Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

Received: October 7, 2013 Accepted after revision: May 24, 2014 Published online: October 30, 2014

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

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 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

© 2014 S. Karger AG, Basel 0302–282X/14/0702–0077$39.50/0 E-Mail [email protected] www.karger.com/nps

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. © 2014 S. Karger AG, Basel

Jerzy Samochowiec and Agnieszka Samochowiec contributed equally to this work.

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

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

Key Words Alcohol-related phenotypes · Alcohol dependence · Genetic risk factors

While alcohol consumption at moderate levels is socially widely accepted in many cultures, excessive drinking ultimately leading to alcohol dependence is a severe health problem both at the individual and large-scale socioeconomic levels. According to World Health Organization 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 standard drinks a week. It is estimated that every fourth person in Europe aged 15–64 regularly consumes alcohol at harmful levels, and over 14 million people are alcohol dependent [1]. The US data from the 2001–2002 National Epidemiological Survey on Alcohol and Related Conditions 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 dependence is 12.5% in the USA. Alcohol use disorders (AUDs) can be defined as ‘maladaptive patterns of alcohol use leading to clinically significant impairment or stress’ [3]. Diagnostic criteria, both from the Diagnostic and Statistical Manual of Mental Disorders [4] and from the International Classification of Diseases [5] divide AUDs into alcohol dependence and abuse/harmful use. The major change proposed for DSMV (and introduced in May 2013) has replaced the 2 separate DSM-IV (substance-specific) categories of dependence and abuse with a single (substance-specific) category, 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 psychological effects in individuals who consume large amounts of alcohol [7]. The discrepancy between the widespread occurrence of risky or harmful alcohol consumption and the frequent, yet substantially lower, prevalence of clinical alcohol dependence raises the question why certain individuals become dependent on alcohol, sometimes in a rather short time, while others do not seem to be affected by 78

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

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 considerable interindividual variability, and it is shaped by both including environmental (e.g. cultural, social political, religious, economic, legal including alcohol accessibility, price and social norms) and genetic factors [8–11]. The biological processes that make one person more susceptible to addiction than another are subject to intense investigations. Twin and adoption studies quantify the heritable component at 50–60% [12–14]. While in early adolescence the initiation and use of alcohol and other addictive drugs is primarily influenced by environmental, i.e. familial and social, factors, the role of environmental influences progressively decreases during the transition to adulthood, when the impact of genetic 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 genegene and gene-environment (GxE) interactions. To distinguish between environmental and genetic factors, adoption and twin studies have been conducted. Adoption studies demonstrate that adoptees more closely resemble their biological parents than their adoptive parents in terms of susceptibility to alcohol dependence [16– 19]. Twin studies reveal greater concordance between monozygotic as compared to dizygotic twins, highlighting a major genetic impact [10, 19–21]. (Note: importantly, not all monozygotic cotwins of alcohol-dependent patients are alcohol-dependent themselves, also highlighting 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 alcohol dependence comes from animal research. In selectively bred rodent lines, a considerable proportion of heritability in the development of addiction-related behavior has been observed, including strong preference for alcohol over water, willingness to work for alcohol, sensitivity to the hypnotic or activating effects of alcohol and to withdrawal, and demonstrations that alcohol is rewarding 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 predisposition to alcohol dependence [27–30]. The environmental contributions to alcohol-related phenotypes include peer influence [31–35], availability Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

Introduction

Genetics of Alcohol Dependence

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 studies. 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 influences 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-language articles published before July 2013 (December 2013 in case of GWAS) in peer-reviewed journals were accepted. Articles were identified and reviewed by 2 of the authors, and their final decision was made by consensus. In the case of GWAS of alcohol dependence, all relevant articles 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 dependence were candidate gene studies targeting coding variations in genes that metabolize alcohol. Alcohol is mostly metabolized in the liver, in a 2-step reaction: oxidation to acetaldehyde, which is further oxidized to acetate. The first step is primarily to be catalyzed by alcohol dehydrogenases (ADHs), a reaction accompanied by reduction 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. Humans 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 glutamate to lysine at position 504 (NCBI accession No.: rs671), Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

79

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 alcohol drinking is thought to contribute to the risk of alcohol dependence. The alcohol dependence risk grows almost linearly with the frequency of binge drinking [42]. Data from the National Epidemiologic Survey on Alcohol 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 behaviors 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 externalizing (e.g. antisocial personality disorder, conduct disorder 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, externalizing behavior, affective instability and antisocial personality disorder. Multidimensional typologies differentiate alcoholism by symptom severity. Typologies have commonly been derived from clinically based samples of alcohol-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 transmission, 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 biologically more directly related to the majority of the genetic 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, neuroendocrine and neuroimaging phenotypes predictive of AUD and other types of addiction [55–60].

