Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Feb 1.
Published in final edited form as: Alcohol Clin Exp Res. 2015 Jul 14;39(8):1292–1311. doi: 10.1111/acer.12785

Drosophila and Caenorhabditis elegans as Discovery Platforms for Genes Involved in Human Alcohol Use Disorder

Mike Grotewiel 1, Jill C Bettinger 1
PMCID: PMC4656040  NIHMSID: NIHMS737907  PMID: 26173477

Abstract

Background

Despite the profound clinical significance and strong heritability of alcohol use disorder (AUD), we do not yet have a comprehensive understanding of the naturally occurring genetic variance within the human genome that drives its development. This lack of understanding is likely to be due in part to the large phenotypic and genetic heterogeneities that underlie human AUD. As a complement to genetic studies in humans, many laboratories are using the invertebrate model organisms (iMOs) Drosophila melanogaster (fruit fly) and Caenorhabditis elegans (nematode worm) to identify genetic mechanisms that influence the effects of alcohol (ethanol) on behavior. While these extremely powerful models have identified many genes that influence the behavioral responses to alcohol, in most cases it has remained unclear whether results from behavioral–genetic studies in iMOs are directly applicable to understanding the genetic basis of human AUD.

Methods

In this review, we critically evaluate the utility of the fly and worm models for identifying genes that influence AUD in humans.

Results

Based on results published through early 2015, studies in flies and worms have identified 91 and 50 genes, respectively, that influence 1 or more aspects of behavioral responses to alcohol. Collectively, these fly and worm genes correspond to 293 orthologous genes in humans. Intriguingly, 51 of these 293 human genes have been implicated in AUD by at least 1 study in human populations.

Conclusions

Our analyses strongly suggest that the Drosophila and C. elegans models have considerable utility for identifying orthologs of genes that influence human AUD.

Keywords: Sensitivity, Tolerance, Behavior, Genetics, Human


The Invertebrate Model organisms (iMOs) Drosophila melanogaster (fruit fly, hereafter Drosophila or fly) and Caenorhabditis elegans (nematode worm, hereafter C. elegans or worm) have become major experimental platforms for identifying genes, genetic pathways, and mechanisms related to the effects of alcohol on the nervous system and behavior. The extent to which genetic findings from iMOs are directly relevant to the genetics of human alcohol use disorder (AUD), however, has not been fully resolved. Despite an important but somewhat limited number of individual reports of orthologous genes influencing both alcohol-related behavior in iMOs and AUD in humans (e.g., Lasek et al., 2011b; Mathies et al., 2015; Morozova et al., 2009; Schumann et al., 2011), fundamental differences between invertebrate and human studies raise reasonable questions regarding the overall translational potential of genetic information from worms and flies. In this review, we comprehensively address this key issue.

BRIEF OVERVIEW OF THE GENETICS OF HUMAN AUD

Humans have deliberately produced and consumed ethanol (EtOH, hereafter alcohol) for 10,000 to 12,000 years (Dietrich et al., 2012). The original motivations for producing alcohol were probably quite varied and could have included the need for clean sources of hydration, a mechanism to bring individuals together for cultural festivals, and a form of payment for laborers (Dietrich et al., 2012). Although moderate alcohol consumption is associated with some health benefits (Spanagel, 2009), heavy consumption of alcohol contributes to a number of serious diseases and other societal problems that together lead to >5% of the global burden of disease and almost 6% of all deaths worldwide (WHO, 2014).

Individuals with AUD can exhibit a number of negative behavioral and physiological alcohol-related phenotypes that include alcohol abuse and alcohol dependence (NIAAA, dynamic). A diagnosis of AUD is warranted when an individual meets any 2 of 11 alcohol use-related criteria in the Diagnostic and Statistical Manual of Mental Disorders-5 (DSM-5) within a single 12-month period (American Psychiatric Association, 2013). Nearly 11% of all individuals in the United States will meet the criteria for AUD within the previous year (Edenberg and Foroud, 2013) and the lifetime risk for developing AUD approaches 25% (Haeny et al., 2014; Nery et al., 2014), although the lifetime risk of alcohol consumption leading to obvious harmful dysfunction might be much lower (Wakefield and Schmitz, 2014). Regardless of the analysis method, however, it is clear that AUD has an enormous, negative impact on human health across the globe.

The heritability of AUD is ~50% (Verhulst et al., 2014), suggesting that an in-depth understanding of the underlying genetic causes of AUD could greatly facilitate risk diagnosis and possibly successful treatment of affected individuals. Although genetic linkage, association, and other types of studies have generated suggestive evidence implicating a large number of genes in AUD or related disorders, to date, only genes encoding several major enzymes involved in the metabolic disposition of alcohol (ADH1A, ADH1B, ADH1C, and ALDH2) have been causally associated with AUD in multiple studies (Edenberg and Foroud, 2013, 2014; Rietschel and Treutlein, 2013). Thus, despite the significant negative health consequences of AUD and its heritability, we do not yet have a detailed understanding of the genes that drive alcohol abuse and other disorders related to problematic alcohol consumption.

INVERTEBRATE MODEL IN ALCOHOL RESEARCH

Flies and worms are the main iMOs currently being used to investigate the genetics of alcohol-related behavior. Fundamental advantages of these 2 iMOs include relatively low costs, high-throughput genetic analyses, and suites of powerful tools to manipulate the functions of individual genes along with a host of molecular and bioinformatic resources that facilitate genomic analyses. Importantly, there is considerable conservation between gene products in the iMOs and humans. In particular, much of the major molecular machinery that supports nervous system function, including several neurotransmitter systems, is structurally and functionally similar in iMOs and humans (Heberlein et al., 2004; Kaletta and Hengartner, 2006; Rodan and Rothenfluh, 2010a; Scholz and Mustard, 2011).

Behavioral responses to acute alcohol exposure in iMOs and humans are also conserved overall (Devineni and Heberlein, 2013; Rothenfluh et al., 2014). Low doses of alcohol elicit locomotor or psychomotor stimulation, while moderate doses of alcohol produce sedation in flies, worms, and humans. Tolerance to alcohol (a blunted effect of the drug after prolonged or repeated exposure) is also observed in both iMOs and humans. A number of assays for measuring the effects of alcohol on behavior have been described for both flies and worms.

In flies, alcohol sedation and/or the effects of alcohol on postural control can be assessed by exposing flies to alcohol vapor (which progressively increases their internal alcohol as occurs in humans when drinking alcohol) and then monitoring the ability of flies to move or remain standing over time (e.g., Bhandari et al., 2009; Lasek et al., 2011a; Maples and Rothenfluh, 2011; Rothenfluh et al., 2006; Sandhu et al., 2015; Schumann et al., 2011; Wen et al., 2005). Additionally, recovery from alcohol sedation is also a useful behavioral end point in flies (e.g., Cowmeadow et al., 2005; Ogueta et al., 2010). The locomotor stimulating effects of alcohol in flies can be assessed by exposing flies to alcohol vapor in conjunction with computer-based data analysis of video recordings of individual fly movement (Wolf et al., 2002). Furthermore, flies will preferentially consume food containing alcohol, allowing measurement of both volume and frequency of alcohol consumption (Devineni and Heberlein, 2009; Ja et al., 2007; Pohl et al., 2012; Shohat-Ophir et al., 2012; Xu et al., 2012), and this preference for alcohol can be modified by experience (Peru Y Colón de Portugal et al., 2014). Alcohol can act as a rewarding substance in flies (Kaun et al., 2011). Fly larvae can develop cognitive dependence on EtOH which can be measured as a decrease in learning ability in alcohol-dependent larvae that are undergoing withdrawal from alcohol (Robinson et al., 2012). Rapid tolerance to alcohol can be measured by assessing the ability of the drug to sedate flies during 2 alcohol exposures separated by a recovery period (flies are more resistant during the second exposure due to adaptations in the nervous system) (e.g., Bhandari et al., 2009; Chan et al., 2014; Cowmeadow et al., 2005; Scholz et al., 2000). Readers are directed to several comprehensive reviews on Drosophila as a model for alcohol behavior for additional details and discussion (Devineni and Heberlein, 2013; Kaun et al., 2012; Morozova et al., 2012; Robinson and Atkinson, 2013; Rodan and Rothenfluh, 2010b; Rothenfluh et al., 2014; Scholz and Mustard, 2011).

The behavioral effects of alcohol in worms are most often assessed by exposing the animals to alcohol and then tracking their locomotion either on an agar surface (crawling) or in a liquid medium (swimming) (e.g., Alaimo et al., 2012; Davies et al., 2003; Morgan and Sedensky, 1995; Speca et al., 2010). Tissue alcohol concentrations, which are substantially lower than exogenous concentrations, continue to rise slowly over the course of at least 50 minutes of exposure (Alaimo et al., 2012). Additional behavioral assays determine the effects of alcohol on egg-laying or hypercontraction of the body wall muscles (Davies et al., 2003; Hawkins et al., 2015). Worms develop acute functional tolerance to alcohol over the course of a 30-minute continuous exposure, which is observed as a decrease in locomotor sedation caused by alcohol despite increasing tissue alcohol concentrations (e.g., Davies et al., 2004; Jee et al., 2013; Mathies et al., 2015; Raabe et al., 2014). In addition, worms can develop chronic tolerance to alcohol, which is observed by withdrawal-induced clumping behavior after 20 hours of exposure (Davies et al., 2004), by withdrawal-induced tremor after 4 hours of exposure (Jee et al., 2013) or by withdrawal-induced increases in omega turns without accompanying reversals after 6 to 48 hours of exposure (Mitchell et al., 2010). Like flies, worms can also express complicated behavioral changes in response to alcohol; they can learn state dependently (Bettinger and McIntire, 2004) and can develop a preference for alcohol when they are exposed to it for 4 hours in the presence of a food source (Lee et al., 2009).

Studies in flies and worms have contributed substantially to our understanding of molecular–genetic mechanisms that influence the effects of alcohol on the nervous system and behavior. To compile all genetic manipulations (and therefore genes) that are important for alcohol-related behavior in flies and worms, we performed extensive searches of PubMed through 2014 (using combinations of the search terms EtOH, alcohol, Drosophila, C. elegans, names of individual investigators, etc.) and supplemented these searches with lists of genes obtained from several recent reviews (Davies and Bettinger, 2014; Kaun et al., 2012; Morozova et al., 2012; Rodan and Rothenfluh, 2010a; Rothenfluh et al., 2014) in addition to a recent publication from one of the authors (Mathies et al., 2015). Together, studies in flies and worms have identified 91 and 50 genes, respectively, that influence behavioral responses to alcohol (Tables 1 and 2). The 50 worm genes are orthologous to 50 genes in flies (identified by DIOPT scores ≥3 [Hu et al., 2011] and BLASTP searches [Altschul et al., 1997]). Of these 50 genes, only 7 (Adh, Clic, Dop1R1, iav, NPFR, Sir2, and slo) have been reported to influence behavioral responses to alcohol in flies. Similarly, the 91 fly genes that influence alcohol-related behavior are orthologous to 92 worm genes and only 8 of these (exc-4, exl-1, dop-4, npr-1, osm-9, sir-2.1, slo-1, and sodh-1) have been reported to be important for alcohol-related behaviors in worms. Presumably not all genes have been tested in both invertebrate models, and therefore, the overlap between genes in flies and worms described here probably underestimates the true genetic congruence in these 2 species.

Table 1.

