releasing factor 
norepinephrine systems in mediating the effects of early
experience on the development of behavioral and
endocrine responses to  stress. Biol  Psychiatry
1999;46(9):115366.
[818]   Gray TS,  Bingaman EW. The amygdala: corticotropin-
releasing  factor, steroids, and stress. Crit Rev Neurobiol
1996;10:15568.
[819]   Lavicky J, Dunn  AJ. Corticotropin-releasing factor
stimulates catecholamine release in hypothalamus and
prefrontal  cortex in freelymoving rats as assessed by
microdialysis. J Neurochem  1993;60:60212.
[820]   Valentino RJ, Foote  SL. Corticotropin-releasing hormone
increases tonic but not sensory-evoked activity of
noradrenergic locus coeruleus neurons in
unanesthetized rats. J Neurosci  1988;8:101625.
[821]   Van  der Kooy D, Koda LY, McGinty JF, Gerfen CR, Bloom
FE. The organization of projections  from the cortex,
amygdala and hypothalamus to  the nucleus of the
solitary tract  in rat. J Comp Neurol 1984;224:124.
[822]   Plotsky PM, Cunningham  ET, Widmaier EP.
Catecholaminergic modulation of corticotropin-releasing
factor and adrenocorticotropin  secretion. Endocr Rev
1989;10:43758.
[823]   Plotsky PM. Pathways to  the secretion  of
adrenocorticotrop: A view from the portal. J
Neuroendocrinol 1991;3:19.
[824]   Piazza PV, Le Moal ML. Pathophysiological  basis of
vulnerability to drug abuse: Role of an interaction
between stress, glucocorticoids,  and dopaminergic
neurons.  Annu Rev Pharmacol  Toxicol 1996;36:35978.
[825]   Kreek MJ, Koob GF. Drug dependence: stress and
dysregulation of brain  reward pathways. Drug Alcohol
Depend 1998;51:2347.
[826]   Shaham Y, Shalev  U, Lu L, De Wit H, Stewart J. The
reinstatement model of drug relapse: history,
methodology, and major ndings. Psychopharm
2003;168:320.
[827]   Aston-Jones G, Harris GC. Brain substrates  for increased
drug seeking during protracted  withdrawal.
Neuropharmacol 2004;47(Suppl.  1):1679.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   313
[828]   Shaham Y, Stewart J. Stress reinstates heroin-seeking in
drug-free animals: an effect mimicking heroin,  not
withdrawal.  Psychopharmacol 1995;119:33441.
[829]   Erb S, Shaham Y, Stewart J. Stress reinstates cocaine-
seeking after prolonged  extinction  and a drug-free
period. Psychopharmacol 1996;128:40812.
[830]   Le AD, Quan B, Juzytch  W, Fletcher  PJ, Joharchi N,
Shaham Y. Reinstatement of alcohol-seeking by priming
injections  of alcohol and exposure to  stress in  rats.
Psychopharmacol 1998;135:16974.
[831]   Ahmed SH, Walker JR, Koob GF. Persistent increase in the
motivation  to take heroin in rats with a history of drug
escalation.  Neuropsychopharmacol  2000;22:41321.
[832]   Martin-Fardon R, Ciccocioppo  R, Massi M, Weiss F.
Nociceptin prevents stress-induced  ethanol- but not
cocaine-seeking behavior in rats. Neuroreport
2000;11:193943.
[833]   Shaham Y, Funk D, Erb S, Brown TJ, Walker CD, Stewart J.
Corticotropin releasing factor, but not corticosterone, is
involved in stress-induced relapse to heroin-seeking  in
rats. J Neurosci  1997;17:260514.
[834]   Erb S, Shaham Y, Stewart J. The role of corticotropin
releasing factor and corticosterone in stress-induced and
cocaine-induced relapse to cocaine seeking in rats. J
Neurosci  1998;18:552936.
[835]   Lu L, Ceng X, Huang M. Corticotropin  releasing  factor
receptor type I mediates stress-induced relapse to opiate
dependence in rats. Neuroreport 2000;11:23738.
[836]   Lu L, Liu D, Ceng  X. Corticotropin releasing factor
receptor  type I mediates  stress-induced  relapse to
cocaine-conditioned place preference  in rats. Eur  J
Pharmacol  2001;415:2038.
[837]   Erb S, Salmaso N, Rodaros D, Stewart J. A role for the CRF-
containing  pathway from central nucleus of the
amygdala  to bed nucleus of the stria terminalis  in the
stress-induced  reinstatement of cocaine seeking in rats.
Psychopharmacol 2001;158:3605.
[838]   Shaham Y, Erb S, Leung S, Buczek Y, Stewart J. CP-
154,526, a selective, non-peptide  antagonist  of the
corticotropin-releasing factor1 receptor attenuates
stress-induced  relapse  to drug seeking in cocaine- and
heroin-trained  rats. Psychopharmacol (Berl)
1998;137:18490.
[839]   Kathleen  TB, Rajita  S. The neurobiological  effects of
chronic stress. Am J Psychiatry  2005;162:148393.
[840]   Piazza  PV, Le Moal M. The role of stress in drug
self-administration. Trends Pharmacol Sci  1998;19:
6774.
[841]   Cooney  NL, Litt MD, Morse PA, Bauer LO, Gaupp L.
Alcohol cue reactivity,  negative-mood reactivity, and
relapse in treated alcoholic  men. J Abnorm  Psychol
1997;106:24350.
[842]   Sinha R, Talih M, Malison R, Cooney N, Anderson GM,
Kreek MJ. Hypothalamic-pituitary-adrenal axis and
sympatho-adrenomedullary responses  during stress-
induced  and drug cue-induced  cocaine craving states.
Psychopharmacology  (Berl) 2003;170:6272.
[843]   Caldji C, Tannenbaum B, Sharma S, Francis D, Plotsky
PM, Meaney MJ. Maternal care  during infancy regulates
the development  of neural systems mediating  the
expression of behavioral  fearfulness  in adulthood in the
rat. Proc Natl Acad Sci USA 1998;95:533540.
[844]   Heim C, Nemeroff CB. The impact  of early adverse
experiences on  brain systems involved in the
pathophysiology of anxiety and affective disorders. Biol
Psychiatry 1999;46:150922.
[845]   Kalinicheva M, Easterling KW, Plotsky PM, Holtzman SG.
Long-lasting  changes  in stress-induced  corticosterone
response and anxiety-like behaviors as a consequence of
neonatal maternal separation in  LongEvans rats.
Pharmacol  Biochem Behav 2002;73:13140.
[846]   Ladd CO, Huot  RL, Thrivikraman  KV, Nemeroff  CB,
Plotsky  PM. Long-term adaptations  in glucocorticoid
receptor  and mineralocorticoid  receptor  mRNA and
negative feedback on the hypothalamo-pituitary-adrenal
axis following  neonatal maternal separation. Bio
Psychiatry  2004;55:36775.
[847]   Liu D, Caldji C, Sharma S, Plotsky PM, Meaney MJ.
Inuence of neonatal rearing conditions on  stress-
induced  adrenocorticotropin responses and
norepinepherine release in the hypothalamic
paraventricular nucleus. J Neuroendocrinol 2000;12:512.
[848]   Meaney MJ. Maternal care,  gene expression, and the
transmission of individual differences in stress reactivity
across generations. Annu  Rev Neurosci  2001;24:
116192.
[849]   Plotsky  PM, Thrivikraman  KV, Nemeroff CB, Caldji C,
Sharma  S, Meaney MJ. Long-term consequences of
neonatal rearing on central corticotropin-releasing factor
systems in adult male rat offspring.
Neuropsychopharmacology  2005;30:2192204.
[850]   Vazquez  DM, Eskandari R, Phelka  A, Lopez JF. Impact  of
maternal deprivation on brain corticotropin-releasing
hormone circuits: prevention of CRH receptor-2 mRNA
changes  by desipramine treatment.
Neuropsychopharmacology  2003;28:898909.
[851]   Meaney MJ, Brake  W, Gratton  A. Environmental
regulation of the development  of mesolimbic dopamine
systems: a neurobiological  mechanism  for vulnerability
to drug abuse? Psychoneuroendocrinology 2002;27:
12738.
[852]   Higley JD, Thompson  WW, Champoux  M, Goldman D,
Hasert MF, Kraemer GW, et al. Paternal  and maternal
genetic and environmental contributions  to
cerebrospinal  uid monoamine  metabolites  in rhesus
monkeys  (Macaca  mulatta). Arch Gen Psychiatry
1993;50:61523.
[853]   Plotsky  PM, Owens MH, Nemeroff CB. Neuropeptide
alterations  in affective disorders.   In: Bloom FE, Kupfer
DJ, editors. Psychopharmacology: the fourth generation
of progress. New York: Raven Press; 1995. p. 97181.
[854]   Arborelius L, Owens  MJ, Plotsky  PM, Nemeroff CB. The
role of corticotrophin-releasing factor in  depression and
anxiety disorders. J Endocrinol 1999;160:112.
[855]   Sinha R. How does stress increase risk of drug abuse and
relapse? Psychopharmacology  2001;158:34359 [647]
Goeders NE (1997) A neuroendocrine  role in cocaine
reinforcement. Psychoneuroendocrinology 22:237259.
[856]   Goeders NE. The HPA axis and cocaine  reinforcement.
Psychoneuroendocrinology  2002;27:1333.
[857]   Tremblay  LK, Naranjo CA,  Cardenas L, Herrmann  N,
Busto UE. Probing brain reward system function in major
depressive disorder: altered response to
dextroamphetamine. Arch  Gen Psychiatry 2002;59:
40916.