Genes Implicated in the Neurobiology of Addiction The positive reinforcing effects of alcohol are generally 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 serotonin [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 example, 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 80

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

studies have linked genetic variations in the dopamine transporter 1 gene to the severity of withdrawal symptoms [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 association of the Taq1A allele of the dopamine D2 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 finding, 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 cautioned 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 genetic association studies, namely that inconclusive results can result from linkage disequilibrium with other variants 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 actually located downstream of the DRD2 gene, within the adjacent and closely linked ANKK1 (ankyrin repeat and kinase domain containing 1) gene region [93, 94]. Therefore, associations of Taq1A with alcohol dependence might result from linkage disequilibrium with other causative genetic variations in either DRD2 or ANKK1 or both. A recent family study showed that a different genetic variation in DRD2 (rs6277, a synonymous single nucleotide polymorphism) that affects mRNA stability [95] did indeed show an association with alcohol dependence, 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 family 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 investigate the biological processes underlying these associations. Such investigations might also help to identify intermediate phenotypes that are more closely related to the genetic variations than a clinical phenotype per se. In the Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 carrying the ALDH2*2 allele, consumption of even small amounts of alcohol leads to accumulation of acetaldehyde to toxic levels resulting in symptoms like flushing, tachycardia and nausea, similar to the effects of alcohol in patients taking the ALDH2 inhibitor disulfiram. The genes of the ADHs 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 encoded 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 important 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’ metabolism 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.

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 model [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 associations: NPY2R was associated with alcohol dependence and alcohol withdrawal symptoms, and NPY5R was associated with seizures during alcohol withdrawal [130]. Other genetic association studies of peptide neurotransmitter systems have found evidence for an association of AUDs with polymorphisms of the brain-derived neurotrophic factor [131] and the tachykinin receptor 3 [132]. In light of the multitude of identified risk variants to date, it must be kept in mind that there are numerous examples of associations that cannot be replicated. This problem has occasionally led to scepticism about the usefulness 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 resulting low power, poorly matched control groups, data-driven 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 arbitrary 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 cascades, and potential risk mechanisms at receptor level might be compensated at the level of intracellular signaling. Therefore, genes affecting intracellular signaling in response to receptor activation could also potentially affect 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

Genetics of Alcohol Dependence

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

81

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

case of DRD2 and ANKK1, it has meanwhile been demonstrated 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 candidate gene studies of alcohol dependence and alcohol-related phenotypes have focused on molecules involved in neurotransmitter pathways interacting with dopaminergic transmission. These include genes involved in cholinergic neurotransmission, such as the muscarinic cholinergic receptor 2 (CHRM2) [102] and the α5-subunit of the nicotinergic cholinergic receptor (CHRNA5) [103–105], components of the GABAergic system like the α2-subunit of the GABA-A receptor (GABRA2) [106] and GABA receptor genes [107–110], genes coding for components of glutamatergic transmission like the metabotropic glutamate receptor 8 (GRM8) [111], genes involved in serotonergic neurotransmission like the solute carrier family 6 member 4 (SLC6A4) gene that encodes the serotonin transporter [112, 113], and components of the endocannabinoid system such as cannabinoid receptor 1. In addition to these genetic variants, which all affect small-molecule neurotransmitters, numerous genetic association studies have been conducted on genes related to peptide neurotransmitters. Most notably, the opioid system 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 endogenous κ-opioid agonist (pro-)dynorphin. The δ-opioid receptor is encoded by OPRD1, and its primary ligand metenkephalin 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 extensively studied in relation to alcohol dependence [85]. Results have been inconclusive and partly conflicting, and a meta-analysis concluded that there was no reliable evidence 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].

Gene-Environment Interactions The association of single molecular-genetic variations with psychiatric disorders including AUDs is modulated by nongenetic influences, and the interplay between specific candidate genes and environmental factors has been subject to numerous GxE interaction studies [32]. Environmental determinants that enhance the risk for AUD differ in both proximity to the disorder and its mechanism. 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 influence 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 inhibit 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 inconsistent results in association studies like the DRD2/ANKK1 gene region, 5-HTTLPR, MAOA, catechol-O-methyltransferase, GABRA2 and the corticotropin-releasing hormone receptor 1 (CRHR1) [138–142]. Therefore, GxE interactions might contribute to the complexity of genetic associations, and systematic assessment of environmental 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 disorders exert their influences in the interaction with environmental 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 Higuchi 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 heterozy82

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

gous for the ALDH2*2 soared from 2.5 to 13% [30]. Given 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 environmental influences, most likely social changes favoring alcohol consumption. In the same study, no alcohol-dependent patients homozygous for the ALDH2*2 were identified, although 120 patients would have been expected based on the allele frequency. It was concluded that the protective influence of the ALDH2 *2 allele was so substantial in homozygotes that further modulation by environmental influences did not occur [30].

Genetic Linkage Studies

Genetic linkage studies provide an unbiased methodology toward identifying genetic variations that affect the risk for a complex genetic disease such as alcohol dependence. The linkage approach is employed to identify genetic risk variants for a disorder in families with several affected members. Linkage studies test markers distributed 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 without 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 psychiatric disorders where common variants are unlikely to have large effects [143]. On the other hand, linkage studies 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 earliest 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 population 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 Affected Sib Pair Study of Alcohol Dependence that additionally provided weaker, suggestive evidence for linkage on chromosomes 1q, 13q and 22q for alcohol dependence, on 2q, 9q and 19p for symptom count [147]. FurSamochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 dependence.

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 manner. 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 approaches; (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 genome 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 contributing 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 psychiatric disorders like schizophrenia or bipolar disorder [158–160]. The GWAS approach is not without limitations, however. Due to the risk of false positives resulting from the extraordinary amount of multiple testing inherent 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 Genetics of Alcohol Dependence

large sample sizes are required to obtain the necessary statistical power. Moreover, GWAS are designed to identify relatively common polymorphisms associated with the risk for a disease. Rare genetic variants with potentially 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 candidate genes for AUDs, alcohol-related phenotypes and comorbid 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 information about novel candidate genes for alcohol dependence, including genes involved in gene regulation, cell signaling, 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 alcohol-related phenotypes, including both clinical features like withdrawal severity and basic behavioral and neurophysiological measures (table 1, third part). Several risk loci identified by GWAS confirm previously observed associations of genes involved in alcohol metabolism with a risk for alcohol dependence and drinking behavior. For example, Frank et al. [162] detected genome-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 association of this chromosomal region with alcohol dependence 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 repeatedly been associated with alcohol consumption levels and risk for alcohol dependence (see above). This association 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 Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

83

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 analysis combining the diagnostic phenotype with an electrophysiological variable (amplitude of the P300, an eventrelated potential related to the processing of rare or novel 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 maximum 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 chromosomal loci.

Authors, year

Number of patients/controls

Identified gene loci

GWAS of alcohol dependence Johnson et al. COGA [161], 2006

120 patients, 160 controls

51 gene loci remain significant after Monte Carlo simulationbased 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 PHF3PTP4A1 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 alcoholdependent patients

Genome-wide significance for NKAIN1-SERINC2 in subjects of European, but not African descent 471 SNPs nominally associated, 53 survived region- and cohortwide 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

84

Study name

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

Table 1. Overview of GWAS on alcohol-related disorders

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. n.a. [204], 2011

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

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

GWAS of alcohol-related phenotypes Wang et al. COGA [176], 2013 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

Genetics of Alcohol Dependence

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

85

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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.

86

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

sults of transcriptome-wide expression analyses showing that the transcripts of these genes were involved in regulatory mechanisms of other alcohol-related candidate genes [172, 173]. At the system level, it has been suggested that the related endophenotypes are more directly related to the molecular effect of a gene variant than the clinical phenotype [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 endophenotypes show a weaker statistical association with the clinical phenotype of interest, but might be more directly related to the underlying pathophysiological mechanisms. 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 individuals) as a replication sample. They identified genetic 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 activity.

From Genes to Pathways

The genes associated with alcohol dependence in both GWAS and replicated candidate-based association studies are, at first sight, a rather heterogeneous group, with their products being involved in diverse biological functions, including (peripheral) alcohol metabolism, neurotransmission, 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 epitope with yet unknown function [178]. However, expression 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 Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 dependence 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 reliably replicated subsequently. A reason for the inconclusive results of most GWAS might be insufficient statistical power resulting from required corrections for multiple testing. Means to circumvent this problem include metaanalyses 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 variants in the ADH gene cluster and additionally identified a putatively regulatory variant rs1437396, located between 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 extragenic or located in thus far poorly characterized genes. To understand the biological processes that underlie the observed 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, expression quantitative trait locus studies allow the identification of both cis and trans regulatory associations of genetic variations with gene expression levels. Zuo et al. [172, 173] combined the GWAS approach with expression 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-

Summary and Future Perspectives

Considerable progress has been made in the research of genetic contributions to alcohol dependence. For several genetic variations, a prominent role in the risk for developing alcohol-related disorders is largely undisputed. Most prominently, several functional variations in genes involved in alcohol metabolism, namely the ADH and ALDH genes, play a major role in modulating alcohol-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 effects of multiple genes with minor individual impact. However, the importance of alcohol-metabolizing enzymes becomes evident as most robust associations in Europeans have been observed in the ADH gene cluster. With recent advances in high-throughput genotyping Genetics of Alcohol Dependence

and bioinformatics, GWAS in large samples become feasible 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. lower heritability) and (b) advanced analytic techniques that make use of both array and next-generation sequencing technologies [193]. A comprehensive discussion of studies 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 potential 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 semiquantitative 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], although 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 within 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 remains to be done to elucidate the neural mechanisms underlying 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 variants and other rare genetic variations, and gene expression data. As we learn more about the genetic framework of AUD, we find that different genetic variants lead to varying responses to treatment options, which might improve our ability to design individualized therapy concepts [135, 199]. One example for the potential pharmacogeNeuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

87

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 distribution of a priori defined sets of genes between populations of interest. Joslyn et al. observed that level of alcohol response phenotypes (i.e. self-report on the effects of ethanol, subjective high assessment, body sway [180]) showed a systematic distribution difference in 173 genes, including CDH13 and XRCC5, which had been identified as susceptibility genes for ethanol sensitivity and alcohol dependence elsewhere [161, 181, 182]. Multiple genes identified in the GSEA were related to glutamatergic signaling, a finding well in line with previous results of animal studies and candidate gene analyses [183, 184]. Additionally, the muscarinic receptor (CHRM2) has been replicated in 2 GWAS [185, 186] and several other well-replicated candidate 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 transmitter systems. Many of these variants do not only affect the risk for AUD, but also have an influence on other substance-related disorders (e.g. GABRA2, CHRM2, NFKB1) [68, 106, 132, 135, 187].

netic approaches is the association of the OPRM1 Asn40Asp variation with the efficacy of the opioid antagonist 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 outcomes being a promising approach in the near future. More generally, from a clinical perspective, distinct phenotype characterization of a vast number of individuals including data on AUDs and alcohol-related phenotypes, comorbid psychiatric and medical disorders as well

as therapy response is needed to facilitate the identification of patients that might benefit from certain therapeutic lines. The above considerations refer both to pharmacotherapeutic and to psychotherapeutic approaches. The identification of reliable genetic markers predicting the response to psychotherapy, medications and their combination remains a great scientific challenge for the future. Acknowledgements This work was supported by the Deutsche Forschungsgemeinschaft (SFB 779, TP A8). We thank Claudia Hägele for her helpful comments on the paper.

References

88

11 12

13 14

15

16

17

18

19

20

hol dependence risk in a national twin sample: consistency of findings in women and men. Psychol Med 1997;27:1381–1396. Kałwa A: Impulsivity and decision making in alcohol-addicted individuals. Psychiatr Pol 2013;47:325–334. Kendler KS, Heath AC, Neale MC, Kessler RC, Eaves LJ: A population-based twin study of alcoholism in women. JAMA 1992; 268: 1877–1882. McGue M: The behavioral genetics of alcoholism. Curr Dir Psychol Sci 1999;8:109–115. Prescott CA, Kendler KS: Genetic and environmental contributions to alcohol abuse and dependence in a population-based sample of male twins. Am J Psychiatry 1999;156:34–40. 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. Bohman M, Sigvardsson S, Cloninger CR: Maternal inheritance of alcohol abuse. Crossfostering analysis of adopted women. Arch Gen Psychiatry 1981;38:965–969. Cloninger CR, Bohman M, Sigvardsson S: Inheritance of alcohol abuse. Cross-fostering analysis of adopted men. Arch Gen Psychiatry 1981;38:861–868. 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. Heath AC: Twin studies reveal greater concordance between monozygotic (MZ) than between dizygotic twins (DZ). Alc Health Res World 1995;19:166–171. Kendler KS, Neale MC, Heath AC, Kessler RC, Eaves LJ: A twin-family study of alcoholism in women. Am J Psychiatry 1994; 151: 707–715.

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

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 predisposition 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 alcoholism: neurobiology of high alcohol-drinking 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. Behav Genet 2002;32:363–388. 26 Whatley VJ, Johnson TE, Erwin VG: Identification and confirmation of quantitative trait loci regulating alcohol consumption in congenic strains of mice. Alcohol Clin Exp Res 1999;23:1262–1271. 27 Edenberg HJ: The genetics of alcohol metabolism: role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health 2007;30:5–13. 28 Hurley TD, Edenberg HJ, Li T-K: Pharmacogenomics of alcoholism; in Licinio J, Wong M-L (eds): Pharmacogenomics: The Search for Individual Therapies. Weinheim, WileyVCH, 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 dehydrogenase genotypes and alcoholism in Chinese men. Am J Hum Genet 1991;48:677–681.

Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 dependence syndrome, 30 years later: a commentary. The 2006 H. David Archibald lecture. Addiction 2007;102:1522–1530. 4 American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, ed 4. Washington, American Psychiatric Association, 1994. 5 World Health Organization: International Classification of Disease, ed 10. Geneva, WHO, 1993. 6 American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders (DSM-5). Washington, American Psychiatric Association, 2013. 7 American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders. Washington, American Psychiatric Association, 1987. 8 Buscemi L, Turchi C: An overview of the genetic susceptibility to alcoholism. Med Sci Law 2011;51(suppl 1):S2–S6. 9 Dragan WŁ, Oniszczenko W, Czerski PM, Dmitrzak-Weglarz M: Family-based association study of dopaminergic gene polymorphisms and EAS temperamental traits. Psychiatr Pol 2013;47:185–195. 10 Heath AC, Bucholz KK, Madden PA, Dinwiddie SH, Slutske WS, Bierut LJ, Statham DJ, Dunne MP, Whitfield JB, Martin NG: Genetic and environmental contributions to alco-

Genetics of Alcohol Dependence

44 Grant BF, Stinson FS, Dawson DA, Chou SP, Dufour MC, Compton W, Pickering RP, Kaplan K: Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders: results from the national 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: results from the national epidemiologic survey on alcohol and related conditions. Arch Gen Psychiatry 2004;61:361–368. 46 Szewczuk-Bogusławska M, Flisiak-Antonijczuk 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 subjects. 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 alcoholism characterized by Cloninger’s and Lesch’s typologies. Ann Acad Med Stetin 2010;56:33–39. 50 Samochowiec J, Kucharska-Mazur J, Grzywacz A, Pelka-Wysiecka J, Mak M, Samochowiec A, Bienkowski P: Genetics of Lesch’s typology of alcoholism. Prog Neuropsychopharmacol Biol Psychiatry 2008;32:423–427. 51 Heinz A, Friedel E, Müller DJ, Puls I, Wrase J: Genetic research with intermediate phenotypes: phenocopies, perspectives and pitfalls. Addiction 2007;102:1696–1697. 52 Mayfield RD, Harris RA, Schuckit MA: Genetic factors influencing alcohol dependence. Br J Pharmacol 2008;154:275–287. 53 Puls I, Gallinat J: The concept of endophenotypes in psychiatric diseases meeting the expectations? Pharmacopsychiatry 2008; 41 (suppl 1):S37–S43. 54 Samochowiec J, Grzywacz A, Kucharska-Mazur J, Samochowiec A, Horodnicki J, PelkaWysiecka J, Syrek S: Family-based and casecontrol association studies of glutamate receptor GRIK3 Ser310Ala polymorphism in Polish patients and families with alcohol dependence. Neurosci Lett 2006;396:159–162. 55 Bevilacqua L, Goldman D: Genes and addictions. 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 personality traits in alcohol-dependent individuals: a pilot case control study. Neuropsychobiology 2011;64:38–46. 58 Rietschel M, Treutlein J: The genetics of alcohol dependence. Ann NY Acad Sci 2013;1282: 39–70. 59 Schuckit MA: An overview of genetic influences 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 deficiency 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 dehydrogenase (ALDH2) gene responsible for alcoholinduced flushing increases turnover of the enzyme tetramers in a dominant fashion. J Clin Invest 1996;98:2027–2032. 64 Grochans E, Grzywacz A, Małecka I, Samochowiec A, Karakiewicz B, Samochowiec J: Research on associations between selected polymorphisms of genes DRD2, 5HTT, GRIK3, ADH4 and alcohol dependence syndrome. Psychiatr Pol 2011;45:325–335. 65 Kimura Y, Nishimura FT, Abe S, Fukunaga T, Tanii H, Saijoh K: Polymorphisms in the promoter region of the human class II alcohol dehydrogenase (ADH4) gene affect both transcriptional 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, Samochowiec 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 phenotypes: results from a larger multicenter association study. Addict Biol 2011;16:323–333. 69 Heinz A, Beck A, Wrase J, Mohr J, Obermayer K, Gallinat J, Puls I: Neurotransmitter systems in alcohol dependence. Pharmacopsychiatry 2009;42(suppl 1):S95–S101.

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

89

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

30 Higuchi S, Matsushita S, Imazeki H, Kinoshita T, Takagi S, Kono H: Aldehyde dehydrogenase genotypes in Japanese alcoholics. Lancet 1994;343:741–742. 31 Kendler KS, Gardner C, Dick DM: Predicting alcohol consumption in adolescence from alcohol-specific and general externalizing genetic risk factors, key environmental exposures and their interaction. Psychol Med 2011;41:1507–1516. 32 Young-Wolff KC, Enoch M-A, Prescott CA: The influence of gene-environment interactions on alcohol consumption and alcohol use disorders: a comprehensive review. Clin Psychol Rev 2011;31:800–816. 33 Guo G, Elder GH, Cai T, Hamilton N: Geneenvironment 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 interaction 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 environmental influences on substance use across development. Twin Res Hum Genet 2007;10:315–326. 36 Dick DM, Rose RJ, Viken RJ, Kaprio J, Koskenvuo M: Exploring gene-environment interactions: socioregional moderation of alcohol use. J Abnorm Psychol 2001;110:625–632. 37 Rose RJ, Dick DM, Viken RJ, Kaprio J: Geneenvironment interaction in patterns of adolescent drinking: regional residency moderates 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 alcohol 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 recommended drinking limits. Alcohol Clin Exp Res 2005;29:902–908. 43 Błachut M, Badura-Brzoza K, Jarzab M, Gorczyca P, Hese RT: Dual diagnosis in psychoactive substance abusing or dependent persons. Psychiatr Pol 2013;47:335–352.

90

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 human dopamine D2 receptor gene in alcoholism. JAMA 1990;263:2055–2060. 84 Wang F, Simen A, Arias A, Lu Q-W, Zhang H: A large-scale meta-analysis of the association between the ANKK1/DRD2 TAQ1A polymorphism and alcohol dependence. Hum Genet 2013;132:347–358. 85 Dick DM, Wang JC, Plunkett J, Aliev F, Hinrichs A, Bertelsen S, Budde JP, Goldstein EL, Kaplan D, Edenberg HJ, Nurnberger J, Hesselbrock 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 D2 receptor (DRD2) with alcoholism. Alcohol Clin Exp Res 1998;22:505–512. 87 Gelernter J, Goldman D, Risch N: The A1 allele at the D2 dopamine receptor gene and alcoholism. A reappraisal. JAMA 1993; 269: 1673–1677. 88 Grzywacz A, Jasiewicz A, Małecka I, Suchanecka A, Grochans E, Karakiewicz B, Samochowiec A, Bieńkowski P, Samochowiec J: Influence of DRD2 and ANKK1 polymorphisms 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 D2 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 D2 receptor gene and alcohol dependence in the German population. Alcohol 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-control studies and evidence of publication bias. Mol Psychiatry 2007;12:454–461. 92 Slatkin M: Linkage disequilibrium – understanding the evolutionary past and mapping the medical future. Nat Rev Genet 2008; 9: 477–485.

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

93 Neville MJ, Johnstone EC, Walton RT: Identification 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 D2 (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, Mukhopadhyay I: Dopaminergic mutations: within-family association and linkage in multiplex alcohol 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: Influence of DRD2 and ANKK1 genotypes on apomorphine-induced growth hormone (GH) response in alcohol-dependent patients. Prog Neuropsychopharmacol Biol Psychiatry 2010;34:45–49. 98 Yang B-Z, Kranzler HR, Zhao H, Gruen JR, Luo X, Gelernter J: Association of haplotypic variants in DRD2, ANKK1, TTC12 and NCAM1 to alcohol dependence in independent 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-Consuegra I, Jimenez-Arriero MA, Hoenicka J: The ANKK1 protein associated with addictions has nuclear and cytoplasmic localization and shows a differential response of Ala239Thr to apomorphine. Neurotox Res 2011;20:32–39. 101 Hoenicka J, Quinones-Lombrana A, EspanaSerrano L, Alvira-Botero X, Kremer L, PerezGonzalez R, Rodriguez-Jimenez R, JimenezArriero MA, Ponce G, Palomo T: The ANKK1 gene associated with addictions is expressed in astroglial cells and upregulated by apomorphine. Biol Psychiatry 2010;67:3–11. 102 Luo X, Kranzler HR, Zuo L, Wang S, Blumberg HP, Gelernter J: CHRM2 gene predisposes to alcohol dependence, drug dependence and affective disorders: results from an extended case-control structured association 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.

Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

70 Heinz A, Goldman D, Gallinat J, Schumann G, Puls I: Pharmacogenetic insights to monoaminergic dysfunction in alcohol dependence. Psychopharmacology (Berl) 2004;174: 561–570. 71 Samochowiec J: The role of dopamine neurotransmission on the development of ethanol dependence and course of abstinence syndrome. 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 ethanol withdrawal in rats. Pharmazie 1995; 50: 815–818. 73 Samochowiec J, Kucharska-Mazur J, Kaminski R, Smolka M, Rommelschpacher H, Wernicke C, Tymicz A, Schmidt LG: Norepinephrine transporter gene polymorphism is not associated with susceptibility to alcohol dependence. 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 Neuropsychiatr Genet 2003;117B:51–56. 75 Hendershot CS, Lindgren KP, Liang T, Hutchison KE: COMT and ALDH2 polymorphisms moderate associations of implicit drinking motives with alcohol use. Addict Biol 2012;17:192–201. 76 Samochowiec J, Kucharska-Mazur J, Grzywacz A, Jabłoński M, Rommelspacher H, Samochowiec 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 dependence. 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, Hesselbrock 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-methyltransferase. 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 Taiwan. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:457–461. 80 Mokrovic G, Matosic A, Hranilovic D, Stefulj J, Novokmet M, Oreskovic D, Balija M, Marusic 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 sequence variation in the dopamine transporter gene polymorphism DAT 1 in alcohol dependence. Psychiatr Pol 2008;42:443–452.

Genetics of Alcohol Dependence

113 Van der Zwaluw CS, Engels RCME, Vermulst AA, Rose RJ, Verkes RJ, Buitelaar J, Franke B, Scholte RHJ: A serotonin transporter 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, opiate, 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-dependent individuals. Alcohol 2001;24:129–135. 117 Smolka M, Sander T, Schmidt LG, Samochowiec J, Rommelspacher H, Gscheidel N, Wendel B, Hoehe MR: Mu-opioid receptor variants and dopaminergic sensitivity in alcohol withdrawal. Psychoneuroendocrinology 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, Samochowiec 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: Association of micro-opioid receptor (OPRM1) gene polymorphism with response to naltrexone 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, Kuperman S, Porjesz B, Begleiter H, Foroud T, Edenberg HJ: Association of the kappa-opioid system with alcohol dependence. Mol Psychiatry 2006;11:1016–1024. 123 Carr LG, Foroud T, Bice P, Gobbett T, Ivashina J, Edenberg H, Lumeng L, Li TK: A quantitative trait locus for alcohol consumption 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-preferring and -nonpreferring rats. Neuroscience 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 preference. 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 neuropeptide Y levels. Nature 1998;396:366–369. 127 Lappalainen J, Kranzler HR, Malison R, Price LH, Van Dyck C, Rosenheck RA, Cramer J, Southwick S, Charney D, Krystal J, Gelernter J: A functional neuropeptide Y Leu7Pro polymorphism associated with alcohol 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 neuropeptide Y (NPY) gene associated with alcohol dependence. Alcohol Clin Exp Res 2005; 29: 702–707. 129 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, Nurnberger JI, Tischfield JA, Porjesz B, Edenberg HJ, Foroud T: Neuropeptide Y receptor genes are associated with alcohol dependence, 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 dependence. Alcohol Clin Exp Res 2008;32:1023– 1030. 133 Grzywacz A, Kucharska-Mazur J, Samochowiec J: Association studies of dopamine D4 receptor gene exon 3 in patients with alcohol dependence. Psychiatr Pol 2008; 42: 453–461. 134 Cardon LR, Bell JI: Association study designs 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, Kuperman S, Porjesz B, Schuckit MA, Tischfield JA, Almasy LA, Nurnberger JI, Foroud T: Association of NFKB1, which encodes a subunit of the transcription factor NF-kappaB, with alcohol dependence. Hum Mol Genet 2008;17:963–970.

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

91

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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-aspartate receptor 1 and 2b subunits are associated with alcoholism-related traits. Biol Psychiatry 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, Begleiter H, Porjesz B, Dick DM: Association of GABRA2 with drug dependence in the collaborative study of the genetics of alcoholism 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: Association of GABRG3 with alcohol dependence. 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 between GABRA1 and drinking behaviors in the collaborative study on the genetics of alcoholism sample. Alcohol Clin Exp Res 2006;30:1101–1110. 109 Sander T, Ball D, Murray R, Patel J, Samochowiec 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 dependence. Alcohol Clin Exp Res 1999;23:427–431. 110 Winterer G, Mahlberg R, Smolka MN, Samochowiec 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 electroencephalogram voltage in normal subjects 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, Hesselbrock 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, Samochowiec J: The influence of parents personality and DRD4 and 5HTT gene polymorphisms on predisposition to alcohol dependence in their sons. Psychiatr Pol 2011; 45: 337–347.

92

147 Prescott CA, Sullivan PF, Kuo P-H, Webb BT, Vittum J, Patterson DG, Thiselton DL, Myers JM, Devitt M, Halberstadt LJ, Robinson VP, Neale MC, van den Oord EJ, Walsh D, Riley BP, Kendler KS: Genomewide linkage study in the Irish Affected Sib Pair Study of Alcohol Dependence: evidence for a susceptibility region for symptoms of alcohol dependence on chromosome 4. Mol Psychiatry 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, Hesselbrock V, Crowe R, Schuckit M, Porjesz B, Begleiter H, Reich T: Alcoholism susceptibility 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, Somberg LK, Page G, Krystal JH, Gelernter J: Confirmation and fine mapping of the chromosome 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, Thiselton DL, van den Oord EJ, Walsh D, Kendler KS, Prescott CA: Identification of susceptibility loci for alcohol-related traits in the Irish Affected Sib Pair Study of Alcohol Dependence. Alcohol Clin Exp Res 2006; 30: 1807–1816. 152 Easton DF, Pooley KA, Dunning AM, Pharoah 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 association 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, Masson G, Jakobsdottir M, Thorlacius S, Helgason A, Aben KK, Strobbe LJ, Albers-Akkers 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 receptorpositive 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, Hubner R, Wild R, Rowan A, Fielding S, Howarth K, Consortium C, Silver A, Atkin W, Muir K, Logan R, Kerr D, Johnstone E,

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

155

156

157

158 159 160

161

162

163

Sieber O, Gray R, et al: A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat Genet 2007;39:984–988. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, ProkuninaOlsson 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, Goldstein JL, et al: A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 2007; 316:1341–1345. 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, Meyre D, Polychronakos C, Froguel P: A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 2007; 445:881–885. McPherson R, Pertsemlidis A, Kavaslar N, Stewart A, Roberts R, Cox DR, Hinds DA, Pennacchio LA, Tybjaerg-Hansen A, Folsom AR, Boerwinkle E, Hobbs HH, Cohen JC: A common allele on chromosome 9 associated with coronary heart disease. Science 2007;316:1488–1491. Duan J, Sanders AR, Gejman PV: Genomewide approaches to schizophrenia. Brain Res Bull 2010;83:93–102. Craddock N, Sklar P: Genetics of bipolar disorder. Lancet 2013;381:1654–1662. Lee KW, Woon PS, Teo YY, Sim K: Genome wide association studies (GWAS) and copy number variation (CNV) studies of the major psychoses: what have we learnt? Neurosci Biobehav Rev 2012;36:556–571. 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 Genetics of Alcoholism. Am J Med Genet B Neuropsychiatr Genet 2006; 141B:844– 853. Frank J, Cichon S, Treutlein J, Ridinger M, Mattheisen M, Hoffmann P, Herms S, Wodarz N, Soyka M, Zill P, Maier W, Mössner R, Gaebel W, Dahmen N, Scherbaum N, Schmäl C, Steffens M, Lucae S, Ising M, Müller-Myhsok B, Nöthen MM, Mann K, Kiefer F, Rietschel M: Genome-wide significant association between alcohol dependence and a variant in the ADH gene cluster. Addict Biol 2012;17:171–180. 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.

Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

136 Samochowiec A, Grzywacz A, Kaczmarek L, Bienkowski P, Samochowiec J, Mierzejewski P, Preuss UW, Grochans E, Ciechanowicz A: Functional polymorphism of matrix metalloproteinase-9 (MMP-9) gene in alcohol dependence: family and case control study. Brain Res 2010;1327:103–106. 137 Sher KJ, Grekin ER, Williams NA: The development 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 association of the serotonin 5-HT1B G681C polymorphism 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, Rommelspacher 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 regulatory MAOA gene promoter polymorphism in antisocial and anxious-depressive alcoholics. J Neural Transm 2000;107:681–689. 142 Baars MY, Müller MJ, Gallhofer B, Netter P: Relapse (number of detoxifications) in abstinent male alcohol-dependent patients as related to personality traits and types of tolerance to frustration. Neuropsychobiology 2013;67:241–248. 143 Risch N, Merikangas K: The future of genetic studies of complex human diseases. Science 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, Sullivan PF: Genomewide association studies: history, 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 American 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, Hesselbrock 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, Begleiter H: Genome-wide search for genes affecting the risk for alcohol dependence. Am J Med Genet 1998;81:207–215.

Genetics of Alcohol Dependence

173

174

175

176

177

178

179

180 181

182

Zhang X-Y, Deng H-W, Krystal JH, Zhang F, Luo X: Genome-wide association study of alcohol dependence implicates KIAA0040 on chromosome 1q. Neuropsychopharmacology 2012;37:557–566. 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 dependence identified by genome-wide association study. PLoS One 2011;6:e26726. Meyer-Lindenberg A, Weinberger DR: Intermediate phenotypes and genetic mechanisms of psychiatric disorders. Nat Rev Neurosci 2006;7:818–827. Mier D, Kirsch P, Meyer-Lindenberg A: Neural substrates of pleiotropic action of genetic variation in COMT: a meta-analysis. Mol Psychiatry 2010;15:918–927. 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, Johnson EO, Kang S, Kapoor M, Kramer J, Kuperman S, Madden PA, Manz N, Martin NG, McClintick JN, Montgomery GW, Nurnberger 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. 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: Genome-wide association study of theta band event-related oscillations identifies serotonin receptor gene HTR7 influencing risk of alcohol dependence. Am J Med Genet B Neuropsychiatr Genet 2011;156B:44–58. 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. 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. Schuckit MA: Low level of response to alcohol as a predictor of future alcoholism. Am J Psychiatry 1994;151:184–189. MacLaren EJ, Bennett B, Johnson TE, Sikela JM: Expression profiling identifies novel candidate genes for ethanol sensitivity QTLs. Mamm Genome 2006;17:147–156. 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, Wichmann 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 association 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 Psychiatry 2008;65:826–838. 184 Spanagel R: Alcoholism: a systems approach from molecular physiology to addictive behavior. 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 identification. 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 association analysis of symptoms of alcohol dependence in the molecular genetics of schizophrenia (MGS2) control sample. Alcohol Clin Exp Res 2011;35:963–975. 187 Edenberg HJ, Dick DM, Xuei X, Tian H, Almasy L, Bauer LO, Crowe RR, Goate A, Hesselbrock V, Jones K, Kwon J, Li T-K, Nurnberger JI, O’Connor SJ, Reich T, Rice J, Schuckit MA, Porjesz B, Foroud T, Begleiter H: Variations in GABRA2, encoding the alpha 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: Genetic 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 alcohol dependence (in Polish). Psychiatr Pol 2013;47:117–126. 190 Sobański JA: Contemporary research directions in ‘Polish psychiatry’ (Psychiatria Polska) 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 association studies: emerging approaches. Nat Rev Genet 2010;11:259–272. 192 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, Cambridge 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 association studies of alcohol dependence and substance use disorders. Curr Psychiatry Rep 2011;13:147–155.

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

93

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

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 regulating multiple metabolic phenotypes in a Japanese 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 associated to alcohol dependence and is the major genetic determinant of ‘daily maximum 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, Mullersman JE, Liu Y, Pan Y: Family-based association 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 genomewide 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, Howells W, Bertelsen S, Hinrichs AL, Almasy L, Breslau N, Culverhouse RC, Dick DM, Edenberg HJ, Foroud T, Grucza RA, Hatsukami D, Hesselbrock V, Johnson EO, Kramer 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, Hesselbrock V, Hinrichs A, Kramer J, Kuperman S, Nurnberger JI, Rice JP, Schuckit MA, Taylor R, Todd Webb B, Tischfield JA, Porjesz B, Foroud T: Genome-wide association 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, Pergadia ML, Goate AM, Lind PA, McEvoy BP, Schrage AJ, Grant JD, Chou Y-L, Zhu R, Henders AK, Medland SE, Gordon SD, Nelson EC, Agrawal A, Nyholt DR, Bucholz KK, Madden PAF, Montgomery GW: A quantitative-trait genome-wide association study of alcoholism risk in the community: findings 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 association study of alcohol dependence: significant findings in African- and EuropeanAmericans 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,

94

202

203

204

205

alcohol dependence. Int J Neuropsychopharmacol 2013;16:271–278. Zuo L, Wang K, Zhang XY, Krystal JH, Li CS, Zhang F, Zhang H, Luo X: NKAIN1SERINC2 is a functional, replicable and genome-wide significant risk gene region specific for alcohol dependence in subjects of European descent. Drug Alcohol Depend 2013;129:254–264. 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. Schumann G, Coin LJ, Lourdusamy A, Charoen P, Berger KH, Stacey D, Desrivières 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: Genomewide association and genetic functional studies identify autism susceptibility candidate 2 gene (AUTS2) in the regulation of alcohol consumption. Proc Natl Acad Sci USA 2011; 108: 7119–7124. Baik I, Cho NH, Kim SH, Han B-G, Shin C: Genome-wide association studies identify

Neuropsychobiology 2014;70:77–94 DOI: 10.1159/000364826

206

207

208

209

genetic loci related to alcohol consumption in Korean men. Am J Clin Nutr 2011; 93: 809–816. 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 behavior. Chin Med J (Engl) 2012; 125: 1127– 1134. Kapoor M, Wang JC, Wetherill L, Le N, Bertelsen S, Hinrichs AL, Budde J, Agrawal A, Bucholz K, Dick D, Harari O, Hesselbrock V, Kramer J, Nurnberger JI Jr, Rice J, Saccone N, Schuckit M, Tischfield J, Porjesz B, Edenberg HJ, Bierut L, Foroud T, Goate A: A meta-analysis of two genome-wide association studies to identify novel loci for maximum number of alcoholic drinks. Hum Genet 2013;132:1141–1151. 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. Wang K-S, Liu X, Zhang Q, Wu L-Y, Zeng M: Genome-wide association study identifies 5q21 and 9p24.1 (KDM4C) loci associated with alcohol withdrawal symptoms. J Neural Transm 2012;119:425–433.

Samochowiec /Samochowiec /Puls / Bienkowski /Schott  

 

 

 

 

Downloaded by: Göteborgs Universitet 130.241.16.16 - 1/11/2018 6:20:22 PM

196 Palmer RHC, McGeary JE, Francazio S, Raphael BJ, Lander AD, Heath AC, Knopik VS: The genetics of alcohol dependence: advancing 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 contribution of genetics to addiction therapy approaches. 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, Martin NG, Willemsen G, de Geus EJC, Vogelzangs N, Penninx BW, Whitfield JB, Montgomery 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

Genetics of alcohol dependence: a review of clinical studies.

Alcohol dependence is a common severe psychiatric disorder with a multifactorial etiology. Since the completion of the human genome project and with t...
3MB Sizes 0 Downloads 5 Views