Genes that Influence Alcohol-Related Behavior in Flies

Gene (aka) Function Genetic manipulation Behavioral assay Measure Effect Citation
AcCoAS Acetyl CoA synthesis LOF mutant Booz-o-mat ACT Decrease Kong et al. (2010a)
Adh Alcohol dehydrogenase LOF mutants Booz-o-mat % Sed Increase Wolf et al. (2002)
LOF mutants Unnamed Locomotor activity Decrease Grell et al. (1968)
LOF mutant Inebriometer MET, Rap Tol (MET) Decrease Cavener (1979)
LOF mutant Larval preference Larvae on alcohol agar Decrease Ogueta et al. (2010)
Akt1 (Akt) Serine/threonine kinase LOF mutant Sedation Time to Sed Increase Eddison et al. (2011)
RNAi Sedation Time to Sed Increase
Overexpression Sedation Time to Sed Decrease
Alk Receptor tyrosine kinase LOF mutants Booz-o-mat Time to Sed Increase Lasek et al. (2011b)
amn PACAP-like neuropeptide LOF mutants Inebriometer MET Decrease Moore et al. (1998)
LOF mutants Booz-o-mat ACT, % Sed Increase Wolf et al. (2002)
LOF mutant Sedation Time to Sed Decrease Peru Y Colon de Portugal et al. (2012)
apt Myb/SANT-containing transcription factor LOF mutants Sedation Time to Sed Increase McClure and Heberlein (2013)
RNAi Sedation Time to Sed Increase
Arf51F (Arf6) Small G protein LOF mutants Sedation Time to Sed Decrease Peru Y Colón de Portugal et al. (2012)
Arfip Dynactin binding LOF mutant Sedation Time to Sed Decrease Peru Y Colón de Portugal et al. (2012)
aru Epidermal growth factor receptor substrate LOF mutants Inebriometer MET, Time to Sed Decrease Eddison et al. (2011)
LOF mutants Sedation MET, Time to Sed Decrease
RNAi Inebriometer MET, Time to Sed Decrease
RNAi Sedation MET, Time to Sed Decrease
Bacc Ribosomal RNA-binding domain protein LOF mutants Sedation Time to Sed Increase Chen et al. (2013)
RNAi Sedation Time to Sed Increase
bsk Serine–threonine kinase LOF mutant Booz-o-mat ACT Increase Kapfhamer et al. (2012)
Bx (dLmo) Zinc-finger protein, LIM-type LOF mutant Sedation Time to Sed Decrease Lasek et al. (2011a)
GOF mutant Sedation Time to Sed Increase
CASK Molecular scaffold LOF mutants Inebriometer Rap Tol (MET) Decrease Maiya et al. (2012)
Cdc42 Small G protein GOF transgenic Sedation % Sed Increase Rothenfluh et al. (2006)
chico Insulin receptor substrate LOF mutant Inebriometer MET Decrease Corl et al. (2005)
Clic Numerous proposed LOF mutants eRING Time to Sed Increase Bhandari et al. (2012)
LOF mutants Sedation Time to Sed Increase Chan et al. (2014)
RNAi Sedation Time to Sed Increase
Crz Neuropeptide RNAi Sedation Time to Sed Increase McClure and Heberlein (2013)
LOF mutant Sedation recovery Time to Rec Increase Sha et al. (2014)
CrzR Corazonin receptor RNAi Sedation recovery Time to Rec Increase Sha et al. (2014)
cyc Transcription factor LOF mutant Sedation Rap Tol (Time to Sed) Decrease Pohl et al. (2013)
Cyp1 Peptidyl-prolyl cis-trans isomerase activity RNAi Inebriometer MET Increase Morozova et al. (2011)
dally Glypican LOF mutant Video ACT Decrease Joslyn et al. (2011)
LOF mutant Sedation Time to Sed Decrease
DAT Dopamine transporter LOF mutant Video ACT Decrease Kong et al. (2010b)
Overexpression Video ACT Increase
dlg1 Membrane-associated guanylate kinase LOF mutants Inebriometer Rap Tol, Chron Tol (MET) Decrease Maiya et al. (2012)
dlp Glypican LOF mutant Video ACT Decrease Joslyn et al. (2011)
LOF mutant Sedation Time to Sed Decrease
LOF mutant Sedation Rap Tol Increase
Dop1R1 Dopamine receptor LOF mutant Video ACT Decrease Bainton et al. (2000)
LOF mutant Video ACT Decrease Kong et al. (2010b)
Egfr Epidermal growth factor receptor Overexpression Sedation Time to Sed Increase Corl et al. (2009)
RNAi, inhibitor Sedation Time to Sed Decrease
elm P22 calcineurin B LOF mutant Inebriometer MET Decrease LaFerriere et al. (2008)
Fas2 Ig-domain adhesion molecule LOF mutants Inebriometer MET Decrease Cheng et al. (2001)
Fng UDP-glycosyltransferase RNAi Inebriometer Rap Tol (MET) Increase Morozova et al. (2011)
FOXO Transcription factor Overexpression Inebriometer MET Decrease Corl et al. (2005)
Fs Negative regulator of activin receptor signaling GOF mutant Inebriometer MET Increase Morozova et al. (2011)
GABA-B-R1 Metabotropic GABA receptor RNAi Sedation recovery Sed Rec Time Decrease Dzitoyeva et al. (2003)
Antagonist Sedation recovery Sed Rec Time Decrease
Agonist Sedation recovery Rap Tol (Sed Rec Time) Decrease
H15 T-box transcription factor RNAi Inebriometer MET Increase Morozova et al. (2011)
hang Zinc-finger protein LOF mutant Inebriometer Rap Tol (MET) Decrease Scholz et al. (2005)
LOF mutant eRING Rap Tol (Time to Sed) Decrease Bhandari et al. (2009)
homer Postsynaptic scaffolding LOF mutant Sedation Time to Sed Decrease Urizar et al. (2007)
LOF mutant Sedation Rap Tol (Time to Sed) Decrease
hppy Ste20 kinase LOF mutants Video, Sedation Time to Sed Increase Corl et al. (2009)
LOF mutant Sedation Time to Sed Increase Eddison et al. (2011)
Hsp26 Heat shock protein Transposons Sedation Rap Tol (Time to Sed) Decrease Awofala et al. (2011)
RNAi Sedation Rap Tol (Time to Sed) Decrease
Overexpression Sedation Rap Tol (Time to Sed) Increase
htl FGF receptor LOF mutant Video ACT Decrease King et al. (2014)
iav Cation channel LOF mutant Inebriometer MET Decrease Scholz (2005)
InR Insulin receptor LOF mutants Inebriometer MET Decrease Corl et al. (2005)
Jwa Microtubule-binding protein Antisense RNA Inebriometer Rap Tol (MET) Decrease Li et al. (2008)
Overexpression Sedation recovery Rap Tol (Sed Rec Time) Increase
KCNQ Voltage-dependent potassium channel LOF mutant Sedation Time to Sed Decrease Cavaliere et al. (2012)
RNAi Sedation Time to Sed Decrease
Overexpression Sedation Rap Tol (Time to Sed) Increase
Men Malate metabolism LOF mutant Inebriometer MET Increase Morozova et al. (2009)
RNAi Inebriometer MET Increase Morozova et al. (2011)
Mlc-c Myosin light chain RNAi Inebriometer MET Decrease Morozova et al. (2011)
moody G protein-coupled receptor LOF mutant Inebriometer MET Increase Bainton et al. (2005)
mys Integrin beta subunit LOF mutants eRING Time to Sed Decrease Bhandari et al. (2009)
LOF mutants eRING Rap Tol (Time to Sed) Increase
NmdaR1 NMDA receptor LOF mutant Inebriometer Rap Tol, Chron Tol (MET) Decrease Kaun et al. (2011)
NPF Neuropeptide F Overexpression Sedation Time to Sed Decrease Wen et al. (2005)
NPFR Neuropeptide F receptor RNAi Sedation Time to Sed Increase Wen et al. (2005)
RNAi CAFÉ Alcohol Preference Increase Shohat-Ophir et al. (2012)
Orco Odorant receptor co-receptor LOF mutant Booz-o-mat ACT Increase Kong et al. (2010a)
LOF mutants Alcohol trap Alcohol Odor Preference Decrease Schneider et al. (2012)
Osi9 Unknown GOF mutant Inebriometer MET Increase Morozova et al. (2011)
par-1 Serine–threonine kinase LOF mutant Video ACT Suppresses tao LOF King et al. (2011)
Pdk1 PIP3-dependent serine/threonine kinase Overexpression Sedation Time to Sed Decrease Eddison et al. (2011)
per CYC/CLK stability LOF mutant Sedation Time to Sed No circadian effect van der Linde and Lyons (2011)
LOF mutant Sedation Rap Tol (Time to Sed) Decrease
Pi3K21B (p60) PI3K inhibitory subunit Overexpression Inebriometer MET Decrease Corl et al. (2005)
Pi3K92E (p110) PI3K catalytic subunit Overexpression Sedation Time to Sed Decrease Eddison et al. (2011)
Dominant negative Sedation Time to Sed Increase
Pka-C1 (DCO) PKA-catalytic subunit 1 LOF mutants Inebriometer MET Decrease Moore et al. (1998)
LOF mutants Inebriometer MET Decrease Rodan et al. (2002)
Pka-R2 Protein kinase A regulatory subunit LOF mutant Sedation Time to Sed Increase Park et al. (2000)
Pkc98E Protein kinase C calcium independent RNAi Sedation Time to Sed Increase Chen et al. (2008)
RNAi Sedation Time to Sed Increase Chen et al. (2010)
psq Helix-loop-helix protein LOF mutant Inebriometer MET Decrease Morozova et al. (2009)
LOF mutant Inebriometer MET Decrease Morozova et al. (2011)
Pten Phosphatidylinositol 3,4,5-triphosphate phosphatase Overexpression Sedation Time to Sed Increase Eddison et al. (2011)
puc Tyrosine phosphatase LOF mutant Booz-o-mat ACT Decrease Kapfhamer et al. (2012)
Rac1 Small GTPase GOF transgenic Sedation % Sed Decrease Rothenfluh et al. (2006)
Rheb Small GTPase Overexpression Sedation Time to Sed Decrease Eddison et al. (2011)
rho Ligand-activated peptidase LOF mutant Sedation Time to Sed Decrease Corl et al. (2009)
Rho1 Small GTPase GOF transgenic Sedation % Sed Decrease Rothenfluh et al. (2006)
RhoGAP18B Rho Gap LOF mutants Video ACT Increase Rothenfluh et al. (2006)
Sedation % Sed Decrease
rl ERK kinase Overexpression Sedation Time to Sed Increase Corl et al. (2009)
rut Ca2+/calmodulin-sensitive adenylyl cyclase LOF mutants Inebriometer MET Decrease Moore et al. (1998)
LOF mutants Booz-o-mat ACT, % Sed Increase Wolf et al. (2002)
LOF mutant CAFÉ Alcohol Preference Decrease Xu et al. (2012)
S Chaperone LOF mutant Sedation Time to Sed Decrease Corl et al. (2009)
sca Fibrinogen LOF mutants Conditioned preference Alcohol preference Decrease Kaun et al. (2011)
SCAP Unknown GOF mutant Inebriometer MET Increase Morozova et al. (2011)
scb Integrin alpha subunit LOF mutants eRING Time to Sed Decrease Bhandari et al. (2009)
LOF mutants eRING Rap Tol (Time to Sed) Increase
sgl UDP-glucose 6-dehydrogenase LOF mutant Inebriometer MET Increase Morozova et al. (2011)
shi Dynamin LOF mutants Sedation recovery Rap Tol (Sed Rec Time) Decrease Krishnan et al. (2012)
Sip1 Unknown GOF mutant Inebriometer MET Decrease Morozova et al. (2011)
Sir2 Histone deacetylase LOF mutant Booz-o-mat ACT Decrease Kong et al. (2010a)
LOF mutant Booz-o-mat % Sed Decrease
LOF mutant Booz-o-mat Rap Tol (% Sed) Decrease
slo Calcium-activated potassium channel LOF mutants Sedation recovery Rap Tol (Sed Rec Time) Decrease Cowmeadow et al. (2005)
Overexpression Sedation Recovery Rap Tol (Sed Rec Time) Induced Rap Tol Cowmeadow et al. (2006)
LOF mutant Seizure induction Seizure Threshold (V) Decrease Ghezzi et al. (2012)
spi Epidermal growth factor receptor ligand Overexpression Sedation Time to Sed Increase Corl et al. (2009)
Spn27A Serine-type endopeptidase inhibitor LOF mutant Booz-o-mat ACT Decrease Kong et al. (2010a)
Syn Synapsin LOF mutant Inebriometer Rap Tol (MET) Increase Godenschwege et al. (2004)
Syx1A Syntaxin LOF mutant Sedation recovery Rap Tol (Sed Rec Time) Decrease Krishnan et al. (2012)
Tao Serine–threonine kinase LOF mutant Booz-o-mat ACT Decrease King et al. (2011)
tay AUTS2 ortholog LOF mutant Sedation Time to Sed Increase Schumann et al. (2011)
RNAi Sedation Time to Sed Increase
Tbh Tyramine hydroxylase LOF mutant Inebriometer Rap Tol (MET) Decrease Scholz et al. (2000)
LOF mutant Inebriometer Rap Tol (MET) Decrease Berger et al. (2004)
LOF mutant Inebriometer Rap Tol (MET) Decrease Scholz et al. (2005)
LOF mutant Sedation Rap Tol (Time to Sed) Decrease Awofala et al. (2011)
LOF mutants Alcohol trap Odor Preference Decrease Schneider et al. (2012)
tim CYC/CLK stability LOF mutant Sedation Rap Tol (Time to Sed) Decrease Pohl et al. (2013)
tra Female-specific mRNA splicing protein Overexpression (♂) Sedation Time to Sed Decrease Devineni and Heberlein (2012)
RNAi (♀) Sedation Time to Sed Increase
Tre1 G protein-coupled receptor GOF mutant Inebriometer MET Increase Morozova et al. (2011)
unc-13 Calmodulin binding; diacylglycerol binding LOF mutant CAFÉ Alcohol Preference Increase Das et al. (2013)
tank EI24/PIG8 ortholog LOF mutant, RNAi Booz-o-mat Time to Sed Increase Devineni et al. (2013)
w ABC transporter LOF mutant eRING Time to Sed Decrease Chan et al. (2014)
RNAi eRING Time to Sed Decrease Chan et al. (2014)
LOF mutant Fly Bar Time to LORR Increase van der Linde et al. (2014)

Columns are the gene name abbreviations, a brief functional description of the gene product, the type of genetic manipulation that was used (LOF, loss of function; RNAi, RNA interference, or overexpression), the name of the assay used to assess alcohol-related behavior, the behavioral end point that was measured in the assay (ACT, alcohol-stimulated locomotor activity or locomotor activity in the presence of alcohol; % Sed, % of flies sedated after a defined exposure to alcohol; MET, mean elution time; Rap Tol and Chron Tol, rapid tolerance and chronic tolerance, respectively, to the behavioral measure indicated in parentheses; Time to Sed, duration of alcohol exposure required to sedate flies to a defined level; Sed Rec Time, time required for flies to recover from sedation; Alcohol Preference, alcohol drinking preference; Time to LORR, alcohol exposure time required for flies to lose a defined amount of their ability to right themselves; Seizure Threshold (V), voltage of electric shock required to produce seizures in a defined fraction of subjects; Alcohol Odor Preference, fraction of flies captured in a trap with alcohol odor vapor), the effect of the genetic manipulation on the behavioral measure, and the citations for the relevant publications.

Table 2.