[858]   Bremner  JD, Licinio  J, Darnell A, Krystal JH, Owens  MJ,
Southwick SM, et al. Elevated CSF corticotropin-releasing
factor concentrations in posttraumatic  stress disorder.
Am J Psychiatry 1997;154:6249.
[859]   Baker DG, West SA, Nicholson WE, Ekhator NN, Kasckow
JW, Hill KK, et al. Serial CSF corticotropin-releasing
hormone levels and adrenocortical activity in combat
veterans  with posttraumatic stress disorder. Am J
Psychiatry  1999;156:5858.
[860]   Widerlov E, Bissette G, Nemeroff CB. Monoamine
metabolites.  corticotropin releasing factor and
somatostatin  as CSF markers in depressed patients.  J
Affect Disord 1988;14:99107.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 314
[861]   Nemeroff C, Widerlov E, Bissette G, Walleus H, Karlsson I,
Eklund K, et al. Elevated  concentrations of CSF
corticotropin-releasing factor-like immunoreactivity in
depressed patients. Science  1984;226:13424.
[862]   Plotsky P, Owens M, Nemeroff C.
Psychoneuroendocrinology of depression. Psychiatr Clin
North Am 1998;21:293307.
[863]   Sillaber I, Rammes G, Zimmermann  S, Mahal B,
Zieglgansberger W, Wurst W, et al. Enhanced anddelayed
stress-induced  alcohol drinking in mice lacking
functional CRH1 receptors. Science  2002;296:9313.
[864]   Timpl P, Spanagel R, Sillaber I, Kresse A, Reul JM, Stalla
GK,  et al. Impaired stress response and reduced anxiety
in  mice lacking  a functional corticotropin-releasing
hormone  receptor  1. Nat Genet  1998;19:1626.
[865]   McEwen BS, Weiss JM, Schwartz LS. Selective retention of
corticosterone by limbic structures in rat brain.  Nature
1968;220:9112.
[866]   Sanchez MM, Young LJ, Plotsky PM, Insel TR. Distribution
of corticosteroid receptors in the rhesus brain:  relative
absence of glucocorticoid receptors in the hippocampal
formation. J Neurosci  2000;20:465768.
[867]   Amaral  DG, Price JL, Pitkanen A, Carmichael ST.
Anatomical organization of the primate amygdaloid
complex.   In: Aggleton JP, editor. The amygdala:
neurobiological aspects of emotion, memory, and mental
dysfunction. New York: Wiley-Liss Inc.; 1992. p. 166.
[868]   Halgren  E. Emotional  neurophysiology of the amygdala
within  the context of human  cognition.   In: Aggleton JP,
editor. The amygdala: neurobiological aspects of
emotion, memory, and mental dysfunction. New York:
Wiley-Liss;  1992. p. 191228.
[869]   LeDoux JE. Emotional  memory systems in the brain.
Behav Brain Res 1993;58:6979.
[870]   Harris  GC, Aston-Jones G. Involvement  of D2 dopamine
receptors in the nucleus accumbens  in the opiate
withdrawal syndrome. Nature 1994;371:1557.
[871]   Pacak  K, Palkovits M, Kopin IJ, Goldstein DS. Stress-
induced norepinephrine release in the hypothalamic
paraventricular nucleus and pituitaryadrenocortical
and sympathoadrenal  activity:  in vivo microdialysis
studies.  Front Neuroendocrinol 1995;16:89150.
[872]   Elman I, Lukas SE, Karlsgodt KH, Gasic GP, Breiter HC.
Acute  cortisol administration  triggers craving in
individuals with  cocaine dependence. Psychopharmacol
Bull 2003;37:849.
[873]   Sinha R, Fuse T, Aubin  LR, OMalley SS. Psychological
stress, drug-related cues and cocaine  craving.
Psychopharmacology  (Berl)  2000;152:1408.
[874]   Piazza PV, Le Moal  M. The role of stress in drug
self-administration.  Trends Pharmacol Sci 1998;19:
6774.
[875]   Saal D, Dong Y, Bonci A, Malenka RC. Drugs of abuse and
stress trigger a common synaptic adaptation in
dopamine neurons.  Neuron 2003;37:57782.
[876]   Barrot M, Marinelli M, Abrous DN, Rouge-Pont F, Le Moal
M, Piazza PV. The dopaminergic hyper-responsiveness of
the shell of the nucleus accumbens is hormone
dependent. Eur J Neurosci  2000;12:9739.
[877]   Marinelli M, Aouizerate B, Barrot M, Le Moal M, Piazza PV.
Dopamine-dependent responses to morphine depend on
glucocorticoid receptors.  Proc Natl  Acad Sci USA
1998;95:77427.
[878]   Rouge-Pont  F, Deroche V, Le Moal M, Piazza PV.
Individual differences in stress-induced  dopamine
release in the nucleus accumbens  are inuenced by
corticosterone. Eur J Neurosci  1998;10:39037.
[879]   Deroche V, Caine S, Heyser  C, Polis I, Koob G, Gold L.
Differences in the liability to self-administer intravenous
cocaine  between C57BL/6 and BALB/cByJ mice.
Pharmacol Biochem  Behav 1997;57:42940.
[880]   Kosten  TA, Miserendino MJD, Kehoe P. Enhanced
acquisition of cocaine self-administration in  adult rats
with  neonatal stress experience.. Brain  Res 2000;875:
4450.
[881]   Virkkunen M. Urinary free cortisol secretion in habitually
violent offenders. Acta Psychiatr Scand 1985;72:404.
[882]   Tennes  K, Avitable N, Welles R. Behavioral  correlate of
excreted catecholamines and cortisol  in second-grade
children.  J Am Acad Child Psychiatry 1986;25:76470.
[883]   King RJ, Jones J, Scheur JW, Curtis D, Zarcone VP. Plasma
cortisol correlates of impulsivity and substance abuse.
Person Indiv Diff 1990;11:28791.
[884]   Vanyukov MM, Moss HB, Plail JA, Blackson T, Mezzich AC,
Tarter RE. Antisocial symptoms in  preadolescent  boys
and their parents: associations with cortisol. Psychiatry
Res 1993;46:917.
[885]   Moss HB, Vanyukov MM, Martin CS. Salivary cortisol
responses  and the risk for substance abuse in pre-
pubertal  boys. Biol Psychiatry 1995;38:54755.
[886]   Moss HB, Vanyukov MM, Yao JK, Kirillova  GP. Salivary
cortisol responses  in pre-pubertal  boys: the effects of
parental  substance abuse and association with drug use
behavior during adolescence. Biol Psychiatry
1999;45:12939.
[887]   Wei Q, Lu X-Y, Liu L, Schafer G, Shieh K-R, Burke S, et al.
Glucocorticoid receptor overexpression in forebra: a
mouse model of increased  emotional  lability. PNAS
2004;101:118516.
[888]   Deroche-Gamonet V, Sillaber I, Aouizerate B, Izawa R,
Jaber M, Ghozland S, et al. The glucocorticoid receptor as
a potential target to reduce  cocaine abuse. J Neurosci
2003;23:478590.
[889]   William  RL. Cortisol secretion  patterns in  addiction and
addiction risk. Int J Physiology 2006;59:195202.
[890]   Gold PW, Chrousos GP. Organization of the stress system
and its dysregulation in  melancholic and atypical
depression: high vs low CRH/NE states. Mol Psychiatry
2002;7:25475.
[891]   David AM, Gabe B, David JE, April SG, Angelica H, Shuaike
M, et al. Role of brain  norepinephrine in the behavioral
response  to stress. Prog Neuro-Psychopharmacol  Biol
Psychiatry 2005;29:121424.
[892]   Woodward DJ, Moises HC, Waterhouse BD, Yeh HH,
Cheun  JE. Modulatory  actions of norepinephrine on
neural circuits. Adv Exp Med Biol 1991;287:193208.
[893]   Aston-Jones G, Bloom FE. Norepinephrine-containing
locus  coeruleus neurons  in behaving rats exhibit
pronounced responses to non-noxious  environmental
stimuli. J Neurosci  1981;1:887900.
[894]   Foote SL,  Aston-Jones G, Bloom FE. Impulse activity  of
locus coeruleus neurons in awake rats and monkeys is a
function  of sensory stimulation  and arousal. Proc Natl
Acad Sci USA 1980;77:30337.
[895]   Feenstra MG. Dopamine and noradrenaline release in the
prefrontal  cortex in relation to unconditioned  and
conditioned  stress and reward. Prog Brain  Res
2000;126:13363. climbing bers, Exp Neurol 1977;55:
26988.
[896]   Feenstra MG,  Teske G, Botterblom MH, de Bruin JP.
Dopamine  and noradrenaline release in the prefrontal
cortex  of rats during classical  aversive  and appetitive
conditioning to a contextual stimulus: interference by
novelty  effects. Neurosci  Lett 1999;272:17982.
[897]   Feenstra MG,  Vogel M, Botterblom MH, Joosten RN, de
Bruin JP. Dopamine  and noradrenaline  efux in the rat
prefrontal  cortex after classical  aversive  conditioning  to
an auditory cue. Eur J Neurosci  2001;13:10514.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   315
[898]   Craig WB, Barry  DW. The locus coeruleusnoradrenergic
system: modulation  of behavioral  state and state-
dependent  cognitive processes. Brain Res Rev 2003;42:
3384.
[899]   Higheld D, Yap J, Grimm JW, Shaleve U, Shaham Y.
Repeated lofexidine  treatment  attenuates stress-
induced,  but not drug cues-induced reinstatement of a
heroin-cocaine mixture (speedball) seeking in  rats.
Neuropsychopharmacology  2001;25:32031.
