Skip to main content
Alcohol and Alcoholism (Oxford, Oxfordshire) logoLink to Alcohol and Alcoholism (Oxford, Oxfordshire)
. 2016 Sep 26;51(5):507–514. doi: 10.1093/alcalc/agw046

Overview of the Genetics of Alcohol Use Disorder

Elisabeth A Tawa 1, Samuel D Hall 1, Falk W Lohoff 1,*
PMCID: PMC5004749  PMID: 27445363

Abstract

Aims

Alcohol Use Disorder (AUD) is a chronic psychiatric illness characterized by harmful drinking patterns leading to negative emotional, physical, and social ramifications. While the underlying pathophysiology of AUD is poorly understood, there is substantial evidence for a genetic component; however, identification of universal genetic risk variants for AUD has been difficult. Recent efforts in the search for AUD susceptibility genes will be reviewed in this article.

Methods

In this review, we provide an overview of genetic studies on AUD, including twin studies, linkage studies, candidate gene studies, and genome-wide association studies (GWAS).

Results

Several potential genetic susceptibility factors for AUD have been identified, but the genes of alcohol metabolism, alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH), have been found to be protective against the development of AUD. GWAS have also identified a heterogeneous list of SNPs associated with AUD and alcohol-related phenotypes, emphasizing the complexity and heterogeneity of the disorder. In addition, many of these findings have small effect sizes when compared to alcohol metabolism genes, and biological relevance is often unknown.

Conclusions

Although studies spanning multiple approaches have suggested a genetic basis for AUD, identification of the genetic risk variants has been challenging. Some promising results are emerging from GWAS studies; however, larger sample sizes are needed to improve GWAS results and resolution. As the field of genetics is rapidly developing, whole genome sequencing could soon become the new standard of interrogation of the genes and neurobiological pathways which contribute to the complex phenotype of AUD.

Short summary

This review examines the genetic underpinnings of Alcohol Use Disorder (AUD), with an emphasis on GWAS approaches for identifying genetic risk variants. The most promising results associated with AUD and alcohol-related phenotypes have included SNPs of the alcohol metabolism genes ADH and ALDH.

INTRODUCTION

Alcohol Use Disorder (AUD) is a chronic psychiatric condition characterized by drinking patterns that lead to detrimental emotional, physical, and social outcomes. The Centers for Disease Control and Prevention (CDC) has reported that alcohol use contributes to approximately 88,000 deaths annually in the United States (Stahre et al., 2014), reflecting high morbidity and mortality. To diagnose individuals with AUD, the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (Mizokawa et al., 2013) utilizes 11 criteria pertaining to excessive alcohol use, alcohol abuse, and alcohol dependence. The range of symptoms encompassed in the criteria for AUD diagnosis, including drinking more or for longer than intended or continuing to drink despite psychological or health problems, for instance, demonstrates the disorder's heterogeneous clinical presentation.

Recent investigations of the intersection of AUD with epidemiological factors and comorbid psychiatric disorders indicate the high and rising prevalence of AUD in the United States. Findings from the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC) Waves 1 and 2 have revealed that lifetime DSM-IV AUD is as high as 36.7% in persons aged 30–44, 42.0% in men compared to women (19.5%), and 34.1% in non-Hispanic whites (Grant et al., 2016). Furthermore, AUD frequently co-occurs with other psychiatric disorders, including mood and anxiety disorders (Regier et al., 1990), post-traumatic stress disorder (Sampson et al., 2015), and other substance use disorders (Kessler et al., 1997). According to the Epidemiologic Catchment Area Study, roughly half of individuals with AUD have a ‘dual diagnosis’, or comorbid psychiatric disorder (Regier et al., 1990), and the National Comorbidity Study presented the finding that ¾ of men and women with AUD met lifetime criteria for the diagnosis of a psychiatric illness (Kessler et al., 1997). These data highlight the heterogeneity of AUD and overlap with other psychiatric disorder that often also have strong genetic heritability estimates.

The etiology of AUD, which encompasses a variety of behavioral, environmental, psychological, and physiological factors, may be genetically predisposed. Lifetime drinking history is an important behavioral risk factor, which includes the age of first alcohol use, average number of drinks per day, and number of years of heavy drinking (Grant and Dawson, 1998). Environmental factors, such early life stressors (including physical or sexual abuse) increase the risk for AUD later in life (Enoch, 2011). High trait anxiety and other psychological factors are associated with an increased risk for developing AUD (Poikolainen, 2000). Physiological factors, such as alcohol withdrawal, also influence the risk of developing AUD (Becker and Mulholland, 2014). In addition to family history, all of the above risk factors for AUD have been previously predicted to have an underlying genetic cause. Approximately 50% of the risk for developing AUD is due to genetics, while the remaining percent may be due to either environmental factors, or gene-environment interactions. Therefore, a genetic predisposition to the addictive effects of alcohol, combined with other environmental risk factors, may cause harmful lifetime drinking patterns and AUD development. The current review provides an overview of the diverse types of genetic studies conducted on AUD, with an emphasis on recent genome-wide association studies (GWAS). Because AUD is a common psychiatric disorder characterized by a complex array of traits, GWAS may provide an effective experimental approach to identifying associated genetic variants. Further exploration of SNPs and other genetic variants associated with AUD could be used to advise at-risk individuals and develop more effective pharmacological interventions.

METHODS

A PubMed search was conducted for original GWAS published from 1/1/2005 to 2/1/2016 using a combination of the terms ‘genetics’, ‘GWAS’, ‘alcohol dependence’, and/or ‘alcohol use disorder’. These initial papers and recent reviews on the genetics of alcohol dependence were used to identify additional literature on twin studies, linkage studies, and candidate gene association studies of AUD. The GWAS section of the current paper focuses on findings with a P-value < 10−8.