Genes that Influence Alcohol-Related Behavior in Worms

Gene Function Genetic manipulation Behavioral assay Measure Effect Citation
aex-3 RAB3 GTP exchange factor LOF mutant Crawling (400 mM) Crawling speed at 20 minutes Resistant Kapfhamer et al. (2008)
alh-13 Aldehyde dehydrogenase RNAi Crawling (400 mM) Crawling speed at 10 minutes Sensitive Alaimo et al. (2012)
alh-6 Aldehyde dehydrogenase RNAi Crawling (400 mM) Crawling speed at 10 minutes Sensitive Alaimo et al. (2012)
bbs-1 BBS1 LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Increased IS; Enhanced AFT Bettinger et al. (2012)
cat-1 Vesicular monoamine transporter LOF mutant State-dependent learning SDL Fails to develop SDL Bettinger and McIntire (2004)
cat-2 Tyrosine hydroxylase LOF mutant State-dependent learning; Alcohol preference SDL; Alcohol preference Fails to develop SDL Does not develop Alcohol preference Bettinger and McIntire (2004), Lee et al. (2009)
cha-1 Cholineacetyltransferase LOF mutant Hypercontraction Hypercontraction and Recovery Resistant Hawkins et al. (2015)
ctbp-1 Transcriptional co-repressor LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Increased IS; Decreased AFT Bettinger et al. (2012)
dgk-1 Diacylglycerol kinase LOF mutant Crawling (500 mM)
 Crawling (400 mM)
Crawling speed at 20 minutes; Crawling speed; initial sensitivity and AFT Resistant; Decrease in IS Bettinger et al. (2012), Davies et al. (2003)
dpff-1 Component of SWI/SNF chromatin remodeling complex RNAi Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS Mathies et al. (2015)
dop-4 Dopamine receptor (D1-like) LOF mutant Alcohol disinhibition of crawling gait in liquid Body posture in liquid (500 mM) Resistant to Alcohol disinhibition of crawling gait in liquid Topper et al. (2014)
eat-6 Alpha subunit Na+/K+-ATPase LOF mutant Hypercontraction Hypercontraction and recovery No recovery from hypercontraction Hawkins et al. (2015)
egl-3 Proprotein convertase LOF mutant Alcohol withdrawal-induced “unaccompanied” omega turns (350 mM) Number of omega turns without a preceding reversal at 5 and 40 minutes No increase in “unaccompanied” omega turns Mitchell et al. (2010)
exc-4 CLIC; numerous proposed LOF mutant Crawling (400 mM), 10, 20, 30, 40, 50 minutes Crawling speed at 10, 20, 30, 40, 50 minutes Decrease in IS at 10 minutes; trend toward decrease in AFT Bhandari et al. (2012)
exl-1 CLIC; numerous proposed LOF mutant Crawling (400 mM), 10, 20, 30, 40, 50 minutes Crawling speed at 10, 20, 30, 40, 50 minutes Enhanced AFT Bhandari et al. (2012)
fat-1 Omega-3 fatty acid acyl desaturase LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Increased IS; No AFT Raabe et al. (2014)
fat-3 Delta-6 fatty acid desaturase LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Raabe et al. (2014)
fat-4 Delta-5 fatty acid desaturase LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Raabe et al. (2014)
let-526 Component of SWI/SNF chromatin remodeling complex RNAi Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS Mathies et al. (2015)
lips-7 Triacylglycerol lipase LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS; Enhanced AFT Bettinger et al. (2012)
nca-1 NALCN-related leak channel LOF mutant Swimming (400 mM) Frequency of body bends at 10 minutes Sensitive Speca et al. (2010)
nca-2 NALCN-related leak channel LOF mutant Swimming (400 mM) Frequency of body bends at 10 minutes Sensitive Speca et al. (2010)
nhr-49 Nuclear hormone receptor LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Bettinger et al. (2012)
npr-1 Neuropeptide Y-like GPCR LOF mutants Crawling (500 mM)
 Crawling (400 mM)
Crawling speed at 10, 30, 50 minutes; initial sensitivity and AFT
 Crawling speed at 10 and 30 minutes; initial sensitivity and AFT
Decreased IS; Enhanced AFT; Decreased IS; Enhanced AFT Bettinger et al. (2012), Davies et al. (2004)
osm-9 TRPV channel LOF mutant Crawling (500 mM) Crawling speed at 10, 30, 50 minutes; initial sensitivity and AFT Partial suppression of npr-1 enhanced AFT Davies et al. (2004)
pag-3 Zinc-finger transcription factor LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Bettinger et al. (2012)
pbrm-1 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased AFT Mathies et al. (2015)
phf-10 Component of SWI/SNF chromatin remodeling complex RNAi Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Mathies et al. (2015)
rab-3 Small molecular weight GTP-binding protein LOF mutant Dispersal assay (200 and 400 mM); Crawling (400 mM) Movement toward food; Crawling speed at 20 minutes Fast dispersal; Resistant Kapfhamer et al. (2008)
sbp-1 Transcription factor LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS; No AFT Bettinger et al. (2012)
seb-3 CRF receptor-like GPCR LOF mutant
  GOF mutant
Crawling (500 mM)
 Withdrawal induced tremor (400 mM)
Crawling speed at 10, 30, 50 minutes, AFT; Number of animals with tremor after withdrawal from 4-hour exposure LOF: Decreased AFT, GOF: Increased AFT; LOF: Resistant to tremor Jee et al. (2013)
sir-2.1 Transcription factor LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS; Enhanced AFT Bettinger et al. (2012)
slo-1 BK voltage and calcium-sensitive large conductance K+ channel LOF mutants Crawling (500 mM)
 Egg laying (500 mM)
 Crawling (400 mM)
Crawling speed at 20 minutes; Number of eggs laid on Alcohol; Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Resistant; Resistant; Decreased IS; Decreased AFT Bettinger et al. (2012), Davies et al. (2003), Dillon et al. (2013)
sodh-1 Alcohol dehydrogenase LOF mutants Crawling (200 mM); Crawling (400 mM) Crawling speed at 10 and 50 minutes; Crawling speed at 10 and 50 minutes; initial sensitivity and AFT Resistant; Resistant Alaimo et al. (2012)
swsn-1 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Mathies et al. (2015)
swsn-2.1 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Mathies et al. (2015)
swsn-2.2 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS; No AFT Mathies et al. (2015)
swsn-3 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Increased IS; No AFT Mathies et al. (2015)
swsn-4 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT No AFT Mathies et al. (2015)
swsn-6 Component of SWI/SNF chromatin remodeling complex RNAi Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS; No AFT Mathies et al. (2015)
swsn-7 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; initial sensitivity and AFT Decreased IS Mathies et al. (2015)
swsn-9 Component of SWI/SNF chromatin remodeling complex LOF mutant Crawling (400 mM) Crawling speed at 10 and 30 minutes; Initial sensitivity and AFT No AFT Mathies et al. (2015)
tph-1 Tryptophan hydroxylase LOF mutant Alcohol preference Alcohol preference Decrease in preference Lee et al. (2009)
unc-17 Vesicular acetylcholine transporter LOF mutant Hypercontraction Hypercontraction and Recovery Resistant to Hypercontraction Hawkins et al. (2015)
unc-18 SM protein LOF mutant Swimming (400 mM) Number of thrashes at 10 minutes Resistant Graham et al. (2009)
unc-25 Glutamic acid decarboxylase LOF mutant Hypercontraction Hypercontraction and Recovery Resistant to Hypercontraction Hawkins et al. (2015)
unc-47 GABA vesicular transporter LOF mutant Hypercontraction Hypercontraction and Recovery Resistant to Hypercontraction Hawkins et al. (2015)
unc-63 Nicotinic acetylcholine receptor LOF mutant Hypercontraction Hypercontraction and Recovery Resistant to Hypercontraction Hawkins et al. (2015)
unc-79 Interacts with voltage insensitive cation leak channels LOF mutant Immobility over a dose curve; Swimming (400 mM) EC50 (immobility) at 5 minutes
 Frequency of body bends at 10 minutes
Resistant; Sensitive Morgan and Sedensky (1995), Speca et al. (2010)
unc-80 Interacts with voltage insensitive cation leak channels LOF mutant Swimming (400 mM) Frequency of body bends at 10 minutes Sensitive Speca et al. (2010)

Columns are the gene name abbreviations, a brief functional description of the gene product, the type of genetic manipulation that was used (LOF, loss of function mutation; GOF, gain of function mutation; RNAi, RNA interference), the name of the assay used to assess alcohol-related behavior, the behavioral end point that was measured in the assay (Crawling speed, locomotion speed on an agar surface; Swimming, number of body bends or thrashes in a liquid medium; SDL, state-dependent learning; Alcohol preference; Hypercontraction; Alcohol disinhibition of crawling gait in a liquid assay; Alcohol withdrawal-induced “unaccompanied” omega turns, omega turns observed without accompanying reversal; Dispersal assay; Alcohol withdrawal-induced tremor; Egg-laying; Immobility in liquid), the effect of the genetic manipulation on the behavioral measure, and the citations for the relevant publications.

The large number of genes influencing alcohol response behaviors uniquely contributed by flies and worms highlights the combined analytical power of these iMOs for further understanding molecular–genetic mechanisms underlying behavioral responses to alcohol. The uniquely contributed genes represent a potentially rich resource for exploring conserved gene function in the context of acute behavioral responses to alcohol. Importantly, it is possible that some molecular–genetic processes may be more amenable for study in one model versus the other. The overlap of genes that influence alcohol response behaviors in both iMOs is also informative; the genes identified in both flies and worms encode a diverse set of proteins that mediate membrane flux of potassium (slo family) and other cations (iav/osm-9 family). Additionally, these gene products participate in histone deacetylation (Sir2 family), alcohol metabolism (Adh/sodh-1 family), and dopamine signaling (Dop1R1/dop-4 family) or have incompletely characterized functions (Clic family). Readers are referred to several excellent recent reviews for additional details on the molecular function of the genes in flies and worms (Davies and Bettinger, 2014; Kaun et al., 2012; Morozova et al., 2012; Rodan and Rothenfluh, 2010a; Rothenfluh et al., 2014).

GENETICS OF ALCOHOL BEHAVIOR IN INVERTEBRATE MODEL ORGANISM AND HUMAN AUD

The large number of genes identified in iMO studies (Tables 1 and 2) highlight the power of the fly and worm model systems for investigating fundamental molecular-genetic mechanisms that influence behavioral responses to alcohol. The utility of the iMOs for identifying orthologs of individual genes or conserved genetic pathways involved in human AUD, however, has not been systematically evaluated. In fact, as counterpoints to the conservation in behavioral responses to alcohol and the molecular underpinnings of nervous system function in humans and iMOs discussed above, there are a number of notable differences between studies in invertebrates and humans that could, in principle, lead to disparate findings across species. For example, studies in humans examine naturally occurring genetic variation that has undergone natural selection during evolution and (typically) determine how that variation is associated with problems related to chronic alcohol exposure that often lasts for years. Studies in iMOs are often just the opposite. Genetic variance in iMOs is usually generated in the laboratory, although naturally occurring genetic variation in EtOH responsiveness in worms and flies has been assessed in some studies (Davies et al., 2004a; Morozova et al., 2009). Most studies in iMOs have used genetic manipulations that are the most severe that still allow the organism to live and perform basic tasks like locomotion, and—since they are maintained in a laboratory setting—these genetic manipulations largely escape the forces of evolution. Alcohol exposure in iMOs is also typically acute (lasting minutes to hours). The alcohol-related behaviors routinely assessed in iMOs (sedation, tolerance, locomotor activation) are fundamentally distinct from and simpler than the phenotypes analyzed in humans (alcohol abuse, alcohol dependence, alcohol craving, etc.) because iMO phenotypic end points are devoid of human social influences and occur in response to forced exposure to alcohol. Consequently, the experimental questions that are addressed in iMOs and humans are substantially different. Studies in humans typically address questions such as: What are the naturally occurring genetic variants that are associated with an AUD, or an endophenotype of an AUD, in response to chronic, voluntary alcohol intake in a defined population? In contrast, studies in iMOs typically investigate questions such as: What are the genes required for normal behavioral responses to forced acute alcohol exposure?

The intrinsic differences between studies in iMOs and humans raise the question of the utility of using flies and worms for identifying individual genes and genetic pathways relevant for AUD. In this review, we address this issue by assessing the overlap—at the level of individual genes— between genetic results from studies on alcohol-related behaviors in iMOs and studies on AUD in humans. Additionally, we determine whether behavioral–genetic studies on alcohol in iMOs have identified conserved genetic pathways relevant to human AUD.

To assess the overlap between genetic findings in iMOs and humans at the level of individual genes, it was necessary to use common gene symbols for all orthologs, regardless of origin. We therefore identified human orthologs of fly and worm genes that influence alcohol-related behavior using DIOPT (Hu et al., 2011), FlyBase (Wilson et al., 2008), OrthoDB (Kriventseva et al., 2015), BLASTP (Altschul et al., 1997), and g:Profiler (Reimand et al., 2011) in addition to our recognition of well-established biochemical activities of gene products (e.g., the alcohol dehydrogenase [ADH]-encoding genes). Genes predicted to be conserved between iMOs and humans by these approaches were visually inspected and unconvincing orthologs (i.e., genes whose gene products were judged to be poorly conserved between humans and iMOs) were ignored. In practice, orthologs with fairly conservative scores (≥3) in DIOPT (which in turn uses several bioinformatic databases) were considered convincing.

The collection of genes that influence alcohol-related behavior in flies and worms (Tables 1 and 2) correspond to 293 unique orthologous genes in humans (hereafter iMO–human genes or orthologs; Table 3). Twenty-two of the iMO–human genes are derived from studies in both flies and worms, while 182 and 89 of the iMO–human genes are exclusively from studies in flies and worms, respectively. The identification of orthologs in some cases can be somewhat challenging (cf. Hu et al., 2011), and therefore, the list of genes in Table 3 should be viewed as highly representative of the current sum of iMO–human orthologs as opposed to being definitive for the inclusion or exclusion of any single potential iMO–human gene.

Table 3.

Invertebrate Model Organism (iMO)–Human Genes: Human Orthologs of Genes that Influence Alcohol-Related Behaviors in iMOs

Human gene iMO source Human gene iMO source Human gene iMO source Human gene iMO source Human gene iMO source Human gene iMO source
ABCG2 Fly CLIC1 Fly, Worm FGFR4 Fly LMO1 Fly PIK3CD Fly SLC6A3 Fly
ACSS1 Fly CLIC2 Fly, Worm FOXO1 Fly LMO2 Fly PIK3CG Fly SLC9A3R1 Fly
ACSS2 Fly CLIC3 Fly, Worm FOXO3 Fly LMO3 Fly PIK3R1 Fly SLC9A3R2 Fly
ACTL6A Worm CLIC4 Fly, Worm FOXO4 Fly LTK Fly PIK3R2 Fly SMARCA2 Worm
ACTL6B Worm CLIC5 Fly, Worm FOXO6 Fly MADD Worm PIK3R3 Fly SMARCA4 Worm
ADCY1 Fly CLIC6 Fly, Worm FST Fly MAP4K1 Fly PPIA Fly SMARCC1 Worm
ADH1A Fly, Worm COX6C Fly FSTL3 Fly MAP4K2 Fly PPIF Fly SMARCC2 Worm
ADH1B Fly, Worm CRYAA Fly GABBR1 Fly MAP4K3 Fly PRAF2 Fly SMARCD1 Worm
ADH1C Fly, Worm CRYAB Fly GAD1 Worm MAP4K5 Fly PRKACA Fly SMARCD2 Worm
AKT1 Fly CSAD Worm GAD2 Worm MAPK1 Fly PRKACB Fly SMARCD3 Worm
AKT2 Fly CSNK1A1 Fly GADL1 Worm MAPK10 Fly PRKACG Fly SMARCE1 Worm
AKT3 Fly CSNK1D Fly GFI1 Worm MAPK3 Fly PRKAR2A Fly SREBF1 Worm
ALDH family (19 genes) Worm CSNK1E Fly GFI1B Worm MAPK8 Fly PRKAR2B Fly SREBF2 Worm
CTBP1 Worm GPC1 Fly MAPK9 Fly PRKCE Fly STX1A Fly
ALK Fly CTBP2 Worm GPC2 Fly MARK1 Fly PRKCH Fly STX1B Fly
ARF6 Fly DBH Fly GPC3 Fly MARK2 Fly PSMD1 Fly STX2 Fly
ARFIP1 Fly DGKQ Worm GPC4 Fly MARK3 Fly PTEN Fly STX3 Fly
ARFIP2 Fly DLG1 Fly GPC5 Fly MARK4 Fly RAB3A Worm STX4 Fly
ARID1A Worm DLG2 Fly GPC6 Fly ME1 Fly RAB3B Worm STXBP1 Worm
ARID1B Worm DLG3 Fly GPR84 Fly ME2 Fly RAB3C Worm STXBP2 Worm
ARID2 Worm DLG4 Fly GRIN1 Fly ME3 Fly RAB3D Worm STXBP3 Worm
ARL6IP5 Fly DNM1 Fly HNF4A Worm MFNG Fly RAC1 Fly SYN1 Fly
ARNTL Fly DNM2 Fly HNF4G Worm MTNR1A Fly RAC2 Fly SYN2 Fly
ARNTL2 Fly DNM3 Fly HOMER1 Fly MTNR1B Fly RAC3 Fly SYN3 Fly
ATP12A Worm DPF1 Worm HOMER2 Fly MYL1 Fly RFNG Fly TAF4 Fly
ATP1A1 Worm DPF2 Worm HOMER3 Fly MYL3 Fly RHBDL1 Fly TAF4B Fly
ATP1A2 Worm DPF3 Worm HPGD Fly MYL4 Fly RHBDL2 Fly TAOK1 Fly
ATP1A3 Worm DRD1 Fly, Worm IGF1R Fly MYL6 Fly RHBDL3 Fly TAOK2 Fly
ATP1A4 Worm DRD5 Fly INSR Fly MYL6B Fly RHEB Fly TAOK3 Fly
ATP4A Worm DUSP10 Fly INSRR Fly NALCN Worm RHEBL1 Fly TBX20 Fly
AUTS2 Fly EGFR Fly IRS1 Fly NAT10 Fly RHOA Fly TH Worm
BBS1 Worm EI24 Fly IRS2 Fly NCAM1 Fly RHOB Fly TIMELESS Fly
BRD7 Worm EPS8 Fly IRS4 Fly NCAM2 Fly RHOC Fly TPH1 Worm
BRD9 Worm EPS8L1 Fly ITGB1 Fly NPY Fly SCAP Fly TPH2 Worm
CASK Fly EPS8L2 Fly ITGB2 Fly NPY1R Fly, Worm SDC1 Fly TRPV1 Fly, Worm
CDC42 Fly EPS8L3 Fly ITGB3 Fly NPY2R Fly, Worm SDC2 Fly TRPV2 Fly, Worm
CHAT Worm ERBB2 Fly ITGB5 Fly NPY4R Fly, Worm SDC3 Fly TRPV3 Fly, Worm
CHP1 Fly ERBB3 Fly ITGB7 Fly PBRM1 Worm SDC4 Fly TRPV4 Fly, Worm
CHP2 Fly ERBB4 Fly KCNMA1 Fly, Worm PCSK2 Worm SGK1 Fly TRPV5 Fly, Worm
CHRNA1 Worm FADS1 Worm KCNQ1 Fly PDPK1 Fly SGK2 Fly TRPV6 Fly, Worm
CHRNA2 Worm FADS2 Worm KCNQ2 Fly PER1 Fly SIRT1 Fly, Worm UGDH Fly
CHRNA3 Worm FADS3 Worm KCNQ3 Fly PER2 Fly SIRT3 Worm UNC13A Fly
CHRNA4 Worm FBRSL1 Fly KCNQ4 Fly PER3 Fly SLC18A1 Worm UNC13B Fly
CHRNA6 Worm FGFR1 Fly KCNQ5 Fly PHF10 Worm SLC18A2 Worm UNC13C Fly
CHRNB2 Worm FGFR2 Fly KCNU1 Fly, Worm PIK3CA Fly SLC32A1 Worm UNC79 Worm
CHRNB4 Worm FGFR3 Fly LFNG Fly PIK3CB Fly SLC6A2 Fly UNC80 Worm

