ArticlePDF AvailableLiterature Review

Genetics of Alcohol Dependence: A Review of Clinical Studies

Authors:

Abstract and Figures

Background/aims: Alcohol dependence is a common severe psychiatric disorder with a multifactorial etiology. Since the completion of the human genome project and with the increased availability of high-throughput genotyping, multiple genetic risk factors for substance-related disorders, including alcohol dependence, have been identified, but not all results could be replicated. Methods: We systematically review the clinical literature on genetic risk factors for alcohol dependence and alcohol-related phenotypes, including candidate gene-based studies, linkage studies and genome-wide association studies (GWAS). Results: Irrespectively of the methodology employed, the most robust findings regarding genetic risk factors for alcohol dependence concern genetic variations that affect alcohol metabolism. GWAS confirm the importance of the alcohol dehydrogenase gene cluster on chromosome 4 in the genetic risk for alcohol dependence with multiple variants that exert a small, but cumulative influence. A single variant with strong influence on individual risk is the aldehyde dehydrogenase 2 ALDHD2*2 variant common in Asian populations. Other robust associations have been found with previously uncharacterized genes like KIAA0040, and such observations can lead to the identification of thus far unknown signaling pathways. Converging evidence also points to a role of glutamatergic, dopaminergic and serotonergic neurotransmitter signaling in the risk for alcohol dependence, but effects are small, and gene-environment interactions further increase the complexity. Conclusion: With few exceptions like ALDH2*2, the contribution of individual genetic variants to the risk for alcohol-related disorders is small. However, the concentration of risk variants within neurotransmitter signaling pathways may help to deepen our understanding of the underlying pathophysiology and thereby contribute to develop novel therapeutic strategies.
Content may be subject to copyright.
E-Mail karger@karger.com
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
Genetics of Alcohol Dependence:
A Review of Clinical Studies
Jerzy Samochowiec a Agnieszka Samochowiec b Imke Puls d
Przemyslaw Bienkowski c Björn H. Schott d, e
a Department of Psychiatry, Pomeranian Medical University, and
b Institute of Psychology, Department of
Clinical Psychology, University of Szczecin, Szczecin , and
c Institute of Psychiatry and Neurology, Warsaw , Poland;
d Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin , and
e Leibniz Institute for Neurobiology, Magdeburg , Germany
multiple variants that exert a small, but cumulative influence.
A single variant with strong influence on individual risk is the
aldehyde dehydrogenase 2 ALDHD2 * 2 variant common in
Asian populations. Other robust associations have been
found with previously uncharacterized genes like KIAA0040 ,
and such observations can lead to the identification of thus
far unknown signaling pathways. Converging evidence also
points to a role of glutamatergic, dopaminergic and seroto-
nergic neurotransmitter signaling in the risk for alcohol de-
pendence, but effects are small, and gene-environment in-
teractions further increase the complexity. Conclusion: With
few exceptions like ALDH2 * 2, the contribution of individual
genetic variants to the risk for alcohol-related disorders is
small. However, the concentration of risk variants within neu-
rotransmitter signaling pathways may help to deepen our
understanding of the underlying pathophysiology and there-
by contribute to develop novel therapeutic strategies.
© 2014 S. Karger AG, Basel
Key Words
Alcohol-related phenotypes · Alcohol dependence ·
Genetic risk factors
Abstract
Background/Aims: Alcohol dependence is a common severe
psychiatric disorder with a multifactorial etiology. Since the
completion of the human genome project and with the in-
creased availability of high-throughput genotyping, multiple
genetic risk factors for substance-related disorders, including
alcohol dependence, have been identified, but not all results
could be replicated. Methods: We systematically review the
clinical literature on genetic risk factors for alcohol depen-
dence and alcohol-related phenotypes, including candidate
gene-based studies, linkage studies and genome-wide asso-
ciation studies (GWAS). Results: Irrespectively of the method-
ology employed, the most robust findings regarding genetic
risk factors for alcohol dependence concern genetic varia-
tions that affect alcohol metabolism. GWAS confirm the im-
portance of the alcohol dehydrogenase gene cluster on chro-
mosome 4 in the genetic risk for alcohol dependence with
Received: October 7, 2013
Accepted after revision: May 24, 2014
Published online: October 30, 2014
Björn H. Schott, MD, PhD
Department of Psychiatry and Psychotherapy
Campus Charité Mitte, Charité Universitätsmedizin Berlin
Charitéplatz 1, DE–10117 Berlin (Germany)
E-Mail bjoern.schott @ charite.de
© 2014 S. Karger AG, Basel
0302–282X/14/0702–0077$39.50/0
www.karger.com/nps
Jerzy Samochowiec and Agnieszka Samochowiec contributed equal-
ly to this work.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
78
Introduction
While alcohol consumption at moderate levels is so-
cially widely accepted in many cultures, excessive drink-
ing ultimately leading to alcohol dependence is a severe
health problem both at the individual and large-scale so-
cioeconomic levels. According to World Health Organi-
zation recommendations, the limit of risky consumption
is for men not to consume more than 4 standard drinks a
day or 28 standard drinks a week and for women not to
consume more than 2 standard drinks a day or 14 stan-
dard drinks a week. It is estimated that every fourth per-
son in Europe aged 15–64 regularly consumes alcohol at
harmful levels, and over 14 million people are alcohol de-
pendent
[1] . The US data from the 2001–2002 National
Epidemiological Survey on Alcohol and Related Condi-
tions showed a 12-month prevalence of 3.8% for alcohol
dependence defined by DSM-IV: 5.4% in men and 2.3%
in women
[2] . The lifetime prevalence of alcohol depen-
dence is 12.5% in the USA.
Alcohol use disorders (AUDs) can be defined as ‘mal-
adaptive patterns of alcohol use leading to clinically sig-
nificant impairment or stress’
[3] . Diagnostic criteria,
both from the Diagnostic and Statistical Manual of Men-
tal Disorders [4] and from the International Classification
of Diseases [5] divide AUDs into alcohol dependence and
abuse/harmful use. The major change proposed for DSM-
V (and introduced in May 2013) has replaced the 2 sepa-
rate DSM-IV (substance-specific) categories of depen-
dence and abuse with a single (substance-specific) cate-
gory, substance use disorder (SUD). The criteria for SUD
merge the previous lists of 7 criteria for dependence and
4 criteria for abuse into a single list of 11 criteria. SUD is
now additionally defined by the presence of craving,
while the criterion of recurrent legal problems has been
discarded. Severity of the illness is graded by the number
of criteria met: 0–1, no diagnosis; 2–3, mild SUD; 4–5,
moderate SUD; 6 or more, severe SUD. Tolerance and
withdrawal remain as symptoms of SUDs
[6] .
Alcohol dependence is defined across all versions of
the DSM, despite the recent changes in its definition, as a
disorder characterized by physiological and psychologi-
cal effects in individuals who consume large amounts of
alcohol
[7] .
The discrepancy between the widespread occurrence
of risky or harmful alcohol consumption and the fre-
quent, yet substantially lower, prevalence of clinical alco-
hol dependence raises the question why certain individu-
als become dependent on alcohol, sometimes in a rather
short time, while others do not seem to be affected by
regular alcohol consumption. There is nowadays little
doubt that alcohol dependence is a disorder of complex
etiology with multiple risk factors contributing to it. The
susceptibility to alcohol dependence is subject to consid-
erable interindividual variability, and it is shaped by both
including environmental (e.g. cultural, social political, re-
ligious, economic, legal including alcohol accessibility,
price and social norms) and genetic factors
[8–11] . The
biological processes that make one person more suscep-
tible to addiction than another are subject to intense in-
vestigations. Twin and adoption studies quantify the her-
itable component at 50–60%
[12–14] .
While in early adolescence the initiation and use of al-
cohol and other addictive drugs is primarily influenced
by environmental, i.e. familial and social, factors, the role
of environmental influences progressively decreases dur-
ing the transition to adulthood, when the impact of ge-
netic contribution reaches its peak
[15] . This observation
suggests that adolescence may be the optimal time point
for educational interventions.
A complex pattern of genetic associations indicates a
combined contribution of many genes as well as gene-
gene and gene-environment (GxE) interactions. To dis-
tinguish between environmental and genetic factors,
adoption and twin studies have been conducted. Adop-
tion studies demonstrate that adoptees more closely re-
semble their biological parents than their adoptive par-
ents in terms of susceptibility to alcohol dependence
[16–
19] . Twin studies reveal greater concordance between
monozygotic as compared to dizygotic twins, highlight-
ing a major genetic impact
[10, 19–21] . (Note: important-
ly, not all monozygotic cotwins of alcohol-dependent pa-
tients are alcohol-dependent themselves, also highlight-
ing the influence of environmental factors and GxE
interactions in the etiology of the disorder.) A third line
of evidence for a substantial genetic contribution to alco-
hol dependence comes from animal research. In selec-
tively bred rodent lines, a considerable proportion of her-
itability in the development of addiction-related behavior
has been observed, including strong preference for alco-
hol over water, willingness to work for alcohol, sensitiv-
ity to the hypnotic or activating effects of alcohol and to
withdrawal, and demonstrations that alcohol is reward-
ing even in the presence of food and water
[22–26] . A
fourth line of evidence comes from early genetic studies
in humans that demonstrated genetic variations in genes
related to alcohol metabolism that affect the predisposi-
tion to alcohol dependence
[27–30] .
The environmental contributions to alcohol-related
phenotypes include peer influence
[31–35] , availability
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
79
of alcohol [31, 32, 36–38] , early first drink [32, 39] and
marital status
[32, 40] . Religious belief has been shown
to affect alcohol initiation
[32, 41] . The pattern of alco-
hol drinking is thought to contribute to the risk of alco-
hol dependence. The alcohol dependence risk grows al-
most linearly with the frequency of binge drinking [42] .
Data from the National Epidemiologic Survey on Alco-
hol and Related Conditions confirm the finding
[3, 42] .
However, one should remember that the pattern of
drinking can be modulated by both environmental and
genetic factors.
Risk loci for AUDs can be grouped into genes involved
in alcohol pharmacokinetics and pharmacodynamics,
and genes moderating addiction-related traits and behav-
iors such as sensation seeking, impulsivity, externalizing
behaviors and disinhibition. Our insight into the causes
of alcoholism is limited due to heterogeneity of its clinical
features, including a frequent co-occurrence with other
addictions and also with other psychiatric disorders, both
internalizing (e.g. depression and anxiety) and external-
izing (e.g. antisocial personality disorder, conduct disor-
der and attention deficit hyperactivity disorder)
[43–46] .
This heterogeneity may also be reflected by various AUD
subtypes used in current typologies and in the diverse
patterns of psychiatric and medical comorbidity observed
among AUD patients. Empirically derived typologies
typically identify 2–5 consistent subtypes classified by
their most prominent symptoms: early onset, externaliz-
ing behavior, affective instability and antisocial personal-
ity disorder. Multidimensional typologies differentiate
alcoholism by symptom severity. Typologies have com-
monly been derived from clinically based samples of al-
cohol-dependent persons, as the precise identification of
genetic loci has, at least so far, proven to be too complex
a task to accomplish
[47–50] . The genetic contribution to
the risk for AUDs is subject to nonmendelian transmis-
sion, and recent studies have concluded that there is a
wide range of genetic factors that might influence the risk
for AUDs
[51–54] .
Intermediate phenotypes – or endophenotypes – of
AUDs have been identified that are believed to be bio-
logically more directly related to the majority of the ge-
netic risk factors compared to the disorder itself. At least
3 reliable endophenotypes have been identified for AUDs:
(a) alcohol-induced skin flushing reaction associated
with lower AUD risk;
(b) a low response level to alcohol;
(c) electrophysiological, psychological, neuroendo-
crine and neuroimaging phenotypes predictive of AUD
and other types of addiction
[55–60] .
There are 3 main techniques used to identify genetic
variations affecting alcohol dependence: candidate gene
studies, linkage studies and genome-wide association
studies (GWAS). The following review is based on a
search for PubMed-indexed articles reporting such stud-
ies. Candidate gene studies of alcohol dependence were
obtained from PubMed (http://www.ncbi.nlm.nih.gov/
pubmed) using the term ‘candidate gene’ combined with
‘alcohol dependence’, ‘alcohol use disorder’ or ‘alcohol
addiction’. Similarly, we searched for PubMed-indexed
articles describing linkage-based studies of genetic influ-
ences on alcohol dependence. Finally, we searched for
PubMed-indexed articles describing GWAS of alcohol
dependence by using the term ‘GWAS’ combined with
‘alcohol dependence’, ‘alcohol use disorder’ or ‘alcohol
addiction’. In each thus identified article, the reference
list was reviewed for similar studies. Only English-lan-
guage articles published before July 2013 (December 2013
in case of GWAS) in peer-reviewed journals were accept-
ed. Articles were identified and reviewed by 2 of the au-
thors, and their final decision was made by consensus. In
the case of GWAS of alcohol dependence, all relevant ar-
ticles identified in PubMed met the above criteria and
were included in this review.
Candidate Gene Studies
Genes Related to Alcohol Metabolism
The earliest genetic association studies in alcohol de-
pendence were candidate gene studies targeting coding
variations in genes that metabolize alcohol. Alcohol is
mostly metabolized in the liver, in a 2-step reaction: oxi-
dation to acetaldehyde, which is further oxidized to ace-
tate. The first step is primarily to be catalyzed by alcohol
dehydrogenases (ADHs), a reaction accompanied by re-
duction of NAD
+ to NADH. The second step is mainly to
be catalyzed by aldehyde dehydrogenases (ALDHs), with
reduction of another molecule of NAD
+ to NADH. Hu-
mans have 7 ADHs, with ADH1 being most important for
ethanol oxidation. ADH1 consists of 3 subunits, α, β and
γ, which are encoded by the genes ADH1A, ADH1B and
ADH1C. ALDHs constitute a group of enzymes, out of
which ALDH2 is primarily involved in the oxidation of
acetaldehyde. Variations in these genes have long been
known to affect the risk for alcoholism
[27–30, 61] .
A variation in the ALDH2 gene has striking effects on
alcohol metabolism. ALDH2 encodes the mitochondrially
localized ALDH2. An amino acid substitution from gluta-
mate to lysine at position 504 (NCBI accession No.: rs671),
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
80
called ALDH2 * 2, leads to a drastic activity reduction of the
catalytically active subunit and additionally increases the
turnover of the tetrameric enzyme. The ALDH2 * 2 variant
is dominant, and the presence of at least 1 allele renders
the ALDH2 catalytic activity (measured in vitro ) below
the usual limit of detection
[62, 63] . In individuals carry-
ing the ALDH2 * 2 allele, consumption of even small
amounts of alcohol leads to accumulation of acetaldehyde
to toxic levels resulting in symptoms like flushing, tachy-
cardia and nausea, similar to the effects of alcohol in pa-
tients taking the ALDH2 inhibitor disulfiram.
The genes of the ADH s that catalyze the first step in
ethanol metabolism, clustered at chromosome 4, also
harbor genetic variations that strongly affect the risk for
alcohol-related disorders. After low to moderate intake of
alcohol, the ADH1 enzyme which consists of subunits en-
coded by ADH1A, ADH1B and ADH1C genes plays the
major role in alcohol metabolism
[27–29, 64] . At higher
(toxic) levels of alcohol, the ADH4 enzyme (encoded by
the ADH4 gene on chromosome 4) becomes more impor-
tant in alcohol metabolism
[64] . Genetic variations of
ADH4 have been linked to alcohol elimination
[65] , as
well as the risk for alcohol dependence in several different
populations
[66–68] . ADH7, located in the esophagus
and stomach lining, can contribute to ‘first pass’ metabo-
lism and thereby influence blood alcohol concentration
[27] .
However, these successfully replicated findings do not
explain risk differences in European populations in which
the alleles with the strongest effects are rare.
Genes Implicated in the Neurobiology of Addiction
The positive reinforcing effects of alcohol are gener-
ally believed to be critically mediated by the mesolimbic
reward pathway, a dopaminergic projection from the
ventral tegmental area to the nucleus accumbens that is
further modulated by several other neurotransmitters. In
addition to its effect on the dopaminergic system, alcohol
can affect the principal neurotransmitter glutamate and
γ-aminobutyric acid (GABA) as well as neuromodulatory
transmitters including acetylcholine, opioids and sero-
tonin
[69–74] .
It is therefore conceivable that genetic variations of the
dopaminergic system have been in the focus of candidate
gene-based association studies for a long time. For ex-
ample, genetic variations in the dopamine-metabolizing
enzymes catechol-O-methyltransferase and monoamine
oxidase A (MAOA) have been found to be associated with
the risk for alcohol dependence
[75–77] , although these
results could not be uniformly replicated
[78–80] . Other
studies have linked genetic variations in the dopamine
transporter 1 gene to the severity of withdrawal symp-
toms
[81, 82] .
One key problem in many candidate gene studies is the
poor replicability, which becomes evident in markedly
mixed results. Perhaps the best-known example is the as-
sociation of the Taq1A allele of the dopamine D
2 receptor
gene ( DRD2-Taq1A; rs1800497) with alcohol disorder,
which was first reported in a small study in 1990
[83, 84] .