RESULTS

Twin studies

Research using family, adoption, and twin studies was the first to demonstrate the role of genetics in AUD. The Australian twin-family study of alcohol use disorder (OZALC) found a greater concordance of alcohol dependence in monozygotic (56% for males) compared to dizygotic twins (33% for males) and a heritability estimate of 64% (Heath et al., 1997). More recent twin studies have established that AUD heritability ranges from 40% to 70% (Enoch and Goldman, 2001; Agrawal and Lynskey, 2008; Kendler et al., 2012) , with similar heritability estimates in both males and females (Heath et al., 1997; Prescott et al., 1999). One sample using male twins from the Vietnam Era Twin Registry reported different heritability estimates for 23 symptoms of alcohol dependence, further highlighting the heterogeneity of AUD (Slutske et al., 1999). Overall, the heritability estimates of these studies strongly suggest that genetic components contribute to AUD; however, given that the concordance rates are below 50%, other factors such as rare somatic de novo mutations, environmental influences, and gene-environment interactions might contribute to the remaining heritability.

Linkage studies

Because of the epidemiological evidence for genetic factors in AUD, the field has hoped for a straightforward identification of AUD risk alleles. The first comprehensive investigation into the genetics of AUD used linkage studies. Linkage studies are useful for identifying broad regions of the genome associated with large increases in risk for a disorder. The term linkage refers to the observation that two genetic markers on the same chromosome are often inherited together. This approach uses families with multiple affected members to determine chromosomal regions with genetic risk variants. Previous linkage studies have been most successful in rare autosomal dominant diseases with high penetrance, such as cystic fibrosis.

The first linkage studies on alcohol dependence from the Collaborative Study on the Genetics of Alcoholism (COGA) (Reich et al., 1998) and a sib-pair study from a Southwest American Indian tribe (Long et al., 1998) reported a broad risk locus on chromosome 4q. This region contains the genes that encode the isoforms of alcohol dehydrogenase. Additional studies have implicated chromosomal regions containing GABA-A (Wang et al., 2004) and CHRM2 (Wang et al., 2004) among other variants (Edenberg and Foroud, 2014). However, these linkage studies were unable to identify specific genetic risk variants associated with AUD and instead, found chromosomal regions with multiple linkage peaks. These results highlight the complex, polygenetic biology of AUD.

Candidate gene association studies

Case/control association studies can be performed to test candidate genes within chromosomal regions identified through linkage studies or based on neurological plausibility. These studies can detect small increases in genetic risk variants associated with a disorder. However, many candidate gene studies on AUD have found results with small effect sizes and are often difficult to replicate.

Based on previous linkage studies, the strongest associations have been identified in the alcohol metabolism genes, alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH). Alcohol metabolism is a two-step process where ethanol is first oxidized to acetaldehyde by ADH and then further oxidized to acetate by ALDH. Accumulation of the toxic intermediate acetaldehyde can cause adverse physiological symptoms, including flushing syndrome, tachycardia, and nausea. The rate at which acetaldehyde is produced and converted to the waste product acetate is influenced by genetic variations encoding the isoenzymes of ADH and ALDH. Individuals with isoforms of ADH that oxidize ethanol at a faster rate and/or isoforms of ALDH that oxidize acetaldehyde at a slower rate are protected against AUD due to the unpleasant effects that result from acetaldehyde accumulation.

There are several isoforms of ADH that metabolize alcohol in the liver (ADH1A, ADH1B, ADH1C, and ADH4-7). A majority of alcohol oxidation is performed by the alcohol dehydrogenase isoform ADH1B. Individuals with the ADH1B*2 single-nucleotide polymorphism (SNP) rs1229984 (Arg48His) metabolize alcohol at a much faster rate than those with the ADH1B*1 variant. ADH1B*2 is more common in East Asians than other populations and has been found to have a protective effect on the risk for developing AUD (Luczak et al., 2006; Edenberg, 2007; Li et al., 2012). Although rare outside of East Asian populations, the ADH1B*2 variant was also found to have a protective effect in a sample of European-Americans (Bierut et al., 2012). In addition, the ADH1B*3 variant was associated with a decreased risk of AUD in African-American and Native American samples (Wall et al., 2003; Edenberg et al., 2006). While ADH1B is the primary enzyme, the isoforms ADH1A and ADH1C contribute to alcohol metabolism, but at lower concentrations of ethanol. Several studies have found a protective effect of the ADH1C*2 SNP rs698 (Ile350Val) (Biernacka et al., 2013b).

Variations in the isoforms of ALDH are also associated with the risk for AUD. A majority of the oxidation of acetaldehyde to acetate is performed by the acetaldehyde dehydrogenase isoform ALDH2. Individuals with the ALDH2*2 SNP rs671 (Glu504Lys) metabolize acetaldehyde at a much slower rate. Slow metabolism of acetaldehyde creates an unpleasant alcohol flushing syndrome, which is only found in East Asian populations. Therefore, the ALDH2*2 variant has been associated with a decreased risk of AUD (Luczak et al., 2006; Hurley and Edenberg, 2012; Li et al., 2012). When combined, both the ADH and ALDH2 variants are highly protective against the risk of developing AUD.

Although alcohol metabolizing enzyme polymorphisms are the most consistent candidate genes, genetic variants in several neurotransmitter systems have been associated with AUD. These include the glutamate receptor (GIRK1), gamma-aminobutyric acid receptor (GABA-A), D2 dopamine receptor (DRD2), dopamine transporter (SLC6A3), serotonin transporter (SLC6A4), tryptophan hydroxylase 1 (TPH1), catechol-O-methyltransferase (COMT), cholinergic muscarinic receptor (CHRM2), and u-opioid receptor (OPRM1, among others) (Edenberg et al., 2004; Ray and Hutchison, 2004; Wang et al., 2004; Munafo et al., 2007; Kranzler et al., 2009; McHugh et al., 2010; Tammimaki and Mannisto, 2010; Du et al., 2011; Xu and Lin, 2011; Chen et al., 2012). However, many of these genetic risk variants are dependent on gene-environment interactions and replication of findings has been difficult due to the generally small expected effect size, lack of power, and clinical and genetic heterogeneity.