[900]   Drug addiction and allostasis. Koob  GF, Le Moal M,
Schulkin J, editors. Allostasis, homeostasis, and the costs
of physiological  adaptation. New York: Cambridge
University Press; 2004. p. 15063.
[901]   Soloman  L, Lanteri C, Glowinski J, Tassin J-P. Behavioral
sensitization to  amphetamine  results from an
uncoupling  between noradrenergic and serotonergic
neurons.  Proc Nat Acad  Sci USA 2006;103:747681.
[902]   Hariri AR, Holmes A. Genetics of emotional regulation:
the role of the serotonin transporter in neural function.
Trends Cogn Sci 2006;10(4):18291.
[903]   Perez de CI, Ibanez A, Saiz-Ruiz  J, Fernandez-Piqueras J.
Genetic contribution to  pathological  gambling:  possible
association between a functional DNA polymorphism at
the serotonin transporter gene (5-HTTT) and affected
men. Pharmacogenetics 1999;9:397400.
[904]   Neumeister A, Konstantinidis A, Stastny J, Schwarz  M,
Vitouch O, Willeit M, et al. Association between serotonin
transporter gene promoter polymorphism (5HTTLPR) and
behavioral  responses to tryptophan depletion in healthy
women with  and without  family history of depression.
Arch  Gen Psychiatry 2002;59:61320.
[905]   Heinz A, Braus DF, Smolka MN, Wrase J, Pulse I, Herman
D, et al. Amygdala-prefrontal coupling depends on  a
genetic variation of the serotonin  transporter.  Nat
Neurosci  2004;8:201.
[906]   David SP, Murthy NV, Rabiner EA, Munafo MR, Johnstone
EC, Jacob R, et al. A functional genetic variation of the
serotonin  (5-HT) transporter affects 5-HT1A receptor
binding in humans.  J Neurosci  2005;25(10):258690.
[907]   Pezawas L, Meyer-Linderberg A, Drabant EM, Verchinisky
BA, Munoz KE, et al. 5-HTTLPR  polymorphism impacts
human  cingulate-amygdala interactions: a genetic
susceptibility mechanism  for depression. Nat Neurosci
2005;8:82834.
[908]   Jacobs N, Kenis G, Peeters F, Deron C, Vlietinch R, van Os
J. Stress-related negative affectivity and genetically
altered serotonin  transporter  function: evidence  of
synergism  in shaping risk of depression. Arch Gen
Psychiatry 2006;63:98996.
[909]   Serretti  A, Calati R, Mandelli L, De Ronchi  D. Serotonin
transporter gene variants and behavior: a comprehensive
review. Curr Drug Targets 2006;7:165969.
[910]   Frodl T, Schule C, Schmitt G, Born C, Baghai T, Zill P, et al.
Association  of the brain-derived neurotrophic  factor
val66met  polymorphism with  reduced hippocampal
volumes in major  depression. Arch Gen Psychiatry
2007;64:4106.
[911]   Vazquez DM, Lopez JF, vanHoers H, Watson SJ, Levine S.
Maternal deprivation regulates serotonin  1A and 2A
receptors  in the infant rat. Brain  Res 2000;855:
7682.
[912]   Vazquez DM, Eskandari R, Zimmer CA, Levine S, Lopez JF.
Brain  5-HT receptor  system in the stressed infant rat:
implications for vulnerability  to substance abuse.
Psychoneuroendocrinology  2002;27:24572.
[913]   Gartside SE, Johnson  DA, Leitch MM, Troakes C, Ingram
CD. Early life adversity programs changes in central 5-HT
neuronal function in adulthood. Eur J Neurosci
2003;17:24018.
[914]   Arborelius L, Hawks BW, Owens MJ, Plotsky  PM,
Nemeroff CB. Increased responsiveness of presumed
5-HT cells to citalopram in  adult rats subjected
to prolonged  maternal separation  relative to
brief separation.  Psychopharmacology  2004;176:
24855.
[915]   Caspi A, Sugden  K, Moftt  TE, Taylor A, Craig IW,
Harrington HL, et al. Inuence of life stress on
depression: moderation by a polymorphism in the 5-htt
gene. Science  2003;301:3869.
[916]   Barr CS, Newman TK, Lindell S, Shannon  C, Champoux
M, Lesch KP,  et al. Interaction  between serotonin
transporter  gene variation  and rearing condition  in
alcohol preference and consumption in female primates.
Arch Gen Psychiatry  2004;61:114652.
[917]   Barr CS, Newman TK, Lindell S, Shannon C, champoux K,
Lesch KP, et al. Serotonin  transporter  gene variation  is
associated with  alcohol sensitivity  in rhesus macaques
exposed to early-life stress. Alcohol Clin Exp  Res
2003;27(5):8127.
[918]   Covalt J, Tennen  H, Armeli S, Conner TS,  Herman AI,
Cillessen  AHN, et al. Interactive effects of the serotonin
transporter  5-HTTLPR  polymorphism and stressful life
events on college student drinking and drug use. Biol
Psychiatry  2007;61(5):60916.
[919]   Jokela M, Keltikangas-Jarvinen L, Kivimaki M, Puttonen S,
Elovainio  M, Rontu  R, et al. Serotonin receptor 2a gene
and the inuence of childhood  maternal nurturance  on
adulthood depressive  symptoms. Arch Gen Psychiatry
2007;64:35660.
[920]   Mantere  T, Tupala E, Hall H, Sa rkioja T, Ra sa nen P,
Bergstro m K, et al. Serotonin transporter distribution and
density in the cerebral cortex of alcoholic  and
nonalcoholic comparison subjects: a whole-hemisphere
autoradiography study. Am J Psychiatry 2002;159:
599606.
[921]   Pfefferbaum  A, Sullivan EV, Mathalon  DH, Lim KO.
Frontal lobe volume loss observed  with magnetic
resonance imaging  in older chronic alcoholics. Alcohol
Clin Exp Res 1997;21:5219.
[922]   Pfefferbaum  A, Sullivan EV, Rosenbloom MJ, Mathalon
DH, Lim KO. A controlled  study of cortical gray  matter
and ventricular  changes in alcoholic men over a 5-year
interval.  Arch Gen Psychiatry  1998;55:90512.
[923]   Abercrombie  HC, Schaefer SM, Larson CL, Oakes TR,
Lindgren KA, Holden JE, et al. Metabolic rate in the right
amygdala  predicts negative affect in depressed patients.
Neuroreport 1998;9:33017.
[924]   Stutzmann  GE, LeDoux JE. GABAergic antagonists block
the inhibitory effects of serotonin in the lateral
amygdala:  a mechanism  for modulation  of sensory
inputs related  to fear conditioning.  J Neurosci  1999;19.
RC8.
[925]   DAquila  PS, Collu  M, Pani L, Gessa GL, Serra  G.
Antidepressant-like effect of selective D1 receptor
agonists in the behavioural despair model animal model
of depression. Eur J Pharmacol 1994;262:10711.
[926]   Papp M, Klimek V, Willner P. Parallel changes  in
dopamine  D2 receptor binding in limbic  forebrain
associated with  chronic mild stress-induced  anhedonia
and its reversal by imipramine. Psychopharmacology
(Berl) 1994;115:4416.
[927]   Pruessner JC, Champagne  F, Meaney MJ, Dagher A.
Dopamine release in response to a psychological stress in
humans and its relationship to early life maternal care. J
Neurosci  2004;24:282531.
[928]   Boadie WD, Charles  BN. The role of dopamine in  the
pathophysiology of depression. Arch Gen Psychiatry
2007;64:32737.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 316
[929]   Oswald LM, Wong  DF, McCaul  M, Zhou Y, Kuwabara  H,
Choi  L, et al. Relationships among ventral striatal
dopamine release,  cortisol  secretion, and subjective
responses to amphetamine.  Neuropsychopharmacology
2005;30:82132.
[930]   Gobbi G, Bambico FR, Mangieri R, Bortolato M,
Campolongo P, Solinas M, et al. Antidepressant-like
activity and modulation  of brain monoaminergic
transmission by blockade  of anandamide hydrolysis.
Proc Natl Acad  Sci 2005;102:186205.
[931]   Walker  JM, Holmes PV, Crystal JD, Duranti A, Tontini A,
Mor M, et al. An endocannabinoid mechanism for stress-
induced analgesia.  Nature 2005;435:110812.
[932]   Viveros MP, Marco EM, File SE. Endocannabinoid  system
and stress and anxiety responses. Pharmacol  Biochem
Behav 2005;81:33142.
[933]   La Rana A, Calignano A, Giustino M, Tattoli M, Palmery V,
Cuomo. et al. Modulation of anxiety through blockade of
anandamide hydrolysis. Nat Med 2003;9:7681.
[934]   Bortolato M, Campolongo P, Mangieri RA, Scattoni ML,
Frau R, Trezza V, et al. Anxiolytic-like properties of the
anandamide transport  inhibitor AM404.
Neuropsychopharmacology 2006;31:26529.
[935]   Patel S, Hillard CJ. Pharmacological evaluation  of
cannabinoid receptor ligands in  a mouse model of
anxiety: further evidence for an anxiolytic  role for
endogenous cannabinoid signaling. J Pharmacol Exp Ther
2006;318:30411.
[936]   Fride E, Suris R, Weidenfeld J, Mechoulam R. Differential
response to acute and repeated stress in cannabinoid
CB1 receptor knockout  newborn and adult mice. Behav
Pharmacol 2005;16:43140.
[937]   Hill MN, Patel S, Carrier EJ, Rademacher DJ, Ormerod BK,
Hillard CJ, et al. Downregulation  of endocannabinoid
signaling  in the hippocampus  following  chronic
unpredictable stress. Neuropsychopharmacology
2005;30:50815.