Columns are the human orthologs of genes originally identified in the iMO source indicated (fly, Worm, or both).

Unfortunately, there is no consensus set of human AUD genes that can be used to determine which of the iMO–human genes (Table 3) have been implicated in human AUD. Thus, the 293 iMO–human genes were queried against a set of 732 human genes compiled from (i) several comprehensive reviews on the genetics of AUD (Edenberg and Foroud, 2013, 2014; Palmer et al., 2012; Rietschel and Treutlein, 2013; Schuckit, 2014) and (ii) genes in the HuGe Navigator (Yu et al., 2008) identified by the search terms “alcoholism.” A small number of these 732 human genes have established roles in AUD, whereas the remaining genes have been implicated in AUD by smaller scale studies, single studies only, etc. Although very few of the 732 genes were implicated by studies that observed formal statistical significance of association with AUD, we included all 732 genes in our analyses in an attempt to capture the broad landscape of genetic findings from studies in humans.

Of the 293 iMO–human genes (Table 3), 83 were found among our compiled set of 732 human AUD genes. Based on literature reviews, only 51 of these had been implicated in 1 or more aspects of AUD (Table 4). The remaining 32 genes were among the HuGe Navigator “alcoholism” genes, but had no experimentally supported connection to AUD and were ignored. Hereafter, the 51 genes in Table 4 are referred to as iMO–human-AUD genes because (i) their respective orthologs influence behavioral responses to alcohol in iMOs and (ii) they have been implicated in human AUD. As expected, ALK, AUTS2, GPC5, LMO1, and several SWI/SNF orthologs—genes previously described within single reports as modulators of alcohol behavior in iMOs and human AUD—were among the 51 iMO–human-AUD genes. Seven of the iMO–human-AUD genes were derived from studies in both flies and worms, while 23 and 21 were orthologs of genes identified exclusively in fly and worm studies (Table 4).

Table 4.

Invertebrate Model Organism (iMO)–Human-Alcohol Use Disorder (AUD) Genes: Orthologs of Genes that Influence Alcohol-Related Behavior in iMOs and also have been Implicated in Human AUD

Human gene Function Fly ortholog Worm ortholog Human genetics Human phenotype Citations
ADH1A Alcohol dehydrogenase Adh sodh-1 SNP(s) associated (gene cluster) AD Birley et al. (2009), Kuo et al. (2008), Luo et al. (2006), Park et al. (2013), Zuo et al. (2013)
ADH1B Alcohol dehydrogenase Adh sodh-1 SNP(s) associated and suggested associated (gene cluster) AD, alcohol intake Birley et al. (2009), Duell et al. (2012), Kuo et al. (2008), Li et al. (2011), Luo et al. (2006), Park et al. (2013), Zuo et al. (2013)
ADH1C Alcohol dehydrogenase Adh sodh-1 SNP(s) associated (gene cluster) AD Birley et al. (2009), Kuo et al. (2008), Li et al. (2012), Zuo et al. (2013)
ALDH2 Aldehyde dehydrogenase alh-6, alh-13 SNP(s) associated with protection Alcohol drinking, AD Ayhan et al. (2015), Edenberg (2007), Peng et al. (2014), Thomasson et al. (1991)
ALDH1A1 Aldehyde dehydrogenase alh-6, alh-13 SNP(s) associated AD Crawford et al. (2014), Ehlers et al. (2004), Lind et al. (2008)
ALDH1B1 Aldehyde dehydrogenase alh-6, alh-13 SNP associated with protection AD, alcohol induced hypersensitivity Bjerregaard et al. (2014), Linneberg et al. (2010)
ALDH5A1 Aldehyde dehydrogenase alh-6, alh-13 “leading edge gene” SREF, BSA, SHAS Joslyn et al. (2010)
ALK Receptor tyrosine kinase, insulin receptor family dAlk SNP(s) associated Low LR Lasek et al. (2011b)
ARL6IP5 Microtubule binding protein jwa SNP(s) associated AD Edenberg et al. (2010)
ARNTL Helix-loop-helix transcription factor cyc Suggestive SNP(s) associated AC Kovanen et al. (2010)
ARNTL2 Helix-loop-helix transcription factor cyc Suggestive SNP(s) associated AA Kovanen et al. (2010)
AUTS2 Unknown tay SNP(s) associated AC, max drinks Kapoor et al. (2013), Schumann et al. (2011)
BRD7 Component of SWI/SNF chromatin remodeling complex swsn-9 SNP associated AD Mathies et al. (2015)
CHRNA1 Acetylcholine receptor unc-63 “leading edge gene” SREF, BSA, SHAS Joslyn et al. (2010)
CHRNA2 Acetylcholine receptor unc-63 “leading edge gene” SREF, BSA, SHAS Joslyn et al. (2010)
CHRNA3 Acetylcholine receptor unc-63 SNP(s) in the cluster A6 B3 A5 A3 B4 associated “leading edge gene” AD, SREF, BSA, SHAS Choquet et al. (2013), Haller et al. (2014), Hallfors et al. (2013), Joslyn et al. (2010), Schlaepfer et al. (2008)
CHRNA4 Acetylcholine receptor unc-63 “leading edge gene”; SNP(s) associated SREF, BSA, SHAS Alcohol use Ehringer et al. (2007), Joslyn et al. (2010)
CHRNB2 Acetylcholine receptor unc-63 SNP associated Initial response to alcohol Ehringer et al. (2007)
CHRNB4 Acetylcholine receptor unc-63 SNP(s) in cluster A5 A3 B4 associated LR Choquet et al. (2013)
CTBP2 Transcriptional co-repressor ctbp-1 SNP associated AD Lind et al. (2010)
DBH Dopamine beta-hydroxylase Tbh SNP associated AD in women, alcoholism Kohnke et al. (2006), Preuss et al. (2013)
DLG1 Synaptic scaffold dlg1 SNP(s) associated in gene set analysis LR Joslyn et al. (2010)
DLG4 Synaptic scaffold dlg1 SNP(s) associated in gene set analysis LR Joslyn et al. (2010)
DRD1 Dopamine receptor DOP1R1 dop-4 SNP(s) associated AD, AUD problems Batel et al. (2008), Kim et al. (2007), Prasad et al. (2013)
DRD5 Dopamine receptor DOP1R1 SNP associated AD disinhibitory factor score Hack et al. (2011)
GABBR1 Gamma-aminobutyric acid receptor GABA-B-R1 SNP(s) associated in gene set analysis and other AD Kertes et al. (2011), Reimers et al. (2012)
Human gene Function Fly ortholog Worm ortholog Human genetics Human phenotype Citations
GAD1 Glutamate decarboxylase unc-25 SNP(s) associated IS, AD age of onset, AD males, AC Joslyn et al. (2010), Kuo et al. (2009), Loh el et al. (2006), Tabakoff et al. (2009)
GAD2 Glutamate decarboxylase unc-25 SNP(s) associated; “leading edge gene” AD, SREF, BSA, SHAS Joslyn et al. (2010), Lappalainen et al. (2007)
GPC5 Cell-surface heparin sulfate proteoglycan dlp, dally SNP(s) associated Body-sway Joslyn et al. (2010)
GRIN1 Glutamate receptor NMDAR1 SNP(s) associated in gene set analysis and other AD Karpyak et al. (2012), Wernicke et al. (2003)
IGF1R Insulin-like growth factor receptor InR SNP(s) associated in gene set analysis LR Joslyn et al. (2010)
ITGB2 Integrin beta subunit mys SNP(s) associated in gene set analysis LR Joslyn et al. (2010)
KCNMA1 Voltage and calcium-sensitive potassium channel slo slo-1 Suggestive SNP(s) associated AD Kendler et al. (2011)
KCNQ5 Voltage-gated potassium channel KCNQ Suggestive SNP(s) associated AD Kendler et al. (2011)
LMO1 LIM domain transcriptional regulator Bx SNP(s) associated Max drinks Kapoor et al. (2013)
MARK1 Microtubule-associated protein kinase par-1 SNP(s) associated AD comorbid with nicotine dependence Lind et al. (2010)
NALCN Sodium leak channel nca-1, nca-2 SNP associated AD Wetherill et al. (2014)
NCAM1 Immunoglobulin family cell adhesion molecule Fas2 SNP(s) associated AD Yang et al. (2007, 2008)
NPY Neuropeptide Y NPF SNP(s) associated or suggestive associations AD, alcoholism, AW Bhaskar et al. (2013), Ilveskoski et al. (2001), Lappalainen et al. (2002), Mottagui-Tabar et al. (2005), Okubo and Harada (2001)
NPY2R Neuropeptide Y receptor NPFr npr-1 SNP(s) associated AD, AW, other Wetherill et al. (2008)
PER2 Transcriptional repressor per SNP(s) associated AC with sleep problems Comasco et al. (2010)
PIK3R1 Phosphoinositide-3-kinase, regulatory subunit 1 (alpha) Pi3K21B SNP(s) associated AC in males, lifetime prevalence of drunkenness, max drinks Desrivieres et al. (2008)
SLC18A2 Monoamine transporter cat-1 SNP(s) associated AD Fehr et al. (2013), Schwab et al. (2005)
SLC6A2 Norepinephrine transporter DAT SNP(s) associated Alcoholism Clarke et al. (2012)
SLC6A3 Dopamine transporter DAT SNP(s) associated Alcoholism, AC, withdrawal seizures Bhaskar et al. (2012), Du et al. (2011), Lind et al. (2009)
SMARCA2 Component of SWI/SNF chromatin remodeling complex swsn-4 SNP(s) associated AD Mathies et al. (2015)
TH Tyrosine hydroxylase cat-2 SNP associated AD Dahmen et al. (2005)
TPH1 Tryptophan hydroxylase tph-1 SNP(s) associated AD Mokrovic et al. (2008), Sun et al. (2005)
TPH2 Tryptophan hydroxylase tph-1 SNP(s) associated AC Agrawal et al. (2011)
TRPV1 Nonselective cation channel; capsaicin receptor iav osm-9 SNP(s) associated Whole-mouth alcohol intensity Allen et al. (2014)
UNC79 Unknown unc-79 SNP associated AD and Nicotine dependence comorbidity Lind et al. (2010)

Columns are the human gene symbol, a brief description of the function of the gene product, the relevant Drosophila or Caenorhabditis elegans orthologs, the genetic evidence from human studies implicating the gene in some aspect of AUD, the phenotypes (AD, alcohol dependence; SREF, self report of the effects of alcohol; BSA, body sway anterior/posterior; SHAS, subjective high assessment scale; LR, level of response; AC, alcohol consumption; IS, initial sensitivity; AW, alcohol withdrawal) that were investigated in humans, and the citations for the human studies.

Assuming an ideal set of circumstances, a quantitative statistical assessment could be performed to determine whether the 51 iMO–human-AUD gene set is larger than would be expected by chance. For example, given 293 iMO–human genes and 732 human genes in the query sample, and assuming 21,000 total human genes (Harrow et al., 2012), one would expect 293/21,000 × 732 ≈ 10 genes in the iMO–human-AUD set by random chance alone. As the observed iMO–human-AUD set contains 51 genes, this would correspond to an approximately 5-fold overrepresentation. Unfortunately, analyses of this type are not strictly valid for several reasons. First, the sets of 293 iMO–human and 732 human alcoholism-related genes queried for overlap are not independent, which is evidenced by published connections between studies in iMOs and humans for several individual genes (the ADH family, AUTS2, ALK, GPC5, GABBR1, NPY, etc.). Second, not all of the genes appearing in the HuGe Navigator have been associated with alcoholism, alcohol dependence, or other forms of AUD. Specifically, we found that of 83 genes in the HuGe Navigator evaluated as part of this review, 32 had neither a statistically significant nor a nominally significant association with AUD. Thus, based on our experience, approximately 40% of the “alcoholism” genes in the HuGe Navigator are not associated with AUD, leaving the number of genes with a reported connection to AUD in the navigator closer to approximately 450. Finally, genes tested in iMOs, but found to not have connections to alcohol response behaviors, are typically not reported, adding further uncertainty to statistical analysis of the number of genes in Table 4. These intrinsic features of the published data make a formal statistical analysis of the overlap impossible.

A small number of the 51 iMO–human-AUD genes (Table 4) have well-established roles in 1 or more features of AUD. Human ADH1A, ADH1B, ADH1C, ALDH2, ALD-DH18A1, and ALDH4A1, genes that encode key alcohol-metabolizing enzymes, are associated with alcohol dependence, alcohol intake, and other related phenotypes in numerous human studies (Table 4). The human genes CHRNA3, DRD1, GAD1, NPY, and SLC18A2 have also been implicated in AUD by multiple studies in humans (Table 4). Given that these human genes are the most or among the most widely accepted for having roles in AUD, it is noteworthy that the invertebrate orthologs of all of these genes influence behavioral responses to acute alcohol exposure (Tables 1 and 2). The remaining 40 iMO–human-AUD genes have been implicated in human AUD by smaller scale or single studies (Table 4). Although it is not clear at this time whether these 40 genes have bona fide roles in human AUD given the lack of replication of the associations in independent populations, the results from studies on the invertebrate orthologs of these genes suggest that additional human studies are warranted.