Genome-wide association studies

Unlike candidate gene studies, which require prior knowledge about the neurobiology underlying potential genetic risk variants, GWAS provide high resolution analysis of the entire genome without an a priori hypothesis. GWAS are used to analyze several hundred thousand or millions of SNPs across the genome to identify differences in genotype frequencies between case-controlled individuals, without selecting for only a few specific genes. This is a useful approach for identifying genetic risk variants in complex disorders where small effect sizes are expected. GWAS on other complex phenotypes, such as rheumatoid arthritis, have been successful in identifying SNPs associated with disease processes (Suzuki et al., 2011). Promising results are also emerging from GWAS on mental illnesses, such as schizophrenia and bipolar disorder (Moskvina et al., 2009; Williams et al., 2011; Schizophrenia Working Group of the Psychiatric Genomics, 2014). Thus, this strategy may also be successful in identifying the genetic underpinnings of AUD.

In 2006, Johnson et al. analyzed 104,268 SNPs from 120 patients and 160 controls in a European-American population from the COGA sample (Johnson et al., 2006). This study identified 51 chromosomal regions in the genome that harbor at least three genetic risk markers, including genes of the cell adhesion molecules cadherin 11 (CDH11) and cadherin 13 (CDH13). However, these results were only nominally significant (P = 3.4 x 10−4). In 2009, the first positive GWAS analysis of alcohol dependence, published by Treutlein et al. (2009), examined 487 alcohol dependent patients and 1358 controls from a German population (Treutlein et al., 2009). This study found genome-wide significance for two intergenic loci on chromosome 2q35 (rs7590720 and rs1344694) close to the gene for peroxisomal trans-2-enoyl-CoA reductase (PECR). A follow-up study with 1024 patients and 966 controls replicated 15 significant SNPs, including rs11640875 in CDH13, rs1614972 in ADH1C, and rs13273672 in the GATA binding protein 4 (GATA4) (Table 1).

Table 1.

Positive findings from GWAS on alcohol use disorder and alcohol-related phenotypes

Author Phenotype Gene/SNP P Value Sample Ethnicity
Baik et al., 2011 Alcohol Consumption C12ORF24 (rs2074356) P  = 9.49e−59 1721 Korean Men
Biernacka et al., 2013a,b Alcohol Dependence KEGG pathway ID 72- Synthesis and degredation of keton bodies P  = 0.003 SAGE (2544) European- and African-American
Bierut et al., 2010 Alcohol Dependence GABRA2 P  < 0.05 SAGE (3829) European- and African American
PBX/knotted 1 homeobox 2, PKNOX2 P  = 1.93e−07
Chen et al., 2012 Alcohol Drinking Ankyrin repeat domain 7, ANKRD 7, and Cytokine-like 1, CYTL1, (rs6466686-rs4295599-rs12531086) (halotype) P  = 6.51e−8 904 Caucasian
Frank et al., 2012 Alcohol Dependence ALDH2 (rs671) P  = 1.27e−08 3501 German descent
ADH1 between ADH1B and ADH1C (rs1789891) P  = 1.27e−08
Gelernter et al., 2014 Alcohol Dependence ADH1B (rs1229984) P  = 1.17e−31 379 European Americans, European- and African American
ADH1B (rs1789882) P  = 6.33e−17 3318 African Americans
(total = 16,087)
ADH1C (Thr151Thr) P  = 4.94e−10
Between MTIF2 and CCDC88A on chromosome 2 (rs1437396) P  = 1.17e−10
Johnson et al., 2006 Alcohol Dependence 51 gene loci, including CDH11, CDH13 P  = 0.00034 COGA (280) European-American
Lind et al., 2010 Alcohol and Nicotine co-dependence Near MAP/microtubule affinity-regulating kinase 1, MARK1 P  = 1.90e−09 1087 Australian
Near DEAD (Asp-Glu-Ala-Asp) box helicase 6, DDX6 P = 2.6e−09
KIAA1409 P  = 4.86e−08
Lind et al., 2010 Alcohol and nicotine co-dependence Near semaphorin 3E, SEMA3E P  = 6.23e−06 OZALC (2386) Australian and Dutch
Park et al., 2013 Alcohol Dependence ALDH2 (rs671) P  = 8.42e−08 396 Korean
ADH1B (rs1229984) P  = 2.63e−21
ADH7 (rs1442492) P  = 6.28e−8
Quillen et al., 2014 Alcohol Dependence ALDH2 (rs671) P  = 4.55e−08 595 Chinese
Schumann et al., 2011 Alcohol Consumption AUTS2 (rs6943555) P  = 4e−08 12 population-based samples (26,316) European
Takeuchi et al., 2011 Alcohol Consumption ALDH2 (rs 671) P  = 3.6e−211 2974 drinkers, 1521 occasional drinkers, 1351 non-drinkers Japanese
ADH1B (rs1229984) P  = 3.6e−4
Treutlein et al., 2009 Alcohol Dependence Near peroxisomal trans-2-enoyl-CoA reductase, PECR (rs7590720 and rs1344694) P  = 9.72e−09 1845 German
CDH13 (rs11640875) P  = 1.84e−5 2020 German
ADH1C (rs1614972) P  = 1.41e−4
GATA4 (rs13273672) P  = 4.75e−4
Wang et al., 2011 Alcohol Dependence DSCMAL1 P  < 10e−08 COGA, OZALC 272 nuclear families European-American and Australian
Wang et al., 2011 Alcohol Dependence near endothelin receptor type B, EDNRB P  = 8.51e−06 COGA, OZALC 272 nuclear families European-American, African-American, and Australian
TPARP, CYFIP2, THEMIS, PSG11 P  = 2.31e−5
Wang et al., 2011 Alcohol Dependence KIAA0040, THSD7B, NRD1 P  = 1.86e−07 COGA (1594), SAGE (1669), OZALC (3334) European-American, African-American, and Australian
Wang et al., 2013 Alcohol Dependence Symptom Count 3 SNPs in C15ORF53 gene P  = 4.5e−8 COGA (2322) European-American
Zuo et al., 2011 Alcohol Dependence PHD finger protein 3, PHF3, - Protein tyrosine phosphatase type IVA 1, PTP4A1, locus P  < 10e−4 COGA, SAGE (4116) European- and African American
Zuo et al., 2012a,b Alcohol Dependence KIAA0040 P  =2 .8e−07 COGA, SAGE (4116) European- and African-American
Zuo et al., 2012a,b Alcohol and Nicotine co-dependence SH3 domain binding protein 5, SH3BP5 P  = 6.9e−6 SAGE (3143) European- and African American
Nuclear receptor subfamily 2, group C, member 2, NR2C2 P  = 5.3e−4
Plasminogen-like B2, PLGLB2 P  = 3.1e−08
Zuo et al., 2013 Alcohol Dependence NKAIN1-SERINC2 P  = 1.7e−07 COGA, SAGE (2927) European- and African-American
Zuo et al., 2013 Alcohol and Nicotine co-dependence IPO11-HTR1A region on chromosome 5q P  = 6.2e−9 COGA, SAGE (2214) European- and African-American