[938]   Martn M, Ledent C, Parmentier M, Maldonado R,
Valverde O. Involvement of CB1 cannabinoid receptors in
emotional behaviour. Psychopharmacology
2002;159:37987.
[939]   Uriguen L, Perez-Rial S, Ledent C, Palomo T, Manzanares
J. Impaired action of anxiolytic drugs in mice decient in
cannabinoid CB(1) receptors.  Neuropharmacology
2004;46:96673.
[940]   Haller J, Varga B, Ledent C, Freund TF. CB1 cannabinoid
receptors mediate  anxiolytic  effects: convergent  genetic
and pharmacological evidence  with CB1-specic agents.
Behav Pharmacol  2004;15:299304.
[941]   Gaetani S, Cuomo V, Piomelli D. Anandamide hydrolysis:
a new target for anti-anxiety drugs? Trends Mol Med
2003;9:4748.
[942]   Subhash CP. Anxiety and alcohol abuse disorders: a
common role for CREB and its target, the neuropeptide Y
gene. Trends Pharmacol Sci 2003;24:45660.
[943]   Spanagel R, Montkowski A, Allingham K, Shoaib  M,
Holsboer1 F, Landgraf R. Anxiety: a potential predictor of
vulnerability to the initiation of ethanol self-
administration  in rats. Psychopharmacology
1995;122:36973.
[944]   Stewart RB, Gatto  GJ, Lumeng L, Li T-K, Murphy  JM.
Comparison of alcohol-preferring P and -nonpreferring
NP rats on tests relating to anxiety and on the anxiolytic
effects of ethanol.  Alcohol 1993;10:110.
[945]   Colombo G, Agabio R, Lobina C, Reali R, Zocchi A, Fadda F,
et al. Sardinian alcohol-preferring rats: a genetic animal
model of anxiety. Physiol Behav 1995;57:11815.
[946]   Salimov RM, McBride WJ. Performance in the cross-maze
and slip funnel tests of four pairs of rat lines selectively-
bred for divergent alcohol drinking behavior. Addict Biol
1996;1:27380.
[947]   Ehlers CL, Somes C, Lumeng L, Li TK. Electrophysiological
response  to neuropeptide  Y (NPY): in alcohol-nave
preferring and non-preferring  rats. Pharmacol  Biochem
Behav  1999;63:2919.
[948]   Thiele TE, Willis B, Stadler  J, Reynolds JG, Bernstein IL,
McKnight GS. High ethanol consumption and low
sensitivity to ethanol-induced sedation in protein kinase
A-mutant mice. J Neurosci  2000;20. p. RC75.
[949]   Pandey SC, Roy A, Zhang H. The decreased
phosphorylation  of cyclic adenosine  monophosphate
(cAMP) response element  binding (CREB) protein in the
central amygdala acts as a molecular substrate  for
anxiety  related to ethanol withdrawal in  rats. Alcohol
Clin Exp  Res 2003;27:396409.
[950]   Nestler EJ, Carlezon Jr WA. The mesolimbic dopamine
reward circuit in depression. Biol Psychiatry
2006;59:11519.
[951]   Dudman JT, Eaton ME, Rajadhyaksha A, Macias W, Taher
M, Barczak A, et al. Dopamine D1 receptors mediate CREB
phosphorylation  via phosphorylation  of the NMDA
receptor at Ser897-NR1.  J Neurochem 2003;87:92234.
[952]   Conti AC, Blendy JA. Regulation of antidepressant activity
by cAMP response  element binding proteins. Mol
Neurobiol  2004;30:14355.
[953]   Carlezon Jr WA, Duman RS, Nestler EJ. The many faces of
CREB. Trends  Neurosci  2005;28:43645.
[954]   Dong  Y, Saal D, Marie H, Xu W, Nestler EJ, Malenka  RC.
CREB-mediated regulation of excitability and synaptic
transmission in  nucleus accumbens  neurons. Soc
Neurosci  Abs 2004;577:2.
[955]   Newton  SS, Thome J, Wallace T, Shirayama Y, Dow A,
Schlesinger  L, et al. Inhibition  of CREB or dynorphin in
the nucleus accumbens produces an antidepressant-like
effect. J Neurosci  2002;22:1088390.
[956]   Thoenen  H. Neurotrophins  and neuronal  plasticity.
Science  1995;270:5938.
[957]   Thoenen  H. Neurotrophins  and activity-dependent
plasticity. Prog Brain Res 2000;128:18391.
[958]   Bibel M, Barde YA. Neurotrophins:  key regulators of cell
fate and cell shape in  the vertebrate nervous system.
Genes  Dev 2000;14:291937.
[959]   Poo MM. Neurotrophins as synaptic modulators. Nat Rev
Neurosci  2001;2:2432.
[960]   Carter AR, Chen C, Schwartz PM, Segal RA. Brain-derived
neurotrophic factor modulates cerebellar plasticity and
synaptic ultrastructure. J Neurosci  2002;22:131627.
[961]   Monteggia LM, Barrot M, Powell CM, Berton O, Galanis V,
Gemelli T, et al. Essential role of brain-derived
neurotrophic factor in adult hippocampal function.  Proc
Natl Acad  Sci USA 2004;29:1082732.
[962]   Jiang X, Xu K, Hoberman J, Tian F, Marko AJ, Waheed JF,
et al. BDNF variation and mood disorders: a novel
functional promoter polymorphism and Val 66Met are
associated with anxiety but  have opposing effects.
Neuropsychopharmacology  2005;30:135361.
[963]   Yan Q, Radeke MJ, Matheson CR, Talvenheimo J, Welcher
AA, Feinstein  SC. Immunocytochemical  localization  of
TrkB in the central nervous system of the adult rat. J
Comp  Neurol 1997;378:13557.
[964]   Seroogy KB, Lundgren KH, Tien TM, Guthrie KM, Isackson
PJ, Gall CM. Dopaminergic neurons in rat ventral
midbrain express brain-derived  neurotrophic  factor
and neurotrophin-3 mRNAs. J Comp Neurol 1994;342:
32134.
[965]   Sokoloff P, Guillin O, Diaz J, Carroll P, Griffon N. Brain-
derived neurotrophic factor  controls dopamine D3
receptor expression: implications for
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   317
neurodevelopmental psychiatric disorders. Neurotox Res
2002;4:6718.
[966]   Guillin  O, Diaz  J, Carroll  P, Griffon  N, Schwartz JC,
Sokoloff  P. BDNF controls dopamine  D3 receptor
expression and triggers behavioural sensitisation. Nature
2001;411:869.
[967]   Ivy AS, Rodriguez FG, Garcia C, Chen MJ, Russo-Neustadt
AA. Noradrenergic and serotonergic blockade  inhibits
BDNF mRNA. activation following  exercise and
antidepressant. Pharmacol  Biochem  Behav 2003;75:818.
[968]   Garcia C, Chen MJ, Garza  AA, Cotman CW, Russo-
Neustadt A. The inuence of specic noradrenergic and
serotonergic lesions on the expression of hippocampal
brain-derived neurotrophic  factor transcripts following
voluntary  physical activity.  Neuroscience 2003;119:
72132.
[969]   Mo ssner R, Daniel S, Albert D, Heils A, Okladnova  O,
Schmitt  A, et al. Serotonin  transporter function is
modulated  by brain-derived neurotrophic factor (BDNF)
but not nerve growth factor (NGF). Neurochem Int
2000;36:197202.
[970]   Horger BA, Iyasere CA, Berhow MT, Messer CJ, Nestler EJ,
Taylor JR. Enhancement of locomotor  activity and
conditioned  reward to cocaine  by brain-derived
neurotrophic factor. J Neurosci  1999;19:411022.
[971]   Hall FS, Drgonova J, Goeb M, Uhl GR. Reduced behavioral
effects of cocaine in heterozygous brain-derived
neurotrophic factor (BDNF) knockout  mice.
Neuropsychopharmacology  2003;28:148590.
[972]   Lu L, Dempsey J, Liu SY, Bossert JM, Shaham Y. A single
infusion of brain-derived neurotrophic  factor into the
ventral tegmental area induces long-lasting potentiation
of cocaine seeking after withdrawal.  J Neurosci
2004;24:160411.
[973]   Hensler JG, Ladenheim EE, Lyons  WE. Ethanol
consumption and serotonin-1A  (5-HT1A) receptor
function in heterozygous BDNF (/) mice. J Neurochem
2003;85:113947.
[974]   McGough NN, He DY, Logrip ML, Jeanblanc J, Phamluong
K, Luong K, et al. RACK1 and brain-derived neurotrophic
factor: a homeostatic pathway that regulates  alcohol
addiction.  J Neurosci  2004;24:105425.
[975]   Meredith  GE, Callen  S, Scheurer  DA. Brain  derived
neurotrophic factor expression is increased in the rat
amygdala,  piriform cortex and hypothalamus following
repeated amphetamine administration.. Brain Res
2002;949:21827.
[976]   Numan S, Lane-Ladd SB, Zhang  L, Lundgren KH, Russell
DS, Seroogy KB, et al. Differential regulation of
neurotrophin and trk receptor mRNAs in
catecholaminergic nuclei during chronic opiate
treatment  and withdrawal.  J Neurosci  1998;18:107008.
[977]   Grimm JW, Lu L, Hayashi T, Hope B, Su TP, Shaham Y.
Time dependent increases in brain derived neurotrophic
factor protein levels within the mesolimbic  dopamine
system after withdrawal  from cocaine: implications
for incubation of cocaine craving. J Neurosci  2003;23:
7427.
[978]   Butovsky E, Juknat A, Goncharov I, Elbaz J, Eilam R,
Zangen A, et al. In vivo upregulation  of brain derived
neurotrophicfactor  in specic brain  areas by chronic
exposure to  D9-tetrahydrocannabinol. J Neurochem
2005;93:80211.
[979]   Le Foll B, Frances H, Diaz J, Schwartz JC, Sokoloff P. Role
ofthe dopamine D3 receptor in reactivity  to cocaine-
associated cues in mice. Eur J Neurosci  2004;15:201626.
[980]   Lapchak PA, Hefti F. BDNF and NGF treatment in lesioned
rats: Effects on cholinergic function  and weight gain.
Neuroreport 1992;3:4058.
[981]   Pelleymounter MA, Cullen MJ, Wellman CL.
Characteristics of BDNF-induced weight loss. Exp Neurol
1995;131:22938.
[982]   Pierce RC, Pierce-Bancroft AF, Prasad BM. Neurotrophin-3
contributes to the initiation of behavioral sensitization to
cocaine by activating the Ras/Mitogen-activated  protein
kinase signal transduction cascade.  J Neurosci
1999;19:868595.
[983]   Sauer H, Fischer W, Nikkhah  G, Wiegand SJ, Brundin  P,
Lindsay RM, et al. Brain-derived neurotrophic factor
enhances  function rather than survival of intrastriatal
dopamine  cell-rich grafts. Brain  Res 1993;626:3744.
[984]   Nakazato  M, Hashimoto  K, Shimizu E, Kumakiri C,
Koizumi  H, Okamura N, et al. Decreased  levels of serum
brainderived neurotrophic factor in female patients with
eating disorders. Biol Psychiatry 2003;54:48590.
[985]   Pandey CP, Zang H, Roy A, Mishra K. Central and medial
brain-derived  neurotrophic  factor signaling  plays a
critical  role in alcohol-drinking  and anxiety-like
behaviors.  J Neurosci  2006;26(32):832031.
[986]   Pandey SC, Roy A, Zhang H, Xu T. Partial deletion  of the
cAMP response  element-binding  protein gene promotes
alcohol-drinking  behaviors.  J Neurosci 2004;24:502230.
[987]   Yan Q-S, Feng M-J, Yan SE. Different expression of brain-
derived neurotrophic factor in the nucleus accumbens of
alcohol-preferring (P) and -nonpreferring (NP) rats. Brain
Res 2005;1035:2158.
[988]   Karege F, Perret G, Bondol MS, Bertschy  G, Aubry JM.
Decreased  serum brain-derived  neurotrophic  factor
levels in  major depressed patients.  Psychiatry  Res
2002;109:1438.
[989]   Karege F, Schwald M, Cisse M. Post natal prole of brain-
derived neurotrophic  factor in rat brain and platelets.
Neurosci  Lett 2002;328:2614.
[990]   Shimuzu E, Hashimoto K, Koike K, Komatsu N, Kumakiri
C, Nakazato M, et al. Alterations of serum levels of brain
derived neurotrophic factor (BDNF) in depressed patients
with or without  antidepressants. Biol Psychiatry
2003;54:705.
[991]   Aydemir C, Yalcin  ES, Aksaray  S, Kissa C, Yildirim SG,
Uzbay T, et al. Brain-derived neurotrophic factor (BDNF)
changes  in the serum  of depressed women. Progress in
Neuro-Psychopharmacology  and Biological  Psychiatry
2006;30:125660.
[992]   Kenji H, Eiji S, Masaomi  I. Critical  role of brain-derived
neurotrophic factor  in mood disorders. Brain Res Rev
2004;45:10414.
[993]   Duman RS, Vaidya  VA. Molecular  and cellular actions of
chronic electroconvulsive seizures. J ECT 1998;14:18193.
[994]   Gonul AS, Akdeniz F, Taneli  F, Donat O, Eker C, Vahip S.
Effect of treatment on serum brain-derived neurotrophic
factor levels in depressed patients.  Eur Arch Psychiatry
Clin Neurosci  2005;255:2678.
[995]   Aydemir O, Deveci  A, Taneli  F. The effect  of chronic
antidepressant  treatment  on serum brain-derived
neurotrophic factor  levels in depressed patients:  a
preliminary  study. Prog Neuropsychopharmacol Biol
Psychiatry  2005;29(2):2615.
[996]   Nibuya M, Morinobu  RS, Duman RS. Regulation  of BDNF
and trkB m RNA in rat brain by chronic electroconvulsive
seizure and antidepressant drug treatments. J Neurosci
1995;15:753947.
[997]   Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman
RS. Brain-derived neurotrophic factor produces
antidepressant  effects in behavioral  models of
depression. J Neurosci 2002;22:325161.
[998]   Siuciak J, Lewis DR, Wiegand SJ, Lindsday RM.
Antidepressant  like effect of brain-derived neurotrophic
factor (BDNF).  Pharmacol  Biochem Behav  1997;56:1317.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 318
[999]   Smith MA, Makino S, Kwetnansky R, Post RM. Stress and
glucocorticoids  affects the expression of brain derived
neurotrophic factor and neurotrophin- 3 mRNAs in the
hippocampus. J Neurosci  1995;15:176877.
[1000]   Hoshaw B, Malberg JE, Lucki  I. Central administration  of
IGF-I and BDNF leads to long-lasting antidepressant-like
effects. Brain  Res 2005;1037:2048.
[1001]   Eisch  AJ, Bolan os CA,  de Wit J, Simonak RD, Pudiak CM,
Barrot M, et al. Brain-derived neurotrophic  factor in  the
ventral midbrainnucleus accumbens pathway. A role in
depression. Biol Psychiatry  2003;54:9941005.
[1002]   Vaidya VA, Marek GJ, Aghajanian GK, Duman RS. 5-HT2A
receptor-mediated regulation of brain-derived
neurotrophic factor mRNA in the hippocampus  and the
neocortex. J Neurosci  1997;17:278595.
[1003]   Vaidya VA, Terwilliger RM, Duman RS. Role of 5-HT2A.
receptors in the stress-induced  down-regulation of
brain-derived neurotrophic factor expression in rat
hippocampus. Neurosci  Lett 1999;262:14.
[1004]   Murakami S, Imbe H, Morikawa Y, Kubo C, Senba E.
Chronic stress, as well as acute stress, reduces BDNF
mRNA expression in the rat hippocampus but less
robustly. Neuroscience Res 2005;53:12939.
[1005]   Magarinos AM, McEwen  BS. Stress-induced atrophy of
apical  dendrites of hippocampal CA3c neurons:
Comparison of stressors. Neuroscience 1995;69:838.
[1006]   Magarinos AM, McEwen  BS. Stress-induced atrophy of
apical  dendrites of hippocampal CA3c neurons:
involvement of glucocorticoid secretion  and excitatory
amino acid receptors. Neuroscience  1995;69:8998.
[1007]   McEwen  BS. Allostasis  and allostatic load: Implications
for neuropsychopharmacology.
Neuropsychopharmacology  2000;22:10824.
[1008]   Sapolsky RM. Glucocorticoids  and hippocampal atrophy
in neuropsychiatric  disorders. Arch Gen Psychiatry
2000;57:92535.
[1009]   Kask  A, Harro J, von Horsten S, Redrobe JP, Dumont Y,
Quirion R. The neurocircuitry  and receptor subtypes
mediating  anxiolytic-like effects of neuropeptide Y.
Neurosci  Biobehav  2002;26(3):25983.
[1010]   Heilig M, Thorsell A. Brain neuropeptide Y (NPY) in stress
and alcohol dependence. Rev Neurosci 2002;13:8594.
[1011]   Redrobe  JP, Dumont  Y, Quirion R. Neuropeptide  Y (NPY)
and depression: from animal studies to  the human
condition.  Life Sci 2002;71:292137.
[1012]   Rasmusson  AM, et al. Low baseline  and yohimbine-
stimulated plasma neuropeptide Y (NPY) levels in
combat-related PTSD. Biol Psychiatry 2000;47:52639.
[1013]   Bannon AW, Seda J, Carmouche M, Francis  JM, Norman
MH, Karbon B, et al. Behavioral  characterization  of
neuropeptide Y knockout  mice.. Brain Res 2000;868:
7987.
[1014]   Carvajal CC, Vercauteren F, Dumont Y, Michalkiewicz M,
Quirion R. Aged neuropeptide Y transgenic rats are
resistant  to acute stress but maintain spatial and
nonspatial learning. Behav Brain Res 2004;153:
47180.
[1015]   Thorsell A, Michalkiewicz  M, Dumont  Y, Quirion R,
Caberlotto L, Rimondini R, et al. Behavioral  insensitivity
to restraint stress, absent  fear suppression of behavior
and impaired spatial learning  in transgenic rats with
hippocampal neuropeptide Y overexpression.  Proc Natl
Acad  Sci USA 2000;97:128527.
[1016]   Heilig  M. Antisense inhibition  of neuropeptide Y (NPY)-
Y1 receptor expression blocks the anxiolytic-like action
of NPY in amygdala and paradoxically increases feeding.
Regul  Pep 1995;59:2015.
[1017]   Kask  A, Rago L, Harro J. Anxiogenic-like effect of the
neuropeptide Y Y1 receptor antagonist  BIBP3226:
antagonism  with  diazepam. Eur J Pharmacol 1996;317:
34.
[1018]   Kask A, Rago L, Harro J. Anxiogenic-like effect of theNPY
Y1 receptor antagonist BIBP3226 administered into the
dorsal periaquaductal  gray matter in rats. Regul Pep
1998;7576:25562.