A major potential limitation to the approach used to identify the 51 iMO–human-AUD genes (Table 4) is that it is based on overlap at the level of individual orthologs of iMO and human genes. This approach almost certainly would miss key signaling or biochemical pathways in which, for example, a gene encoding a ligand was investigated in iMOs and the gene encoding the orthologous receptor for that ligand was found to be associated with human AUD. In such a case, the iMO data would provide strong evidence for a role of the biochemical process in alcohol-related behavior relevant to human AUD, even in the absence of directly implicating the particular orthologous human gene. Thus, as a complement to our analysis of the overlap between individual iMO and human genes, we visually compared the predicted or known biochemical functions of the iMO genes (Tables 1 and 2) with the functions of genes described in several comprehensive reviews on the genetics of AUD (Edenberg and Foroud, 2013, 2014; Palmer et al., 2012; Rietschel and Treutlein, 2013; Schuckit, 2014). As is true for individual genes, the most compelling evidence for a pathway important in iMOs and humans is for the alcohol-metabolizing machinery of ADH and ALDH. In addition, several other cellular processes also have strong support in iMOs and humans including signaling via dopamine, NPY, growth factors, and potassium channels (Tables 1 and 2). Thus, the 51 iMO–human-AUD genes in Table 4 probably under represent the functional overlap of findings from alcohol studies in iMOs and humans.

SUMMARY AND PERSPECTIVES

The large number of genes that influence alcohol-related behaviors identified in iMOs (Tables 1 and 2) demonstrates the analytical power of flies and worms for investigating molecular–genetic mechanisms underlying nervous system responses to alcohol. This analysis provides strong support for the use of iMOs as key experimental platforms for identifying and subsequently investigating novel genes that modulate alcohol-related behavior. Importantly, our analysis also demonstrates that studies in both flies and worms have independently contributed to our understanding of genetic mechanisms that influence behavioral responses to alcohol. Several major open questions remain regarding the function of many iMO genes identified to date: (i) Which of the genes influence developmental versus adult physiological processes relevant to alcohol-related behavior? (ii) Are there interactions between the gene-driven developmental and adult physiological processes that influence alcohol-related behavior? (iii) Do subsets of genes act in an integrated fashion to influence behavioral responses to alcohol? (iv) Do the genes alter alcohol-related behavioral responses via a common set of neuronal or other cellular mechanisms? (v) What are the major areas of the nervous system in which the genes function and what neurotransmitter systems are modulated by the genes? (vi) Do the genes influence alcohol-related behavior by functioning in non-neuronal cells? (vii) Do the genes influence multiple behavioral responses to alcohol similarly? (viii) Are the gene products direct pharmacological targets of alcohol? (ix) What is the complete complement of genes that is required for normal behavioral responses to alcohol? Flies and worms have contributed substantially to the field, yet much work remains to be done in iMOs on the genetics of alcohol-related behavior.

The numerous orthologs of genes that influence both human AUD and alcohol-related behaviors in iMOs support the concept of conservation of gene function in alcohol responses in humans and iMOs. Thus, additional genetic information gleaned from the fly and worm models should have translational utility for understanding AUD in humans. Given the intrinsic and unavoidable differences between studies in iMOs and humans (see Genetics of Alcohol Behavior in Invertebrate Model Organisms and Human AUD), it is reasonable to expect that not all alcohol behavior genes identified in iMOs will be associated with human AUD. Nevertheless, orthologs of alcohol behavior genes in iMOs (Table 3) might be prime candidates for targeted investigations in studies on human AUD. Conversely, additional orthologs of genes identified in studies on human AUD could be investigated in iMOs to better understand (i) the fundamental alcohol behavioral consequences of altered gene function and (ii) the genetic pathways and gene networks that function in concert with the originally identified human AUD gene. An understanding of these 2 aspects of gene function in iMOs might ultimately provide a more comprehensive appreciation of molecular and cellular mechanisms underlying human AUD. Consequently, the integration of information from behavioral–genetic studies on alcohol in iMOs with genetic findings from humans has the potential to lead to a much deeper understanding of AUD, its diagnosis, and its treatment.

Acknowledgments

This work was supported in part by grants from the National Institute on Alcohol Abuse and Alcoholism (MG and JCB, P20AA017828 and P50AA022357; MG, R01A A020634; JCB, R01AA016837). The authors thank their fellow members of the Virginia Commonwealth University Alcohol Research Center (Ken Kendler, Mike Miles, Todd Webb, and Andrew Davies) and Laura Mathies for helpful discussions and comments on the manuscript. The authors also thank Lara Lewellyn for administrative assistance.

Footnotes

The authors declare that they have no conflicts of interest.