In an effort to increase sample size, several GWAS publications have analyzed genome-wide SNPs from the COGA, OZALC, and the Study of Addiction: Genetics and Environment (SAGE) samples. In 2010, Edenberg et al. performed the first complete GWAS of the COGA. Although this study found no single SNPs with genome-wide significance, it provided converging evidence for a chromosome 11 gene cluster. In 2011, Wang et al. conducted a low density GWAS with 11,120 SNPs from the COGA and OZALC samples and found genome-wide significance at P < 10−8 for DSCMAL1 (Table 1). However, other groups have found only nominally significant SNPs (P > 10−8) or no genome−wide significant markers by analyzing the COGA, OZALC, and SAGE samples (Bierut et al., 2010; Lind et al., 2010; Wang et al., 2011; Zuo et al., 2011, 2012b, 2013; Biernacka et al., 2013a) (Table 1).

The most consistent GWAS findings for AUD have confirmed previously reported associations for genetic risk variants in ADH and ALDH genes. In 2012, Frank et al. identified genome-wide significance for rs1789891 in the ADH1 gene cluster in a German population. This SNP was found to be in linkage disequilibrium with the functional variant ADH1C (Arg272Gln). Park et al. (2013) also found multiple nominally significant SNPs in the ADH gene cluster on chromosome 4q22-q23, as well as genome-wide significance for rs1442492 and rs10516441 in ADH7 and rs671 in ALDH2 in an East Asian sample. Likewise, other GWAS have shown that the ALDH2*2 variant rs671 (Glu504Lys) is associated with a decreased risk of AUD in East Asian populations (Takeuchi et al., 2011; Frank et al., 2012; Quillen et al., 2014). Additionally, GWAS have found that the ADH1B*2 SNP rs1229984 has a protective effect for AUD development (Takeuchi et al., 2011; Park et al., 2013). A recent study by Gelernter et al. (2014) found that the SNP rs1229984 decreased the risk of AUD in a European-American population as well. This study reported that the ADH1C variant (Thr151Thr) and the ADH1B SNP rs1789882 (Arg369Cyc) decreased the risk of AUD in an African-American population (Gelernter et al., 2014) (Table 1).

Several other GWAS have found genome-wide significant SNPs for other alcohol-related phenotypes, such as alcohol and nicotine codependence, and alcohol consumption. When analyzed for alcohol and nicotine comorbidity, Lind et al. (2010) found genome-wide significance for rs7530302 near MAP/microtubule affinity-regulating kinase 1 (MARK1), rs1784300 near DEAD box helicase 6 (DDX6), and rs12882384 in KIAA1409 (Lind et al., 2010). Likewise, Zuo et al. (2012a,b) identified SNPs in SH3 domain binding protein 5 (SH3BP5), nuclear receptor subfamily 2 group C member 2 (NR2C2), Plasminogen-like B2 (PLGLB2), and rs7445832 in IPO11-HRT1A region on chromosome 5q associated with alcohol and nicotine dependence codependence (Zuo et al., 2012a; Zuo et al., 2013). Schumann et al. (2011) identified genome-wide significance of alcohol consumption for AUT2 rs6943555 in a sample of 26,316 individuals from 12 population-based samples (Schumann et al., 2011). Additionally, Baik et al. (2011) measured genome-wide significance of alcohol consumption in a sample of 1,721 males and replicated SNPs on chromosome 12q24, including C12ORF51 rs2074356, which is LD with ALDH2, CCDC63, and MYL2 (Baik et al., 2011). In 2012, Chen et al. found genome-wide significance for SNP clusters in Ankyrin repeat domain 7 (ANKRD7) Cytokine-like 1 (CYTL1) associated with alcohol drinking. A 2013 study by Wang et al. also found significance for three SNPs in the C15ORF53 gene when analyzed for alcohol dependence symptom count (Wang et al., 2013) (Table 1).