[1019]   Kask A, Rago L, Harro J. NPY Y1 receptors  in the dorsal
periaquaductal gray matter regulate anxiety in the social
interaction  test. Neuroreport 1998;9:27316.
[1020]   Kask A, Kivastik T, Rago L, Harro J. Neuropeptide  Y Y1
receptor antagonist BIBP3226 produces conditioned place
aversion  in rats. Prog Neuro Psychopharmacol Biol
Psychiatry 1999;23:70511.
[1021]   Sajdyk  TJ, Vandergriff MG, Gehlert DR. Amygdalar
neuropeptide Y Y-1 receptors mediate the anxiolytic-like
actions  of neuropeptide Y in the social interaction test.
Eur J Pharmacol 1999;368:1437.
[1022]   Gerald C, Walker MW, Criscione L, Gustafson  EL, Batzl-
Hartmann C, Smith KE, et al. A receptor subtype involved
in neuropeptide-Y-induced food intake. Nature (London)
1996;382:16871.
[1023]   Nakajima M, Inui  A, Asakawa A, Momose K, Ueno N,
Teranishi  A, et al. Neuropeptide Y produces anxiety  via
Y2-type receptors.  Peptides 1998;19:35963.
[1024]   Sajdyk  TJ, Schober  DA, Gehlert DR. Neuropeptide  Y
receptor  subtypes in the basolateral nucleus of the
amygdala  modulate anxiogenic  responses  in rats.
Neuropharmacology  2002;43:116572.
[1025]   Sajdyk  TJ, Schober  DA, Smiley  DL, Gehlert DR.
Neuropeptide Y-Y2 receptors mediate  anxiety in the
amygdala.  Pharmacol  Biochem Behav 2002;71:41923.
[1026]   Kask A, Rago L, Harro J. Anxiolytic-like  effect of
neuropeptide Y (NPY) and NPY13-36 microinjected into
vicinity of locus  coeruleus in rats. Brain Res 1998;788:
3458.
[1027]   Zardetto-Smith AM, Gray TS. Organization of peptidergic
and catecholaminergic efferents from the nucleus of the
solitary tract to  the rat amygdala.  Brain Res Bull
1990;25:87587.
[1028]   Allen YS, Roberts  GW, Bloom SR, Crow TJ,  Polak JM.
Neuropeptide Y in the stria terminalis: evidence for an
amygdalofugal projection. Brain  Res 1984;321:35762.
[1029]   Gustafson EL, Smith KE, Durkin MM, Walker MW, Gerald
C, Weinshank R, et al. Distribution of the neuropeptide Y
Y2 receptor mRNA in  rat central nervous system. Brain
Res Mol Brain Res 1997;46:22335.
[1030]   Parker RM, Herzog H. Regional distribution of Y-receptor
subtype mRNAs in rat brain. Eur J Neurosci 1999;11:1431
48. and in Wistar rats after ethanol exposure, Alcohol
Clin Exp Res 1998;22:177882.
[1031]   Wolak ML, De Joseph MR, Cator AD, Mokashi AS,
Browneld  MS, Urban  JH. Comparative distribution of
neuropeptide Y Y1 and Y5 receptors in the rat brain by
using immunohistochemistry. J Comp Neurol
2003;464:285311.
[1032]   Heilig M, Widerlov E. Neuropeptide Y: an overview of
central  distribution,  functional aspects, and possible
involvement in neuropsychiatric  illnesses. Acta
Psychiatr Scand 1990;82:95114.
[1033]   Colmers WF, Bleakman D. Effects of neuropeptide Y on
the electrical properties  of neurons.  Trends  in Neurosci
1994;17:3739.
[1034]   King PJ, Williams G, Doods H, Widdowson PS. Effect of a
selective neuropeptide Y Y2 receptor antagonist,
BIIE0246 on  neuropeptide Y release.  Eur J Pharmacol
2000;396:R13.
[1035]   McDonald AJ, Pearson JC. Coexistence of GABA and
peptide immunoreactivity in  non-pyramidal  neurons of
the basolateral amygdala. Neurosci  Lett  1989;100:538.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   319
[1036]   Chen G, Van Den Pol AN. Multiple NPY receptors coexist
in pre- and postsynaptic sites: inhibition of GABA release
in isolated self-innervating  SCN neurons.  J Neurosci
1996;16:771124.
[1037]   Rainnie  DG, Asprodini EK, Shinnick-Gallagher  P.
Excitatory  transmission in  the basolateral amygdala.  J
Neurophysiol 1991;66:98698.
[1038]   Rainnie  DG, Asprodini EK, Shinnick-Gallagher  P.
Inhibitory  transmission  in the basolateral amygdala. J
Physiol  1991;66:9991009.
[1039]   Ehlers CL, Li T-K, Lumeng L, Hwang BH, Somes C, Jiminez
P, et al. Neuropeptide  Y levels of ethanol-nave alcohol-
preferring and -nonpreferring rats and in  Wistar rats
after ethanol  exposure. Alcohol Clin Exp  Res
1998;22:177882.
[1040]   Hwang BH, Zhang JK, Ehlers CL, Lumeng L, Li TK. Innate
differences of neuropeptide Y (NPY) in  hyothalamic
nuclei and central nucleus of the amygdala between
selectively  bred rats with  high and low alcohol
preference. Alcohol Clin Exp Res 1999;23:102330.
[1041]   Pandey SC, Zhang H, Roy A, Xu T. Decits in amygdaloid
cAMP-responsive element binding protein signaling play
a role in genetic predisposition to anxiety and
alcoholism. J Clin Invest 2005;115:276273.
[1042]   Suzuki  R, Lumeng L, McBridge WJ, Li T-K, Hwang BH.
Reduced neuropeptide Y mRNA expression in the central
nucleus of amygdala of alcohol preferring (P) rats: its
potential involvement in alcohol preference and anxiety.
Brain Res 2004;1014:2514.
[1043]   Badia-Elder NE, Gilpin NW, Stewart RB. Neuropeptide  Y
modulation of ethanol intake: Effects of ethanol drinking
history and genetic background. Peptides 2007;28:33944.
[1044]   Primeaux  SD, Wilson SP, Bray GA, York DA, Wilson MA.
Overexpression of neuropeptide Y in the central nucleus
of the amygdala decreases ethanol self-administration in
anxious  rats. Alcohol Clin Exp Res 2006;30:791801.
[1045]   Primeaux  SD, Wilson SP, Cusick MC, York DA, Wilsom
MA. Effects of altered amygdalar neuropeptide Y
expression on anxiety-related behaviors.
Neuropsychopharmacology  2005;30:158997.
[1046]   Valdez GR, Koob  GF. Allostasis and dysregulation of
corticotropin-releasing factor and neuropeptide Y
systems: implications for the development of
alcoholism. Pharmacol Biochem Behav 2004;79:67189.
[1047]   Khoshbouei H, Cecchi M, Morilak DA. Modulatory effects
of galanin in the lateral bed nucleus of the stria
terminalis  on behavioral and neuroendocrine responses
to acute stress. Neuropsychopharmacology  2002;27:
2534.
[1048]   Bing O, Moller C, Engel JA, Soderpalm B, Heilig M.
Anxiolytic-like action  of centrally administered galanin.
Neurosci  Lett 1993;164:1720.
[1049]   Holmes A, Yang RJ, Crawley JN. Evaluation of an anxiety-
related phenotype in galanin overexpressing transgenic
mice. J Mol Neurosci  2002;18:15165.
[1050]   Kuteeva E, Hokfelt T, Ogren SO. Behavioural
characterisation of young adult transgenic mice
overexpressing  galanin under the PDGF-B  promoter.
Regul Pept 2005;125:6778.
[1051]   Khoshbouei H, Cecchi M, Dove S, Javors M, Morilak DA.
Behavioral  reactivity  to stress: amplication  of stress-
inducednoradrenergic activation elicits a galanin-
mediated anxiolytic  effect in central  amygdala.
Pharmacol  Biochem Behav 2002;71:40717.
[1052]   Holmes A, Picciotto  MRG. A novel therapeutic  target for
depression, anxiety  disorders drug addiction? CNS
Neurol Disord   Drug Targets 2006;5(2):22532.
[1053]   Leibowitz SF,  Avena NM, Chang GQ, Karatayev O, Chau
DT, Hoebel BG. Ethanol  intake increases galanin mRNA
in the hypothalamus and withdrawal  decreases it.
Physiol  Behav 2003;79:10311.
[1054]   Lewis MJ, Johnson DF, Waldman D, Leibowitz SF, Hoebel
BG. Galanin microinjection in the third ventricle
increases voluntary ethanol intake. Alcohol Clin Exp Res
2004;28:18228.
[1055]   Thiele TE, Stewart RB, Badia-Elder NE, Geary N, Massi M,
Leibowitz SF, et al. Overlapping peptide control of alcohol
self-administration  and feeding.  Alcohol Clin Exp Res
2004;28:28894.
[1056]   Belfer HH, McKnight C, Evans  C, Buzas  B, Bollettino A,
Albaugh B, et al. Enoch Association of galanin haplotypes
with alcoholism and anxiety in two ethnically distinct
populations. Mol Psychiatry 2006;11:30111.
[1057]   Mantyh PW, Hunt SP, Maggio JE. Substance P receptors:
localization  by light microscopic autoradiography in rat
brain using [
3
H]SP as the radioligand.  Brain  Res
1984;307:14765.
[1058]   Arai H,  Emson PC.  Regional distribution  of neuropeptide
K and other tachykinins (neurokinin A, neurokinin B and
substance P) in rat central nervous system. Brain  Res
1986;399:2409.
[1059]   Hokfelt T, Johansson O, Holets V, Meister B, Melander T.
Distribution  of neuropeptides with special reference to
their coexistence with classical transmitters.   In: Meltzer
HY, editor. Psychopharmacology: the third generation of
progress. New York: Raven; 1987. p. 40116.
[1060]   Rupniak NMJ, Carlson EC, Harrison T, Oates B, Seward E,
Owen S, et al. Pharmacological blockade  or genetic
deletion of substance P (NK
1
) receptors attenuates
neonatal vocalisation in guinea-pigs and mice.
Neuropharmacology 2000;39:141321.
[1061]   Rupniak  NM, Kramer MS. Discovery of the
antidepressant  and anti-emetic efcacy of substance P
receptor (NK1) antagonists.  Trends Pharmacol  Sci
1999;20:48590.
[1062]   File SE. Anxiolytic  action of a neurokinin1  receptor
antagonist  in the social interaction test. Pharmacol
Biochem Behav 1997;58:74752.
[1063]   File SE. NKP608, an NK1 receptor  antagonist,  has an
anxiolytic  action in the social interaction  test in rats.
Psychopharmacology (Berl) 2000;152:1059.
[1064]   Papp M, Vassout  A, Gentsch C. The NK1- receptor
antagonist  NKP608 has an antidepressant-like effect in
the chronic mild stress model of depression in rats.
Behav Brain  Res 2000;115:1923.
[1065]   Kramer MS, Cutler N, Feighner J, Shrivastava R, Carman J,
Sramek JJ, et al. Distinct mechanism  for antidepressant
activity by blockade  of central substance P receptors.
Science 1998;281:16405.
[1066]   Shirayama Y, Isheda H, Iwata M, Hazama G, Kawahara R,
Duman RS. Stress increases dynorphin
immunoreactivity in limbic brain regions and dynorphin
antagonism  produces antidepressant-like effects. J
Neurochem 2004;90:125868.
[1067]   Mague SD, Pliakas AM, Todtenkopf MS, Tomasiewicz HC,
Zhang Y, Stevens Jr WC, et al. Antidepressant-like effects
of kappa-opioid receptor antagonists in the forced swim
test in rats. J Pharmacol Exp  Ther 2003;305:32330.
[1068]   Terman GW, Drake CT, Simmons ML, Milner TA, Chavkin
C. Opioid modulation of recurrent  excitation in  the
hippocampal  dentate gyrus. J Neurosci  2000;20:437988.
[1069]   Croll SD, Wiegand SJ, Anderson KD, Lindsay RM, Nawa H.
Regulation of neuropeptides in adult rat forebrain by the
neurotrophins BDNF and NGF. Eur  J Neurosci
1994;6:134353.
[1070]   Duman RS, Malberg J, Nakagawa S, DSa C. Neuronal
plasticity and survival in mood disorders. Biol Psychiatry
2000;48:7329.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 320
[1071]   Hiroi N, Graybiel  AM. Atypical and typical neuroleptic
treatments induce  distinct programs of transcription
factor expression in the striatum. J Comp Neurol
1996;374:7083.
[1072]   Moratalla R, Elibol B, Vallejo M, Graybiel  AM. Network-
level changes in expression of inducible Fos-Jun proteins
in the striatum  during chronic cocaine treatment and
withdrawal. Neuron 1996;17:14756.
[1073]   Atkins JB, Chlan-Fourney J, Nye HE, Hiroi N, Carlezon Jr
WA, Nestler EJ. Region-specic induction of deltaFosB by
repeated administration  of typical versus atypical
antipsychotic drugs. Synapse  1999;33:11828.
[1074]   Rodriguez JJ, Garcia DR, Nakabeppu Y, Pickel VM. FosB in
rat striatum: normal regional distribution and enhanced
expression after 6-month  haloperiodol  administration.
Synapse  2001;39:12232.
[1075]   Linda IP, Yuki H, Paula  GU, Michel B, Lisa M, Ronald SD,
et al. Induction of  DFosB in reward-related brain
structures after chronic stress. J Neurosci  2004;24:
10594602.
[1076]   Koob  GF, Britton KT. Neurobiological substrates  for the
anti-anxiety effects of ethanol. In: Begleiter H, Kissin B,
editors.   The pharmacology  of alcohol and alcohol
dependence (series title Alcohol and alcoholism, vol. 2.
New York: Oxford University  Press; 1996.  p.  477506.
[1077]   Liljequist S, Engel JA. The effects of GABA  and
benzodiazepine receptor antagonists on the anti-conict
actions  of diazepam or ethanol. Pharmacol Biochem
Behav 1984;21:5215.
[1078]   Koob  GF, Mendelson WB, Schafer J, Wall TL, Britton KT,
Bloom FE. Picrotoxinin  receptor  ligand blocks anti-
punishment effects of alcohol. Alcohol 1989;5:43743.
[1079]   Koob  GF, Braestrup C, Thatcher-Britton K. The effects of
FG 7142 and RO 15-1788 on the release of punished
responding produced by chlordiazepoxide and ethanol in
the rat. Psychopharmacology 1986;90:1738.
[1080]   Britton KT, Ehlers CL, Koob GF. Is ethanol  antagonist  Ro
15-4513 selective for ethanol.  Science 1988;239:
64850.
[1081]   Boyle AE, Segal R, Smith BR, Amit Z. Bidirectional effects
of GABAergic agonists  and antagonists  on maintenance
of voluntary  ethanol intake  in rats. Pharmacol  Biochem
Behav 1993;46(1):17982.
[1082]   Castelli MP, Pibiri F, Piras AP, Carboni G, Orru`   A, Gessa
GL, et al. Differential G-protein coupling  to GABA
B
receptor in limbic  areas of alcohol-preferring  and -
nonpreferring rats. Eur J Pharmacol  2005;523:6770.
[1083]   Richter RM, Zorrilla EP,  Basso AM, Koob GF, Weiss F.
Altered amygdala CRF release and increased anxiety-like
behavior in  Sardinian alcohol-preferring rats: a
microdialysis and behavioral study. Alcohol Clin Exp Res
2000;24:176572.
[1084]   Cagiano R, Cassano T, Coluccia A, Gaetani S, Giustino A,
Steardo L, et al. Genetic factors involved in the effects of
developmental low-level alcohol induced  behavioral
alterations in rats. Neuropsychopharmacology
2002;26:191203.
[1085]   Cryan JF, Kaupmann K. Dont worry B happy!: a role for
GABA(B)  receptors in anxiety  and depression. Trends
Pharmacol Sci 2005;26:3643.
[1086]   Evenden JL. Varieties  of impulsivity.
Psychopharmacology  1999;146:34861.
[1087]   Van  Gaalen MM, Brueggeman  RJ, Bronius PFC,
Schoffelmeer ANM, Vanderschuren  LJMJ. Behavioral
disinhibition requires dopamine receptor  activation.
Psychopharmacology  2006;187(1):7385.
[1088]   Soubrie   P. Reconciling the role of central serotonin
neurones in human and animal  behaviour. Behav Brain
Sci 1986;9:31964.
[1089]   Linnoila M, Virkkunen  M, Scheinin M, Nuutila A, Rimon
R, Goodwin FK. Low cerebrospinal  uid 5-
hydroxyindolacetic acid concentration  differentiates
impulsive  from nonimpulsive  violent behavior. Life Sci
1983;33:260914.
[1090]   Linnoila M, Virkkunen  M, George T, Higley D. Impulse
control disorders. Int Clin Paychopharmacol 1993;8:536.
[1091]   Kreek MJ, Nielsen DA, LaForge KS. Genes associated with
addiction:  alcoholism, opiate  and cocaine addiction.
Neuromolecular Med 2004;5:85108.
[1092]   Nielsen DA, Matti V, Jaakko L, Monica E, Gerald LB, Jeffrey
CL, et al. A tryptophan  hydroxylase  gene marker for
suicidality  and alcoholism. Arch Gen Psychiatry
1998;55:593602.
[1093]   Coccaro EF, Silverman JM, Klar HM, Horvath TB, Siever LJ.
Familial  correlates  of reduced central serotonergic
system function  in patients  with personality  disorders.
Arch  Gen Psychiatry 1994;51:31824.
[1094]   Jimerson  DC, Lesem MD, Kaye WH, Brewerton TD. Low
serotonin  and dopamine metabolite concentrations in
cerebrospinal uid from bulimic  patients  with frequent
binge episodes. Arch  Gen Psychiatry 1992;49(2):1328.
[1095]   Piazza  PV, Rouge-Pont  F, Deminiere JM, Kharoubi M, Le
Moal M, Simon H. Dopaminergic activity is reduced in the
prefrontal cortex and increased  in the nucleus
accumbens of rats predisposed to develop amphetamine
self-administration. Brain Res 1991;567:16974.
[1096]   Tiihonem J, Keski-Rahkonen A, Loponen M, Mohonen M,
Kajandar J, Allonen T, et al. Brain serotonin 1
A
 receptor
binding in bulimia  nervosa. Biol  Psychiatry  2004;55:
8713.
[1097]   Walter HK, Ursula FB, Guido KF, Angela W, Shannan EH.
Brain  imaging of serotonin  after recovery from anorexia
and bulimia  nervosa. Physiol Behav 2005;86:157.
[1098]   Kaye WH, Frank GK, Bailer UF, Henry SE, Meltzer CC,
Price JC, et al. Serotonin  alterations in anorexia and
bulimia  nervosa: New insights from imaging studies.
Physiol  Behav 2005;85:7381. both 1a and 2a..
[1099]   Frank GK, Kaye  WH. Positron emission tomography
studies in eating disorders: multireceptor brain imaging,
correlates  with behavior and implications for
pharmacotherapy.  Nucl Med Biol 2005;32:75561.
[1100]   Brunner D, Hen R. Insights into the neurobiology of
impulsive  behavior from serotonin  receptor knockout
mice. Ann NY Acad  Sci 1997;836:81105.
[1101]   Rocha BA, Scearce-Levie K, Lucas JJ, Hirori N, Castanan N,
Crabbe JCETAL. Increased  vulnerability  to cocaine in
mice lacking  the serotonin-1B receptor. Nature
1998;393:1759.
[1102]   Boschert U, Amara DA, Segu L, Hen R. The mouse  5-
hydroxytryptamine1B  receptor is localized
predominantly on axon terminals.  Neuroscience
1994;58:16782.
[1103]   Cameron DL, Williams JT. Cocaine inhibits GABA release
in the VTA through endogenous 5-HT. J Neurosci
1994;14:67637.
[1104]   Kaye WH, Frank GK, Meltzer CC, Price JC, McConaha CW,
Crossan PJ, et al. Altered serotonin 2A receptor activity in
women who have recovered from bulimia nervosa. Am J
Psychiatry 2001;158:11525.
[1105]   Bailer UF, Price JC, Meltzer CC, Mathis CA, Frank GK,
Weissfeld L, et al. Altered 5-HT
2A
 receptor  binding after
recovery  from bulimia-type  anorexia nervosa:
relationships to harm avoidance  and drive for thinness.
Neuropsychopharmacology  2004;29:114355.
[1106]   Steiger H, Young SN, Kin NM, Koerner N, Israel M, Lageix
P, et al. Implications of impulsive and affective
symptoms for serotonin  function in  bulimia nervosa.
Psychol Med 2001;31:8595.
b i oc he mi c a l   p ha r ma c ol o gy  7 5  ( 2 0 0 8 )   2 6 6  3 2 2   321
[1107]   Kuikka JT, Tammela L, Karhunen L, Rissanen  A,
Bergstro m KA, Hannu N, et al. Reduced serotonin
transporter  binding in binge eating women.
Psychopharmacology (Berl) 2001;155(3):3104.
[1108]   Tammela LT, Rissanen A, Kuikka JT, Karhunen LJ,
Bergstro m KA, Repo-Tiihonen  E, et al. Treatment
improves  serotonin transporter binding and reduces
binge eating. Psychopharmacology  (Berl)  2003;170(1):
8993.
[1109]   Steiger H, Gauvin  L, Joober R, Israel M, Ng NMK, Kim Y,
et al. Intrafamilial correspondences on  platelet [3H-
]paroxetine-binding indices  in bulimic  probands and
their unaffected rst-degree relatives.
Neuropsychopharmacology  2006;31:178592.
[1110]   Jentsch JD, Taylor JR. Impulsivity resulting from
frontostriatal dysfunction in drug abuse: implications for
the control of behavior by reward-related  stimuli.
Psychopharmacology 1999;146:37390.
[1111]   Jaskiw GE, Karoum  G, Freed WJ, Phillips I, Kleinman JE,
Weinberger  DR. Effect of ibotenic acid lesions of the
medial prefrontal cortex on amphetamine-induced
locomotion  and regional brain  catecholamine
concentrations in  the rat. Brain Res 1990;534:26372.
[1112]   Banks KE, Gratton  A. Possible involvement  of medial
prefrontal cortex in amphetamine-induced sensitization
of mesolimbic dopamine function.  Eur J Pharmacol
1995;282:15767.
[1113]   Schenk S, Horger BA, Peltier R, Shelton  K.
Supersensitivity to the reinforcing  effects of cocaine
following  6-hydroxydopamine lesions to the medial
prefrontal cortex  in rats. Brain  Res 1991;543:2536.
[1114]   Deutch  AY. The regulation of subcortical  dopamine
systems by the prefrontal cortex: interactions  of central
dopamine  systems and the pathogenesis of
schizophrenia. J Neural Transm [Gen Sect] 1992;36:6189.
[1115]   Louilot A, Le Moal M, Simon H. Opposite inuences  of
dopaminergic  pathways to the prefrontal cortex or the
septum on the dopaminergic transmission in the nucleus
accumbens: an in vivo voltammetric study. Neuroscience
1989;29:4556.
[1116]   Piazza PV, Deminiere JM, Maccari S, LeMoal M, Mormede
P, Simon H. Individual vulnerability to drug self-
administration:  action of corticosterone on
dopaminergic  systems as a possible pathophysiological
mechanism.   In: Willner P, Scheel-Kruger J, editors. The
mesolimbic dopamine sytem: from motivation to action.
Chichester: John  Wiley & Sons; 1991. p. 47395.
[1117]   Le Moal M, Simon H. Mesocorticolimbic  dopaminergic
network: functional  and regulatory roles. Physiol Rev
1991;71:155234.
[1118]   Piazza  PV, Rouge-Pont  F, Deminiere JM, Kharoubi M,
LeMoal M, Simon H. Dopaminergic activity is reduced in
the prefrontal cortex and increased in the nucleus
accumbens of rats predisposed to develop amphetamine
sensitization.  Brain  Res 1991;567:16974.
[1119]   Prasad BM, Hochstatter T, Sorg BA. Expresssion of
cocaine sensitization: regulation by the medial prefrontal
cortex. Neuroscience 1999;88:76574.
[1120]   Cole BJ, Robbins TW. Amphetamine  impairs  the
discriminative performance  of rats with  dorsal
noradrenergic bundle lesions on a 5-choice serial
reaction  time task: new evidence  for central
dopaminergic-noradrenergic  interactions.
Psychopharmacology (Berl) 1987;91:45866.
[1121]   Cole BJ, Robbins TW. Effects of 6-hydroxydopamine
lesions of the nucleus accumbens  septi on performance
of a 5-choice serial reaction time task in  rats:
implications for theories of selective attention and
arousal. Behav Brain  Res 1989;33:16579.
[1122]   Harrison  AA, Everitt BJ, Robbins TW. Central 5-HT
depletion enhances  impulsive responding without
affecting  the accuracy  of attentional performance:
interactions  with dopaminergic  mechanisms.
Psychopharmacology (Berl) 1997;133:32942.
[1123]   Dalley JW, Fryer TD, Brichard L, Robinson ESJ, Theobald
DEH, La a ne K, et al. Nucleus  accumbens  D2/3 receptors
predict trait impulsivity and cocaine reinforcement.
Science 2007;315(5816):126770.
[1124]   Lobo DSS, Kennedy JL. The genetics of gambling  and
behavioral  addictions. CNS Spectr 2006;11:9319.
[1125]   Laucht M, Becker  K, Blomeyer  D, Schmidt MH. Novelty
seeking involved in mediating  the association between
the dopamine d4 receptor gene exon  III polymorphism
and heavy drinking in  male adolescents: results from a
high-risk community  sample. Biol Psychiatry  2007;61:
8792.
[1126]   McGeary JE, Esposito-Smythers C, Spirito A, Monti PM.
Associations of the dopamine D4 receptor gene VNTR
polymorphism with  drug use in adolescent psychiatric
inpatients.  Pharmacol  Biochem Behav 2007;86(2):4016.
[1127]   Li T, Xu K, Deng H, Cai G, Liu J, Liu X, et al. Association
analysis  of the dopamine  D4 gene exon  III VNTR and
heroin abuse in Chinese  subjects. Mol Psychiatry
1997;2(5):4136.
[1128]   Shoa C, Li Y, Jiang K, Zang D, Xu Y, Lin Y, et al. Dopamine
D4 receptor polymorphism modulates cue-elicited
heroin craving in Chinese. Psychopharmacology
2006;186(2):18590.
[1129]   Lusher J, Ebersole L, Ball D. Dopamine  D4 receptor gene
and severity of dependence. Addict Biol 2000;5(4):46972.
[1130]   Levitan  RD, Masellis M, Basile VS, Lam RW, Kaplan AS,
Davis C, et al. The dopamine-4 receptor gene associated
with binge eating and weight gain in women  with
seasonal affective disorder: An evolutionary perspective.
Biol Psychiatry  2004;56(9):6659.
[1131]   Sobik L, Hutchison  K, Craighead L. Cue-elicited craving
for food: a fresh approach to the study of binge eating.
Appetite 2005;44(3):25361.
[1132]   Roman T, Bau CHD, Almeida  S, Hutz  MH. Lack of
association of the dopamine D4  receptor gene
polymorphism with alcoholism in a Brazilian population.
Addict Biol 1999;4(2):2037.
[1133]   Franke P, Wang T, Mo then MM, Knapp M, Neith H,
Lichtermann D, et al. Susceptibility for alcoholism: DRD4
exon III polymorphism: a casecontrol and a family-
based association  approach. Addict Biol 2000;5(3):
28995.
[1134]   Franke P, No then MM, Wang T, Knapp M, Lichtermann D,
Neidt H, et al. DRD4 exon III VNTR polymorphism-
susceptibility  factor for heroin dependence? Results of a
case-control  and a family-based association approach.
Mol Psychiatry  2000;5(1):1014.
[1135]   Kirley A, Hawi Z, Daly G, McCarron M, Mullins C, Millar N,
et al. Dopaminergic system genes in adhd:  toward a
biological  hypothesis.  Neuropsychopharmacology
2002;27(4):60719.
[1136]   Lowe N, Kirley A, Hawi Z, Sham P, Wickham H,
Kratochvil  CJ, et al. Joint  analysis  of the drd5 marker
concludes association with attention-decit/
hyperactivity  disorder conned to the predominantly
inattentive and combined subtypes. Am J Hum Genet
2004;74(2):34856.
b i o c he mi c a l   p ha r ma c o l og y  7 5  ( 2 0 0 8 )   2 6 6  3 2 2 322