References

  1. Agrawal A, Lynskey MT, Todorov AA, Schrage AJ, Littlefield AK, Grant JD, Zhu Q, Nelson EC, Madden PA, Bucholz KK, Sher KJ, Heath AC. A candidate gene association study of alcohol consumption in young women. Alcohol Clin Exp Res. 2011;35:550–558. doi: 10.1111/j.1530-0277.2010.01372.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alaimo JT, Davis SJ, Song SS, Burnette CR, Grotewiel M, Shelton KL, Pierce-Shimomura JT, Davies AG, Bettinger JC. Ethanol metabolism and osmolarity modify behavioral responses to ethanol in C. elegans. Alcohol Clin Exp Res. 2012;36:1840–1850. doi: 10.1111/j.1530-0277.2012.01799.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allen AL, McGeary JE, Hayes JE. Polymorphisms in TRPV1 and TAS2Rs associate with sensations from sampled ethanol. Alcohol Clin Exp Res. 2014;38:2550–2560. doi: 10.1111/acer.12527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 1997;25:3389–3402. doi: 10.1093/nar/25.17.3389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5. American Psychiatric Publishing; Washington, DC: 2013. [Google Scholar]
  6. Awofala AA, Jones S, Davies JA. The heat shock protein 26 gene is required for ethanol tolerance in Drosophila. J Exp Neurosci. 2011;5:31–44. [Google Scholar]
  7. Ayhan Y, Gurel SC, Karaca O, Zoto T, Hayran M, Babaoglu M, Yasar U, Bozkurt A, Dilbaz N, Ulug BD, Demir B. Association between ADH1C and ALDH2 polymorphisms and alcoholism in a Turkish sample. Nord J Psychiatry. 2015;69:233–239. doi: 10.3109/08039488.2014.972450. [DOI] [PubMed] [Google Scholar]
  8. Bainton RJ, Tsai LT, Schwabe T, DeSalvo M, Gaul U, Heberlein U. Moody encodes two GPCRs that regulate cocaine behaviors and blood-brain barrier permeability in Drosophila. Cell. 2005;123:145–156. doi: 10.1016/j.cell.2005.07.029. [DOI] [PubMed] [Google Scholar]
  9. Bainton RJ, Tsai LT, Singh CM, Moore MS, Neckameyer WS, Heberlein U. Dopamine modulates acute responses to cocaine, nicotine and ethanol in Drosophila. Curr Biol. 2000;10:187–194. doi: 10.1016/s0960-9822(00)00336-5. [DOI] [PubMed] [Google Scholar]
  10. Batel P, Houchi H, Daoust M, Ramoz N, Naassila M, Gorwood P. A haplotype of the DRD1 gene is associated with alcohol dependence. Alcohol Clin Exp Res. 2008;32:567–572. doi: 10.1111/j.1530-0277.2008.00618.x. [DOI] [PubMed] [Google Scholar]
  11. Berger KH, Heberlein U, Moore MS. Rapid and chronic: two distinct forms of ethanol tolerance in Drosophila. Alcohol Clin Exp Res. 2004;28:1469–1480. doi: 10.1097/01.alc.0000141817.15993.98. [DOI] [PubMed] [Google Scholar]
  12. Bettinger JC, Leung K, Bolling MH, Goldsmith AD, Davies AG. Lipid environment modulates the development of acute tolerance to ethanol in Caenorhabditis elegans. PLoS One. 2012;7:e35192. doi: 10.1371/journal.pone.0035192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bettinger JC, McIntire SL. State-dependency in C. elegans. Genes Brain Behav. 2004;3:266–272. doi: 10.1111/j.1601-183X.2004.00080.x. [DOI] [PubMed] [Google Scholar]
  14. Bhandari P, Hill JS, Farris SP, Costin B, Martin I, Chan CL, Alaimo JT, Bettinger JC, Davies AG, Miles MF, Grotewiel M. Chloride intracellular channels modulate acute ethanol behaviors in Drosophila, Caenorhabditis elegans and mice. Genes Brain Behav. 2012;11:387–397. doi: 10.1111/j.1601-183X.2012.00765.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bhandari P, Kendler KS, Bettinger JC, Davies AG, Grotewiel M. An assay for evoked locomotor behavior in Drosophila reveals a role for integrins in ethanol sensitivity and rapid ethanol tolerance. Alcohol Clin Exp Res. 2009;33:1794–1805. doi: 10.1111/j.1530-0277.2009.01018.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bhaskar LV, Thangaraj K, Kumar KP, Pardhasaradhi G, Singh L, Rao VR. Association between neuropeptide Y gene polymorphisms and alcohol dependence: a case-control study in two independent populations. Eur Addict Res. 2013;19:307–313. doi: 10.1159/000346679. [DOI] [PubMed] [Google Scholar]
  17. Bhaskar LV, Thangaraj K, Wasnik S, Singh L, Raghavendra Rao V. Dopamine transporter (DAT1) VNTR polymorphism and alcoholism in two culturally different populations of south India. Am J Addict. 2012;21:343–347. doi: 10.1111/j.1521-0391.2012.00244.x. [DOI] [PubMed] [Google Scholar]
  18. Birley AJ, James MR, Dickson PA, Montgomery GW, Heath AC, Martin NG, Whitfield JB. ADH single nucleotide polymorphism associations with alcohol metabolism in vivo. Hum Mol Genet. 2009;18:1533–1542. doi: 10.1093/hmg/ddp060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bjerregaard P, Mikkelsen SS, Becker U, Hansen T, Tolstrup JS. Genetic variation in alcohol metabolizing enzymes among Inuit and its relation to drinking patterns. Drug Alcohol Depend. 2014;144:239–244. doi: 10.1016/j.drugalcdep.2014.09.774. [DOI] [PubMed] [Google Scholar]
  20. Cavaliere S, Gillespie JM, Hodge JJ. KCNQ channels show conserved ethanol block and function in ethanol behaviour. PLoS One. 2012;7:e50279. doi: 10.1371/journal.pone.0050279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cavener D. Preference for ethanol in Drosophila melanogaster associated with the alcohol dehydrogenase polymorphism. Behav Genet. 1979;9:359–365. doi: 10.1007/BF01066974. [DOI] [PubMed] [Google Scholar]
  22. Chan RF, Lewellyn L, DeLoyht JM, Sennett K, Coffman S, Hewitt M, Bettinger JC, Warrick JM, Grotewiel M. Contrasting influences of Drosophila white/mini-white on ethanol sensitivity in two different behavioral assays. Alcohol Clin Exp Res. 2014;38:1582–1593. doi: 10.1111/acer.12421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chen J, Wang Y, Zhang Y, Shen P. Mutations in Bacchus reveal a tyramine-dependent nuclear regulator for acute ethanol sensitivity in Drosophila. Neuropharmacology. 2013;67:25–31. doi: 10.1016/j.neuropharm.2012.10.013. [DOI] [PubMed] [Google Scholar]
  24. Chen J, Zhang Y, Shen P. A protein kinase C activity localized to neuropeptide Y-like neurons mediates ethanol intoxication in Drosophila melanogaster. Neuroscience. 2008;156:42–47. doi: 10.1016/j.neuroscience.2008.07.008. [DOI] [PubMed] [Google Scholar]
  25. Chen J, Zhang Y, Shen P. Protein kinase C deficiency-induced alcohol insensitivity and underlying cellular targets in Drosophila. Neuroscience. 2010;166:34–39. doi: 10.1016/j.neuroscience.2009.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cheng Y, Endo K, Wu K, Rodan AR, Heberlein U, Davis RL. Drosophila fasciclin II is required for the formation of odor memories and for normal sensitivity to alcohol. Cell. 2001;105:757–768. doi: 10.1016/s0092-8674(01)00386-5. [DOI] [PubMed] [Google Scholar]
  27. Choquet H, Joslyn G, Lee A, Kasberger J, Robertson M, Brush G, Schuckit MA, White R, Jorgenson E. Examination of rare missense variants in the CHRNA5-A3-B4 gene cluster to level of response to alcohol in the San Diego Sibling Pair study. Alcohol Clin Exp Res. 2013;37:1311–1316. doi: 10.1111/acer.12099. [DOI] [PubMed] [Google Scholar]
  28. Clarke TK, Dempster E, Docherty SJ, Desrivieres S, Lourdsamy A, Wodarz N, Ridinger M, Maier W, Rietschel M, Schumann G. Multiple polymorphisms in genes of the adrenergic stress system confer vulnerability to alcohol abuse. Addict Biol. 2012;17:202–208. doi: 10.1111/j.1369-1600.2010.00263.x. [DOI] [PubMed] [Google Scholar]
  29. Comasco E, Nordquist N, Gokturk C, Aslund C, Hallman J, Oreland L, Nilsson KW. The clock gene PER2 and sleep problems: association with alcohol consumption among Swedish adolescents. Ups J Med Sci. 2010;115:41–48. doi: 10.3109/03009731003597127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Corl AB, Berger KH, Ophir-Shohat G, Gesch J, Simms JA, Bartlett SE, Heberlein U. Happyhour, a Ste20 family kinase, implicates EGFR signaling in ethanol-induced behaviors. Cell. 2009;137:949–960. doi: 10.1016/j.cell.2009.03.020. [DOI] [PubMed] [Google Scholar]
  31. Corl AB, Rodan AR, Heberlein U. Insulin signaling in the nervous system regulates ethanol intoxication in Drosophila melanogaster. Nat Neurosci. 2005;8:18–19. doi: 10.1038/nn1363. [DOI] [PubMed] [Google Scholar]
  32. Cowmeadow RB, Krishnan HR, Atkinson NS. The slowpoke gene is necessary for rapid ethanol tolerance in Drosophila. Alcohol Clin Exp Res. 2005;29:1777–1786. doi: 10.1097/01.alc.0000183232.56788.62. [DOI] [PubMed] [Google Scholar]
  33. Cowmeadow RB, Krishnan HR, Ghezzi A, Al’Hasan YM, Wang YZ, Atkinson NS. Ethanol tolerance caused by slowpoke induction in Drosophila. Alcohol Clin Exp Res. 2006;30:745–753. doi: 10.1111/j.1530-0277.2006.00087.x. [DOI] [PubMed] [Google Scholar]
  34. Crawford A, Dalvie S, Lewis S, King A, Liberzon I, Fein G, Koenen K, Ramesar R, Stein DJ. Haplotype-based study of the association of alcohol and acetaldehyde-metabolising genes with alcohol dependence (with or without comorbid anxiety symptoms) in a Cape Mixed Ancestry population. Metab Brain Dis. 2014;29:333–340. doi: 10.1007/s11011-014-9503-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dahmen N, Volp M, Singer P, Hiemke C, Szegedi A. Tyrosine hydroxylase Val-81-Met polymorphism associated with early-onset alcoholism. Psychiatr Genet. 2005;15:13–16. doi: 10.1097/00041444-200503000-00003. [DOI] [PubMed] [Google Scholar]
  36. Das J, Xu S, Pany S, Guillory A, Shah V, Roman GW. The pre-synaptic Munc13-1 binds alcohol and modulates alcohol self-administration in Drosophila. J Neurochem. 2013;126:715–726. doi: 10.1111/jnc.12315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Davies AG, Bettinger JC. The role of the BK channel in ethanol response behaviors: evidence from model organism and human studies. Front Physiol. 2014;5:1–9. doi: 10.3389/fphys.2014.00346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Davies AG, Bettinger JC, Thiele TR, Judy ME, McIntire SL. Natural variation in the npr-1 gene modifies ethanol responses of wild strains of C. elegans. Neuron. 2004;42:731–743. doi: 10.1016/j.neuron.2004.05.004. [DOI] [PubMed] [Google Scholar]
  39. Davies AG, Pierce-Shimomura JT, Kim H, VanHoven MK, Thiele TR, Bonci A, Bargmann CI, McIntire SL. A central role of the BK potassium channel in behavioral responses to ethanol in C. elegans. Cell. 2003;115:655–666. doi: 10.1016/s0092-8674(03)00979-6. [DOI] [PubMed] [Google Scholar]
  40. Desrivieres S, Krause K, Dyer A, Frank J, Blomeyer D, Lathrop M, Mann K, Banaschewski T, Laucht M, Schumann G. Nucleotide sequence variation within the PI3K p85 alpha gene associates with alcohol risk drinking behaviour in adolescents. PLoS One. 2008;3:e1769. doi: 10.1371/journal.pone.0001769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Devineni AV, Eddison M, Heberlein U. The novel gene tank, a tumor suppressor homolog, regulates ethanol sensitivity in Drosophila. J Neurosci. 2013;33:8134–8143. doi: 10.1523/JNEUROSCI.3695-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Devineni AV, Heberlein U. Preferential ethanol consumption in Drosophila models features of addiction. Curr Biol. 2009;19:2126–2132. doi: 10.1016/j.cub.2009.10.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Devineni AV, Heberlein U. Acute ethanol responses in Drosophila are sexually dimorphic. Proc Natl Acad Sci USA. 2012;109:21087–21092. doi: 10.1073/pnas.1218850110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Devineni AV, Heberlein U. The evolution of Drosophila melanogaster as a model for alcohol research. Annu Rev Neurosci. 2013;36:121–138. doi: 10.1146/annurev-neuro-062012-170256. [DOI] [PubMed] [Google Scholar]
  45. Dietrich O, Heun M, Notroff J, Schmidt K, Zarnkow M. The role of cult and feasting in the emergence of Neolithic communities. New evidence from Gobekli Tepe, south-eastern Turkey. Antiquity. 2012;86:674–695. [Google Scholar]
  46. Dillon J, Andrianakis I, Mould R, Ient B, Liu W, James C, O’Connor V, Holden-Dye L. Distinct molecular targets including SLO-1 and gap junctions are engaged across a continuum of ethanol concentrations in Caenorhabditis elegans. FASEB J. 2013;27:4266–4278. doi: 10.1096/fj.11-189340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Du Y, Nie Y, Li Y, Wan YJ. The association between the SLC6A3 VNTR 9-repeat allele and alcoholism-a meta-analysis. Alcohol Clin Exp Res. 2011;35:1625–1634. doi: 10.1111/j.1530-0277.2011.01509.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Duell EJ, Sala N, Travier N, Munoz X, Boutron-Ruault MC, Clavel-Chapelon F, Barricarte A, Arriola L, Navarro C, Sanchez-Cantalejo E, Quiros JR, Krogh V, Vineis P, Mattiello A, Tumino R, Khaw KT, Wareham N, Allen NE, Peeters PH, Numans ME, Bueno-de-Mesquita HB, van Oijen MG, Bamia C, Benetou V, Trichopoulos D, Canzian F, Kaaks R, Boeing H, Bergmann MM, Lund E, Ehrnstrom R, Johansen D, Hallmans G, Stenling R, Tjonneland A, Overvad K, Ostergaard JN, Ferrari P, Fedirko V, Jenab M, Nesi G, Riboli E, Gonzalez CA. Genetic variation in alcohol dehydrogenase (ADH1A, ADH1B, ADH1C, ADH7) and aldehyde dehydrogenase (ALDH2), alcohol consumption and gastric cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Carcinogenesis. 2012;33:361–367. doi: 10.1093/carcin/bgr285. [DOI] [PubMed] [Google Scholar]
  49. Dzitoyeva S, Dimitrijevic N, Manev H. Gamma-aminobutyric acid B receptor 1 mediates behavior-impairing actions of alcohol in Drosophila: adult RNA interference and pharmacological evidence. Proc Natl Acad Sci USA. 2003;100:5485–5490. doi: 10.1073/pnas.0830111100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Eddison M, Guarnieri DJ, Cheng L, Liu CH, Moffat KG, Davis G, Heberlein U. Arouser reveals a role for synapse number in the regulation of ethanol sensitivity. Neuron. 2011;70:979–990. doi: 10.1016/j.neuron.2011.03.030. [DOI] [PubMed] [Google Scholar]
  51. Edenberg HJ. The genetics of alcohol metabolism: role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health. 2007;30:5–13. [PMC free article] [PubMed] [Google Scholar]
  52. Edenberg HJ, Foroud T. Genetics and alcoholism. Nat Rev Gastroenterol Hepatol. 2013;10:487–494. doi: 10.1038/nrgastro.2013.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Edenberg HJ, Foroud T. Genetics of alcoholism. Handb Clin Neurol. 2014;125:561–571. doi: 10.1016/B978-0-444-62619-6.00032-X. [DOI] [PubMed] [Google Scholar]
  54. 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, Jr, 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. doi: 10.1111/j.1530-0277.2010.01156.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Ehlers CL, Spence JP, Wall TL, Gilder DA, Carr LG. Association of ALDH1 promoter polymorphisms with alcohol-related phenotypes in southwest California Indians. Alcohol Clin Exp Res. 2004;28:1481–1486. doi: 10.1097/01.alc.0000141821.06062.20. [DOI] [PubMed] [Google Scholar]
  56. Ehringer MA, Clegg HV, Collins AC, Corley RP, Crowley T, Hewitt JK, Hopfer CJ, Krauter K, Lessem J, Rhee SH, Schlaepfer I, Smolen A, Stallings MC, Young SE, Zeiger JS. Association of the neuronal nicotinic receptor beta2 subunit gene (CHRNB2) with subjective responses to alcohol and nicotine. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:596–604. doi: 10.1002/ajmg.b.30464. [DOI] [PubMed] [Google Scholar]
  57. Fehr C, Sommerlad D, Sander T, Anghelescu I, Dahmen N, Szegedi A, Mueller C, Zill P, Soyka M, Preuss UW. Association of VMAT2 gene polymorphisms with alcohol dependence. J Neural Transm. 2013;120:1161–1169. doi: 10.1007/s00702-013-0996-y. [DOI] [PubMed] [Google Scholar]
  58. Ghezzi A, Krishnan HR, Atkinson NS. Susceptibility to ethanol withdrawal seizures is produced by BK channel gene expression. Addict Biol. 2012;19:332–337. doi: 10.1111/j.1369-1600.2012.00465.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Godenschwege TA, Reisch D, Diegelmann S, Eberle K, Funk N, Heisenberg M, Hoppe V, Hoppe J, Klagges BR, Martin JR, Nikitina EA, Putz G, Reifegerste R, Reisch N, Rister J, Schaupp M, Scholz H, Schwarzel M, Werner U, Zars TD, Buchner S, Buchner E. Flies lacking all synapsins are unexpectedly healthy but are impaired in complex behaviour. Eur J Neurosci. 2004;20:611–622. doi: 10.1111/j.1460-9568.2004.03527.x. [DOI] [PubMed] [Google Scholar]
  60. Graham ME, Edwards MR, Holden-Dye L, Morgan A, Burgoyne RD, Barclay JW. UNC-18 modulates ethanol sensitivity in Caenorhabditis elegans. Mol Biol Cell. 2009;20:43–55. doi: 10.1091/mbc.E08-07-0689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Grell EH, Jacobson KB, Murphy JB. Alterations of genetic material for analysis of alcohol dehydrogenase isozymes of Drosophila melanogaster. Ann N Y Acad Sci. 1968;151:441–455. doi: 10.1111/j.1749-6632.1968.tb11907.x. [DOI] [PubMed] [Google Scholar]