While the recent use of GWAS to identify the underlying genetic components of AUD has been promising, there are several limitations of GWAS that must be considered. GWAS use a ‘hypothesis-free’ design by genotyping hundreds of thousands to 2 million markers simultaneously in cases and controls. This approach generates large amounts of data and creates issues with regard to multiple testing. The current stringent statistical correction for GWAS is a P value of 10−8. As a result, early GWAS in psychiatric phenotypes yielded negative findings (Sklar et al., 2008; Craddock and Sklar, 2013). In retrospect, those studies (despite sample sizes in the range of 1000–2000) were largely underpowered to detect risk variants of small effect. Current power and sample size estimates for GWAS with effect sizes of 1.05–1.2 range from 30,000 – 120,000 (Owen et al., 2010; Schizophrenia Working Group of the Psychiatric Genomics, 2014). While the use of a stringent P-value for GWAS avoids the detection of false positive findings, it might also miss ‘true’ variants. Recent attempts to address this issue have used pathway analysis and polygenic risk score approaches (Gelernter et al., 2014) but have not been widely applied to AUD genetic analyses.

As the field moves forward, it is important to identify expected findings. AUD is a complex disorder, and likely hundreds if not thousands of genes contribute to its broad and varied phenotype. Therefore, it is unlikely that GWAS will detect genes of large effect . In addition, given the current chip-based methodology of GWAS, this technology by design misses rare de novo mutations or insertion/deletion variants (Stefansson et al., 2008; Walsh et al., 2008; Clarke and Cooper, 2010). Furthermore, several findings have been for intronic SNPs with no clear understanding of their underlying biological relevance. It is expected that GWAS will continue to be the standard of investigation of current genetic efforts to understand AUD. As it has been done for other psychiatric phenotypes, GWAS in AUD will need a collaborative approach in the form of large meta-analyses (Cichon et al., 2009; Sklar et al., 2011). While efforts are ongoing (Dick and Agrawal, 2008), no AUD GWAS meta-analysis currently exists.

A changing definition of the heterogeneous phenotype of AUD may also pose a challenge to identifying genetic variants through GWAS. The above studies used the DSM-IV-TR criteria for alcohol dependence in order to define the phenotype. As the field of psychiatry transitions to the DSM-5 criteria for AUD, there may also be changes in the functional variants identified by GWAS. Future GWAS should focus on the endophenotypes of AUD in order to better understand the genetic connections to specific behavioral symptoms. Likewise, it will be important to separate the role of genetic variants due other substance use disorders and to comorbid psychiatric disorders. Defining specific phenotypes and separating comorbid disorders will be useful in order to parse genetic variants involved in multiple disorders and addictions from those only involved in AUD. Future studies may also focus on pathway analysis in order to better understand the heterogeneous group of variants currently identified by GWAS.

GCTA/GREML methods

To address the ‘missing heritability’ problem, or the fact that variations in single genes have not accounted for much of the heritability in diseases, phenotypes, or behavioral pathologies, researchers have adopted Genome-Wide Complex Trait Analysis (GCTA)/Genomic Restricted Maximum Likelihood (GREML) (Yang et al., 2011). GCTA/GREML, or GCTA, is a statistical method which estimates variance in genetics by quantifying the chance genetic similarity of individuals and comparing their similarity in trait measurements. By quantifying the additive contributions of a subset of genetic variants (SNPs) to a trait's heritability, GCTA can corroborate the findings of GWAS studies. If the GCTA estimate of SNP heritability is consistent with the total genetic heritability, it is implicated that those genetics variants have a causal effect on the observed phenotype (Yang et al., 2011).

In the context of AUD, GCTA could be applied to the subsets of previously discussed SNPs that reached genome-wide significance and were correlated with alcohol-dependent phenotypes. GCTA estimates could be used for diagnostic purposes and provide further insight as to whether variants in ADH and ALDH, among other genes, in fact contribute to the genetic predisposition for AUD.

In a study examining the heritability of behavioral disinhibition, a trait which has been previously linked to substance use disorder development, twin-estimated heritabilities, GCTA-estimated heritabilities, and genome-wide scores were calculated to determine genetics correlations among various indicators of substance use and behavioral inhibition (Vrieze et al., 2013). Vrieze et al. (2013) found that, in biometric twin models, behavioral inhibition was highly genetically correlated with all substance use traits (nicotine use/dependence, alcohol consumption, alcohol dependence, and drug use). Regarding alcohol dependence, heritability was as high as 56%, and the aggregate additive SNP effects estimated by GCTA on the parent sample accounted for 16% of the variance (Vrieze et al., 2013). Hence, Vrieze et al. (2013) found that substance use phenotypes, including those pertaining to alcohol use, and behavioral disinhibition share a genetic etiology, and that measured genetic variants contribute to their heritability.

Another study investigating the heritability of assorted substance dependencies, including alcohol, tobacco, cannabis, and illicit drugs, used GCTA estimates to conclude that common SNPs contribute to at least 20% of the variance in substance dependence vulnerability (Palmer et al., 2015). Because the GWAS findings on substance dependence broadly have been limited, Palmer et al. (2015) demonstrated the efficacy of GCTA in identifying the heritability of substance use disorders via aggregate effects of genetic variants. Overall, GCTA methods may greatly facilitate investigators’ abilities to make causal attributions of common SNPs to complex psychiatric conditions, including alcohol use phenotypes and dependence.