Numerous groups have attempted to replicate this find-
ing, but most replication studies have yielded negative or
inconclusive results
[85–90] . A recent meta-analysis has
suggested a small but significant effect. The authors cau-
tioned that the positive result might have been caused
by publication bias
[91] , but a more recent meta-analysis
by a different group has confirmed the association and
largely ruled out publication bias
[84] . The example of
rs1800497 highlights another important problem of ge-
netic association studies, namely that inconclusive results
can result from linkage disequilibrium with other vari-
ants that might actually be causally related to a phenotype
(Note: linkage disequilibrium refers to the nonrandom
association and coupled inheritance of allelic variants at
two loci that may or may not have a large genetic distance
[92] ). The so-called DRD2 Taq1A polymorphism is actu-
ally located downstream of the DRD2 gene, within the
adjacent and closely linked ANKK1 (ankyrin repeat and
kinase domain containing 1) gene region
[93, 94] . There-
fore, associations of Taq1A with alcohol dependence
might result from linkage disequilibrium with other caus-
ative genetic variations in either DRD2 or ANKK1 or
both. A recent family study showed that a different ge-
netic variation in DRD2 (rs6277, a synonymous single
nucleotide polymorphism) that affects mRNA stability
[95] did indeed show an association with alcohol depen-
dence, while rs1800497 was not significantly associated
[96] . On the other hand, another family-based analysis
showed that the association of the ANKK1/DRD2 region
on chromosome 11q23 with alcohol dependence might in
fact be strongest in a part of the ANKK1 gene that is not
in linkage disequilibrium with DRD2 [35] . A further fam-
ily and case-control study suggested that a gene cluster in
this region consisting of NCAM1, TTC12 and ANKK1
might be involved in the etiology of alcohol dependence
[97–99] . The complexity and the inconclusive character
of these data point to a need for basic research to investi-
gate the biological processes underlying these associa-
tions. Such investigations might also help to identify in-
termediate phenotypes that are more closely related to the
genetic variations than a clinical phenotype per se. In the
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
81
case of DRD2 and ANKK1, it has meanwhile been dem-
onstrated that the two molecules interact at protein levels
and that ANKK1 is upregulated by the dopamine agonist
apomorphine [100, 101] .
Beyond the dopaminergic system, numerous candi-
date gene studies of alcohol dependence and alcohol-re-
lated phenotypes have focused on molecules involved in
neurotransmitter pathways interacting with dopaminer-
gic transmission. These include genes involved in cholin-
ergic neurotransmission, such as the muscarinic cholin-
ergic receptor 2 (CHRM2) [102] and the α
5 -subunit of the
nicotinergic cholinergic receptor (CHRNA5) [103–105] ,
components of the GABAergic system like the α
2 -subunit
of the GABA-A receptor ( GABRA2 )
[106] and GABA re-
ceptor genes
[107–110] , genes coding for components of
glutamatergic transmission like the metabotropic gluta-
mate receptor 8 ( GRM8 )
[111] , genes involved in seroto-
nergic neurotransmission like the solute carrier family 6
member 4 (SLC6A4) gene that encodes the serotonin
transporter
[112, 113] , and components of the endocan-
nabinoid system such as cannabinoid receptor 1.
In addition to these genetic variants, which all affect
small-molecule neurotransmitters, numerous genetic as-
sociation studies have been conducted on genes related to
peptide neurotransmitters. Most notably, the opioid sys-
tem has repeatedly been found to be implicated in alcohol
addiction
[114–118] . There are 3 main opioid receptors
and 3 main genes encoding endogenous ligands. OPRM1
encodes the μ-receptor, and its primary ligand β-endorphin
is cleaved from the proopiomelanocortin protein, which
is encoded by the POMC gene. OPRK1 encodes the
κ-opioid receptor, and PDYN encodes the primary endog-
enous κ-opioid agonist (pro-)dynorphin. The δ-opioid re-
ceptor is encoded by OPRD1, and its primary ligand met-
enkephalin is synthesized as a propeptide, proenkephalin
A, which is encoded by the PENK gene [119] .
The μ-opioid receptor gene OPRM1, and especially the
coding variation Asn40Asp (rs1799971), has been exten-
sively studied in relation to alcohol dependence
[85] . Re-
sults have been inconclusive and partly conflicting, and a
meta-analysis concluded that there was no reliable evi-
dence for association
[120] . On the other hand, a more
recent meta-analysis has provided evidence for a role of
the Asn40Asp variation in the therapeutic response to the
μ-opioid antagonist naltrexone for prevention of relapse
[121] . The κ-opioid system has also been linked to AUDs
with both the PDYN gene coding for the κ-opioid agonist
prodynorphin and the κ-opioid receptor gene OPRK1
showing a genetic association with alcohol dependence
[122] .
Beyond the endogenous opioid system, research has
provided evidence for NPY , which encodes the peptide
neurotransmitter neuropeptide Y, as a candidate gene for
AUD. NPY modulates alcohol dependence in a rat mod-
el
[123–125] , and NPY knockout mice show increased
preference for alcohol
[126] . In humans, some but not all
genetic association studies have found evidence for an
association between polymorphisms in the NPY gene on
chromosome 7 and the risk for AUDs
[127–130] . Three
NPY receptor genes on chromosome 4 have also been
investigated in relation to AUDs, yielding 2 positive as-
sociations: NPY2R was associated with alcohol depen-
dence and alcohol withdrawal symptoms, and NPY5R
was associated with seizures during alcohol withdrawal
[130] . Other genetic association studies of peptide neu-
rotransmitter systems have found evidence for an asso-
ciation of AUDs with polymorphisms of the brain-de-
rived neurotrophic factor
[131] and the tachykinin re-
ceptor 3
[132] .
In light of the multitude of identified risk variants to
date, it must be kept in mind that there are numerous ex-
amples of associations that cannot be replicated. This
problem has occasionally led to scepticism about the use-
fulness of candidate-based association studies. Reasons
contributing to this lack of reproducibility include poor
study design, incorrect assumptions about the underlying
genetic architecture and simple overinterpretations of the
data. The most common errors in association studies of
complex diseases relate to small sample sizes and result-
ing low power, poorly matched control groups, data-driv-
en subgroup analyses (‘double dipping’) and insufficient
statistical control for multiple testing. To some extent, a
positive publication bias and unwarranted ‘candidate
gene’ declaration after identifying an association in an ar-
bitrary genetic region also contribute to poor replicability
[133, 134] .
Moreover, there might also be a biological reason for
heterogeneous results of association studies between
AUDs and genes related to neurotransmitter systems,
namely the fact that most neural cells express multiple
receptors that activate converging complex signaling cas-
cades, and potential risk mechanisms at receptor level
might be compensated at the level of intracellular signal-
ing. Therefore, genes affecting intracellular signaling in
response to receptor activation could also potentially af-
fect the risk for alcohol dependence. For example, the
gene encoding the nuclear factor of κ light polypeptide
gene enhancer in B cells 1 ( NFKB1 ), a transcription factor
activated by growth factor signaling
[135] , and the gene
encoding the matrix metallopeptidase 9, which plays a
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
82
role in the remodeling of the perineuronal extracellular
matrix during synaptic plasticity
[136] , have both been
found to be associated with the risk for alcohol depen-
dence.
Gene-Environment Interactions
The association of single molecular-genetic variations
with psychiatric disorders including AUDs is modulated
by nongenetic influences, and the interplay between spe-
cific candidate genes and environmental factors has been
subject to numerous GxE interaction studies
[32] . Envi-
ronmental determinants that enhance the risk for AUD
differ in both proximity to the disorder and its mecha-
nism. Important risks for alcohol dependence include
prenatal exposure to alcohol, growing up in a home with
an alcoholic parent and being poorly monitored by one’s
parents
[137] . Other environmental factors exert their in-
fluence on an individual’s risk later in life. For instance,
genetic effects on drinking are greater in urban versus
rural residential settings
[36, 37] , as social restrictions in-
hibit development of genetically determined patterns of
behavior. Findings from a Collaborative Study on the
Genetics of Alcoholism (COGA) further suggest that
variation within an AUD susceptibility gene ( GABRA2;
rs279871) also depends on a person’s marital status, with
individuals carrying the high risk variant being less likely
to be married. Marital status also abated the influence of
other variants within GABRA2 [108] .
GxE interactions have been reported for several gene
variants that have previously been found to be associated
with alcohol dependence including genes with inconsis-
tent results in association studies like the DRD2 / ANKK1
gene region, 5-HTTLPR , MAOA , catechol-O-methyl-
transferase, GABRA2 and the corticotropin-releasing
hormone receptor 1 ( CRHR1 )
[138–142] . Therefore, GxE
interactions might contribute to the complexity of genet-
ic associations, and systematic assessment of environ-
mental factors might help to explain the difficulties in
replicating certain results.
Notably, GxE interactions are not limited to genes
with weak associations, but even genetic variations with
strong influence on alcohol-related behaviors and disor-
ders exert their influences in the interaction with envi-
ronmental factors. In fact, one of the most robust GxE
interactions was observed in carriers of the ALDH2 * 2
variant. A substantial GxE interaction with respect to
ALDH2 in the Japanese population was reported by Hi-
guchi et al.
[30] . The protective effect of the ALDH2 * 2
allele seemed to decrease substantially from 1979 to 1992
when the proportion of Japanese AUD patients heterozy-
gous for the ALDH2 * 2 soared from 2.5 to 13%
[30] . Giv-
en the short time interval of observation, a hypothetical
shift in allele frequencies within the Japanese population
could not explain this result. More likely, the protective
effect of the ALDH2 * 2 allele was reduced due to environ-
mental influences, most likely social changes favoring al-
cohol consumption. In the same study, no alcohol-de-
pendent patients homozygous for the ALDH2 * 2 were
identified, although 120 patients would have been expect-
ed based on the allele frequency. It was concluded that the
protective influence of the ALDH2 * 2 allele was so sub-
stantial in homozygotes that further modulation by envi-
ronmental influences did not occur
[30] .
Genetic Linkage Studies
Genetic linkage studies provide an unbiased method-
ology toward identifying genetic variations that affect the
risk for a complex genetic disease such as alcohol depen-
dence. The linkage approach is employed to identify ge-
netic risk variants for a disorder in families with several
affected members. Linkage studies test markers distrib-
uted across the genome, and if one such marker variant is
located near a causal genetic variation, family members
with the phenotype of interest would be expected to share
larger numbers of the same marker allele than those with-
out the phenotype. This provides information about the
broad chromosomal region in which the causal variants
are located, but, in most cases, not about specific genes.
The linkage method is most effective in phenotypes with
a mendelian inheritance pattern, which is rare in psychi-
atric disorders where common variants are unlikely to
have large effects
[143] . On the other hand, linkage stud-
ies are well suited for detecting genetic variants associated
with a major risk increase (at least 5- to 10-fold)
[144] .
Although nowadays largely of historic interest, linkage
studies have provided some useful information regarding
chromosomal risk loci for alcohol dependence. The earli-
est whole-genome linkage studies on alcohol dependence
were reported in 1998
[145, 146] . Large-scale linkage
studies like the COGA
[146] or the study by Long et al.
[145] in a relatively homogeneous Native American pop-
ulation have pointed to a risk locus on chromosome 4q,
which contains the ADH region. Additional evidence for
a risk locus on chromosome 4q was found in the Irish Af-
fected Sib Pair Study of Alcohol Dependence that addi-
tionally provided weaker, suggestive evidence for linkage
on chromosomes 1q, 13q and 22q for alcohol depen-
dence, on 2q, 9q and 19p for symptom count
[147] . Fur-
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
83
ther loci identified in the COGA that were replicated in a
follow-up
[148] include chromosomes 1 and 7.
Other linkage studies have focused on disease-related
phenotypes rather than on the diagnosis per se. An anal-
ysis combining the diagnostic phenotype with an electro-
physiological variable (amplitude of the P300, an event-
related potential related to the processing of rare or nov-
el events) also pointed to a risk locus on chromosome 4q
[149] . A quantitative trait, maximum drinks in a 24-hour
period, also showed linkage on chromosome 4q
[150] , on
chromosome 9 for age of onset, on chromosomes 1 and
11 for initial response to alcohol, on chromosomes 1, 6
and 22 for tolerance, on chromosomes 12 and 18 for max-
imum drinks, and on chromosome 2 for withdrawal
symptoms
[151] . As these linkage studies only provide
information about chromosomal regions associated with
a disorder, a challenge for future research remains the
identification of the specific risk genes at these chromo-
somal loci.
Genome-Wide Association Studies
Recent advances in large-scale array-based genotyping
have opened up the possibility to conduct GWAS that by
now largely replaced linkage studies as the method of
choice for detecting genetic risk loci in an unbiased man-
ner. These studies combine several advantages of linkage
studies and candidate gene-based association studies: (a)
the coverage of the entire genome without the need for
strong a priori hypotheses and of candidate gene ap-
proaches; (b) the high resolution providing information
about single genes; (c) the possibility to include sporadic
and not only familial cases. In GWAS, several hundred
thousands or millions of markers across the entire ge-
nome are analyzed in order to identify differences in allele
or genotype frequencies between individuals positive or
negative for a phenotype of interest.
Since their introduction, GWAS have proven to be
successful in identifying numerous genetic factors con-
tributing to the risk for complex somatic diseases like
breast cancer
[152, 153] , colorectal cancer [154] , type 2
diabetes
[155, 156] or heart disease [157] , but also for psy-
chiatric disorders like schizophrenia or bipolar disorder
[158–160] . The GWAS approach is not without limita-
tions, however. Due to the risk of false positives resulting
from the extraordinary amount of multiple testing inher-
ent to the method, conservative statistical corrections of
the significance level are required that are likely to result
in false-negative data. To overcome this limitation, very
large sample sizes are required to obtain the necessary
statistical power. Moreover, GWAS are designed to iden-
tify relatively common polymorphisms associated with
the risk for a disease. Rare genetic variants with poten-
tially high influence on individual risk might thus be
overlooked by GWAS, but can be still captured by linkage
studies.
Regarding alcohol-related disorders, GWAS have
identified a number of chromosomal regions and candi-
date genes for AUDs, alcohol-related phenotypes and co-
morbid disorders. Johnson et al.
[161] published in 2006
a pioneering GWAS of alcohol dependence from the
COGA. The study was performed on 100,000 markers in
120 patients and 160 healthy controls. Fifty-one clusters
of polymorphisms were identified that provided informa-
tion about novel candidate genes for alcohol dependence,
including genes involved in gene regulation, cell signal-
ing, brain development and cell adhesion. Among these
were the genes of the cell adhesion molecules cadherin 11
(CDH11) and cadherin 13 (CDH13) . In the following
years, several GWAS have been conducted to identify risk
factors for alcohol dependence ( table1 , first part) as well
as genetic modifiers of alcohol consumption in the healthy
population ( table 1 , second part). Additionally, a few
GWAS have been aimed to identify associations with al-
cohol-related phenotypes, including both clinical fea-
tures like withdrawal severity and basic behavioral and
neurophysiological measures ( table1 , third part).
Several risk loci identified by GWAS confirm previ-
ously observed associations of genes involved in alcohol
metabolism with a risk for alcohol dependence and drink-
ing behavior. For example, Frank et al.
[162] detected ge-
nome-wide significance for marker rs1789891 in the
ADH gene cluster on chromosome 4q in a sample of 1,333
patients and 2,168 controls. This finding is well in line
with prior results of linkage analyses reporting an asso-
ciation of this chromosomal region with alcohol depen-
dence and alcohol-related phenotypes
[147] . rs1789891 is
also in high linkage disequilibrium with the functional
ADHC1 Arg272Gln polymorphism that has been shown
to affect alcohol oxidation in vitro. Further genome-wide
evidence for a role of the ADH gene on the chromosome
4q cluster in the risk of alcohol dependence was provided
by Park et al.
[163] , who additionally found genome-wide
significance for the functional ALDH2 rs671 that has re-
peatedly been associated with alcohol consumption levels
and risk for alcohol dependence (see above). This asso-
ciation was also confirmed at the genome-wide level in a
2-stage GWAS for alcohol use in a Japanese population
performed by Takeuchi et al.
[164] . For both, nominally
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
84
Authors, year Study name Number of patients/controls Identified gene loci
GWAS of alcohol dependence
Johnson et al.
[161], 2006
COGA 120 patients, 160 controls 51 gene loci remain significant after Monte Carlo simulation-
based correction, including CDH11, CDH13
FDR threshold less conservative than in later GWAS
Treutlein et al.
[182], 2009
n.a. Initial sample: 487 patients, 1,358 controls;
follow-up sample: 1,024 patients, 966 controls
Genome-wide significance for 2 intergenic loci on chromosome
2q35: rs7590720, rs1344694 in the combined sample
15 nominally significant SNPs were replicated, including SNPs
in CDH13 and ADH1C
Edenberg et al.
[169], 2010
COGA Initial sample: 1,192 patients, 692 controls;
follow-up in 292 pedigrees
No SNP with genome-wide significance
Initial sample and follow-up sample provide converging
evidence for a gene cluster on chromosome 11, encompassing
SLC22A18, PHLDA2, NAP1L4, SNORA54, CARS, OSBPL5
Bierut et al.
[168], 2010
SAGE (includes
COGA, FSCD,
COGEND)
1,897 patients, 1,932 controls; 2 replication
samples
No SNP with replicated genome-wide significance
Replicated nominal significance for GABRA2 SNPs
Lind et al.