  62. Hack LM, Kalsi G, Aliev F, Kuo PH, Prescott CA, Patterson DG, Walsh D, Dick DM, Riley BP, Kendler KS. Limited associations of dopamine system genes with alcohol dependence and related traits in the Irish Affected Sib Pair Study of Alcohol Dependence (IASPSAD) Alcohol Clin Exp Res. 2011;35:376–385. doi: 10.1111/j.1530-0277.2010.01353.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Haeny AM, Littlefield AK, Sher KJ. Repeated diagnoses of lifetime alcohol use disorders in a prospective study: insights into the extent and nature of the reliability and validity problem. Alcohol Clin Exp Res. 2014;38:489–500. doi: 10.1111/acer.12237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Haller G, Kapoor M, Budde J, Xuei X, Edenberg H, Nurnberger J, Kramer J, Brooks A, Tischfield J, Almasy L, Agrawal A, Bucholz K, Rice J, Saccone N, Bierut L, Goate A. Rare missense variants in CHRNB3 and CHRNA3 are associated with risk of alcohol and cocaine dependence. Hum Mol Genet. 2014;23:810–819. doi: 10.1093/hmg/ddt463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hallfors J, Loukola A, Pitkaniemi J, Broms U, Mannisto S, Salomaa V, Heliovaara M, Lehtimaki T, Raitakari O, Madden PA, Heath AC, Montgomery GW, Martin NG, Korhonen T, Kaprio J. Scrutiny of the CHRNA5-CHRNA3-CHRNB4 smoking behavior locus reveals a novel association with alcohol use in a Finnish population based study. Int JMol Epidemiol Genet. 2013;4:109–119. [PMC free article] [PubMed] [Google Scholar]
  66. Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, Barnes I, Bignell A, Boychenko V, Hunt T, Kay M, Mukherjee G, Rajan J, Despacio-Reyes G, Saunders G, Steward C, Harte R, Lin M, Howald C, Tanzer A, Derrien T, Chrast J, Walters N, Balasubramanian S, Pei B, Tress M, Rodriguez JM, Ezkurdia I, van Baren J, Brent M, Haussler D, Kellis M, Valencia A, Reymond A, Gerstein M, Guigo R, Hubbard TJ. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 2012;22:1760–1774. doi: 10.1101/gr.135350.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hawkins E, Martin I, Kondo L, Judy M, Brings V, Chan C, Blackwell G, Bettinger J, Davies A. A novel cholinergic action of alcohol and the development of tolerance to that effect in Caenorhabditis elegans. Genetics. 2015;199:135–149. doi: 10.1534/genetics.114.171884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Heberlein U, Wolf FW, Rothenfluh A, Guarnieri DJ. Molecular genetic analysis of ethanol intoxication in Drosophila melanogaster. Integr Comp Biol. 2004;44:269–274. doi: 10.1093/icb/44.4.269. [DOI] [PubMed] [Google Scholar]
  69. Hu Y, Flockhart I, Vinayagam A, Bergwitz C, Berger B, Perrimon N, Mohr SE. An integrative approach to ortholog prediction for disease-focused and other functional studies. BMC Bioinformatics. 2011;12:357. doi: 10.1186/1471-2105-12-357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ilveskoski E, Kajander OA, Lehtimaki T, Kunnas T, Karhunen PJ, Heinala P, Virkkunen M, Alho H. Association of neuropeptide y polymorphism with the occurrence of type 1 and type 2 alcoholism. Alcohol Clin Exp Res. 2001;25:1420–1422. doi: 10.1097/00000374-200110000-00003. [DOI] [PubMed] [Google Scholar]
  71. Ja WW, Carvalho GB, Mak EM, de la Rosa NN, Fang AY, Liong JC, Brummel T, Benzer S. Prandiology of Drosophila and the CAFE assay. Proc Natl Acad Sci USA. 2007;104:8253–8256. doi: 10.1073/pnas.0702726104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jee C, Lee J, Lim JP, Parry D, Messing RO, McIntire SL. SEB-3, a CRF receptor-like GPCR, regulates locomotor activity states, stress responses and ethanol tolerance in Caenorhabditis elegans. Genes Brain Behav. 2013;12:250–262. doi: 10.1111/j.1601-183X.2012.00829.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. 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. doi: 10.1111/j.1530-0277.2010.01152.x. [DOI] [PubMed] [Google Scholar]
  74. Joslyn G, Wolf FW, Brush G, Wu L, Schuckit M, White RL. Glypican gene GPC5 participates in the behavioral response to ethanol: evidence from humans, mice, and fruit flies. G3 (Bethesda) 2011;1:627–635. doi: 10.1534/g3.111.000976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Kaletta T, Hengartner MO. Finding function in novel targets: C. elegans as a model organism. Nat Rev Drug Discov. 2006;5:387–398. doi: 10.1038/nrd2031. [DOI] [PubMed] [Google Scholar]
  76. Kapfhamer D, Bettinger JC, Davies AG, Eastman CL, Smail EA, Heberlein U, McIntire SL. Loss of RAB-3/A in Caenorhabditis elegans and the mouse affects behavioral response to ethanol. Genes Brain Behav. 2008;7:669–676. doi: 10.1111/j.1601-183X.2008.00404.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kapfhamer D, King I, Zou ME, Lim JP, Heberlein U, Wolf FW. JNK pathway activation is controlled by Tao/TAOK3 to modulate ethanol sensitivity. PLoS One. 2012;7:e50594. doi: 10.1371/journal.pone.0050594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. 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. doi: 10.1007/s00439-013-1318-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Karpyak VM, Geske JR, Colby CL, Mrazek DA, Biernacka JM. Genetic variability in the NMDA-dependent AMPA trafficking cascade is associated with alcohol dependence. Addict Biol. 2012;17:798–806. doi: 10.1111/j.1369-1600.2011.00338.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kaun KR, Azanchi R, Maung Z, Hirsh J, Heberlein U. A Drosophila model for alcohol reward. Nat Neurosci. 2011;14:612–619. doi: 10.1038/nn.2805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Kaun KR, Devineni AV, Heberlein U. Drosophila melanogaster as a model to study drug addiction. HumGenet. 2012;131:959–975. doi: 10.1007/s00439-012-1146-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. 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. doi: 10.1111/j.1530-0277.2010.01427.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kertes DA, Kalsi G, Prescott CA, Kuo PH, Patterson DG, Walsh D, Kendler KS, Riley BP. Neurotransmitter and neuromodulator genes associated with a history of depressive symptoms in individuals with alcohol dependence. Alcohol Clin Exp Res. 2011;35:496–505. doi: 10.1111/j.1530-0277.2010.01366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Kim DJ, Park BL, Yoon S, Lee HK, Joe KH, Cheon YH, Gwon DH, Cho SN, Lee HW, NamGung S, Shin HD. 5′ UTR polymorphism of dopamine receptor D1 (DRD1) associated with severity and temperament of alcoholism. Biochem Biophys Res Commun. 2007;357:1135–1141. doi: 10.1016/j.bbrc.2007.04.074. [DOI] [PubMed] [Google Scholar]
  85. King I, Tsai LT, Pflanz R, Voigt A, Lee S, Jackle H, Lu B, Heberlein U. Drosophila tao controls mushroom body development and ethanol-stimulated behavior through par-1. J Neurosci. 2011;31:1139–1148. doi: 10.1523/JNEUROSCI.4416-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. King IF, Eddison M, Kaun KR, Heberlein U. EGFR and FGFR pathways have distinct roles in Drosophila mushroom body development and ethanol-induced behavior. PLoS One. 2014;9:e87714. doi: 10.1371/journal.pone.0087714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Kohnke MD, Kolb W, Kohnke AM, Lutz U, Schick S, Batra A. DBH*444G/A polymorphism of the dopamine-beta-hydroxylase gene is associated with alcoholism but not with severe alcohol withdrawal symptoms. J Neural Transm. 2006;113:869–876. doi: 10.1007/s00702-005-0365-6. [DOI] [PubMed] [Google Scholar]
  88. Kong EC, Allouche L, Chapot PA, Vranizan K, Moore MS, Heberlein U, Wolf FW. Ethanol-regulated genes that contribute to ethanol sensitivity and rapid tolerance in Drosophila. Alcohol Clin Exp Res. 2010a;34:302–316. doi: 10.1111/j.1530-0277.2009.01093.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Kong EC, Woo K, Li H, Lebestky T, Mayer N, Sniffen MR, Heberlein U, Bainton RJ, Hirsh J, Wolf FW. A pair of dopamine neurons target the D1-like dopamine receptor DopR in the central complex to promote ethanol-stimulated locomotion in Drosophila. PLoS One. 2010b;5:e9954. doi: 10.1371/journal.pone.0009954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Kovanen L, Saarikoski ST, Haukka J, Pirkola S, Aromaa A, Lonnqvist J, Partonen T. Circadian clock gene polymorphisms in alcohol use disorders and alcohol consumption. Alcohol Alcohol. 2010;45:303–311. doi: 10.1093/alcalc/agq035. [DOI] [PubMed] [Google Scholar]
  91. Krishnan HR, Al-Hasan YM, Pohl JB, Ghezzi A, Atkinson NS. A role for dynamin in triggering ethanol tolerance. Alcohol Clin Exp Res. 2012;36:24–34. doi: 10.1111/j.1530-0277.2011.01587.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kriventseva EV, Tegenfeldt F, Petty TJ, Waterhouse RM, Simao FA, Pozdnyakov IA, Ioannidis P, Zdobnov EM. OrthoDB v8: update of the hierarchical catalog of orthologs and the underlying free software. Nucleic Acids Res. 2015;43(Database issue):D250–D256. doi: 10.1093/nar/gku1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kuo PH, Kalsi G, Prescott CA, Hodgkinson CA, Goldman D, Alexander J, van den Oord EJ, Chen X, Sullivan PF, Patterson DG, Walsh D, Kendler KS, Riley BP. Associations of glutamate decarboxylase genes with initial sensitivity and age-at-onset of alcohol dependence in the Irish Affected Sib Pair Study of Alcohol Dependence. Drug Alcohol Depend. 2009;101:80–87. doi: 10.1016/j.drugalcdep.2008.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Kuo PH, Kalsi G, Prescott CA, Hodgkinson CA, Goldman D, van den Oord EJ, Alexander J, Jiang C, Sullivan PF, Patterson DG, Walsh D, Kendler KS, Riley BP. Association of ADH and ALDH genes with alcohol dependence in the Irish Affected Sib Pair Study of alcohol dependence (IASPSAD) sample. Alcohol Clin Exp Res. 2008;32:785–795. doi: 10.1111/j.1530-0277.2008.00642.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. LaFerriere H, Guarnieri DJ, Sitaraman D, Diegelmann S, Heberlein U, Zars T. Genetic dissociation of ethanol sensitivity and memory formation in Drosophila melanogaster. Genetics. 2008;178:1895–1902. doi: 10.1534/genetics.107.084582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. 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. doi: 10.1001/archpsyc.59.9.825. [DOI] [PubMed] [Google Scholar]
  97. Lappalainen J, Krupitsky E, Kranzler HR, Luo X, Remizov M, Pchelina S, Taraskina A, Zvartau E, Rasanen P, Makikyro T, Somberg LK, Krystal JH, Stein MB, Gelernter J. Mutation screen of the GAD2 gene and association study of alcoholism in three populations. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:183–192. doi: 10.1002/ajmg.b.30377. [DOI] [PubMed] [Google Scholar]
  98. Lasek AW, Giorgetti F, Berger KH, Tayor S, Heberlein U. Lmo genes regulate behavioral responses to ethanol in Drosophila melanogaster and the mouse. Alcohol Clin Exp Res. 2011a;35:1600–1606. doi: 10.1111/j.1530-0277.2011.01506.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Lasek AW, Lim J, Kliethermes CL, Berger KH, Joslyn G, Brush G, Xue L, Robertson M, Moore MS, Vranizan K, Morris SW, Schuckit MA, White RL, Heberlein U. An evolutionary conserved role for anaplastic lymphoma kinase in behavioral responses to ethanol. PLoS One. 2011b;6:e22636. doi: 10.1371/journal.pone.0022636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lee J, Jee C, McIntire SL. Ethanol preference in C. elegans. Genes Brain Behav. 2009;8:578–585. doi: 10.1111/j.1601-183X.2009.00513.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Li C, Zhao X, Cao X, Chu D, Chen J, Zhou J. The Drosophila homolog of jwa is required for ethanol tolerance. Alcohol Alcohol. 2008;43:529–536. doi: 10.1093/alcalc/agn045. [DOI] [PubMed] [Google Scholar]
  102. Li D, Zhao H, Gelernter J. Strong association of the alcohol dehydrogenase 1B gene (ADH1B) with alcohol dependence and alcohol-induced medical diseases. Biol Psychiatry. 2011;70:504–512. doi: 10.1016/j.biopsych.2011.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Li D, Zhao H, Gelernter J. Further clarification of the contribution of the ADH1C gene to vulnerability of alcoholism and selected liver diseases. Hum Genet. 2012;131:1361–1374. doi: 10.1007/s00439-012-1163-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Lind PA, Eriksson CJ, Wilhelmsen KC. The role of aldehyde dehydrogenase-1 (ALDH1A1) polymorphisms in harmful alcohol consumption in a Finnish population. Hum Genomics. 2008;3:24–35. doi: 10.1186/1479-7364-3-1-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Lind PA, Eriksson CJ, Wilhelmsen KC. Association between harmful alcohol consumption behavior and dopamine transporter (DAT1) gene polymorphisms in a male Finnish population. Psychiatr Genet. 2009;19:117–125. doi: 10.1097/YPG.0b013e32832a4f7b. [DOI] [PubMed] [Google Scholar]
  106. Lind PA, Macgregor S, Vink JM, Pergadia ML, Hansell NK, de Moor MH, Smit AB, Hottenga JJ, Richter MM, Heath AC, Martin NG, Willemsen G, de Geus EJ, Vogelzangs N, Penninx BW, Whitfield JB, Montgomery GW, Boomsma DI, Madden PA. A genomewide association study of nicotine and alcohol dependence in Australian and Dutch populations. Twin Res Hum Genet. 2010;13:10–29. doi: 10.1375/twin.13.1.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. van der Linde K, Fumagalli E, Roman G, Lyons LC. The FlyBar: Administering alcohol to flies. [Accessed January 5, 2015];J Vis Exp. 2014 87:e50442. doi: 10.3791/50442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. van der Linde K, Lyons LC. Circadian modulation of acute alcohol sensitivity but not acute tolerance in Drosophila. Chronobiol Int. 2011;28:397–406. doi: 10.3109/07420528.2011.577921. [DOI] [PubMed] [Google Scholar]
  109. Linneberg A, Gonzalez-Quintela A, Vidal C, Jorgensen T, Fenger M, Hansen T, Pedersen O, Husemoen LL. Genetic determinants of both ethanol and acetaldehyde metabolism influence alcohol hypersensitivity and drinking behaviour among Scandinavians. Clin Exp Allergy. 2010;40:123–130. doi: 10.1111/j.1365-2222.2009.03398.x. [DOI] [PubMed] [Google Scholar]
  110. Loh el W, Lane HY, Chen CH, Chang PS, Ku LW, Wang KH, Cheng AT. Glutamate decarboxylase genes and alcoholism in Han Taiwanese men. Alcohol Clin Exp Res. 2006;30:1817–1823. doi: 10.1111/j.1530-0277.2006.00218.x. [DOI] [PubMed] [Google Scholar]
  111. Luo X, Kranzler HR, Zuo L, Wang S, Schork NJ, Gelernter J. Diplotype trend regression analysis of the ADH gene cluster and the ALDH2 gene: multiple significant associations with alcohol dependence. Am J Hum Genet. 2006;78:973–987. doi: 10.1086/504113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Maiya R, Lee S, Berger KH, Kong EC, Slawson JB, Griffith LC, Takamiya K, Huganir RL, Margolis B, Heberlein U. DlgS97/SAP97, a neuronal isoform of discs large, regulates ethanol tolerance. PLoS One. 2012;7:e48967. doi: 10.1371/journal.pone.0048967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Maples T, Rothenfluh A. A simple way to measure ethanol sensitivity in flies. J Vis Exp. 2011;48:e2541. doi: 10.3791/2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Mathies LD, Blackwell GG, Austin MK, Edwards AC, Riley BP, Davies AG, Bettinger JC. SWI/SNF chromatin remodeling regulates alcohol response behaviors in Caenorhabditis elegans and is associated with alcohol dependence in humans. Proc Natl Acad Sci USA. 2015;112:3032–3037. doi: 10.1073/pnas.1413451112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. McClure KD, Heberlein U. A small group of neurosecretory cells expressing the transcriptional regulator apontic and the neuropeptide corazonin mediate ethanol sedation in Drosophila. J Neurosci. 2013;33:4044–4054. doi: 10.1523/JNEUROSCI.3413-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Mitchell P, Mould R, Dillon J, Glautier S, Andrianakis I, James C, Pugh A, Holden-Dye L, O’Connor V. A differential role for neuropeptides in acute and chronic adaptive responses to alcohol: behavioural and genetic analysis in Caenorhabditis elegans. PLoS One. 2010;5:e10422. doi: 10.1371/journal.pone.0010422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. 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 Antropol. 2008;32(Suppl 1):127–131. [PubMed] [Google Scholar]
  118. Moore MS, DeZazzo J, Luk AY, Tully T, Singh CM, Heberlein U. Ethanol intoxication in Drosophila: genetic and pharmacological evidence for regulation by the cAMP signaling pathway. Cell. 1998;93:997–1007. doi: 10.1016/s0092-8674(00)81205-2. [DOI] [PubMed] [Google Scholar]
  119. Morgan PG, Sedensky MM. Mutations affecting sensitivity to ethanol in the nematode, Caenorhabditis elegans. Alcohol Clin Exp Res. 1995;19:1423–1429. doi: 10.1111/j.1530-0277.1995.tb01002.x. [DOI] [PubMed] [Google Scholar]