Whole genome sequencing

As the field of genomics is rapidly expanding, with advances in technology and decreases in costs, whole genome sequencing is expected to become feasible in the near future. Although GWAS are much more economical, the financial burden of whole genome sequencing could be outweighed by the value of genetic information obtained. Unlike GWAS, whole-genome sequencing is more likely to identify rare mutations, particularly recessive mutations, in exonic regions of the genome. These coding regions may have a strong impact on disease etiology and shed new light into possible pathophysiological mechanisms (Cirulli and Goldstein, 2010; Ng and Kirkness, 2010; Kato, 2015). Since exome analysis has been successful in identifying de novo and inherited point mutations in autism spectrum disorders, and it has been applied to mood and psychotic disorders, there is hope that exome/whole genome sequencing could be a highly beneficial tool in mapping the genetic architecture of substance use and addiction disorders (Kato, 2015).

CONCLUSIONS

Despite the evidence supporting the prominence of genetic factors in AUD's etiology, the identification of genetic risk variants has been difficult and labor intensive. With recent advances in technology, the most promising results stem from recent GWAS, which have helped to identify new variants in the genetics of AUD. Among the variants identified, the most significant SNPs remain in the alcohol metabolism enzyme genes, ADH and ALDH. Importantly, the prevalence of the various isoforms of ADH and ALDH differs among ethnicities and populations. Therefore, lower alcohol consumption in certain populations, as a result of the protective effect of alcohol metabolism SNPs, may be due to gene-environment interactions.

AUD prevention could be enhanced with a growing knowledge of the disorder's neurobiology and genetics. A growing body of literature on AUD genetics will improve both the understanding of at-risk individuals’ biology and the development of new medications. Although information such as family history can currently be used to identify at-risk individuals, understanding the genetic architecture of AUD could enable us to pinpoint these individuals with greater certainty. Understanding of the genetic risk factors involved could be important to guide personalized treatments of patients who have already developed AUD and to inform the development of new pharmacological and other novel interventions.

Acknowledgments

Funding

The review was supported by intramural research funding from the National Institute on Alcohol Abuse and Alcoholism. There are no competing financial interests.

Conflict of interest statement

None declared.

References

  1. Agrawal A, Lynskey MT (2008) Are there genetic influences on addiction: evidence from family, adoption and twin studies. Addiction 103:1069–81. [DOI] [PubMed] [Google Scholar]
  2. Baik I, Cho NH, Kim SH, et al. (2011) Genome-wide association studies identify genetic loci related to alcohol consumption in Korean men. Am J Clin Nutr 93:809–16. [DOI] [PubMed] [Google Scholar]
  3. Becker HC, Mulholland PJ (2014) Neurochemical mechanisms of alcohol withdrawal. Handb Clin Neurol 125:133–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Biernacka JM, Geske J, Jenkins GD, et al. (2013. a) Genome-wide gene-set analysis for identification of pathways associated with alcohol dependence. Int J Neuropsychopharmacol 16:271–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Biernacka JM, Geske JR, Schneekloth TD, et al. (2013. b) Replication of genome wide association studies of alcohol dependence: support for association with variation in ADH1C. PLoS One 8:e58798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bierut LJ, Agrawal A, Bucholz KK, et al. (2010) A genome-wide association study of alcohol dependence. Proc Natl Acad Sci USA 107:5082–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bierut LJ, Goate AM, Breslau N, et al. (2012) ADH1B is associated with alcohol dependence and alcohol consumption in populations of European and African ancestry. Mol Psychiatry 17:445–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Chen D, Liu F, Yang C, et al. (2012) Association between the TPH1 A218C polymorphism and risk of mood disorders and alcohol dependence: evidence from the current studies. J Affect Disord 138:27–33. [DOI] [PubMed] [Google Scholar]
  9. Cichon S, Craddock N, Daly M, et al. (2009) A framework for interpreting genome-wide association studies of psychiatric disorders The Psychiatric GWAS Consortium Steering Committee. Molecular Psychiatry 14:10–17. [DOI] [PubMed] [Google Scholar]
  10. Cirulli ET, Goldstein DB (2010) Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat Rev Genet 11:415–25. [DOI] [PubMed] [Google Scholar]
  11. Clarke AJ, Cooper DN (2010) GWAS: heritability missing in action. Eur J Hum Genet 18:859–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Craddock N, Sklar P (2013) Genetics of bipolar disorder. Lancet 381:1654–62. [DOI] [PubMed] [Google Scholar]
  13. Dick DM, Agrawal A (2008) The genetics of alcohol and other drug dependence. Alcohol Res Health 31:111–8. [PMC free article] [PubMed] [Google Scholar]
  14. Du Y, Nie Y, Li Y, et al. (2011) The association between the SLC6A3 VNTR 9-repeat allele and alcoholism-a meta-analysis. Alcohol Clin Exp Res 35:1625–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Edenberg HJ. (2007) The genetics of alcohol metabolism: role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health 30:5–13. [PMC free article] [PubMed] [Google Scholar]
  16. Edenberg HJ, Dick DM, Xuei X, et al. (2004) 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 74:705–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Edenberg HJ, Foroud T (2014) Genetics of alcoholism. Handb Clin Neurol 125:561–71. [DOI] [PubMed] [Google Scholar]
  18. Edenberg HJ, Xuei X, Chen HJ, et al. (2006) Association of alcohol dehydrogenase genes with alcohol dependence: a comprehensive analysis. Hum Mol Genet 15:1539–49. [DOI] [PubMed] [Google Scholar]
  19. Enoch MA. (2011) The role of early life stress as a predictor for alcohol and drug dependence. Psychopharmacology (Berl) 214:17–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Enoch MA, Goldman D (2001) The genetics of alcoholism and alcohol abuse. Curr Psychiatry Rep 3:144–51. [DOI] [PubMed] [Google Scholar]
  21. Frank J, Cichon S, Treutlein J, et al. (2012) Genome-wide significant association between alcohol dependence and a variant in the ADH gene cluster. Addict Biol 17:171–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Gelernter J, Kranzler HR, Sherva R, et al. (2014) Genome-wide association study of alcohol dependence:significant findings in African- and European-Americans including novel risk loci. Mol Psychiatry 19:41–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Grant BF, Dawson DA (1998) Age of onset of drug use and its association with DSM-IV drug abuse and dependence: results from the National Longitudinal Alcohol Epidemiologic Survey. J Subst Abuse 10:163–73. [DOI] [PubMed] [Google Scholar]
  24. Grant BF, Saha TD, Ruan WJ, et al. (2016) Epidemiology of DSM-5 drug use disorder: results from the National Epidemiologic Survey on Alcohol and Related Conditions-III. JAMA Psychiatry 73:39–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Heath AC, Bucholz KK, Madden PA, et al. (1997) Genetic and environmental contributions to alcohol dependence risk in a national twin sample: consistency of findings in women and men. Psychol Med 27:1381–96. [DOI] [PubMed] [Google Scholar]
  26. Hurley TD, Edenberg HJ (2012) Genes encoding enzymes involved in ethanol metabolism. Alcohol Res 34:339–44. [PMC free article] [PubMed] [Google Scholar]
  27. Johnson C, Drgon T, Liu QR, et al. (2006) 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 141B:844–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Kato T. (2015) Whole genome/exome sequencing in mood and psychotic disorders. Psychiatry Clin Neurosci 69:65–76. [DOI] [PubMed] [Google Scholar]
  29. Kendler KS, Chen X, Dick D, et al. (2012) Recent advances in the genetic epidemiology and molecular genetics of substance use disorders. Nat Neurosci 15:181–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Kessler RC, Crum RM, Warner LA, et al. (1997) Lifetime co-occurrence of DSM-III-R alcohol abuse and dependence with other psychiatric disorders in the National Comorbidity Survey. Arch Gen Psychiatry 54:313–21. [DOI] [PubMed] [Google Scholar]
  31. Kranzler HR, Gelernter J, Anton RF, et al. (2009) Association of markers in the 3′ region of the GluR5 kainate receptor subunit gene to alcohol dependence. Alcohol Clin Exp Res 33:925–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Li D, Zhao H, Gelernter J (2012) Strong protective effect of the aldehyde dehydrogenase gene (ALDH2) 504lys (*2) allele against alcoholism and alcohol-induced medical diseases in Asians. Hum Genet 131:725–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lind PA, Macgregor S, Vink JM, et al. (2010) A genomewide association study of nicotine and alcohol dependence in Australian and Dutch populations. Twin Res Hum Genet 13:10–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Long JC, Knowler WC, Hanson RL, et al. (1998) 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 81:216–21. [DOI] [PubMed] [Google Scholar]
  35. Luczak SE, Glatt SJ, Wall TL (2006) Meta-analyses of ALDH2 and ADH1B with alcohol dependence in Asians. Psychol Bull 132:607–21. [DOI] [PubMed] [Google Scholar]
  36. Mchugh RK, Hofmann SG, Asnaani A, et al. (2010) The serotonin transporter gene and risk for alcohol dependence: a meta-analytic review. Drug Alcohol Depend 108:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Mizokawa T, Wakisaka Y, Sudayama T, et al. (2013) Role of oxygen holes in Li(x)CoO(2) revealed by soft X-ray spectroscopy. Phys Rev Lett 111:056404. [DOI] [PubMed] [Google Scholar]
  38. Moskvina V, Craddock N, Holmans P, et al. (2009) Gene-wide analyses of genome-wide association data sets: evidence for multiple common risk alleles for schizophrenia and bipolar disorder and for overlap in genetic risk. Mol Psychiatry 14:252–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Munafo MR, Matheson IJ, Flint J (2007) Association of the DRD2 gene Taq1A polymorphism and alcoholism: a meta-analysis of case-control studies and evidence of publication bias. Mol Psychiatry 12:454–61. [DOI] [PubMed] [Google Scholar]
  40. Ng PC, Kirkness EF (2010) Whole genome sequencing. Methods Mol Biol 628:215–26. [DOI] [PubMed] [Google Scholar]
  41. Owen MJ, Craddock N, O'donovan MC (2010) Suggestion of roles for both common and rare risk variants in genome-wide studies of schizophrenia. Arch Gen Psychiatry 67:667–73. [DOI] [PubMed] [Google Scholar]
  42. Palmer RH, Brick L, Nugent NR, et al. (2015) Examining the role of common genetic variants on alcohol, tobacco, cannabis and illicit drug dependence: genetics of vulnerability to drug dependence. Addiction 110:530–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Park BL, Kim JW, Cheong HS, et al. (2013) Extended genetic effects of ADH cluster genes on the risk of alcohol dependence: from GWAS to replication. Hum Genet 132:657–68. [DOI] [PubMed] [Google Scholar]
  44. Poikolainen K. (2000) Risk factors for alcohol dependence: a case-control study. Alcohol Alcohol 35:190–6. [DOI] [PubMed] [Google Scholar]
  45. Prescott CA, Aggen SH, Kendler KS (1999) Sex differences in the sources of genetic liability to alcohol abuse and dependence in a population-based sample of U.S. twins. Alcohol Clin Exp Res 23:1136–44. [DOI] [PubMed] [Google Scholar]
  46. Quillen EE, Chen XD, Almasy L, et al. (2014) 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 165B:103–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Ray LA, Hutchison KE (2004) A polymorphism of the mu-opioid receptor gene (OPRM1) and sensitivity to the effects of alcohol in humans. Alcohol Clin Exp Res 28:1789–95. [DOI] [PubMed] [Google Scholar]
  48. Regier DA, Farmer ME, Rae DS, et al. (1990) Comorbidity of mental disorders with alcohol and other drug abuse. Results from the Epidemiologic Catchment Area (ECA) Study. JAMA 264:2511–8. [PubMed] [Google Scholar]
  49. Reich T, Edenberg HJ, Goate A, et al. (1998) Genome-wide search for genes affecting the risk for alcohol dependence. Am J Med Genet 81:207–15. [PubMed] [Google Scholar]
  50. Sampson L, Cohen GH, Calabrese JR, et al. (2015) Mental Health Over Time in a Military Sample: The Impact of Alcohol Use Disorder on Trajectories of Psychopathology After Deployment. J Trauma Stress 28:547–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Schizophrenia Working Group Of The Psychiatric Genomics, C. (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511:421–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Schumann G, Coin LJ, Lourdusamy A, et al. (2011) Genome-wide association and genetic functional studies identify autism susceptibility candidate 2 gene (AUTS2) in the regulation of alcohol consumption. Proc Natl Acad Sci USA 108:7119–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Sklar P, Ripke S, Scott LJ, et al. (2011) Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nature Genetics 43:977–U162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Sklar P, Smoller JW, Fan J, et al. (2008) Whole-genome association study of bipolar disorder Mol Psychiatry 13:558–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Slutske WS, True WR, Scherrer JF, et al. (1999) The heritability of alcoholism symptoms: ‘indicators of genetic and environmental influence in alcohol-dependent individuals’ revisited. Alcohol Clin Exp Res 23:759–69. [DOI] [PubMed] [Google Scholar]
  56. Stahre M, Roeber J, Kanny D, et al. (2014) Contribution of excessive alcohol consumption to deaths and years of potential life lost in the United States. Prev Chronic Dis 11:E109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Stefansson H, Rujescu D, Cichon S, et al. (2008) Large recurrent microdeletions associated with schizophrenia Nature 455:232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Suzuki A, Kochi Y, Okada Y, et al. (2011) Insight from genome-wide association studies in rheumatoid arthritis and multiple sclerosis. FEBS Lett 585:3627–32. [DOI] [PubMed] [Google Scholar]
  59. Takeuchi F, Isono M, Nabika T, et al. (2011) Confirmation of ALDH2 as a Major locus of drinking behavior and of its variants regulating multiple metabolic phenotypes in a Japanese population. Circ J 75:911–8. [DOI] [PubMed] [Google Scholar]
  60. Tammimaki AE, Mannisto PT (2010) Are genetic variants of COMT associated with addiction. Pharmacogenet Genomics 20:717–41. [DOI] [PubMed] [Google Scholar]
  61. Treutlein J, Cichon S, Ridinger M, et al. (2009) Genome-wide association study of alcohol dependence. Arch Gen Psychiatry 66:773–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Vrieze SI, Mcgue M, Miller MB, et al. (2013) Three Mutually Informative Ways to Understand the Genetic Relationships Among Behavioral Disinhibition, Alcohol Use, Drug Use, Nicotine Use/Dependence, and Their Co-occurrence: Twin Biometry, GCTA, and Genome-Wide Scoring. Behavior Genetics 43:97–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Wall TL, Carr LG, Ehlers CL (2003) Protective association of genetic variation in alcohol dehydrogenase with alcohol dependence in Native American Mission Indians. Am J Psychiatry 160:41–6. [DOI] [PubMed] [Google Scholar]
  64. Walsh T, Mcclellan JM, Mccarthy SE, et al. (2008) Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia. Science 320:539–43. [DOI] [PubMed] [Google Scholar]
  65. Wang JC, Foroud T, Hinrichs AL, et al. (2013) A genome-wide association study of alcohol-dependence symptom counts in extended pedigrees identifies C15orf53. Mol Psychiatry 18:1218–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Wang JC, Hinrichs AL, Stock H, et al. (2004) Evidence of common and specific genetic effects: association of the muscarinic acetylcholine receptor M2 (CHRM2) gene with alcohol dependence and major depressive syndrome. Hum Mol Genet 13:1903–11. [DOI] [PubMed] [Google Scholar]
  67. Wang KS, Liu X, Zhang Q, et al. (2011) A meta-analysis of two genome-wide association studies identifies 3 new loci for alcohol dependence. J Psychiatr Res 45:1419–25. [DOI] [PubMed] [Google Scholar]
  68. Williams HJ, Craddock N, Russo G, et al. (2011) Most genome-wide significant susceptibility loci for schizophrenia and bipolar disorder reported to date cross-traditional diagnostic boundaries. Hum Mol Genet 20:387–91. [DOI] [PubMed] [Google Scholar]
  69. Xu M, Lin Z (2011) Genetic influences of dopamine transport gene on alcohol dependence: a pooled analysis of 13 studies with 2483 cases and 1753 controls. Prog Neuropsychopharmacol Biol Psychiatry 35:1255–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Yang JA, Lee SH, Goddard ME, et al. (2011) GCTA: a tool for genome-wide complex trait analysis. American Journal of Human Genetics 88:76–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Zuo L, Gelernter J, Zhang CK, et al. (2012. a) Genome-wide association study of alcohol dependence implicates KIAA0040 on chromosome 1q. Neuropsychopharmacology 37:557–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Zuo L, Zhang CK, Wang F, et al. (2011) A novel, functional and replicable risk gene region for alcohol dependence identified by genome-wide association study. PLoS One 6:e26726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Zuo L, Zhang F, Zhang H, et al. (2012. b) Genome-wide search for replicable risk gene regions in alcohol and nicotine co-dependence. Am J Med Genet B Neuropsychiatr Genet 159B:437–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Zuo L, Zhang XY, Wang F, et al. (2013) Genome-wide significant association signals in IPO11-HTR1A region specific for alcohol and nicotine codependence. Alcohol Clin Exp Res 37:730–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Alcohol and Alcoholism (Oxford, Oxfordshire) are provided here courtesy of Oxford University Press

RESOURCES