[200], 2010
OZALC Initial sample: 1,224 patients, 1,162 controls
from the Australian Twin Registry; replication
sample: 555 patients, 2,768 controls from the
Netherlands
No replicated genome-wide significance
Meta-analysis of initial and replication samples provides
evidence for risk variants in genes coding ion channels and cell
adhesion molecules
Kendler et al.
[186], 2011
MGS2 3,169 controls from the MGS2 study screened
for symptoms of alcohol dependence
No genome-wide significance
Most significant loci KCNMA1 (Caucasians) and SLC35B4
(African-Americans)
Wang et al.
[166], 2011
COGA, OZALC 272 nuclear families from 116 pedigrees
(COGA); replication in the OZALC cohort
(twin study)
Low density GWAS with 11,120 SNPs and less conservative
threshold (p < 10 3)
Genome-wide significance at p < 10 8 for DSCMAL1
Further replicated loci include TPARP, CYFIP2, THEMIS,
PSG11
Wang et al.
[167], 2011
COGA, SAGE,
OZALC
COGA: 1,025 patients, 569 controls; SAGE: 637
patients, 1,032 controls; replication in OZALC:
1,650 patients, 1,684 controls from 778 nuclear
families
No genome-wide significance
Replicated novel associations with KIAA0040, THSD7B, NRD1
Confirmed association with PKNOX2
Frank et al.
[162], 2012
n.a. 1,333 patients, 2,168 controls Genome-wide significance for rs1789891 in ADH1 gene cluster,
in LD with functional ADH1C Arg272Gln, nominally replicated
in COGA sample
Feasibility of polygenic score analysis was demonstrated
Zuo et al.
[172, 173],
2011, 2012
COGA, SAGE 2,090 patients, 2,026 controls Genome-wide significance for KIAA0040 (chromosome 1q)
Replicated association for 90-Mb region around the PHF3-
PTP4A1 locus on chromosome 6q12
Biernacka et al.
[201], 2013
SAGE 1,165 patients, 1,379 controls Gene set enrichment analysis, not aimed at single genome-wide
significant loci
Pathway analysis points to potential involvement of ketone
metabolism and ligand-receptor interactions
Zuo et al.
[202], 2013
COGA, SAGE Initial sample: 1,409 patients, 1,518 controls;
replication sample: 6,438 European-Australian
family subjects including 1,645 alcohol-
dependent patients
Genome-wide significance for NKAIN1-SERINC2 in subjects of
European, but not African descent
471 SNPs nominally associated, 53 survived region- and cohort-
wide correction, 92 were replicated
Zuo et al.
[203], 2013
SAGE, COGA Initial sample: 818 European-American patients
with alcohol and nicotine codependence, 1,396
controls; 2 replication samples
Genome-wide significant association of combined alcohol and
nicotine dependence with rs7445832 in IPO11-HTR1A region
on chromosome 5q
4 genome-wide significant SNPs in IPO11-HTR1A region
revealed by meta-analysis of discovery and replication samples
Park et al.
[163], 2013
n.a. Initial sample: 117 alcohol-dependent patients,
279 controls; replication sample: 504 patients,
471 controls
Genome-wide significane for rs1442492 and rs10516441 in
ADH7 gene region and ALDH2 rs671
Multiple nominally significant SNPs in ADH gene cluster on
4q22–q23
Table 1. Overview of GWAS on alcohol-related disorders
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
85
Table 1 (continued)
Authors, year Study name Number of patients/controls Identified gene loci
Quillen et al.
[165], 2014
n.a. 352 males, 243 females from an isolated rural
Chinese population
Genome-wide significance for ALDH2*2
ALDH2*2 also shows genome-wide significant association with
maximum alcohol intake and flushing response
Gelernter et al.
[171], 2014
GCD Initial sample: 379 European-Americans, 3,318
African-Americans; replication sample: 1,746
European-Americans, 803 African-Americans;
further replication in SAGE sample and in a
German sample; total n = 16,087 subjects
Replicated genome-wide significance for ADH1B, ADH1C
Novel genome-wide significant risk variant rs1437396 between
MTIF2 and CCDC88A
GWAS of alcohol consumption
Schumann et al.
[204], 2011
n.a. 26,316 individuals from 12 population-based
samples
Genome-wide significance for AUTS2 rs6943555
Nominal significant association with RASGRF2 rs26907
Baik et al.
[205], 2011
n.a. Initial sample: 1,721 males; replication sample:
1,113 males
Replicated genome-wide significance for SNPs on chromosome
12q24, including C12ORF51 rs2074356, C12orf51 in LD with
ALDH2, CCDC63, MYL2
Genome-wide significance, but no replication for SNPs in
CCDC63, OAS3, CUX2 and RPH3A
Takeuchi et al.
[164], 2011
n.a. Initial sample: 733 ever-drinkers, 729
nondrinkers; replication sample: 2,794 drinkers,
1,521 occasional drinkers, 1,351 nondrinkers
genome-wide significance for ALDH2 rs671 in both discovery
and replication sample
replicated nominal significance for ADH1B rs1229984
Chen et al.
[206], 2012
n.a. Initial sample: 904 Caucasians, 3 replication
samples (n = 1,972, 761, 2,955)
Genome-wide significance for SNP clusters in ANKRD7, CYTL1
Most significant SNPs ANKRD7 rs4295599, CYTL1 rs16836497
Heath et al.
[170], 2011
n.a. 8,754 individuals including 2,062 patients No genome-wide significance
Convergent evidence for association of ANKS1A rs2140418 and
TMEM108 rs10935045 with drinking severity
Kapoor et al.
[207], 2013
COGA, SAGE 2,322 subjects from 118 extended families;
2,593 subjects from a case-control study
No genome-wide significance
Meta-analysis suggests association of LMO1 and PLCL1 with
maximum number of drinks
Nominal association of AUTS2, INADL, C15ORF32 and HIP1
with measures of alcohol consumption in meta-analysis
Pan et al.
[208], 2013
COGA, SAGE 1,059 subjects from COGA study, 1,628 subjects
from SAGE study; replication in OZALC
sample
Genome-wide significant association of rs11128951 near SGOL1
with maximum number of drinks
Meta-analysis additionally shows association of rs17144687 near
DTWD2, rs12108602 near NDST4 and rs2128158 in KCNB2
with maximum number of drinks
GWAS of alcohol-related phenotypes
Wang et al.
[176], 2013
COGA 2,322 subjects from 118 extended families;
replication in SAGE and OZALC samples
Genome-wide significance for 3 SNPs in C15ORF53 gene with
alcohol dependence symptom count, albeit only nominally
significant after inflation correction
Wang et al.
[209], 2012
COGA 461 patients, 408 controls No genome-wide significance
4 SNPs nominally associated with alcohol withdrawal severity:
rs770182 (near EFNA5, chromosome 5q21); rs10975990,
rs10758821 and rs1407862 (KDM4C, 9p24.1)
Joslyn et al.
[179], 2010
n.a. 367 healthy individuals with positive family
history for alcohol dependence
Gene set enrichment analysis, not aimed at single genome-wide
significant loci
156 loci pointing to 173 genes, mostly in neuronal signaling
pathways, influence alcohol level of response
Zlojutro et al.
[177], 2011
COGA 1,192 patients, 692 controls; replication in 262
pedigrees (1,195 individuals)
No genome-wide significance, ARID5A rs4907240 and HTR7
rs7916403 nominally associated with novelty-related theta
oscillations
HTR7 rs7916403 nominally associated with diagnosis of alcohol
dependence
FDR = False discovery rate; n.a. = not available; SNP = single-nucleotide polymorphism; SAGE = Study of Addiction: Genetics and Environment;
FSCD = Family Study of Cocaine Dependence; COGEND = Collaborative Genetic Study of Nicotine Dependence; OZALC = Australian Twin-Family Study
of Alcohol Use Disorder; MGS2 = Molecular Genetics of Schizophrenia; GCD = GWAS discovery samples; LD = linkage disequilibrium.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
86
classified drinking behavior (nondrinkers vs. occasional
drinkers vs. drinkers) and level of alcohol consumption,
ALDH2 rs671 came out as the strongest predictor with
genome-wide significance. The most recent replication of
the prominent role for ALDH2 in the modulation of
drinking behavior in East Asians comes from a GWAS by
Quillen et al.
[165] , which was performed in an isolated
rural Chinese sample.
In addition to these highly expected findings, GWAS
have also helped to identify novel risk loci for alcohol de-
pendence and related phenotypes, albeit resulting in
more ambiguous results (see table 1 for a summary).
While some studies did not yield any genetic variations
with genome-wide significance, others identified several
candidate loci, many of which could, however, not be re-
liably replicated subsequently. A reason for the inconclu-
sive results of most GWAS might be insufficient statistical
power resulting from required corrections for multiple
testing. Means to circumvent this problem include meta-
analyses of the literature and reanalyses of pre-existing
original data, both of which increase statistical power. For
example, Wang et al.
[166, 167] combined data sets from
the SAGE, the COGA and the OZALC GWAS
[168–170]
and identified several gene loci not reported in the initial
publications ( table1 , first part). In a very recent GWAS,
Gelernter et al.
[171] included a total of 16,087 subjects,
both newly recruited and from pre-existing samples. The
study yielded strong genome-wide associations with vari-
ants in the ADH gene cluster and additionally identified
a putatively regulatory variant rs1437396, located be-
tween the MTIF2 and CCDC88A genes as a novel risk
variant with replicated genome-wide significance
[171] .
A further scientific challenge posed by GWAS is due
to the identification of numerous associations that have
been observed with genetic variants that are either extra-
genic or located in thus far poorly characterized genes. To
understand the biological processes that underlie the ob-
served associations, it is therefore necessary to further
characterize the impact of the identified variants at both
the molecular and system levels. At a molecular level, ex-
pression quantitative trait locus studies allow the identi-
fication of both cis and trans regulatory associations of
genetic variations with gene expression levels. Zuo et al.
[172, 173] combined the GWAS approach with expres-
sion quantitative trait locus in the analysis of data from
2,090 alcohol-dependent patients and 2,026 controls. The
observed associations of variants in PHD finger protein 3
gene ( PHF3), protein tyrosine phosphatase type IVA,
member 1 gene (PTP4A1) region [173] , and KIAA0040
[172] genes with alcohol dependents corresponded to re-
sults of transcriptome-wide expression analyses showing
that the transcripts of these genes were involved in regu-
latory mechanisms of other alcohol-related candidate
genes
[172, 173] .
At the system level, it has been suggested that the re-
lated endophenotypes are more directly related to the
molecular effect of a gene variant than the clinical pheno-
type
[174, 175] . A potential endophenotype solely based
on clinical criteria is the symptom count as a crude, but
straightforward measure of disease severity that can be
applied in both patients and healthy controls. In a GWAS
directed at alcohol dependence symptom count, Wang et
al.
[176] found converging evidence for a role of the thus
far poorly characterized C15ORF53 gene in the etiology
of alcohol dependence-related phenotypes. Other endo-
phenotypes show a weaker statistical association with the
clinical phenotype of interest, but might be more directly
related to the underlying pathophysiological mecha-
nisms. For example, novelty-related brain oscillations in
the theta band have previously been found to be linked to
alcohol dependence. Zlojutro et al.
[177] performed a
2-stage GWAS on event-related theta oscillations in 1,192
patients and 692 controls, using 262 pedigrees (1,095 in-
dividuals) as a replication sample. They identified ge-
netic variations in the ARID5A gene (rs4907240) and in
the gene coding for the serotonin receptor 7 ( HTR7 ,
rs7916403) to be associated with novelty-related theta ac-
tivity.
From Genes to Pathways
The genes associated with alcohol dependence in both
GWAS and replicated candidate-based association stud-
ies are, at first sight, a rather heterogeneous group, with
their products being involved in diverse biological func-
tions, including (peripheral) alcohol metabolism, neuro-
transmission, intracellular signaling, but also seemingly
unrelated processes like immune function. For example,
the KIAA0040 risk variant identified in a GWAS by Zuo
et al.
[172] and replicated in the meta-analysis by Wang
et al.
[167] , encodes an HLA1-DR11-restricted T cell epi-
tope with yet unknown function
[178] . However, expres-
sion quantitative trait locus analysis has demonstrated
that the expression of the KIAA0040 gene is associated
with the expression of multiple other genes involved in
neurotransmission, including several genes previously
implicated in alcohol dependence and other psychiatric
disorders, such as DRD2-TTC12, DRD3, HTR1B, GRM5
and OPRD1 [172] . Further evidence for a role of genes
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
87
involved in neurotransmission and intracellular signaling
comes from a gene set enrichment analysis (GSEA) on
GWAS data performed by Joslyn et al.
[179] . GSEA is a
computational method that assesses differential distribu-
tion of a priori defined sets of genes between populations
of interest. Joslyn et al. observed that level of alcohol re-
sponse phenotypes (i.e. self-report on the effects of etha-
nol, subjective high assessment, body sway
[180] ) showed
a systematic distribution difference in 173 genes, includ-
ing CDH13 and XRCC5 , which had been identified as sus-
ceptibility genes for ethanol sensitivity and alcohol de-
pendence elsewhere
[161, 181, 182] . Multiple genes iden-
tified in the GSEA were related to glutamatergic signaling,
a finding well in line with previous results of animal stud-
ies and candidate gene analyses
[183, 184] . Additionally,
the muscarinic receptor (CHRM2) has been replicated in
2 GWAS
[185, 186] and several other well-replicated can-
didate genes (i.e. >10 studies), such as GABRA2 [168] ,
MAOA [166] , GRIN2B [179] and ANKK1 [186] , have
emerged in recent GWAS, further confirming the role of
the cholinergic, dopaminergic and GABAergic transmit-
ter systems. Many of these variants do not only affect the
risk for AUD, but also have an influence on other sub-
stance-related disorders (e.g. GABRA2, CHRM2, NFKB1)
[68, 106, 132, 135, 187] .
Summary and Future Perspectives
Considerable progress has been made in the research
of genetic contributions to alcohol dependence. For sev-
eral genetic variations, a prominent role in the risk for
developing alcohol-related disorders is largely undisput-
ed. Most prominently, several functional variations in
genes involved in alcohol metabolism, namely the ADH
and ALDH genes, play a major role in modulating alco-
hol-related behaviors and thereby affect the individual
risk for AUD. This is particularly true for the ALDH2 * 2
variant that strongly predicts drinking behavior in East
Asian populations, with effect sizes only comparable to
the robust association of APOE/TOMM40 with dementia
[188] . In populations of European ancestry, in which the
coding variations in ALDH are far less common, the
greatest proportion of the individual genetic risk is yet
unexplained and probably reflects the accumulating ef-
fects of multiple genes with minor individual impact.
However, the importance of alcohol-metabolizing en-
zymes becomes evident as most robust associations in Eu-
ropeans have been observed in the ADH gene cluster.
With recent advances in high-throughput genotyping
and bioinformatics, GWAS in large samples become fea-
sible that allow the robust detection of variants beyond
the metabolism-related genes
[171] . Additionally, rare
genetic variations that are insufficiently detected by
GWAS as well as gene-gene and GxE interactions
[189–
191] are likely to contribute heritability, phenomena that
are actually common to the genetics of most psychiatric
disorders
[192] .
A more comprehensive characterization of the genetic
susceptibility to AUD will most likely require (a) a more
refined characterization of alcohol-related phenotypes
(e.g. subtypes of alcohol dependence with higher vs. low-
er heritability) and (b) advanced analytic techniques that
make use of both array and next-generation sequencing
technologies
[193] . A comprehensive discussion of stud-
ies applying these approaches is beyond the scope of this
review, but we refer to recent review papers that cover
GWAS of alcohol dependence
[182, 186, 194–196] . A po-
tential future challenge might be posed by the change in
diagnostic criteria with the publication of DSM-V
[6] .
While merging the diagnostic criteria into a single entity
‘substance use disorders’ with varying degrees of severity
might ease the definition of endophenotypes or of semi-
quantitative approaches like symptom count
[176] , great
care must be taken when comparing studies based on
DSM-V with previously published DSM-IV-based work.
Also, GxE interactions must be considered
[189–191] , al-
though keeping in mind that the GxE approach is one of
the most challenging and complex of all conceptual
frameworks applied in the genetic investigation of AUDs
[197] .
From the statistical point of view, lack of power is a
weakness of most genetic investigations in psychiatry, a
problem that is actually shared by many disciplines with-
in neurosciences and contributes to poor replicability of
data
[198] ; very large samples are required to overcome
this essential limitation. Preliminary findings must be
confirmed in independent studies, and much work re-
mains to be done to elucidate the neural mechanisms un-
derlying the genetic associations. Nevertheless, owing to
the emergence of new technologies allowing to study even
larger sample sizes, progress will be swift. The future will
involve studies of epigenetic factors, copy number vari-
ants and other rare genetic variations, and gene expres-
sion data.
As we learn more about the genetic framework of
AUD, we find that different genetic variants lead to vary-
ing responses to treatment options, which might improve
our ability to design individualized therapy concepts
[135, 199] . One example for the potential pharmacoge-
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
88
netic approaches is the association of the OPRM1 As-
n40Asp variation with the efficacy of the opioid antago-
nist naltrexone on relapse prevention
[117] , although it
remains to be established whether this association is of
clinical significance. Irrespective of the actual magnitude
of the effect of OPRM1 Asn40Asp, this observation
should stimulate large-scale studies investigating the role
of genetic markers in the response to pharmacotherapy,
with the possibility to perform GWAS on treatment out-
comes being a promising approach in the near future.
More generally, from a clinical perspective, distinct
phenotype characterization of a vast number of individu-
als including data on AUDs and alcohol-related pheno-
types, comorbid psychiatric and medical disorders as well
as therapy response is needed to facilitate the identifica-
tion of patients that might benefit from certain therapeu-
tic lines. The above considerations refer both to pharma-
cotherapeutic and to psychotherapeutic approaches. The
identification of reliable genetic markers predicting the
response to psychotherapy, medications and their com-
bination remains a great scientific challenge for the fu-
ture.
Acknowledgements
This work was supported by the Deutsche Forschungsgemein-
schaft (SFB 779, TP A8). We thank Claudia Hägele for her helpful
comments on the paper.
References
1 Rehm J, Room R, van den Brink W, Jacobi F:
Alcohol use disorders in EU countries and
Norway: an overview of the epidemiology.
Eur Neuropsychopharmacol 2005;
15: 377–
388.
2 Grant BF, Dawson DA, Stinson FS, Chou SP,
Dufour MC, Pickering RP: The 12-month
prevalence and trends in DSM-IV alcohol
abuse and dependence: United States, 1991–
1992 and 2001–2002. Drug Alcohol Depend
2004;
74: 223–234.
3 Li T-K, Hewitt BG, Grant BF: The alcohol de-
pendence syndrome, 30 years later: a com-
mentary. The 2006 H. David Archibald lec-
ture. Addiction 2007;
102: 1522–1530.
4 American Psychiatric Association: Diagnos-
tic and Statistical Manual of Mental Disor-
ders, ed 4. Washington, American Psychiatric
Association, 1994.
5 World Health Organization: International
Classification of Disease, ed 10. Geneva,
WHO, 1993.
6 American Psychiatric Association: Diagnos-
tic and Statistical Manual of Mental Disorders
(DSM-5). Washington, American Psychiatric
Association, 2013.
7 American Psychiatric Association: Diagnos-
tic and Statistical Manual of Mental Disor-
ders. Washington, American Psychiatric As-
sociation, 1987.
8 Buscemi L, Turchi C: An overview of the ge-
netic susceptibility to alcoholism. Med Sci
Law 2011;
51(suppl 1):S2–S6.
9 Dragan WŁ, Oniszczenko W, Czerski PM,
Dmitrzak-Weglarz M: Family-based associa-
tion study of dopaminergic gene polymor-
phisms and EAS temperamental traits. Psy-
chiatr Pol 2013;
47: 185–195.
10 Heath AC, Bucholz KK, Madden PA, Dinwid-
die SH, Slutske WS, Bierut LJ, Statham DJ,
Dunne MP, Whitfield JB, Martin NG: Genet-
ic and environmental contributions to alco-
hol dependence risk in a national twin sam-
ple: consistency of findings in women and
men. Psychol Med 1997;
27: 1381–1396.
11 Kałwa A: Impulsivity and decision making in
alcohol-addicted individuals. Psychiatr Pol
2013;
47: 325–334.
12 Kendler KS, Heath AC, Neale MC, Kessler
RC, Eaves LJ: A population-based twin study
of alcoholism in women. JAMA 1992;
268:
1877–1882.
13 McGue M: The behavioral genetics of alco-
holism. Curr Dir Psychol Sci 1999;
8: 109–115.
14 Prescott CA, Kendler KS: Genetic and envi-
ronmental contributions to alcohol abuse and
dependence in a population-based sample of
male twins. Am J Psychiatry 1999;
156: 34–40.
15 Kendler KS, Schmitt E, Aggen SH, Prescott
CA: Genetic and environmental influences on
alcohol, caffeine, cannabis, and nicotine use
from early adolescence to middle adulthood.
Arch Gen Psychiatry 2008;
65: 674–682.
16 Bohman M, Sigvardsson S, Cloninger CR:
Maternal inheritance of alcohol abuse. Cross-
fostering analysis of adopted women. Arch
Gen Psychiatry 1981;
38: 965–969.
17 Cloninger CR, Bohman M, Sigvardsson S: In-
heritance of alcohol abuse. Cross-fostering
analysis of adopted men. Arch Gen Psychiatry
1981;
38: 861–868.
18 Sigvardsson S, Bohman M, Cloninger CR:
Replication of the Stockholm Adoption Study
of Alcoholism. Confirmatory cross-fostering
analysis. Arch Gen Psychiatry 1996;
53: 681–
687.
19 Heath AC: Twin studies reveal greater con-
cordance between monozygotic (MZ) than
between dizygotic twins (DZ). Alc Health Res
World 1995;19:166–171.
20 Kendler KS, Neale MC, Heath AC, Kessler
RC, Eaves LJ: A twin-family study of alcohol-
ism in women. Am J Psychiatry 1994;
151:
707–715.
21 Pickens RW, Svikis DS, McGue M, Lykken
DT, Heston LL, Clayton PJ: Heterogeneity in
the inheritance of alcoholism. A study of male
and female twins. Arch Gen Psychiatry 1991;
48: 19–28.
22 Buck KJ, Metten P, Belknap JK, Crabbe JC:
Quantitative trait loci involved in genetic pre-
disposition to acute alcohol withdrawal in
mice. J Neurosci 1997;
17: 3946–3955.
23 Crabbe JC, Phillips TJ, Buck KJ, Cunningham
CL, Belknap JK: Identifying genes for alcohol
and drug sensitivity: recent progress and future
directions. Trends Neurosci 1999;
22: 173–179.
24 McBride WJ, Li TK: Animal models of alco-
holism: neurobiology of high alcohol-drink-
ing behavior in rodents. Crit Rev Neurobiol
1998;
12: 339–369.
25 Murphy JM, Stewart RB, Bell RL, Badia-Elder
NE, Carr LG, McBride WJ, Lumeng L, Li T-K:
Phenotypic and genotypic characterization of
the Indiana University rat lines selectively
bred for high and low alcohol preference. Be-
hav Genet 2002;
32: 363–388.
26 Whatley VJ, Johnson TE, Erwin VG: Identifi-
cation and confirmation of quantitative trait
loci regulating alcohol consumption in con-
genic strains of mice. Alcohol Clin Exp Res
1999;
23: 1262–1271.
27 Edenberg HJ: The genetics of alcohol metabo-
lism: role of alcohol dehydrogenase and alde-
hyde dehydrogenase variants. Alcohol Res
Health 2007;
30: 5–13.
28 Hurley TD, Edenberg HJ, Li T-K: Pharma-
cogenomics of alcoholism; in Licinio J, Wong
M-L (eds): Pharmacogenomics: The Search
for Individual Therapies. Weinheim, Wiley-
VCH, 2003, pp 417–441.
29 Thomasson HR, Edenberg HJ, Crabb DW,
Mai XL, Jerome RE, Li TK, Wang SP, Lin YT,
Lu RB, Yin SJ: Alcohol and aldehyde dehydro-
genase genotypes and alcoholism in Chinese
men. Am J Hum Genet 1991;
48: 677–681.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
89
30 Higuchi S, Matsushita S, Imazeki H, Kinoshi-
ta T, Takagi S, Kono H: Aldehyde dehydroge-
nase genotypes in Japanese alcoholics. Lancet
1994;
343: 741–742.
31 Kendler KS, Gardner C, Dick DM: Predicting
alcohol consumption in adolescence from al-
cohol-specific and general externalizing ge-
netic risk factors, key environmental expo-
sures and their interaction. Psychol Med
2011;
41: 1507–1516.
32 Young-Wolff KC, Enoch M-A, Prescott CA:
The influence of gene-environment interac-
tions on alcohol consumption and alcohol use
disorders: a comprehensive review. Clin Psy-
chol Rev 2011;
31: 800–816.
33 Guo G, Elder GH, Cai T, Hamilton N: Gene-
environment interactions: peers’ alcohol use
moderates genetic contribution to adolescent
drinking behavior. Social Sci Res 2009;
38:
213–224.
34 Harden KP, Hill JE, Turkheimer E, Emery RE:
Gene-environment correlation and interac-
tion in peer effects on adolescent alcohol and
tobacco use. Behav Genet 2008;
38: 339–347.
35 Dick DM, Pagan JL, Viken R, Purcell S,
Kaprio J, Pulkkinen L, Rose RJ: Changing en-
vironmental influences on substance use
across development. Twin Res Hum Genet
2007;
10: 315–326.
36 Dick DM, Rose RJ, Viken RJ, Kaprio J, Ko-
skenvuo M: Exploring gene-environment in-
teractions: socioregional moderation of alco-
hol use. J Abnorm Psychol 2001;
110: 625–632.
37 Rose RJ, Dick DM, Viken RJ, Kaprio J: Gene-
environment interaction in patterns of ado-
lescent drinking: regional residency moder-
ates longitudinal influences on alcohol use.
Alcohol Clin Exp Res 2001;
25: 637–643.
38 Legrand LN, Keyes M, McGue M, Iacono
WG, Krueger RF: Rural environments reduce
the genetic influence on adolescent substance
use and rule-breaking behavior. Psychol Med
2008;
38: 1341–1350.
39 Agrawal A, Grant JD, Littlefield A, Waldron
M, Pergadia ML, Lynskey MT, Madden PAF,
Todorov A, Trull T, Bucholz KK, Todd RD,
Sher K, Heath AC: Developing a quantitative
measure of alcohol consumption for genomic
studies on prospective cohorts. J Stud Alcohol
Drugs 2009;
70: 157–168.
40 Heath AC, Jardine R, Martin NG: Interactive
effects of genotype and social environment on
alcohol consumption in female twins. J Stud
Alcohol 1989;
50: 38–48.
41 Koopmans JR, Slutske WS, van Baal GC,
Boomsma DI: The influence of religion on al-
cohol use initiation: evidence for genotype ×
environment interaction. Behav Genet 1999;
29: 445–453.
42 Dawson DA, Grant BF, Li T-K: Quantifying
the risks associated with exceeding recom-
mended drinking limits. Alcohol Clin Exp
Res 2005;
29: 902–908.
43 Błachut M, Badura-Brzoza K, Jarzab M, Gor-
czyca P, Hese RT: Dual diagnosis in psychoac-
tive substance abusing or dependent persons.
Psychiatr Pol 2013;
47: 335–352.
44 Grant BF, Stinson FS, Dawson DA, Chou SP,
Dufour MC, Compton W, Pickering RP, Ka-
plan K: Prevalence and co-occurrence of sub-
stance use disorders and independent mood
and anxiety disorders: results from the na-
tional epidemiologic survey on alcohol and
related conditions. Arch Gen Psychiatry 2004;
61: 807–816.
45 Grant BF, Stinson FS, Dawson DA, Chou SP,
Ruan WJ, Pickering RP: Co-occurrence of
12-month alcohol and drug use disorders and
personality disorders in the United States: re-
sults from the national epidemiologic survey
on alcohol and related conditions. Arch Gen
Psychiatry 2004;
61: 361–368.
46 Szewczuk-Bogusławska M, Flisiak-Antonijc-
zuk H: Will new diagnostic criteria facilitate
the diagnostic process of ADHD in adults?
Psychiatr Pol 2013;
47: 293–302.
47 Betkowska-Korpała B: Personality in the big
five model and maintaining abstinence after
one year follow-up. Psychiatr Pol 2012;
46:
387–399.
48 Grzywacz A, Małecka I, Korostyński M,
Przewłocki R, Bieńkowski P, Samochowiec J:
GABA-A receptor genes do not play a role in
genetics of Lesch’s typology in Caucasian sub-
jects. Arch Med Sci 2012;
8: 357–361.
49 Samochowiec A: The influence of parents
personality measured by temperamental and
character inventory (TCI) on course of alco-
holism characterized by Cloninger’s and
Lesch’s typologies. Ann Acad Med Stetin
2010;
56: 33–39.
50 Samochowiec J, Kucharska-Mazur J, Grzy-
wacz A, Pelka-Wysiecka J, Mak M, Samo-
chowiec A, Bienkowski P: Genetics of Lesch’s
typology of alcoholism. Prog Neuropsycho-
pharmacol Biol Psychiatry 2008;
32: 423–427.
51 Heinz A, Friedel E, Müller DJ, Puls I, Wrase J:
Genetic research with intermediate pheno-
types: phenocopies, perspectives and pitfalls.
Addiction 2007;
102: 1696–1697.
52 Mayfield RD, Harris RA, Schuckit MA: Ge-
netic factors influencing alcohol dependence.
Br J Pharmacol 2008;
154: 275–287.
53 Puls I, Gallinat J: The concept of endopheno-
types in psychiatric diseases meeting the ex-
pectations? Pharmacopsychiatry 2008;
41
(suppl 1):S37–S43.
54 Samochowiec J, Grzywacz A, Kucharska-Ma-
zur J, Samochowiec A, Horodnicki J, Pelka-
Wysiecka J, Syrek S: Family-based and case-
control association studies of glutamate re-
ceptor GRIK3 Ser310Ala polymorphism in
Polish patients and families with alcohol de-
pendence. Neurosci Lett 2006;
396: 159–162.
55 Bevilacqua L, Goldman D: Genes and addic-
tions. Clin Pharmacol Ther 2009;
85: 359–361.
56 Ducci F, Goldman D: Genetic approaches to
addiction: genes and alcohol. Addiction 2008;
103: 1414–1428.
57 Landgren S, Berglund K, Jerlhag E, Fahlke C,
Balldin J, Berggren U, Zetterberg H, Blennow
K, Engel JA: Reward-related genes and per-
sonality traits in alcohol-dependent individu-
als: a pilot case control study. Neuropsycho-
biology 2011;
64: 38–46.
58 Rietschel M, Treutlein J: The genetics of alco-
hol dependence. Ann NY Acad Sci 2013;
1282:
39–70.
59 Schuckit MA: An overview of genetic influ-
ences in alcoholism. J Subst Abuse Treat 2009;
36:S5–S14.
60 Vetulani J: Neurochemistry of impulsiveness
and aggression. Psychiatr Pol 2013;
47: 103–
115.
61 Stamatoyannopoulos G, Chen SH, Fukui M:
Liver alcohol dehydrogenase in Japanese:
high population frequency of atypical form
and its possible role in alcohol sensitivity. Am
J Hum Genet 1975;
27: 789–796.
62 Crabb DW, Edenberg HJ, Bosron WF, Li TK:
Genotypes for aldehyde dehydrogenase defi-
ciency and alcohol sensitivity. The inactive
ALDH2(2) allele is dominant. J Clin Invest
1989;
83: 314–316.
63 Xiao Q, Weiner H, Crabb DW: The mutation
in the mitochondrial aldehyde dehydroge-
nase (ALDH2) gene responsible for alcohol-
induced flushing increases turnover of the en-
zyme tetramers in a dominant fashion. J Clin
Invest 1996;
98: 2027–2032.
64 Grochans E, Grzywacz A, Małecka I, Samo-
chowiec A, Karakiewicz B, Samochowiec J:
Research on associations between selected
polymorphisms of genes DRD2, 5HTT,
GRIK3, ADH4 and alcohol dependence syn-
drome. Psychiatr Pol 2011;
45: 325–335.
65 Kimura Y, Nishimura FT, Abe S, Fukunaga T,
Tanii H, Saijoh K: Polymorphisms in the pro-
moter region of the human class II alcohol de-
hydrogenase (ADH4) gene affect both tran-
scriptional activity and ethanol metabolism in
Japanese subjects. J Toxicol Sci 2009;
34: 89–
97.
66 Luo X, Kranzler HR, Zuo L, Lappalainen J,
Yang B-z, Gelernter J: ADH4 gene variation is
associated with alcohol dependence and drug
dependence in European Americans: results
from HWD tests and case-control association
studies. Neuropsychopharmacology 2006;
31:
1085–1095.
67 Luo X, Zuo L, Kranzler HR, Wang S, Anton
RF, Gelernter J: Recessive genetic mode of an
ADH4 variant in substance dependence in
African-Americans: a model of utility of the
HWD test. Behav Brain Funct 2008;
4: 42.
68 Preuss UW, Ridinger M, Rujescu D, Samo-
chowiec J, Fehr C, Wurst FM, Koller G, Bondy
B, Wodarz N, Debniak T, Grzywacz A, Soyka
M, Zill P: Association of ADH4 genetic variants
with alcohol dependence risk and related phe-
notypes: results from a larger multicenter as-
sociation study. Addict Biol 2011;
16: 323–333.
69 Heinz A, Beck A, Wrase J, Mohr J, Obermay-
er K, Gallinat J, Puls I: Neurotransmitter sys-
tems in alcohol dependence. Pharmacopsy-
chiatry 2009;
42(suppl 1):S95–S101.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
90
70 Heinz A, Goldman D, Gallinat J, Schumann
G, Puls I: Pharmacogenetic insights to mono-
aminergic dysfunction in alcohol depen-
dence. Psychopharmacology (Berl) 2004;
174:
561–570.
71 Samochowiec J: The role of dopamine neuro-
transmission on the development of ethanol
dependence and course of abstinence syn-
drome. Ann Acad Med Stetin 1994;
40: 195–
217.
72 Samochowiec J, Kleinrok Z, Horodnicki J:
The role of dopamine neurotransmission in
ethanol dependence development and etha-
nol withdrawal in rats. Pharmazie 1995;
50:
815–818.
73 Samochowiec J, Kucharska-Mazur J, Kamin-
ski R, Smolka M, Rommelschpacher H, Wer-
nicke C, Tymicz A, Schmidt LG: Norepineph-
rine transporter gene polymorphism is not
associated with susceptibility to alcohol de-
pendence. Psychiatry Res 2002;
111: 229–233.
74 Winterer G, Smolka M, Samochowiec J, Ziller
M, Mahlberg R, Gallinat J, Rommelspacher
H-P, Herrmann WM, Sander T: Association
of EEG coherence and an exonic GABA(B)R1
gene polymorphism. Am J Med Genet B Neu-
ropsychiatr Genet 2003;
117B:51–56.
75 Hendershot CS, Lindgren KP, Liang T,
Hutchison KE: COMT and ALDH2 polymor-
phisms moderate associations of implicit
drinking motives with alcohol use. Addict
Biol 2012;
17: 192–201.
76 Samochowiec J, Kucharska-Mazur J, Grzy-
wacz A, Jabłoński M, Rommelspacher H, Sa-
mochowiec A, Sznabowicz M, Horodnicki J,
Sagan L, Pełka-Wysiecka J: Family-based and
case-control study of DRD2, DAT, 5HTT,
COMT gene polymorphisms in alcohol de-
pendence. Neurosci Lett 2006;
410: 1–5.
77 Tikkanen R, Sjöberg RL, Ducci F, Goldman D,
Holi M, Tiihonen J, Virkkunen M: Effects of
MAOA-genotype, alcohol consumption, and
aging on violent behavior. Alcohol Clin Exp
Res 2009;
33: 428–434.
78 Foroud T, Wetherill LF, Dick DM, Hessel-
brock V, Nurnberger JI Jr, Kramer J, Tischfield
J, Schuckit M, Bierut LJ, Xuei X, Edenberg HJ:
Lack of association of alcohol dependence
and habitual smoking with catechol-O-meth-
yltransferase. Alcohol Clin Exp Res 2007;
31:
1773–1779.
79 Lu RB, Lee JF, Ko HC, Lin WW, Chen K, Shih
JC: No association of the MAOA gene with
alcoholism among Han Chinese males in Tai-
wan. Prog Neuropsychopharmacol Biol Psy-
chiatry 2002;
26: 457–461.
80 Mokrovic G, Matosic A, Hranilovic D, Stefulj
J, Novokmet M, Oreskovic D, Balija M, Maru-
sic S, Cicin-Sain L: Alcohol dependence and
polymorphisms of serotonin-related genes:
association studies. Coll Anthropol 2008;
32(suppl 1):127–131.
81 Grzywacz A, Samochowiec J: Case-control,
family based and screening for DNA se-
quence variation in the dopamine transporter
gene polymorphism DAT 1 in alcohol depen-
dence. Psychiatr Pol 2008;
42: 443–452.
82 Morozova TV, Goldman D, Mackay TFC,
Anholt RRH: The genetic basis of alcoholism:
multiple phenotypes, many genes, complex
networks. Genome Biol 2012;
13: 239.
83 Blum K, Noble EP, Sheridan PJ, Montgomery
A, Ritchie T, Jagadeeswaran P, Nogami H,
Briggs AH, Cohn JB: Allelic association of hu-
man dopamine D
2 receptor gene in alcohol-
ism. JAMA 1990;
263: 2055–2060.
84 Wang F, Simen A, Arias A, Lu Q-W, Zhang
H: A large-scale meta-analysis of the associa-
tion between the ANKK1/DRD2 TAQ1A
polymorphism and alcohol dependence.
Hum Genet 2013;
132: 347–358.
85 Dick DM, Wang JC, Plunkett J, Aliev F, Hin-
richs A, Bertelsen S, Budde JP, Goldstein EL,
Kaplan D, Edenberg HJ, Nurnberger J, Hes-
selbrock V, Schuckit M, Kuperman S,
Tischfield J, Porjesz B, Begleiter H, Bierut LJ,
Goate A: Family-based association analyses of
alcohol dependence phenotypes across DRD2
and neighboring gene ANKK1. Alcohol Clin
Exp Res 2007;
31: 1645–1653.
86 Edenberg HJ, Foroud T, Koller DL, Goate A,
Rice J, Van Eerdewegh P, Reich T, Cloninger
CR, Nurnberger JI, Kowalczuk M, Wu B, Li
TK, Conneally PM, Tischfield JA, Wu W,
Shears S, Crowe R, Hesselbrock V, Schuckit
M, Porjesz B, Begleiter H: A family-based
analysis of the association of the dopamine D
2
receptor (DRD2) with alcoholism. Alcohol
Clin Exp Res 1998;
22: 505–512.
87 Gelernter J, Goldman D, Risch N: The A
1 al-
lele at the D
2 dopamine receptor gene and al-
coholism. A reappraisal. JAMA 1993;
269:
1673–1677.
88 Grzywacz A, Jasiewicz A, Małecka I, Sucha-
necka A, Grochans E, Karakiewicz B, Samo-
chowiec A, Bieńkowski P, Samochowiec J: In-
fluence of DRD2 and ANKK1 polymor-
phisms on the manifestation of withdrawal
syndrome symptoms in alcohol addiction.
Pharmacol Rep 2012;
64: 1126–1134.
89 Samochowiec J, Ladehoff M, Pelz J, Smolka
M, Schmidt LG, Rommelspacher H, Finckh
U: Predominant influence of the 3 -region of
dopamine D
2 receptor gene (DRD2) on the
clinical phenotype in German alcoholics.
Pharmacogenetics 2000;
10: 471–475.
90 Sander T, Ladehoff M, Samochowiec J, Finckh
U, Rommelspacher H, Schmidt LG: Lack of
an allelic association between polymorphisms
of the dopamine D
2 receptor gene and alcohol
dependence in the German population. Alco-
hol Clin Exp Res 1999;
23: 578–581.
91 Munafò MR, Matheson IJ, Flint J: Association
of the DRD2 gene TAQ1A polymorphism
and alcoholism: a meta-analysis of case-con-
trol studies and evidence of publication bias.
Mol Psychiatry 2007;
12: 454–461.
92 Slatkin M: Linkage disequilibrium – under-
standing the evolutionary past and mapping
the medical future. Nat Rev Genet 2008;
9:
477–485.
93 Neville MJ, Johnstone EC, Walton RT: Iden-
tification and characterization of ANKK1: a
novel kinase gene closely linked to DRD2 on
chromosome band 11q23.1. Hum Mutat
2004;
23: 540–545.
94 Suchanecka A, Grzywacz A, Samochowiec J:
ANKK1 gene in psychiatry. Psychiatr Pol
2011;
45: 349–356.
95 Duan J, Wainwright MS, Comeron JM,
Saitou N, Sanders AR, Gelernter J, Gejman
PV: Synonymous mutations in the human
dopamine receptor D
2 (DRD2) affect mRNA
stability and synthesis of the receptor. Hum
Mol Genet 2003;
12: 205–216.
96 Hill SY, Hoffman EK, Zezza N, Thalamuthu
A, Weeks DE, Matthews AG, Mukhopadhy-
ay I: Dopaminergic mutations: within-fami-
ly association and linkage in multiplex alco-
hol dependence families. Am J Med Genet B
Neuropsychiatr Genet 2008;
147B:517–526.
97 Lucht M, Samochowiec A, Samochowiec J,
Jasiewicz A, Grabe HJ, Geissler I, Rimmbach
C, Rosskopf D, Grzywacz A, Wysiecka JP,
Tybura P, Brzuchalski B, Bieńkowski P: In-
fluence of DRD2 and ANKK1 genotypes on
apomorphine-induced growth hormone
(GH) response in alcohol-dependent pa-
tients. Prog Neuropsychopharmacol Biol
Psychiatry 2010;
34: 45–49.
98 Yang B-Z, Kranzler HR, Zhao H, Gruen JR,
Luo X, Gelernter J: Association of haplotyp-
ic variants in DRD2, ANKK1, TTC12 and
NCAM1 to alcohol dependence in indepen-
dent case control and family samples. Hum
Mol Genet 2007;
16: 2844–2853.
99 Yang B-Z, Kranzler HR, Zhao H, Gruen JR,
Luo X, Gelernter J: Haplotypic variants in
DRD2, ANKK1, TTC12, and NCAM1 are
associated with comorbid alcohol and drug
dependence. Alcohol Clin Exp Res 2008;
32:
2117–2127.
100 Garrido E, Palomo T, Ponce G, Garcia-Con-
suegra I, Jimenez-Arriero MA, Hoenicka J:
The ANKK1 protein associated with addic-
tions has nuclear and cytoplasmic localiza-
tion and shows a differential response of Al-
a239Thr to apomorphine. Neurotox Res
2011;
20: 32–39.
101 Hoenicka J, Quinones-Lombrana A, Espana-
Serrano L, Alvira-Botero X, Kremer L, Perez-
Gonzalez R, Rodriguez-Jimenez R, Jimenez-
Arriero MA, Ponce G, Palomo T: The ANKK1
gene associated with addictions is expressed in
astroglial cells and upregulated by apomor-
phine. Biol Psychiatry 2010;
67: 3–11.
102 Luo X, Kranzler HR, Zuo L, Wang S, Blum-
berg HP, Gelernter J: CHRM2 gene predis-
poses to alcohol dependence, drug depen-
dence and affective disorders: results from
an extended case-control structured associa-
tion study. Hum Mol Genet 2005;
14: 2421–
2434.
103 Grzywacz A, Małecka I, Suchanecka A,
Bieńkowski P, Samochowiec J: Family-based
and case-control study of glutamate receptor
GRIK3 Ser310Ala polymorphism in alcohol
dependence. Eur Addict Res 2013;
19: 55–59.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
91
104 Wang JC, Grucza R, Cruchaga C, Hinrichs
AL, Bertelsen S, Budde JP, Fox L, Goldstein
E, Reyes O, Saccone N, Saccone S, Xuei X,
Bucholz K, Kuperman S, Nurnberger J, Rice
JP, Schuckit M, Tischfield J, Hesselbrock V,
Porjesz B, Edenberg HJ, Bierut LJ, Goate
AM: Genetic variation in the CHRNA5 gene
affects mRNA levels and is associated with
risk for alcohol dependence. Mol Psychiatry
2009;
14: 501–510.
105 Wernicke C, Samochowiec J, Schmidt LG,
Winterer G, Smolka M, Kucharska-Mazur J,
Horodnicki J, Gallinat J, Rommelspacher H:
Polymorphisms in the N-methyl-
D -aspar-
tate receptor 1 and 2b subunits are associ-
ated with alcoholism-related traits. Biol Psy-
chiatry 2003;
54: 922–928.
106 Agrawal A, Edenberg HJ, Foroud T, Bierut
LJ, Dunne G, Hinrichs AL, Nurnberger JI,
Crowe R, Kuperman S, Schuckit MA, Beglei-
ter H, Porjesz B, Dick DM: Association of
GABRA2 with drug dependence in the col-
laborative study of the genetics of alcohol-
ism sample. Behav Genet 2006;
36: 640–650.
107 Dick DM, Edenberg HJ, Xuei X, Goate A,
Kuperman S, Schuckit M, Crowe R, Smith
TL, Porjesz B, Begleiter H, Foroud T: Asso-
ciation of GABRG3 with alcohol depen-
dence. Alcohol Clin Exp Res 2004;
28: 4–9.
108 Dick DM, Plunkett J, Wetherill LF, Xuei X,
Goate A, Hesselbrock V, Schuckit M, Crowe
R, Edenberg HJ, Foroud T: Association be-
tween GABRA1 and drinking behaviors in
the collaborative study on the genetics of al-
coholism sample. Alcohol Clin Exp Res
2006;
30: 1101–1110.
109 Sander T, Ball D, Murray R, Patel J, Samo-
chowiec J, Winterer G, Rommelspacher H,
Schmidt LG, Loh EW: Association analysis of
sequence variants of GABA(A) alpha6, beta2,
and gamma2 gene cluster and alcohol depen-
dence. Alcohol Clin Exp Res 1999;
23: 427–431.
110 Winterer G, Mahlberg R, Smolka MN, Sa-
mochowiec J, Ziller M, Rommelspacher
H-P, Herrmann WM, Schmidt LG, Sander
T: Association analysis of exonic variants of
the GABA(B)-receptor gene and alpha elec-
troencephalogram voltage in normal sub-
jects and alcohol-dependent patients. Behav
Genet 2003;
33: 7–15.
111 Chen ACH, Tang Y, Rangaswamy M, Wang
JC, Almasy L, Foroud T, Edenberg HJ, Hes-
selbrock V, Nurnberger J, Kuperman S,
O’Connor SJ, Schuckit MA, Bauer LO,
Tischfield J, Rice JP, Bierut L, Goate A,
Porjesz B: Association of single nucleotide
polymorphisms in a glutamate receptor gene
(GRM8) with theta power of event-related
oscillations and alcohol dependence. Am J
Med Genet B Neuropsychiatr Genet 2009;
150B:359–368.
112 Samochowiec A, Horodnicki JM, Samo-
chowiec J: The influence of parents person-
ality and DRD4 and 5HTT gene polymor-
phisms on predisposition to alcohol depen-
dence in their sons. Psychiatr Pol 2011;
45:
337–347.
113 Van der Zwaluw CS, Engels RCME, Ver-
mulst AA, Rose RJ, Verkes RJ, Buitelaar J,
Franke B, Scholte RHJ: A serotonin trans-
porter polymorphism (5-HTTLPR) predicts
the development of adolescent alcohol use.
Drug Alcohol Depend 2010;
112: 134–139.
114 Kreek MJ: Opiates, opioids and addiction.
Mol Psychiatry 1996;
1: 232–254.
115 Kreek MJ, Nielsen DA, LaForge KS: Genes
associated with addiction: alcoholism, opi-
ate, and cocaine addiction. Neuromolecular
Med 2004;
5: 85–108.
116 Rommelspacher H, Smolka M, Schmidt LG,
Samochowiec J, Hoehe MR: Genetic analysis
of the mu-opioid receptor in alcohol-depen-
dent individuals. Alcohol 2001;
24: 129–135.
117 Smolka M, Sander T, Schmidt LG, Samo-
chowiec J, Rommelspacher H, Gscheidel N,
Wendel B, Hoehe MR: Mu-opioid receptor
variants and dopaminergic sensitivity in al-
cohol withdrawal. Psychoneuroendocrinol-
ogy 1999;
24: 629–638.
118 Gscheidel N, Sander T, Wendel B, Heere P,
Schmidt LG, Rommelspacher H, Hoehe MR,
Samochowiec J: Five exon 1 variants of mu
opioid receptor and vulnerability to alcohol
dependence. Pol J Pharmacol 2000;
52: 27–
31.
119 Sander T, Gscheidel N, Wendel B, Samo-
chowiec J, Smolka M, Rommelspacher H,
Schmidt LG, Hoehe MR: Human mu-opioid
receptor variation and alcohol dependence.
Alcohol Clin Exp Res 1998;
22: 2108–2110.
120 Arias A, Feinn R, Kranzler HR: Association
of an Asn40Asp (A118G) polymorphism in
the mu-opioid receptor gene with substance
dependence: a meta-analysis. Drug Alcohol
Depend 2006;
83: 262–268.
121 Chamorro AJ, Marcos M, Miron-Canelo JA,
Pastor I, Gonzalez-Sarmiento R, Laso FJ: As-
sociation of micro-opioid receptor (OPRM1)
gene polymorphism with response to nal-
trexone in alcohol dependence: a systematic
review and meta-analysis. Addict Biol 2012;
17: 505–512.
122 Xuei X, Dick D, Flury-Wetherill L, Tian H-J,
Agrawal A, Bierut L, Goate A, Bucholz K,
Schuckit M, Nurnberger J, Tischfield J, Ku-
perman S, Porjesz B, Begleiter H, Foroud T,
Edenberg HJ: Association of the kappa-opi-
oid system with alcohol dependence. Mol
Psychiatry 2006;
11: 1016–1024.
123 Carr LG, Foroud T, Bice P, Gobbett T, Iva-
shina J, Edenberg H, Lumeng L, Li TK: A
quantitative trait locus for alcohol consump-
tion in selectively bred rat lines. Alcohol Clin
Exp Res 1998;
22: 884–887.
124 Spence JP, Liang T, Habegger K, Carr LG:
Effect of polymorphism on expression of the
neuropeptide Y gene in inbred alcohol-pre-
ferring and -nonpreferring rats. Neurosci-
ence 2005;
131: 871–876.
125 Hwang BH, Zhang JK, Ehlers CL, Lumeng L,
Li TK: Innate differences of neuropeptide Y
(NPY) in hypothalamic nuclei and central
nucleus of the amygdala between selectively
bred rats with high and low alcohol prefer-
ence. Alcohol Clin Exp Res 1999;
23: 1023–
1030.
126 Thiele TE, Marsh DJ, Ste Marie L, Bernstein
IL, Palmiter RD: Ethanol consumption and
resistance are inversely related to neuropep-
tide Y levels. Nature 1998;
396: 366–369.
127 Lappalainen J, Kranzler HR, Malison R,
Price LH, Van Dyck C, Rosenheck RA, Cra-
mer J, Southwick S, Charney D, Krystal J,
Gelernter J: A functional neuropeptide Y Le-
u7Pro polymorphism associated with alco-
hol dependence in a large population sample
from the United States. Arch Gen Psychiatry
2002;
59: 825–831.
128 Mottagui-Tabar S, Prince JA, Wahlestedt C,
Zhu G, Goldman D, Heilig M: A novel single
nucleotide polymorphism of the neuropep-
tide Y (NPY) gene associated with alcohol
dependence. Alcohol Clin Exp Res 2005;
29:
702–707.
12
9 Zhu G, Pollak L, Mottagui-Tabar S,
Wahlestedt C, Taubman J, Virkkunen M,
Goldman D, Heilig M: NPY Leu7Pro and
alcohol dependence in Finnish and Swedish
populations. Alcohol Clin Exp Res 2003;
27:
19–24.
130 Wetherill L, Schuckit MA, Hesselbrock V,
Xuei X, Liang T, Dick DM, Kramer J, Nurn-
berger JI, Tischfield JA, Porjesz B, Edenberg
HJ, Foroud T: Neuropeptide Y receptor
genes are associated with alcohol depen-
dence, alcohol withdrawal phenotypes, and
cocaine dependence. Alcohol Clin Exp Res
2008;
32: 2031–2040.
131 Grzywacz A, Samochowiec A, Ciechanowicz
A, Samochowiec J: Family-based study of
brain-derived neurotrophic factor (BDNF)
gene polymorphism in alcohol dependence.
Pharmacol Rep 2010;
62: 938–941.
132 Foroud T, Wetherill LF, Kramer J, Tischfield
JA, Nurnberger JI, Schuckit MA, Xuei X,
Edenberg HJ: The tachykinin receptor 3 is
associated with alcohol and cocaine depen-
dence. Alcohol Clin Exp Res 2008;
32: 1023–
1030.
133 Grzywacz A, Kucharska-Mazur J, Samo-
chowiec J: Association studies of dopamine
D
4 receptor gene exon 3 in patients with al-
cohol dependence. Psychiatr Pol 2008;
42:
453–461.
134 Cardon LR, Bell JI: Association study de-
signs for complex diseases. Nat Rev Genet
2001;
2: 91–99.
135 Edenberg HJ, Xuei X, Wetherill LF, Bierut L,
Bucholz K, Dick DM, Hesselbrock V, Ku-
perman S, Porjesz B, Schuckit MA, Tischfield
JA, Almasy LA, Nurnberger JI, Foroud T:
Association of NFKB1, which encodes a sub-
unit of the transcription factor NF-kappaB,
with alcohol dependence. Hum Mol Genet
2008;
17: 963–970.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
92
136 Samochowiec A, Grzywacz A, Kaczmarek L,
Bienkowski P, Samochowiec J, Mierzejewski
P, Preuss UW, Grochans E, Ciechanowicz A:
Functional polymorphism of matrix metal-
loproteinase-9 (MMP-9) gene in alcohol de-
pendence: family and case control study.
Brain Res 2010;
1327: 103–106.
137 Sher KJ, Grekin ER, Williams NA: The de-
velopment of alcohol use disorders. Annu
Rev Clin Psychol 2005;
1: 493–523.
138 Samochowiec J, Lesch KP, Rottmann M,
Smolka M, Syagailo YV, Okladnova O,
Rommelspacher H, Winterer G, Schmidt
LG, Sander T: Association of a regulatory
polymorphism in the promoter region of the
monoamine oxidase a gene with antisocial
alcoholism. Psychiatry Res 1999;
86: 67–72.
139 Sander T, Ostapowicz A, Samochowiec J,
Smolka M, Rommelspacher H, Winterer G,
Schmidt LG: Evaluation of an allelic associa-
tion of the serotonin 5-HT1B G681C poly-
morphism with antisocial alcoholism in the
German population. Addict Biol 2000;
5:
167–172.
140 Sander T, Samochowiec J, Ladehoff M,
Smolka M, Peters C, Riess O, Rommelspach-
er H, Schmidt LG: Association analysis of
exonic variants of the gene encoding the
GABAB receptor and alcohol dependence.
Psychiatr Genet 1999;
9: 69–73.
141 Schmidt LG, Sander T, Kuhn S, Smolka M,
Rommelspacher H, Samochowiec J, Lesch
KP: Different allele distribution of a regula-
tory MAOA gene promoter polymorphism
in antisocial and anxious-depressive alco-
holics. J Neural Transm 2000;
107: 681–689.
142 Baars MY, Müller MJ, Gallhofer B, Netter P:
Relapse (number of detoxifications) in absti-
nent male alcohol-dependent patients as re-
lated to personality traits and types of toler-
ance to frustration. Neuropsychobiology
2013;
67: 241–248.
143 Risch N, Merikangas K: The future of genet-
ic studies of complex human diseases. Sci-
ence 1996;
273: 1516–1517.
144 Psychiatric GCCC, Cichon S, Craddock N,
Daly M, Faraone SV, Gejman PV, Kelsoe J,
Lehner T, Levinson DF, Moran A, Sklar P, Sul-
livan PF: Genomewide association studies: his-
tory, rationale, and prospects for psychiatric
disorders. Am J Psychiatry 2009;
166: 540–556.
145 Long JC, Knowler WC, Hanson RL, Robin
RW, Urbanek M, Moore E, Bennett PH,
Goldman D: Evidence for genetic linkage to
alcohol dependence on chromosomes 4 and
11 from an autosome-wide scan in an Amer-
ican Indian population. Am J Med Genet
1998;
81: 216–221.
146 Reich T, Edenberg HJ, Goate A, Williams JT,
Rice JP, Van Eerdewegh P, Foroud T, Hes-
selbrock V, Schuckit MA, Bucholz K, Porjesz
B, Li TK, Conneally PM, Nurnberger JI,
Tischfield JA, Crowe RR, Cloninger CR, Wu
W, Shears S, Carr K, Crose C, Willig C, Be-
gleiter H: Genome-wide search for genes af-
fecting the risk for alcohol dependence. Am
J Med Genet 1998;
81: 207–215.
147 Prescott CA, Sullivan PF, Kuo P-H, Webb
BT, Vittum J, Patterson DG, Thiselton DL,
Myers JM, Devitt M, Halberstadt LJ, Robin-
son VP, Neale MC, van den Oord EJ, Walsh
D, Riley BP, Kendler KS: Genomewide link-
age study in the Irish Affected Sib Pair Study
of Alcohol Dependence: evidence for a sus-
ceptibility region for symptoms of alcohol
dependence on chromosome 4. Mol Psychi-
atry 2006;
11: 603–611.
148 Foroud T, Edenberg HJ, Goate A, Rice J,
Flury L, Koller DL, Bierut LJ, Conneally PM,
Nurnberger JI, Bucholz KK, Li TK, Hessel-
brock V, Crowe R, Schuckit M, Porjesz B,
Begleiter H, Reich T: Alcoholism suscepti-
bility loci: confirmation studies in a replicate
sample and further mapping. Alcohol Clin
Exp Res 2000;
24: 933–945.
149 Lappalainen J, Kranzler HR, Petrakis I, Som-
berg LK, Page G, Krystal JH, Gelernter J:
Confirmation and fine mapping of the chro-
mosome 1 alcohol dependence risk locus.
Mol Psychiatry 2004;
9: 312–319.
150 Saccone NL, Kwon JM, Corbett J, Goate A,
Rochberg N, Edenberg HJ, Foroud T, Li TK,
Begleiter H, Reich T, Rice JP: A genome
screen of maximum number of drinks as an
alcoholism phenotype. Am J Med Genet
2000;
96: 632–637.
151 Kuo P-H, Neale MC, Riley BP, Webb BT,
Sullivan PF, Vittum J, Patterson DG, Thisel-
ton DL, van den Oord EJ, Walsh D, Kendler
KS, Prescott CA: Identification of suscepti-
bility loci for alcohol-related traits in the
Irish Affected Sib Pair Study of Alcohol De-
pendence. Alcohol Clin Exp Res 2006;
30:
1807–1816.
152 Easton DF, Pooley KA, Dunning AM, Phar-
oah PDP, Thompson D, Ballinger DG,
Struewing JP, Morrison J, Field H, Luben R,
Wareham N, Ahmed S, Healey CS, Bowman
R, collaborators S, Meyer KB, Haiman CA,
Kolonel LK, Henderson BE, Le Marchand L,
Brennan P, Sangrajrang S, Gaborieau V,
Odefrey F, Shen C-Y, et al: Genome-wide as-
sociation study identifies novel breast cancer
susceptibility loci. Nature 2007;
447: 1087–
1093.
153 Stacey SN, Manolescu A, Sulem P, Rafnar
T, Gudmundsson J, Gudjonsson SA, Mas-
son G, Jakobsdottir M, Thorlacius S, Hel-
gason A, Aben KK, Strobbe LJ, Albers-Ak-
kers MT, Swinkels DW, Henderson BE,
Kolonel LN, Le Marchand L, Millastre E,
Andres R, Godino J, Garcia-Prats MD,
Polo E, Tres A, Mouy M, et al: Common
variants on chromosomes 2q35 and 16q12
confer susceptibility to estrogen receptor-
positive breast cancer. Nat Genet 2007;
39:
865–869.
154 Tomlinson I, Webb E, Carvajal-Carmona L,
Broderick P, Kemp Z, Spain S, Penegar S,
Chandler I, Gorman M, Wood W, Barclay E,
Lubbe S, Martin L, Sellick G, Jaeger E, Hub-
ner R, Wild R, Rowan A, Fielding S, How-
arth K, Consortium C, Silver A, Atkin W,
Muir K, Logan R, Kerr D, Johnstone E,
Sieber O, Gray R, et al: A genome-wide as-
sociation scan of tag SNPs identifies a sus-
ceptibility variant for colorectal cancer at
8q24.21. Nat Genet 2007;
39: 984–988.
155 Scott LJ, Mohlke KL, Bonnycastle LL, Willer
CJ, Li Y, Duren WL, Erdos MR, Stringham
HM, Chines PS, Jackson AU, Prokunina-
Olsson L, Ding C-J, Swift AJ, Narisu N, Hu
T, Pruim R, Xiao R, Li X-Y, Conneely KN,
Riebow NL, Sprau AG, Tong M, White PP,
Hetrick KN, Barnhart MW, Bark CW, Gold-
stein JL, et al: A genome-wide association
study of type 2 diabetes in Finns detects mul-
tiple susceptibility variants. Science 2007;
316: 1341–1345.
156 Sladek R, Rocheleau G, Rung J, Dina C, Shen
L, Serre D, Boutin P, Vincent D, Belisle A,
Hadjadj S, Balkau B, Heude B, Charpentier
G, Hudson TJ, Montpetit A, Pshezhetsky
AV, Prentki M, Posner BI, Balding DJ, Mey-
re D, Polychronakos C, Froguel P: A ge-
nome-wide association study identifies nov-
el risk loci for type 2 diabetes. Nature 2007;
445: 881–885.
157 McPherson R, Pertsemlidis A, Kavaslar N,
Stewart A, Roberts R, Cox DR, Hinds DA,
Pennacchio LA, Tybjaerg-Hansen A, Fol-
som AR, Boerwinkle E, Hobbs HH, Cohen
JC: A common allele on chromosome 9 as-
sociated with coronary heart disease. Sci-
ence 2007;
316: 1488–1491.
158 Duan J, Sanders AR, Gejman PV: Genome-
wide approaches to schizophrenia. Brain Res
Bull 2010;
83: 93–102.
159 Craddock N, Sklar P: Genetics of bipolar dis-
order. Lancet 2013;
381: 1654–1662.
160 Lee KW, Woon PS, Teo YY, Sim K: Genome
wide association studies (GWAS) and copy
number variation (CNV) studies of the ma-
jor psychoses: what have we learnt? Neurosci
Biobehav Rev 2012;
36: 556–571.
161 Johnson C, Drgon T, Liu Q-R, Walther D,
Edenberg H, Rice J, Foroud T, Uhl GR:
Pooled association genome scanning for
alcohol dependence using 104,268 SNPs:
validation and use to identify alcoholism
vulnerability loci in unrelated individuals
from the Collaborative Study on the Ge-
netics of Alcoholism. Am J Med Genet B
Neuropsychiatr Genet 2006;
141B:844–
853.
162 Frank J, Cichon S, Treutlein J, Ridinger M,
Mattheisen M, Hoffmann P, Herms S,
Wodarz N, Soyka M, Zill P, Maier W, Möss-
ner R, Gaebel W, Dahmen N, Scherbaum N,
Schmäl C, Steffens M, Lucae S, Ising M, Mül-
ler-Myhsok B, Nöthen MM, Mann K, Kiefer
F, Rietschel M: Genome-wide significant as-
sociation between alcohol dependence and a
variant in the ADH gene cluster. Addict Biol
2012;
17: 171–180.
163 Park BL, Kim JW, Cheong HS, Kim LH, Lee
BC, Seo CH, Kang TC, Nam YW, Kim GB,
Shin HD, Choi IG: Extended genetic effects
of ADH cluster genes on the risk of alcohol
dependence: from GWAS to replication.
Hum Genet 2013;
132: 657–668.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Genetics of Alcohol Dependence Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
93
164 Takeuchi F, Isono M, Nabika T, Katsuya T,
Sugiyama T, Yamaguchi S, Kobayashi S,
Ogihara T, Yamori Y, Fujioka A, Kato N:
Confirmation of ALDH2 as a major locus of
drinking behavior and of its variants regulat-
ing multiple metabolic phenotypes in a Japa-
nese population. Circ J 2011;
75: 911–918.
165 Quillen EE, Chen XD, Almasy L, Yang F, He
H, Li X, Wang XY, Liu TQ, Hao W, Deng
HW, Kranzler HR, Gelernter J: ALDH2 is as-
sociated to alcohol dependence and is the
major genetic determinant of ‘daily maxi-
mum drinks’ in a GWAS study of an isolated
rural Chinese sample. Am J Med Genet B
Neuropsychiatr Genet 2014;
165B:103–110.
166 Wang K-S, Liu X, Aragam N, Jian X, Mul-
lersman JE, Liu Y, Pan Y: Family-based as-
sociation analysis of alcohol dependence in
the COGA sample and replication in the
Australian Twin-Family Study. J Neural
Transm 2011;
118: 1293–1299.
167 Wang K-S, Liu X, Zhang Q, Pan Y, Aragam
N, Zeng M: A meta-analysis of two genome-
wide association studies identifies 3 new loci
for alcohol dependence. J Psychiatr Res
2011;
45: 1419–1425.
168 Bierut LJ, Agrawal A, Bucholz KK, Doheny
KF, Laurie C, Pugh E, Fisher S, Fox L, How-
ells W, Bertelsen S, Hinrichs AL, Almasy L,
Breslau N, Culverhouse RC, Dick DM,
Edenberg HJ, Foroud T, Grucza RA, Hatsu-
kami D, Hesselbrock V, Johnson EO, Kra-
mer J, Krueger RF, Kuperman S, Lynskey M,
Mann K, Neuman RJ, et al: A genome-wide
association study of alcohol dependence.
Proc Natl Acad Sci USA 2010;
107: 5082–
5087.
169 Edenberg HJ, Koller DL, Xuei X, Wetherill
L, McClintick JN, Almasy L, Bierut LJ,
Bucholz KK, Goate A, Aliev F, Dick D, Hes-
selbrock V, Hinrichs A, Kramer J, Kuper-
man S, Nurnberger JI, Rice JP, Schuckit MA,
Taylor R, Todd Webb B, Tischfield JA,
Porjesz B, Foroud T: Genome-wide associa-
tion study of alcohol dependence implicates
a region on chromosome 11. Alcohol Clin
Exp Res 2010;
34: 840–852.
170 Heath AC, Whitfield JB, Martin NG, Perga-
dia ML, Goate AM, Lind PA, McEvoy BP,
Schrage AJ, Grant JD, Chou Y-L, Zhu R,
Henders AK, Medland SE, Gordon SD, Nel-
son EC, Agrawal A, Nyholt DR, Bucholz KK,
Madden PAF, Montgomery GW: A quanti-
tative-trait genome-wide association study
of alcoholism risk in the community: find-
ings and implications. Biol Psychiatry 2011;
70: 513–518.
171 Gelernter J, Kranzler HR, Sherva R, Almasy
L, Koesterer R, Smith AH, Anton R, Preuss
UW, Ridinger M, Rujescu D, Wodarz N, Zill
P, Zhao H, Farrer LA: Genome-wide asso-
ciation study of alcohol dependence: signifi-
cant findings in African- and European-
Americans including novel risk loci. Mol
Psychiatry 2014;
19: 41–49.
172 Zuo L, Gelernter J, Zhang CK, Zhao H, Lu L,
Kranzler HR, Malison RT, Li C-SR, Wang F,
Zhang X-Y, Deng H-W, Krystal JH, Zhang
F, Luo X: Genome-wide association study of
alcohol dependence implicates KIAA0040
on chromosome 1q. Neuropsychopharma-
cology 2012;
37: 557–566.
173 Zuo L, Zhang CK, Wang F, Li CS, Zhao H,
Lu L, Zhang XY, Lu L, Zhang H, Zhang F,
Krystal JH, Luo X: A novel, functional and
replicable risk gene region for alcohol de-
pendence identified by genome-wide asso-
ciation study. PLoS One 2011;
6:e26726.
174 Meyer-Lindenberg A, Weinberger DR: In-
termediate phenotypes and genetic mecha-
nisms of psychiatric disorders. Nat Rev Neu-
rosci 2006;
7: 818–827.
175 Mier D, Kirsch P, Meyer-Lindenberg A:
Neural substrates of pleiotropic action of ge-
netic variation in COMT: a meta-analysis.
Mol Psychiatry 2010;
15: 918–927.
176 Wang JC, Foroud T, Hinrichs AL, Le NX,
Bertelsen S, Budde JP, Harari O, Koller DL,
Wetherill L, Agrawal A, Almasy L, Brooks
AI, Bucholz K, Dick D, Hesselbrock V, John-
son EO, Kang S, Kapoor M, Kramer J, Ku-
perman S, Madden PA, Manz N, Martin NG,
McClintick JN, Montgomery GW, Nurn-
berger JI Jr, et al: A genome-wide association
study of alcohol-dependence symptom
counts in extended pedigrees identifies
C15ORF53. Mol Psychiatry 2013;
18: 1218–
1224.
177 Zlojutro M, Manz N, Rangaswamy M, Xuei
X, Flury-Wetherill L, Koller D, Bierut LJ,
Goate A, Hesselbrock V, Kuperman S,
Nurnberger J, Rice JP, Schuckit MA, Foroud
T, Edenberg HJ, Porjesz B, Almasy L: Ge-
nome-wide association study of theta band
event-related oscillations identifies sero-
tonin receptor gene HTR7 influencing risk
of alcohol dependence. Am J Med Genet B
Neuropsychiatr Genet 2011;
156B:44–58.
178 Peng W, Wang HY, Miyahara Y, Peng G,
Wang R-F: Tumor-associated galectin-3
modulates the function of tumor-reactive T
cells. Cancer Res 2008;
68: 7228–7236.
179 Joslyn G, Ravindranathan A, Brush G,
Schuckit M, White RL: Human variation in
alcohol response is influenced by variation
in neuronal signaling genes. Alcohol Clin
Exp Res 2010;
34: 800–812.
180 Schuckit MA: Low level of response to alco-
hol as a predictor of future alcoholism. Am J
Psychiatry 1994;
151: 184–189.
181 MacLaren EJ, Bennett B, Johnson TE, Sikela
JM: Expression profiling identifies novel
candidate genes for ethanol sensitivity
QTLs. Mamm Genome 2006;
17: 147–156.
182 Treutlein J, Cichon S, Ridinger M, Wodarz
N, Soyka M, Zill P, Maier W, Moessner R,
Gaebel W, Dahmen N, Fehr C, Scherbaum
N, Steffens M, Ludwig KU, Frank J, Wich-
mann HE, Schreiber S, Dragano N, Sommer
WH, Leonardi-Essmann F, Lourdusamy A,
Gebicke-Haerter P, Wienker TF, Sullivan
PF, Nöthen MM, et al: Genome-wide asso-
ciation study of alcohol dependence. Arch
Gen Psychiatry 2009;
66: 773–784.
183 Schumann G, Johann M, Frank J, Preuss U,
Dahmen N, Laucht M, Rietschel M, Rujescu
D, Lourdusamy A, Clarke T-K, Krause K,
Dyer A, Depner M, Wellek S, Treutlein J,
Szegedi A, Giegling I, Cichon S, Blomeyer D,
Heinz A, Heath S, Lathrop M, Wodarz N,
Soyka M, Spanagel R, Mann K: Systematic
analysis of glutamatergic neurotransmission
genes in alcohol dependence and adolescent
risky drinking behavior. Arch Gen Psychia-
try 2008;
65: 826–838.
184 Spanagel R: Alcoholism: a systems approach
from molecular physiology to addictive be-
havior. Physiol Rev 2009;
89: 649–705.
185 Dick DM, Aliev F, Wang JC, Grucza RA,
Schuckit M, Kuperman S, Kramer J, Hinrichs
A, Bertelsen S, Budde JP, Hesselbrock V,
Porjesz B, Edenberg HJ, Bierut LJ, Goate A:
Using dimensional models of externalizing
psychopathology to aid in gene identifica-
tion. Arch Gen Psychiatry 2008;
65: 310–318.
186 Kendler KS, Kalsi G, Holmans PA, Sanders
AR, Aggen SH, Dick DM, Aliev F, Shi J,
Levinson DF, Gejman PV: Genomewide as-
sociation analysis of symptoms of alcohol
dependence in the molecular genetics of
schizophrenia (MGS2) control sample. Al-
cohol Clin Exp Res 2011;
35: 963–975.
187 Edenberg HJ, Dick DM, Xuei X, Tian H, Al-
masy L, Bauer LO, Crowe RR, Goate A, Hes-
selbrock V, Jones K, Kwon J, Li T-K, Nurn-
berger JI, O’Connor SJ, Reich T, Rice J,
Schuckit MA, Porjesz B, Foroud T, Begleiter
H: Variations in GABRA2, encoding the al-
pha 2 subunit of the GABA(A) receptor, are
associated with alcohol dependence and
with brain oscillations. Am J Hum Genet
2004;
74: 705–714.
188 Sullivan PF, Daly MJ, O’Donovan M: Genet-
ic architectures of psychiatric disorders: the
emerging picture and its implications. Nat
Rev Genet 2012;
13: 537–551.
189 Bieńkowski P: Pharmacological features of
naltrexone and its use in the treatment of al-
cohol dependence (in Polish). Psychiatr Pol
2013;
47: 117–126.
190 Sobański JA: Contemporary research direc-
tions in ‘Polish psychiatry’ (Psychiatria Pol-
ska) on the basis of publications in the years
2010–2012. A preliminary report. Psychiatr
Pol 2012;
46: 691–707.
191 Thomas D: Gene-environment-wide asso-
ciation studies: emerging approaches. Nat
Rev Genet 2010;
11: 259–272.
1
92 Plomin R: Missing heritability, polygenic
scores, and gene-environment correlation. J
Child Psychol Psychiatry 2013;
54: 1147–1149.
193 Nurnberger JI Jr, Berrettini WH: Principles
of Psychiatric Genetics. Cambridge, Cam-
bridge Medicine, 2012.
194 Kimura M, Higuchi S: Genetics of alcohol
dependence. Psychiatry Clin Neurosci 2011;
65: 213–225.
195 Treutlein J, Rietschel M: Genome-wide as-
sociation studies of alcohol dependence and
substance use disorders. Curr Psychiatry
Rep 2011;
13: 147–155.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
Samochowiec /Samochowiec /Puls /
Bienkowski
/Schott
Neuropsychobiology 2014;70:77–94
DOI: 10.1159/000364826
94
196 Palmer RHC, McGeary JE, Francazio S, Ra-
phael BJ, Lander AD, Heath AC, Knopik VS:
The genetics of alcohol dependence: advanc-
ing towards systems-based approaches.
Drug Alcohol Depend 2012;
125: 179–191.
197 Edenberg HJ: Common and rare variants in
alcohol dependence. Biol Psychiatry 2011;
70: 498–499.
198 Button KS, Ioannidis JPA, Mokrysz C,
Nosek BA, Flint J, Robinson ESJ, Munafò
MR: Power failure: why small sample size
undermines the reliability of neuroscience.
Nat Rev Neurosci 2013;
14: 365–376.
199 Edenberg HJ, Kranzler HR: The contribu-
tion of genetics to addiction therapy ap-
proaches. Pharmacol Ther 2005;
108: 86–93.
200 Lind PA, Macgregor S, Vink JM, Pergadia
ML, Hansell NK, de Moor MHM, Smit AB,
Hottenga J-J, Richter MM, Heath AC, Mar-
tin NG, Willemsen G, de Geus EJC, Vogel-
zangs N, Penninx BW, Whitfield JB, Mont-
gomery GW, Boomsma DI, Madden PAF: A
genomewide association study of nicotine
and alcohol dependence in Australian and
Dutch populations. Twin Res Hum Genet
2010;
13: 10–29.
201 Biernacka JM, Geske J, Jenkins GD, Colby C,
Rider DN, Karpyak VM, Choi DS, Fridley
BL: Genome-wide gene-set analysis for
identification of pathways associated with
alcohol dependence. Int J Neuropsycho-
pharmacol 2013;
16: 271–278.
202 Zuo L, Wang K, Zhang XY, Krystal JH, Li
CS, Zhang F, Zhang H, Luo X: NKAIN1-
SERINC2 is a functional, replicable and ge-
nome-wide significant risk gene region spe-
cific for alcohol dependence in subjects of
European descent. Drug Alcohol Depend
2013;
129: 254–264.
203 Zuo L, Zhang XY, Wang F, Li CS, Lu L, Ye
L, Zhang H, Krystal JH, Deng HW, Luo X:
Genome-wide significant association signals
in IPO11-HTR1A region specific for alcohol
and nicotine codependence. Alcohol Clin
Exp Res 2013;
37: 730–739.
2
04 Schumann G, Coin LJ, Lourdusamy A,
Charoen P, Berger KH, Stacey D, Des-
rivières S, Aliev FA, Khan AA, Amin N,
Aulchenko YS, Bakalkin G, Bakker SJ,
Balkau B, Beulens JW, Bilbao A, de Boer
RA, Beury D, Bots ML, Breetvelt EJ, Cauchi
S, Cavalcanti-Proença C, Chambers JC,
Clarke T-K, Dahmen N, et al: Genome-
wide association and genetic functional
studies identify autism susceptibility can-
didate 2 gene (AUTS2) in the regulation of
alcohol consumption. Proc Natl Acad Sci
USA 2011;
108: 7119–7124.
205 Baik I, Cho NH, Kim SH, Han B-G, Shin C:
Genome-wide association studies identify
genetic loci related to alcohol consumption
in Korean men. Am J Clin Nutr 2011;
93:
809–816.
206 Chen X-D, Xiong D-H, Yang T-L, Pei Y-F,
Guo Y-F, Li J, Yang F, Pan F, Tan L-J, Yan
H, Liu X-G, Lei S-F, Li X, Ning L-L, Zhu X-Z,
Levy S, Kranzler HR, Farrer LA, Gelernter J,
Recker RR, Deng H-W: ANKRD7 and Cytl1
are novel risk genes for alcohol drinking be-
havior. Chin Med J (Engl) 2012;
125: 1127–
1134.
207 Kapoor M, Wang JC, Wetherill L, Le N, Ber-
telsen S, Hinrichs AL, Budde J, Agrawal A,
Bucholz K, Dick D, Harari O, Hesselbrock
V, Kramer J, Nurnberger JI Jr, Rice J, Sac-
cone N, Schuckit M, Tischfield J, Porjesz B,
Edenberg HJ, Bierut L, Foroud T, Goate A:
A meta-analysis of two genome-wide asso-
ciation studies to identify novel loci for max-
imum number of alcoholic drinks. Hum
Genet 2013;
132: 1141–1151.
208 Pan Y, Luo X, Liu X, Wu LY, Zhang Q, Wang
L, Wang W, Zuo L, Wang KS: Genome-wide
association studies of maximum number of
drinks. J Psychiatr Res 2013;
47: 1717–1724.
209 Wang K-S, Liu X, Zhang Q, Wu L-Y, Zeng
M: Genome-wide association study identi-
fies 5q21 and 9p24.1 (KDM4C) loci associ-
ated with alcohol withdrawal symptoms. J
Neural Transm 2012;
119: 425–433.
Downloaded by:
Charité - Universitätsmedizin Berlin
193.175.73.217 - 11/3/2014 3:08:22 PM
... As enzimas codificadas pelos genes pertencentes à classe I da ADH (ADH1A, ADH1B e ADH1C) apresentam maiores atividades e são as principais responsáveis pela metabolização hepática do álcool. Portanto, variantes nestes genes, principalmente em ADH1B e ADH1C, são amplamente estudadas no contexto genético do TUA 9,12,17,20 . ...
... Isso ocorre, pois o acetaldeído causa uma sintomatologia desconfortável, principalmente náuseas, fazendo com que o indivíduo não consuma grandes quantidades de substâncias alcoólicas [8][9][10][11][12][13]20,21 . Quanto maior a concentração sérica de acetaldeído, maiores serão os efeitos adversos e menor a probabilidade de continuidade do consumo de álcool 8,10,11,[15][16][17][18][19][20][21] . O gene ADH1C também apresenta SNPs associados ao desenvolvimento do TUA 8,14,16 . ...
... Os avanços científicos recentes no estudo de genomas estão possibilitando o entendimento de aspectos genéticos em diferentes níveis no organismo humano e relacionados ao desenvolvimento do TUA. Polimorfismos em genes associados a comportamentos impulsivos, desinibição e compulsividade foram relacionados previamente ao TUA [13][14][15][16][17][18][19] . Entretanto, diversos estudos relatam a importância de uma transformação bioquímica essencial na metabolização do álcool e que é fundamental na eliminação da molécula do organismo para evitar efeitos adversos mais severos e também o TUA 20,21 . ...
... As enzimas codificadas pelos genes pertencentes à classe I da ADH (ADH1A, ADH1B e ADH1C) apresentam maiores atividades e são as principais responsáveis pela metabolização hepática do álcool. Portanto, variantes nestes genes, principalmente em ADH1B e ADH1C, são amplamente estudadas no contexto genético do TUA 9,12,17,20 . ...
... Isso ocorre, pois o acetaldeído causa uma sintomatologia desconfortável, principalmente náuseas, fazendo com que o indivíduo não consuma grandes quantidades de substâncias alcoólicas [8][9][10][11][12][13]20,21 . Quanto maior a concentração sérica de acetaldeído, maiores serão os efeitos adversos e menor a probabilidade de continuidade do consumo de álcool 8,10,11,[15][16][17][18][19][20][21] . O gene ADH1C também apresenta SNPs associados ao desenvolvimento do TUA 8,14,16 . ...
... Os avanços científicos recentes no estudo de genomas estão possibilitando o entendimento de aspectos genéticos em diferentes níveis no organismo humano e relacionados ao desenvolvimento do TUA. Polimorfismos em genes associados a comportamentos impulsivos, desinibição e compulsividade foram relacionados previamente ao TUA [13][14][15][16][17][18][19] . Entretanto, diversos estudos relatam a importância de uma transformação bioquímica essencial na metabolização do álcool e que é fundamental na eliminação da molécula do organismo para evitar efeitos adversos mais severos e também o TUA 20,21 . ...
Article
Full-text available
O transtorno por uso de álcool (TUA) é influenciado pela genética, principalmente na metabolização do etanol. Os genes da álcool desidrogenase (ADH1B/ADH1C), enzima que transforma o etanol, apresentam SNPs (single nucleotide polymorphisms) que resultam em isoenzimas com diferentes taxas catalíticas. Estudos demonstraram que os SNPs Arg48His, Arg370Cys, Arg272Gln e Ile350Val contribuem para o TUA. Este artigo revisou os estudos que investigaram SNPs em ADH1B (Arg48His/Arg370Cys) e ADH1C (Arg272Gln/Ile350Val), bem como avaliou as variações nas frequências alélicas desses genes e a influência no TUA nas diferentes populações no mundo. As frequências alélicas dos polimorfismos nos genes ADH1B e ADH1C foram comparadas entre diferentes regiões do mundo. A revisão foi realizada através de plataformas eletrônicas como NCBI/PUBMED, Google Scholar, SciELO, Science Direct, MEDLINE, LILACS e Scopus. As coletas dos dados referentes as frequências dos polimorfismos nos genes ADH1B e ADH1C foram realizadas a partir do banco de dados Ensembl Genome Browser 92 com os seguintes descritores: rs1229984, rs2066702, rs1693482 e rs698. As frequências alélicas dos polimorfismos foram comparadas pelos testes qui-quadrado de Pearson e exato de Fisher, considerando um nível de significância pré-estabelecido para o erro alfa de 5% (p
... На цей час виявлено понад 50 генів ризику розвитку алкоголізму, прояв яких різною мірою залежить від чинників навколишнього середовища -ситуації в сім'ї, колі товаришів та суспільстві (Iyer-Eimerbrink and Nurnberger, 2014). Дослідники виявили дві групи таких генів: гени, які контролюють утилізацію алкоголю в організмі, та гени, які визначають нейропсихічні функції (Samochowiec et al., 2014). В утилізації алкоголю в організмі беруть участь ферменти алкоголь-дегідрогеназа та альдегід-дегідрогеназа. ...
... Генів, які причетні до розвитку алкоголізму, багато, але кожен із них окремо чинить незначний вплив (Iyer-Eimerbrink and Nurnberger, 2014;Samochowiec et al., 2014). Повною мірою несприятлива сукупність таких генів може проявитися при наявності провокуючих чинників навколишнього середовища, одним із яких є неблагополучна родина. ...
... Furthermore, there is evidence of associations of the A1 allele with psychiatric disorders such as addictionsmost notably alcohol dependence (for a meta-analysis, see Wang et al. 2013; for reviews, see Samochowiec et al. 2014 andKoeneke et al. 2020)-and ADHD (for a metaanalysis, see Pan et al. 2015). In addition, it was initially hypothesized that there was an advantage of the A1 allele in schizophrenia disorders in terms of lower risk (Dubertret et al. 2004) and better response to haloperidol (Schafer et al. 2001). ...
Article
Full-text available
Dopaminergic neurotransmission plays a pivotal role in appetitively motivated behavior in mammals, including humans. Notably, action and valence are not independent in motivated tasks, and it is particularly difficult for humans to learn the inhibition of an action to obtain a reward. We have previously observed that the carriers of the DRD2/ANKK1 TaqIA A1 allele, that has been associated with reduced striatal dopamine D2 receptor expression, showed a diminished learning performance when required to learn response inhibition to obtain rewards, a finding that was replicated in two independent cohorts. With our present study, we followed two aims: first, we aimed to replicate our finding on the DRD2/ANKK1 TaqIA polymorphism in a third independent cohort (N = 99) and to investigate the nature of the genetic effects more closely using trial-by-trial behavioral analysis and computational modeling in the combined dataset (N = 281). Second, we aimed to assess a potentially modulatory role of prefrontal dopamine availability, using the widely studied COMT Val108/158Met polymorphism as a proxy. We first report a replication of the above mentioned finding. Interestingly, after combining all three cohorts, exploratory analyses regarding the COMT Val108/158Met polymorphism suggest that homozygotes for the Met allele, which has been linked to higher prefrontal dopaminergic tone, show a lower learning bias. Our results corroborate the importance of genetic variability of the dopaminergic system in individual learning differences of action-valence interaction and, furthermore, suggest that motivational learning biases are differentially modulated by genetic determinants of striatal and prefrontal dopamine function.
... The etiology of brain disorders caused by drug abuse is complex, involving a combination of factors that include individual genotype, social environment, and age or developmental stage (Samochowiec et al., 2014;Wall et al., 2016). In general, drugs act by changing neuronal function at molecular, cellular, circuitry, macro-structural, and numerous other functional levels leading to physiological and behavioral alterations (Berridge, 2017;Volkow et al., 2016). ...
Article
Full-text available
Drug abuse and brain disorders related to drug comsumption are public health problems with harmful individual and social consequences. The identification of therapeutic targets and precise pharmacological treatments to these neuropsychiatric conditions associated with drug abuse are urgently needed. Understanding the link between neurobiological mechanisms and behavior is a key aspect of elucidating drug abuse-related targets. Due to various molecular, biochemical, pharmacological, and physiological features, the zebrafish (Danio rerio) has been considered a suitable vertebrate for modeling complex processes involved in drug abuse responses. In this review, we discuss how the zebrafish has been successfully used for modeling neurobehavioral phenotypes related to drug abuse and review the effects of opioids, cannabinoids, alcohol, nicotine, and psychedelic drugs on the central nervous system (CNS). Moreover, we summarize recent advances in zebrafish-based studies and outline potential advantages and limitations of the existing zebrafish models to explore the neurochemical bases of drug abuse and addiction. Finally, we discuss how the use of zebrafish models may present fruitful approaches to provide valuable clinically translatable data.
... 8 In fact, several candidate studies assessing the risk loci for AD were designed to target gene variants related to alcohol metabolism or neurobiology. [9][10][11][12][13] Recently, a number of genome-wide association studies (GWASs) have investigated genetic markers of AD, including the genomic region of chromosome ...
Article
Full-text available
Background Alcohol dependence (AD) is a common disorder that is influenced by genetic as well as environmental factors. A previous genome-wide association study (GWAS) of the Korean population performed by our research group identified a number of genes, including BRCA1-associated protein (BRAP) and protein arginine methyltransferase 8 (PRMT8), as novel genetic markers of AD. Methods The present investigation was a fine-mapping follow-up study of 459 AD and 455 non-AD subjects of Korean descent to determine the associations between BRAP and PRMT8 polymorphisms and AD. The Alcohol Use Disorders Identification Test (AUDIT) was administered to screen for the degree of AD risk in the subjects and 58 genetic variants, 5 for BRAP and 53 for PRMT8, were genotyped for subsequent association analyses. Results In the present case–control analysis, BRAP rs3782886 showed the most significant association signal with a risk of AD (P=1.29×10⁻¹⁶, Pcorr =7.74×10⁻¹⁶, OR =0.19). There were also significant differences in the overall and subcategory scores for the BRAP genetic variants, including rs3782886 (P=9.94×10⁻³¹, Pcorr =5.96×10⁻³⁰ at rs3782886 for the overall AUDIT score). However, the genetic effects of PRMT8 polymorphisms observed in our previous GWAS were not replicated in the present study (minimum P=0.0005, Pcorr >0.05, OR =0.30 at rs4766139 in the recessive model). Furthermore, the single-nucleotide polymorphisms of PRMT8 were not associated with the overall and subcategory AUDIT scores. Conclusion The present findings suggest that the genetic variants of BRAP may contribute to a predisposition for an alcohol use disorder.
... However, most GWAS have been performed for alcohol dependence (Tawa et al., 2016). These GWAS have highlighted some risk genes that had already emerged through candidate gene studies, like some members of the ADH family and ALDH2, encoding alcohol and aldehyde dehydrogenases, respectively (Samochowiec et al., 2014). Others have emerged that were not a priori candidates for the disorder, pointing at new functions that may be related to its etiology. ...
Article
Background: Substance dependence is a chronic and relapsing disorder explained by genetic and environmental risk factors. The aim of our study is to replicate previous genome-wide significant (GWS) hits identified in substance dependence in general or in cocaine dependence in particular using an independent sample from Spain. Methods: We evaluated, in a Spanish sample of 1711 subjects with substance dependence (1011 of them cocaine dependent) and 1719 control individuals, three SNPs identified as GWS in previous studies: rs1868152 and rs2952621 (located near LINC02052 and LINC01854, respectively), associated with substance dependence, and rs2629540 (in the first intron of FAM53B), associated with cocaine dependence. Results: We replicated the association between rs2952621 and substance dependence under the dominant model (P = 0.020), with the risk allele (T) being the same in our sample and in those two reported previously. We then performed a meta-analysis of the two samples used in the original study that reported the association of rs2952621 with substance dependence (Collaborative Studies on Genetics of Alcoholism (COGA) and Study of Addiction: Genetics and Environment (SAGE)) together with our Spanish sample. The meta-analysis of 3747 cases and 4043 controls confirmed the association (OR = 1.26, 95% CI = 1.15-1.39). Conclusions: The rs2952621 variant, located downstream from the yet uncharacterized gene LINC01854, is associated with substance dependence in our Spanish sample. Further research is needed to understand its contribution to the susceptibility to substance dependence.
Article
Objectives This study examined the changes in the retinal nerve fiber layer (RNFL) thickness and the macular thickness in Chinese patients with alcohol dependency and ascertaining the influence of optic cup volume on cognitive functioning. Methods A total of 26 alcohol-dependent patients and 53 age- and gender-matched healthy controls were enrolled in the study. All subjects underwent Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) measurement and were scanned via spectral-domain optical coherence tomography (OCT). Results Compared with the healthy controls, the alcohol-dependent patients returned lower scores across all subscales and total RBANS scores. The total thickness of the RNFL of the left eye, temporal and nasal RNFLthickness of both eyes were thinner in the alcohol-dependent patients (all p < 0.05). In terms of macular thickness, eight macular regions and the average thickness of the right eye and three of the left eye were thinner in the alcohol-dependent patients than in the healthy controls. The linear regression analysis indicated that a higher alcohol consumption was associated with thinner RNFL and macular thickness especially in temporal and inferior quadrant region after controlling for smoker status, age, BMI, cholesterol level and AKT level. Conclusions A higher alcohol consumption was significantly associated with thinner RNFL and macular thickness, indicating that alcohol is a potential risk factor affecting RNFL thickness and macular thickness. Meanwhile, the increase in optic cup volume was associated with the reduced cognitive functioning of the alcohol-dependent patients.
Article
Introduction: A relationship between alcohol dependence (AD) and calcium/calmodulin-dependent protein kinase IV (CAMKIV) has been reported in a whole genome study of Korean AD patients. The purpose of the present study is to compare the frequency of CAMKIV genotypes and alleles between AD and control subjects in Korea. Methods: The present study includes 281 AD patients and 139 control subjects. Seven single nucleotide polymorphism of CAMKIV gene known to show significant separation ratio in Asians were searched in SNP database and previous studies related to CAMKIV gene. Polymerase chain reaction and restriction fragment length polymorphism techniques were used to analyze genotype of CAMKIV gene SNPs. Results: Major TT genotype and T allele frequencies of rs 25917 in AD patients were significantly higher than those of control subjects (genotype frequency, p=0.002; allele frequency, p=0.001). Major CC genotype and C allele frequencies of rs 117590959 in AD patients were also significantly higher than those of control subjects (genotype frequency, p<0.001; allele frequency, p=0.001). Major genotypes of rs25917 (p=0.002, odd ratio: 3.13, 95% CI: 1.54-6.38) and rs11790959 (p=0.002, odd ratio: 3.22, 95% CI: 1.52-6.81) showed significantly higher odds ratios associated with AD than minor genotypes in logistic regression. Discussion: These results suggest that CAMKIV might be a candidate AD gene. Further research is needed to determine the precise relationship between CAMKIV and AD and the function of each SNP.
Article
Full-text available
Alcohol dependence (alcoholism) is accompanied by evidence of tolerance, withdrawal (physiological dependence), or compulsive behavior related to alcohol use. Studies of strain and individual differences using animal models for acute physiological dependence liability are useful means to identify potential genetic determinants of liability in humans. Behavioral and quantitative trait analyses were conducted using animal models for high risk versus resistance to acute physiological dependence. Using a two-step genetic mapping strategy, loci on mouse chromosomes 1, 4, and 11 were mapped that contain genes that influence alcohol withdrawal severity. In the aggregate, these three risk markers accounted for 68% of the genetic variability in alcohol withdrawal. Candidate genes in proximity to the chromosome 11 locus include genes encoding the α 1 , α 6 , and γ 2 subunits of type-A receptors for the inhibitory neurotransmitter, GABA. In addition, suggestive linkage is indicated for two loci on mouse chromosome 2, one near Gad1 encoding glutamic acid decarboxylase, and the other near the El2 locus which influences the seizure phenotype in the neurological mutant strain El. The present analyses detect and map some of the loci that increase risk to develop physiological dependence and may facilitate identification of genes related to the development of alcoholism. Syntenic conservation between human and mouse chromosomes suggests that human homologs of genes that increase risk for physiological dependence may localize to 1q21–q32, 2q24–q37/11p13, 9p21–p23/1p32–p22.1, and 5q32–q35.
Article
Persistent tobacco use and excessive alcohol consumption are major public health concerns worldwide. Both alcohol and nicotine dependence (AD, ND) are genetically influenced complex disorders that exhibit a high degree of comorbidity. To identify gene variants contributing to one or both of these addictions, we first conducted a pooling-based genomewide association study (GWAS) in an Australian population, using Illumina Infinium 1M arrays. Allele frequency differences were compared between pooled DNA from case and control groups for: (1) AD, 1224 cases and 1162 controls; (2) ND, 1273 cases and 1113 controls; and (3) comorbid AD and ND, 599 cases and 488 controls. Secondly, we carried out a GWAS in independent samples from the Netherlands for AD and for ND. Thirdly, we performed a meta-analysis of the 10, 000 most significant AD- and ND-related SNPs from the Australian and Dutch samples. In the Australian GWAS, one SNP achieved genomewide significance (p < 5 x 10 -8 ) for ND (rs964170 in ARHGAPlOon chromosome 4, p = 4.43 x 10” 8 ) and three others for comorbid AD/ND (rs7530302 near MARK1 on chromosome 1 ( p = 1.90 x 10 -9 ), rs1784300 near DDX6 on chromosome 11 (p = 2.60 x 10 -9 ) and rs12882384 in KIAA1409 on chromosome 14 (p = 4.86 x 10 -8 )). None of the SNPs achieved genomewide significance in the Australian/Dutch meta-analysis, but a gene network diagram based on the top-results revealed overrepre-sentation of genes coding for ion-channels and cell adhesion molecules. Further studies will be requirec before the detailed causes of comorbidity between AC and ND are understood.
Article
Background: Very little information is available on the co-occurrence of different personality disorders (PDs) and alcohol and drug use disorders in the U.S. population. Objective: To present national data on sex differences in the co-occurrence of Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) alcohol and drug use disorders and 7 of the 10 DSM-IV PDs. Design: Face-to-lace interviews conducted in the 2001-2002 National Epidemiologic Survey on Alcohol and Related Conditions (N = 43,093). Setting: The United States and the District of Columbia, including Alaska and Hawaii. Participants: Household and group-quarters residents, age 18 and older. Results: Among individuals with a current alcohol use disorder, 28.6 percent (95 percent confidence interval [CI], 26.7-30.6) had at least one PD, whereas 47.7 percent (95 percent CI, 43.9-51.6) of those with a current drug use disorder had at least one PD. Further, 16.4 percent (95 percent CI, 15.1-17.6) of individuals with at least one PD had a current alcohol use disorder, and 6.5 percent (95 percent CI, 5.7-7.3) had a current drug use disorder. Associations between PDs and alcohol and drug use disorders were overwhelmingly positive and significant (P <.05). Overall, alcohol use disorders were most strongly related to antisocial (odds ratio [OR], 4.8; 95 percent CI, 4.1-5.6), histrionic (OR, 4.7; 95 percent CI 3.&-5.8), and dependent (OR, 3.0; 95 percent CI, 1.9-4.8) PDs. Drug use disorders also were more highly associated with antisocial (OR, 11.8; 95 percent CI, 9.7-14.3), histrionic (OR, 8.0; 95 percent CI, 6.0-10.7), and dependent (OR, 11.6; 95 percent CI, 7.1-19. 1) PDs. Associations between obsessive-compulsive, histrionic, schizoid, and antisocial PDs and specific alcohol and drug use disorders were significantly stronger (P <.04) among women than men, whereas the association between dependent PD and drug dependence was significantly greater (P <.04) among men than women. Conclusions: The co-occurrence of PDs with alcohol and drug use disorders is pervasive in the U.S. population. Results highlight the need for further research on the underlying structure of these disorders and the treatment implications of these disorders when comorbid.
Article
Genetic linkage analysis of rats that were selectively bred for alcohol preference identified a chromosomal region that includes the neuropeptide Y (NPY) gene. Alcohol-preferring rats have lower levels of NPY in several brain regions compared with alcohol-non-preferring rats. We therefore studied alcohol consumption by mice that completely lack NPY as a result of targeted gene disruption. Here we report that NPY-deficient mice show increased consumption, compared with wild-type-mice, of solutions containing 65, 10% and 20% (v/v) ethanol. NPY-deficient mice are also less sensitive to the sedative/hypnotic effects of ethanol, as shown by more rapid recovery from ethanol-induced sleep, even though plasma ethanol concentrations do not differ significantly from those of controls. In contrast, transgenic mice that overexpress a marked NPY gene in neurons that usually express it have a lower preference for ethanol and are more sensitive to the sedative/hypnotic effects of this drug than controls. These data are direct evidence that alcohol consumption and resistance are inversely related to NPY levels in the brain.
Article
Breast cancer exhibits familial aggregation, consistent with variation in genetic susceptibility to the disease. Known susceptibility genes account for less than 25% of the familial risk of breast cancer, and the residual genetic variance is likely to be due to variants conferring more moderate risks. To identify further susceptibility alleles, we conducted a two-stage genome-wide association study in 4,398 breast cancer cases and 4,316 controls, followed by a third stage in which 30 single nucleotide polymorphisms (SNPs) were tested for confirmation in 21,860 cases and 22,578 controls from 22 studies. We used 227,876 SNPs that were estimated to correlate with 77% of known common SNPs in Europeans at r(2) > 0.5. SNPs in five novel independent loci exhibited strong and consistent evidence of association with breast cancer (P < 10(-7)). Four of these contain plausible causative genes (FGFR2, TNRC9, MAP3K1 and LSP1). At the second stage, 1,792 SNPs were significant at the P < 0.05 level compared with an estimated 1,343 that would be expected by chance, indicating that many additional common susceptibility alleles may be identifiable by this approach.
Article
Alcohol dependence is a leading cause of morbidity and premature death. Several lines of evidence suggest a substantial genetic component to the risk for alcoholism: sibs of alcoholic probands have a 3–8 fold increased risk of also developing alcoholism, and twin heritability estimates of 50–60% are reported by contemporary studies of twins. We report on the results of a six-center collaborative study to identify susceptibility loci for alcohol dependence. A genome-wide screen examined 291 markers in 987 individuals from 105 families. Two-point and multipoint nonparametric linkage analyses were performed to detect susceptibility loci for alcohol dependence. Multipoint methods provided the strongest suggestions of linkage with susceptibility loci for alcohol dependence on chromosomes 1 and 7, and more modest evidence for a locus on chromosome 2. In addition, there was suggestive evidence for a protective locus on chromosome 4 near the alcohol dehydrogenase genes, for which protective effects have been reported in Asian populations. Am. J. Med. Genet. (Neuropsychiatr. Genet.) 81:207–215, 1998.