  120. Morozova TV, Ayroles JF, Jordan KW, Duncan LH, Carbone MA, Lyman RF, Stone EA, Govindaraju DR, Ellison RC, Mackay TF, Anholt RR. Alcohol sensitivity in Drosophila: translational potential of systems genetics. Genetics. 2009;183:733–745. 1SI–12SI. doi: 10.1534/genetics.109.107490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Morozova TV, Goldman D, Mackay TF, Anholt RR. The genetic basis of alcoholism: multiple phenotypes, many genes, complex networks. Genome Biol. 2012;13:239. doi: 10.1186/gb-2012-13-2-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Morozova TV, Mackay TF, Anholt RR. Transcriptional networks for alcohol sensitivity in Drosophila melanogaster. Genetics. 2011;187:1193–1205. doi: 10.1534/genetics.110.125229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. 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. doi: 10.1097/01.alc.0000164365.04961.b1. [DOI] [PubMed] [Google Scholar]
  124. Nery FG, Miranda-Scippa A, Nery-Fernandes F, Kapczinski F, Lafer B. Prevalence and clinical correlates of alcohol use disorders among bipolar disorder patients: results from the Brazilian Bipolar Research Network. Compr Psychiatry. 2014;55:1116–1121. doi: 10.1016/j.comppsych.2014.02.006. [DOI] [PubMed] [Google Scholar]
  125. NIAAA (dynamic) Alcohol and Your Health. [Accessed January 5, 2015]; Available at: http://www.niaaa.nih.gov/alcohol-health/overview-alcohol-consumption/alcohol-use-disorders.
  126. Ogueta M, Cibik O, Eltrop R, Schneider A, Scholz H. The influence of Adh function on ethanol preference and tolerance in adult Drosophila melanogaster. Chem Senses. 2010;35:813–822. doi: 10.1093/chemse/bjq084. [DOI] [PubMed] [Google Scholar]
  127. Okubo T, Harada S. Polymorphism of the neuropeptide Y gene: an association study with alcohol withdrawal. Alcohol Clin Exp Res. 2001;25(6 Suppl):59S–62S. doi: 10.1097/00000374-200106001-00014. [DOI] [PubMed] [Google Scholar]
  128. Palmer RH, 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. doi: 10.1016/j.drugalcdep.2012.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. 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. doi: 10.1007/s00439-013-1281-8. [DOI] [PubMed] [Google Scholar]
  130. Park SK, Sedore SA, Cronmiller C, Hirsh J. Type II cAMP-dependent protein kinase-deficient Drosophila are viable but show developmental, circadian, and drug response phenotypes. J Biol Chem. 2000;275:20588–20596. doi: 10.1074/jbc.M002460200. [DOI] [PubMed] [Google Scholar]
  131. Peng Q, Gizer IR, Libiger O, Bizon C, Wilhelmsen KC, Schork NJ, Ehlers CL. Association and ancestry analysis of sequence variants in ADH and ALDH using alcohol-related phenotypes in a Native American community sample. Am J Med Genet B Neuropsychiatr Genet. 2014;165B:673–683. doi: 10.1002/ajmg.b.32272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Peru Y Colón de Portugal RL, Acevedo SF, Rodan AR, Chang LY, Eaton BA, Rothenfluh A. Adult neuronal Arf6 controls ethanol-induced behavior with Arfaptin downstream of Rac1 and RhoGAP18B. J Neurosci. 2012;32:17706–17713. doi: 10.1523/JNEUROSCI.1944-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Peru Y Colón de Portugal, Ojelade SA, Penninti PS, Dove RJ, Nye MJ, Acevedo SF, Lopez A, Rodan AR, Rothenfluh A. Long-lasting, experience-dependent alcohol preference in Drosophila. Addict Biol. 2014;19:392–401. doi: 10.1111/adb.12105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Pohl JB, Baldwin BA, Dinh BL, Rahman P, Smerek D, Prado FJ, III, Sherazee N, Atkinson NS. Ethanol preference in Drosophila melanogaster is driven by its caloric value. Alcohol Clin Exp Res. 2012;36:1903–1912. doi: 10.1111/j.1530-0277.2012.01817.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Pohl JB, Ghezzi A, Lew LK, Robles RB, Cormack L, Atkinson NS. Circadian genes differentially affect tolerance to ethanol in Drosophila. Alcohol Clin Exp Res. 2013;37:1862–1871. doi: 10.1111/acer.12173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Prasad P, Ambekar A, Vaswani M. Case-control association analysis of dopamine receptor polymorphisms in alcohol dependence: a pilot study in Indian males. BMC Res Notes. 2013;6:418. doi: 10.1186/1756-0500-6-418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Preuss UW, Wurst FM, Ridinger M, Rujescu D, Fehr C, Koller G, Bondy B, Wodarz N, Soyka M, Zill P. Association of functional DBH genetic variants with alcohol dependence risk and related depression and suicide attempt phenotypes: results from a large multicenter association study. Drug Alcohol Depend. 2013;133:459–467. doi: 10.1016/j.drugalcdep.2013.07.002. [DOI] [PubMed] [Google Scholar]
  138. Raabe R, Mathies L, Davies A, Bettinger J. The omega-3 fatty acid eicosapentaenoic acid is required for normal alcohol response behaviors in C. elegans. PLoS One. 2014;9:e105999. doi: 10.1371/journal.pone.0105999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Reimand J, Arak T, Vilo J. g:Profiler—a web server for functional interpretation of gene lists (2011 update) Nucleic Acids Res. 2011;39(Web Server issue):W307–W315. doi: 10.1093/nar/gkr378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Reimers MA, Riley BP, Kalsi G, Kertes DA, Kendler KS. Pathway based analysis of genotypes in relation to alcohol dependence. Pharmacogenomics J. 2012;12:342–348. doi: 10.1038/tpj.2011.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Rietschel M, Treutlein J. The genetics of alcohol dependence. Ann N Y Acad Sci. 2013;1282:39–70. doi: 10.1111/j.1749-6632.2012.06794.x. [DOI] [PubMed] [Google Scholar]
  142. Robinson BG, Atkinson NS. Is alcoholism learned? Insights from the fruit fly. Curr Opin Neurobiol. 2013;23:529–534. doi: 10.1016/j.conb.2013.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Robinson BG, Khurana S, Kuperman A, Atkinson NS. Neural adaptation leads to cognitive ethanol dependence. Curr Biol. 2012;22:2338–2341. doi: 10.1016/j.cub.2012.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Rodan AR, Kiger JA, Jr, Heberlein U. Functional dissection of neuroanatomical loci regulating ethanol sensitivity in Drosophila. J Neurosci. 2002;22:9490–9501. doi: 10.1523/JNEUROSCI.22-21-09490.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Rodan AR, Rothenfluh A. The genetics of behavioral alcohol responses in Drosophila. Int Rev Neurobiol. 2010a;91:25–51. doi: 10.1016/S0074-7742(10)91002-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Rodan AR, Rothenfluh A, editors. International Review of Neurobiology. Functional Plasticity and Genetic Variation: Insights into the Neurobiology of Alcoholism. Academic Press; New York: 2010b. The Genetics of Behavioral Alcohol Responses in Drosophila. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Rothenfluh A, Threlkeld RJ, Bainton RJ, Tsai LT, Lasek AW, Heberlein U. Distinct behavioral responses to ethanol are regulated by alternate RhoGAP18B isoforms. Cell. 2006;127:199–211. doi: 10.1016/j.cell.2006.09.010. [DOI] [PubMed] [Google Scholar]
  148. Rothenfluh A, Troutwine BR, Ghezzi A, Atkinson NS. The genetics of alcohol responses of invertebrate model systems. In: Noronha A, Cui C, Harris RA, Crabbe JC, editors. Neurobiology of Alcohol Dependence. Academic Press; London: 2014. pp. 467–495. [Google Scholar]
  149. Sandhu S, Kollah A, Lewellyn L, Chan RF, Grotewiel M. An inexpensive, scalable assay for measuring ethanol sedation sensitivity and rapid tolerance in Drosophila. [Accessed January 5, 2015];J Vis Exp. 2015 98:e52676. doi: 10.3791/52676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Schlaepfer IR, Hoft NR, Collins AC, Corley RP, Hewitt JK, Hopfer CJ, Lessem JM, McQueen MB, Rhee SH, Ehringer MA. The CHRNA5/A3/B4 gene cluster variability as an important determinant of early alcohol and tobacco initiation in young adults. Biol Psychiatry. 2008;63:1039–1046. doi: 10.1016/j.biopsych.2007.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Schneider A, Ruppert M, Hendrich O, Giang T, Ogueta M, Hampel S, Vollbach M, Buschges A, Scholz H. Neuronal basis of innate olfactory attraction to ethanol in Drosophila. PLoS One. 2012;7:e52007. doi: 10.1371/journal.pone.0052007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Scholz H. Influence of the biogenic amine tyramine on ethanol-induced behaviors in Drosophila. J Neurobiol. 2005;63:199–214. doi: 10.1002/neu.20127. [DOI] [PubMed] [Google Scholar]
  153. Scholz H, Franz M, Heberlein U. The hangover gene defines a stress pathway required for ethanol tolerance development. Nature. 2005;436:845–847. doi: 10.1038/nature03864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Scholz H, Mustard JA. Invertebrate models of alcoholism. Curr Top Behav Neurosci. 2011;13:433–457. doi: 10.1007/7854_2011_128. [DOI] [PubMed] [Google Scholar]
  155. Scholz H, Ramond J, Singh CM, Heberlein U. Functional ethanol tolerance in Drosophila. Neuron. 2000;28:261–271. doi: 10.1016/s0896-6273(00)00101-x. [DOI] [PubMed] [Google Scholar]
  156. Schuckit MA. A brief history of research on the genetics of alcohol and other drug use disorders. J Stud Alcohol Drugs Suppl. 2014;75(Suppl 17):59–67. doi: 10.15288/jsads.2014.s17.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Schumann G, Coin LJ, Lourdusamy A, Charoen P, Berger KH, Stacey D, Desrivieres 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-Proenca C, Chambers JC, Clarke TK, Dahmen N, de Geus EJ, Dick D, Ducci F, Easton A, Edenberg HJ, Esko T, Fernandez-Medarde A, Foroud T, Freimer NB, Girault JA, Grobbee DE, Guarrera S, Gudbjartsson DF, Hartikainen AL, Heath AC, Hesselbrock V, Hofman A, Hottenga JJ, Isohanni MK, Kaprio J, Khaw KT, Kuehnel B, Laitinen J, Lobbens S, Luan J, Mangino M, Maroteaux M, Matullo G, McCarthy MI, Mueller C, Navis G, Numans ME, Nunez A, Nyholt DR, Onland-Moret CN, Oostra BA, O’Reilly PF, Palkovits M, Penninx BW, Polidoro S, Pouta A, Prokopenko I, Ricceri F, Santos E, Smit JH, Soranzo N, Song K, Sovio U, Stumvoll M, Surakk I, Thorgeirsson TE, Thorsteinsdottir U, Troakes C, Tyrfingsson T, Tonjes A, Uiterwaal CS, Uitterlinden AG, van der Harst P, van der Schouw YT, Staehlin O, Vogelzangs N, Vollenweider P, Waeber G, Wareham NJ, Waterworth DM, Whitfield JB, Wichmann EH, Willemsen G, Witteman JC, Yuan X, Zhai G, Zhao JH, Zhang W, Martin NG, Metspalu A, Doering A, Scott J, Spector TD, Loos RJ, Boomsma DI, Mooser V, Peltonen L, Stefansson K, van Duijn CM, Vineis P, Sommer WH, Kooner JS, Spanagel R, Heberlein UA, Jarvelin MR, Elliott P. 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. 2011;108:7119–7124. doi: 10.1073/pnas.1017288108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Schwab SG, Franke PE, Hoefgen B, Guttenthaler V, Lichtermann D, Trixler M, Knapp M, Maier W, Wildenauer DB. Association of DNA polymorphisms in the synaptic vesicular amine transporter gene (SLC18A2) with alcohol and nicotine dependence. Neuropsychopharmacology. 2005;30:2263–2268. doi: 10.1038/sj.npp.1300809. [DOI] [PubMed] [Google Scholar]
  159. Sha K, Choi SH, Im J, Lee GG, Loeffler F, Park JH. Regulation of ethanol-related behavior and ethanol metabolism by the Corazonin neurons and Corazonin receptor in Drosophila melanogaster. PLoS One. 2014;9:e87062. doi: 10.1371/journal.pone.0087062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Shohat-Ophir G, Kaun KR, Azanchi R, Heberlein U. Sexual deprivation increases ethanol intake in Drosophila. Science. 2012;335:1351–1355. doi: 10.1126/science.1215932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Spanagel R. Alcoholism: a systems approach from molecular physiology to addictive behavior. Physiol Rev. 2009;89:649–705. doi: 10.1152/physrev.00013.2008. [DOI] [PubMed] [Google Scholar]
  162. Speca DJ, Chihara D, Ashique AM, Bowers MS, Pierce-Shimomura JT, Lee J, Rabbee N, Speed TP, Gularte RJ, Chitwood J, Medrano JF, Liao M, Sonner JM, Eger EI, II, Peterson AS, McIntire SL. Conserved role of unc-79 in ethanol responses in lightweight mutant mice. PLoS Genet. 2010;6:e1001057. doi: 10.1371/journal.pgen.1001057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Sun HS, Fann CS, Lane HY, Chang YT, Chang CJ, Liu YL, Cheng AT. A functional polymorphism in the promoter region of the tryptophan hydroxylase gene is associated with alcohol dependence in one aboriginal group in Taiwan. Alcohol Clin Exp Res. 2005;29:1–7. doi: 10.1097/01.alc.0000150635.51934.6d. [DOI] [PubMed] [Google Scholar]
  164. Tabakoff B, Saba L, Printz M, Flodman P, Hodgkinson C, Goldman D, Koob G, Richardson HN, Kechris K, Bell RL, Hubner N, Heinig M, Pravenec M, Mangion J, Legault L, Dongier M, Conigrave KM, Whitfield JB, Saunders J, Grant B, Hoffman PL WHO/ISBRA Study on State and Trait Markers of Alcoholism. Genetical genomic determinants of alcohol consumption in rats and humans. BMC Biol. 2009;7:70. doi: 10.1186/1741-7007-7-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. 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. [PMC free article] [PubMed] [Google Scholar]
  166. Topper S, Aguilar S, Topper V, Elbel E, Pierce-Shimomura J. Alcohol disinhibition of behaviors in C. elegans. PLoS One. 2014;9:e92965. doi: 10.1371/journal.pone.0092965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Urizar NL, Yang Z, Edenberg HJ, Davis RL. Drosophila homer is required in a small set of neurons including the ellipsoid body for normal ethanol sensitivity and tolerance. J Neurosci. 2007;27:4541–4551. doi: 10.1523/JNEUROSCI.0305-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Verhulst B, Neale MC, Kendler KS. The heritability of alcohol use disorders: a meta-analysis of twin and adoption studies. Psychol Med. 2014;45:1061–1072. doi: 10.1017/S0033291714002165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Wakefield JC, Schmitz MF. How many people have alcohol use disorders? Using the harmful dysfunction analysis to reconcile prevalence estimates in two community surveys. Front Psychiatry. 2014;5:10. doi: 10.3389/fpsyt.2014.00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Wen T, Parrish CA, Xu D, Wu Q, Shen P. Drosophila neuropeptide F and its receptor, NPFR1, define a signaling pathway that acutely modulates alcohol sensitivity. Proc Natl Acad Sci USA. 2005;102:2141–2146. doi: 10.1073/pnas.0406814102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. 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. doi: 10.1016/s0006-3223(03)00072-6. [DOI] [PubMed] [Google Scholar]
  172. Wetherill L, Kapoor M, Agrawal A, Bucholz K, Koller D, Bertelsen SE, Le N, Wang JC, Almasy L, Hesselbrock V, Kramer J, Nurnberger JI, Jr, Schuckit M, Tischfield JA, Xuei X, Porjesz B, Edenberg HJ, Goate AM, Foroud T. Family-based association analysis of alcohol dependence criteria and severity. Alcohol Clin Exp Res. 2014;38:354–366. doi: 10.1111/acer.12251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Wetherill L, Schuckit MA, Hesselbrock V, Xuei X, Liang T, Dick DM, Kramer J, Nurnberger JI, Jr, 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. doi: 10.1111/j.1530-0277.2008.00790.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. WHO. [Accessed January 5, 2015];Global status report on alcohol and health 2014. 2014 Available at: http://www.who.int/substance_abuse/publications/global_alcohol_report/en/
  175. Wilson RJ, Goodman JL, Strelets VB, FlyBase C. FlyBase: integration and improvements to query tools. Nucleic Acids Res. 2008;36(Database issue):D588–D593. doi: 10.1093/nar/gkm930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Wolf FW, Rodan AR, Tsai LT, Heberlein U. High-resolution analysis of ethanol-induced locomotor stimulation in Drosophila. J Neurosci. 2002;22:11035–11044. doi: 10.1523/JNEUROSCI.22-24-11035.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Xu S, Chan T, Shah V, Zhang S, Pletcher SD, Roman G. The propensity for consuming ethanol in Drosophila requires rutabaga adenylyl cyclase expression within mushroom body neurons. Genes Brain Behav. 2012;11:727–739. doi: 10.1111/j.1601-183X.2012.00810.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Yang BZ, 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. doi: 10.1093/hmg/ddm240. [DOI] [PubMed] [Google Scholar]
  179. Yang BZ, 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. doi: 10.1111/j.1530-0277.2008.00800.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Yu W, Gwinn M, Clyne M, Yesupriya A, Khoury MJ. A navigator for human genome epidemiology. Nat Genet. 2008;40:124–125. doi: 10.1038/ng0208-124. [DOI] [PubMed] [Google Scholar]
  181. Zuo L, Zhang H, Malison RT, Li CS, Zhang XY, Wang F, Lu L, Lu L, Wang X, Krystal JH, Zhang F, Deng HW, Luo X. Rare ADH variant constellations are specific for alcohol dependence. Alcohol Alcohol. 2013;48:9–14. doi: 10.1093/alcalc/ags104. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES