CADM2 is implicated in impulsive personality and numerous other traits by genome- and phenome-wide association studies in humans, with further support from studies of Cadm2 mutant mice ======================================================================================================================================================================================== * Sandra Sanchez-Roige * Mariela V Jennings * Hayley H A Thorpe * Jazlene E Mallari * Lieke C van der Werf * Sevim B Bianchi * Calvin Lee * Travis T Mallard * Samuel A Barnes * Jin Yi Wu * Amanda M Barkley-Levenson * Ely C Boussaty * Cedric E Snethlage * Danielle Schafer * Zeljana Babic * Boyer D Winters * Katherine E Watters * Thomas Biederer * the 23andMe Research Team * James Mackillop * David N Stephens * Sarah L Elson * Pierre Fontanillas * Jibran Y Khokhar * Jared W Young * Abraham A Palmer ## ABSTRACT **Background** Impulsivity is a multidimensional heritable phenotype that broadly refers to the tendency to act prematurely and is associated with multiple forms of psychopathology, including substance use disorders. **Methods** We performed genome-wide association studies (**GWAS**) of eight impulsive personality traits from the Barratt Impulsiveness Scale and the short UPPS-P Impulsive Personality Scale (N=123,509-133,517 23andMe research participants of European ancestry), and a measure of Drug Experimentation (N=130,684). Because these GWAS implicated the gene *CADM2*, we next performed a phenome-wide study (**PheWAS**) of several of the implicated variants in *CADM2* in a multi-ancestral 23andMe cohort (N=3,229,317, European; N=579,623, Latin American; N=199,663, African American). Finally, we produced *Cadm2* mutant mice and tested them using a battery of analogous behavioral tasks. **Results** In humans, impulsive personality traits showed modest chip-heritability (∼6-11%), and moderate genetic correlations (rg=.20-.50) with other personality traits, and various psychiatric and medical traits. We replicated associations from earlier GWAS of these traits and found novel associations including *DRD2, CRHR1, FOXP2, TCF4, PTPRF*. PheWAS for *CADM2* variants identified associations with 378 traits in European participants, and 47 traits in Latin American participants, replicating associations with risky behaviors, cognition and BMI, and revealing novel associations including allergies, anxiety, irritable bowel syndrome, and migraine. *Cadm2* mutant mice recapitulated some of the associations found in humans, including impulsivity, cognition, and BMI. **Conclusions** Our results further delineate the role of *CADM2* in impulsivity and numerous other psychiatric and somatic traits across ancestries, with further support from studies of *Cadm2* mutant mice. ## INTRODUCTION Impulsivity is a multifaceted psychological construct that has been broadly defined as thoughts or actions that are “poorly conceived, prematurely expressed, unduly risky or inappropriate to the situation, and that often result in undesirable consequences” (1). Impulsivity has been repeatedly associated with numerous psychiatric diseases, including ADHD and substance use disorders (2,3). We previously performed genome-wide association studies (**GWAS**) of impulsive personality traits (n=21,806-22,861) using two of the most widely used impulsivity questionnaires, the Barratt Impulsiveness Scale (**BIS-11**; 3 traits) and the Impulsive Personality Scale (**UPPS-P**; 5 traits), as well as a measure of Drug Experimentation (4). These traits were partially genetically correlated, suggesting that each impulsivity domain is governed by overlapping but distinct biological mechanisms (4,5). Our work also identified significant genetic correlations between impulsivity and numerous psychiatric and substance use traits, in line with the NIMH Research Domain Criteria (**RDoC**), proposing impulsivity as a transdiagnostic endophenotype for psychopathology (6). The cell adhesion molecule 2 (*CADM2*) gene, which was the most robustly implicated gene in our prior GWAS of impulsivity (4), has also been extensively implicated in other risky and substance use behaviors (7). *CADM2* mediates synaptic plasticity and is enriched in the frontal cortex and striatum, which are regions that regulate reward and inhibitory processes. We and others have implicated this gene in traits that may underlie disinhibition in humans, supporting the observed genetic correlations between impulsivity and personality (8), educational attainment (9), cognition (10), risk-taking (11), substance use (4,10,12–15), externalizing psychopathology (16), neurodevelopmental disorders (17,18), physical activity (23), reproductive health (20,21), metabolic traits (19), and BMI (22), among others (see GWAS Catalog [www.ebi.ac.uk/gwas/](https://www.ebi.ac.uk/gwas/)). *Cadm2* knockout mice have previously been assessed for body weight and energy homeostasis (24) but have never been behaviorally characterized for measures of impulsivity or related behaviors. Here, we took three approaches to elucidate genetic factors related to impulsivity. First, we collaborated with 23andMe, Inc., to extend upon our earlier GWAS of impulsivity (4) by increasing our sample size approximately 6-fold (n=123,509-133,517). Second, we performed a phenome-wide study (**PheWAS**) of the 5 single nucleotide polymorphisms (SNPs) in and around *CADM2* that have been most strongly implicated by the current and prior GWAS. PheWAS were conducted in three ancestral groups (N=3,229,317, European; N=579,623, Latin American; N=199,663, African American) from the 23andMe research cohort, examining close to 1,300 traits, most with no published GWAS. Finally, we extensively phenotyped a mouse model of a *Cadm2* mutant allele. We tested mice that expressed a null allele Cadm2tm1a(KOMP)Mbp on an otherwise isogenic (inbred) background (C57BL/6N) under homogeneous environmental conditions, in a broad battery of behavioral tasks that included analogous human measures of risk-taking and impulsivity, substance use, cognition and BMI. ## MATERIALS AND METHODS ### Human Studies #### GWAS cohort and phenotypes We analyzed data from a cohort of up to 133,517 male and female research participants of European ancestry, a subset of which were analyzed in our prior publications (4,13,13,25,26). All participants were drawn from the research participant base of 23andMe, Inc., a direct-to-consumer genetics company, and were not compensated for their participation. Participants provided informed consent and participated in the research online, under a protocol approved by the external AAHRPP-accredited IRB, Ethical & Independent Review Services ([www.eandireview.com](https://www.eandireview.com)). During 4 months in 2015 and 14 months from 2018-2020, participants responded to a survey that, depending on branching logic, included up to 139 questions pertaining to aspects of impulsivity and substance use and abuse. To measure impulsive personality, we used five subscales from the 20-item Impulsive Behavior Scale [**UPPS-P** (27,28); (lack of) Premeditation, (lack of) Perseverance, Positive Urgency, Negative Urgency, and Sensation Seeking (**Table S1**)]. Each subscale includes 4-items and yields integer scores from 4 to 16. We also administered the Barratt Impulsiveness Scale [**BIS-11** (29); a 30-item questionnaire that measures Attentional, Motor, and Nonplanning impulsiveness (**Table S1**)]. Lastly, we measured Drug Experimentation, defined as the number of 11 different classes of drugs an individual has used (see **Table S1**; tobacco [cigarettes, cigars, chewing tobacco], alcohol, marijuana, cocaine, methamphetamine, LSD/magic mushrooms, ecstasy, prescription stimulants [taken not as prescribed; e.g., Ritalin, Adderall, Strattera], prescription painkillers [taken not as prescribed; e.g., Vicodin, Oxycontin], heroin, opium); this measure yields scores from 0 to 11, and was adapted from the PhenX toolkit [[https://www.phenxtoolkit.org/](https://www.phenxtoolkit.org/); (30)]. We scored UPPS-P, BIS-11 and Drug Experimentation as previously described (4). We used quantile normalization, since some scores were not normally distributed (**Figures S1-3**). Only individuals identified as European ancestry based on empirical genotype data (31) were included in this study. Basic demographic information about this sample is presented in **Table S2**. We used Pearson correlation coefficients (*r*) to measure the phenotypic relationships between impulsivity subscales and demographics. #### Genome-wide association and secondary analyses DNA extraction and genotyping were performed on saliva samples by CLIA-certified and CAP-accredited clinical laboratories of Laboratory Corporation of America. Quality control, imputation, and genome-wide analyses were performed by 23andMe. Briefly, participants were genotyped on one of five Illumina genotyping platforms, containing between 550,000 to 950,000 variants, for a total of 1.6 million genotyped variants. Samples that failed to reach 98.5% call rate were re-analyzed. Genotyping quality controls included discarding variants with a Hardy-Weinberg *p*<1.00E−20, batch effects (ANOVA *p*<1.00E-20), or a call rate of <90% (33,35,36). About 64.4M variants were then imputed against the Haplotype Reference Consortium (**HRC**) panel, augmented by a single unified imputation reference panel combining the May 2015 release of the 1000 Genomes Phase 3 haplotypes with the UK10K imputation reference panel, for variants not present in the HRC. Imputed variants with low imputation quality (r2<0.5 averaged across batches or a minimum r2<0.3), or with evidence of batch effects (*p*<1.00E-50) were removed (33,35). A total of 1.3M genotyped and 30.5M imputed variants passed the pre- and post GWAS quality controls. We furthermore filtered out variants with minor allele frequency (**MAF**) <0.1%, which are extremely sensitive to quantitative trait over-dispersion, reducing to 14.1M variants available for follow-up analyses (**Table S3**; see (32,33) for further information). Principal components were computed using ∼65,000 high-quality genotyped variants present in all five genotyping platforms. 23andMe’s analysis pipeline performs logistic regression assuming an additive model for allelic effects (**Supplementary Material**). A maximal set of unrelated individuals was chosen for the analysis using a segmental identity-by-descent (**IBD**) estimation algorithm (32) to ensure that only unrelated individuals were included in the sample. Individuals were defined as related if they shared more than 700□cM IBD, including regions where the two individuals shared either one or both genomic segments IBD. This level of relatedness (∼20% of the genome) corresponds to approximately the minimal expected sharing between first cousins in an outbred population. Covariates included age (inverse-normal transformed), sex, the top five principal genotype components, and indicator variables for genotyping platforms. *p*-values were corrected for genomic control. We used the FUMA web-based platform (version 1.3.6a) and MAGMA v1.08 (33,34) to further explore the functional consequences of the loci identified in the GWAS and to conduct gene-based analyses, respectively. MAGMA uses Ensembl (build 85) to map SNPs to 19,773 protein-coding genes. We used a Bonferroni correction based on the number of genes tested (*p*<2.53E-06). We used LDSC (35) to calculate genetic correlations (rg) between UPPS-P, BIS and Drug Experimentation, and 96 selected traits informed by prior literature across the following categories: substance use, personality, psychiatric, metabolic, education, lifestyle, health, pain, cognitive, longevity, reproductive, and sleep. #### Phenome-wide association scan (PheWAS) in 23andMe We performed a PheWAS for 5 *CADM2* SNPs (rs993137, rs62263923, rs11708632, rs818219, rs6803322) using up to 1,291 well-curated self-reported phenotypes from 23andMe research participants of European (N≤3,229,317), Latin American (N≤579,623) and African American (N≤199,663) ancestries. We excluded traits with <1,000 responses (11.99%, 3.42%, and 6.61% from the European, Latin American, and African American cohorts), based on a prior simulation study for PheWAS power analysis (36). Ancestry was determined by analyzing local ancestry (31), as described in detail in the **Supplementary Material**. Only the 3 largest ancestry groups were considered; smaller ancestry groups (e.g., East Asian, South Asian, Middle Eastern & North African) were not included due to sample size. The variants were selected based on our current results and previous literature (**Table S4**). Genotyped and imputed variant statistics in PheWAS are shown in **Table S5**. An overview of the data collection process has been previously described (37). The traits were distributed among 20 phenotypic categories (e.g., cognitive, autoimmune, psychiatric, etc.). For case-control comparisons, we computed association test results by logistic regression. For quantitative and categorical traits, association tests were performed by linear regression. All regression analyses were performed using R version 3.2.2. We assumed additive allelic effects and included covariates for age (as determined by participant date of birth), sex, and the top five ancestry-specific principal components. We used a 5% FDR correction for multiple testing. ### Mouse Studies #### Subjects, behavioral characterization, and statistical analyses Our *Cadm2* mutant mice were produced via *in vitro* fertilization, which was performed at the University of California San Diego, Moores Cancer Center, Transgenic Mouse Core. We used the JM8.N4 cryosperm line (CSD70565 KOMP), which carries a floxed null allele in the *Cadm2* gene (see **Figure S30**), on a C57BL/6N background. We then crossed the floxed null allele line with a constitutive CRE driver line (Stock# 014094; The Jackson Laboratory), yielding a global constitutive null allele. We used a heterozygous x heterozygous (**HET**) breeding scheme, which produced homozygous (**HOM**) mutant *Cadm2* mice and their HET and wildtype (**WT**) littermates. Mice were genotyped using allele-specific polymerase chain reaction on ear notch tissue followed by gel electrophoresis (38). CADM2 protein expression levels were quantified by western blotting (**Figure S31**). Five separate cohorts of male and female mice were used for these studies. See **Supplementary Material** for a more detailed description of the tasks and analyses of main variables. HOM, HET, and WT littermates were tested in parallel. Mice were between 4 and 6 months of age at the time of testing, unless otherwise stated. Lighting, housing and feeding conditions for each cohort are shown in the **Supplementary Material**. Procedures were approved by the University of California San Diego Institutional Animal Care and Use Committee. The UCSD animal facility meets all federal and state requirements for animal care and was approved by the American Association for Accreditation of Laboratory Animal Care. Procedures from cohort 2 were conducted in accordance with the Canadian Council on Animal Care and were approved by the University of Guelph Institutional Animal Care and Use Committee. #### Cohort 1 - Motivation, inhibition, and risk-taking behavior The first cohort (WT=25, HET=30, HOM=3) was used to examine risky behavior or “choice” impulsivity, behavioral flexibility, prepulse inhibition (**PPI**) of the acoustic startle response, and general exploration. The primary measure of choice impulsivity included risky behavior via the mouse Iowa gambling task [**IGT**, (39,40)]. Because *Cadm2* has been previously associated with alcohol consumption, we used a within-subjects design to assess risky behavior under acute doses of ethanol (0, 0.5, 1g/kg), as previously described (41). In addition, we evaluated motivation, as measured by a Progressive Ratio Breakpoint task [**PBRT**, (42,43)], and behavioral flexibility, as measured by a Probabilistic Reversal Learning task [**PRL**, (44)]. General exploration was measured via the Behavioral pattern monitor [**BPM**, (45,46)]. PPI took place in eight startle chambers (SR-LAB; San Diego Instruments, San Diego, California, USA), using previously published protocols (47,48). Data from PRBT, PRL, IGT, BPM and PPI were subjected to a univariate ANOVA with sex and genotype as between-subject factors. Data from the IGT ethanol challenge were analyzed using a repeated measure ANOVA with drug as a within-subject factor and genotype and sex as between-subject factors. The sample size of the HOM mice deviated from the expected Mendelian frequency (N=3) for no known reason; therefore, we excluded this group from the analyses of this cohort. #### Cohort 2 - Motoric impulsivity The second cohort (WT=13; HET=14, HOM=12) was used to examine “motoric” impulsivity via the 5-choice serial reaction time task [**5CSRTT** (41,49)]. Reinforcer preference, baseline and long ITI performance were analyzed using univariate ANOVA for parametric data or Kruskal-Wallis ANOVA for nonparametric data with genotype as the between-subject factor. Reduced stimulus duration (**RSD**) and variable ITI (**vITI**) performance were analyzed with genotype and stimulus duration (RSD) or stimulus delay (vITI) as the between- and within-subject factors, respectively, using two-way repeated-measures ANOVA for parametric data or two-way between-within subjects ANOVA on trimmed means for nonparametric data. Due to the low number of HOM females (N=4), sex was not included in the analyses of this cohort. #### Cohort 3 - General locomotion, anxiety-like behavior, and ethanol consumption The third cohort (WT=22; HET=44, HOM=12) was used to measure general locomotion, anxiety-like behavior, and ethanol consumption. Precisely, we assessed general locomotion via the Open Field (50–53), and anxiety-like behavior via the elevated plus maze [**EPM** (54)] and light-dark box [**LDB** (55)]. Lastly, we measured acute voluntary ethanol consumption using a drinking-in-the-dark paradigm [**DID**, (56)]. OFT, EPM and LDB data were assessed for normality and genotype effects were analyzed by ANOVA, with between-subject factors of genotype and sex. For the DID, ethanol drinking data were averaged and converted from milliliters to g ethanol/kg body weight for analysis. Drinking data were analyzed using ANOVA, with between-subject factors of genotype and sex. #### Cohort 4 - Body weight Considering the previous role of *Cadm2* on energy homeostasis (24), the fourth cohort (WT=29, HT=54, HOM=17) was used to assess body weight changes from adolescence (week 5) to late adulthood (week 35). Data were analyzed using linear mixed models with genotype and sex as dependent variables, and subject as a random factor. #### Cohort 5 - Dendrite morphology As *Cadm2* is important for synapse organization (24), the fifth cohort (WT=3, HET=3, HOM=3) was used to quantify dendritic spines from medium spiny neurons in the nucleus accumbens (**NAc**), which is one of the core regions for impulsive behavior (57). Only male mice were tested. ImageJ (Version 2.0.0-rc-69/1.52p) was used to analyze secondary and tertiary dendrites and score spine types. The results of the dendritic spine were averaged per 10μm for each mouse to standardize measures, using a classification criteria described previously (58). Statistical analyses were performed using ANOVA with genotype as a between-subjects factor. Statistical analysis was conducted using SPSS 26.0 or 28.0 (IBM, Armonk, NY), the Bio-Medical Data Package (for BPM data) or RStudio (RStudio, PBC, Boston, MA). For all analyses, outliers deviating more than three times the interquartile range from quartile 1 or quartile 3 were excluded from the analysis. A *p*<0.05 was required for results to be considered statistically significant. Post-hoc differences were assessed using Bonferroni or Tukey’s honestly significant difference with a *p*<0.05. All analyses were performed with the researchers blind to the genotype. ## RESULTS ### Genome-wide association analyses and secondary analyses Self-reported impulsivity and Drug Experimentation scores are shown in **Table S6**. We found that ∼6-11% of the phenotypic variation of these traits can be explained by common variants (**Table S7**). We identified 21 genome-wide significant associations (*p*<5.0E-08) for UPPS-P (5 traits), BIS (3 traits), and Drug Experimentation (**Figure 1**; **Figures S4-21**; **Table S8**). Although we tested 9 traits, in keeping with the standards of the field, we did not adjust the significance threshold. We also detected several nominal associations (*p*<1.0E-06, **Table S8**); we discuss some of the most interesting ones below. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/03/2022.01.29.22270095/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2022/05/03/2022.01.29.22270095/F1) Figure 1. Porcupine plot displaying 21 genome-wide significant hits for all impulsivity facets and Drug Experimentation. *CADM2* was consistent across 3/8 impulsivity facets [Sensation Seeking (UPPS-P), Motor and Nonplanning impulsivity (BIS-11)] at a genome-wide association level, and with 3 more impulsivity facets [Attentional (BIS-11), Negative Urgency and Premeditation (UPPS-P]), at a gene-based level (**Table S8**). *CADM2* was also associated with risky behavior, such as Drug Experimentation ### GWAS of UPPS-P #### Premeditation We detected one significant hit (rs2958162, *p*=2.50E-10), located on chromosome 18 in the *TCF4* gene, which encodes a helix-loop-helix transcription factor and is widely expressed throughout the body and during development. Polymorphisms in *TCF4* have been associated with risk-taking and adventurousness (15), alcohol consumption (59), schizophrenia (60), depression (61,62), and neuroticism (63,64) (**Table S9**); *TCF4* is also a non-GWAS candidate gene for other psychiatric and neurological conditions (65). We noted two interesting nominal associations: rs774880622 (*p*=2.10E-07) on chromosome 11, near the *DRD2* gene, which has been extensively studied in relation to reward and is a candidate gene for many psychiatric disorders, particularly substance use disorders [e.g., (66)]; and rs72819189 (*p*=9.70E-07), on chromosome 2, near the *AFF3* gene, which was a robust candidate gene associated with externalizing psychopathology (16). #### Perseverance We detected one significant association (rs5943997, *p*=1.50E-8) in the *POLA1* gene on the X chromosome. *POLA1* has been related to blood traits (65) and neurodevelopmental disorders (67), but its association with impulsivity is novel. We note a nominal association with rs10401120 (*p*=6.80E-07), in the *TCF4* gene. #### Positive Urgency We identified one potential significant hit (rs143987963, *p*=4.30E-08) on chromosome 12, near the genes *MDM1* and *RAP1B*, but inspection of the locus zoom plot (**Figure S9**) does not support a robust association. #### Negative Urgency We detected three significant hits: rs4840542 (*p*=1.60E-09), on chromosome 8, in the *XKR6* gene; rs5008475 (*p*=4.90E-09), on chromosome 5, near *TMEM161B* and *MEF2C*; and rs7829975, on chromosome 8, near *SGK223* and *CLDN23* (*p*=5.00E-09). Variants in strong LD with rs4840542 and rs7829975 are highly pleiotropic, and have been previously associated with several traits (**Table S9**), including body mass index (**BMI**) (68,69), neuroticism (70,71), depression (72), blood pressure, and alcohol consumption (73). *XKR6* was also implicated in a recent GWAS of externalizing (74), and a GWAS of anxiety and depression (72). We note nominal associations near the corticotropin receptor gene (*CRHR1*; rs2532373, *p*=9.10E-08), which is a robust candidate gene in relation to stress, depression and anxiety disorders; with *CDH13* (rs426583, *p*=8.10E-07), which has been previously associated with methamphetamine response in both humans (75) and rats (76); and with *NRXN1* (rs10651842, *p*=7.10E-07), which has previously been associated with educational attainment (9) and BMI (69). #### Sensation Seeking We detected 5 genome-wide significant associations. First, we replicated our previously reported (4) association with a SNP near *CADM2* (rs11288859, *p*=2.10E-09). We also detected an association with a SNP in *TCF4* (rs2958178, *p*=3.80E-12). We identified a significant hit in *CACNA2D1* (rs38547, *p*=2.10E-08) on chromosome 18. *CACNA2D1* has been previously associated with feeling nervous (70), and levels of sex hormone-binding globulin (77). Furthermore, we found a significant association (rs1605379, *p*=3.80E-08) on chromosome 16, near *CYLD* and *SALL1*. SNPs in strong LD with rs1605379 have been previously identified for risk-taking, adventurousness, and smoking initiation (**Table S9**). Lastly, we found a significant association (rs12600879, *p*=4.10E-08) on chromosome 17, near *TBX21* and *OSBPL7*. Variants in strong LD with rs12600879 have been associated with BMI (78), but the finding in relation to impulsivity is novel. ### GWAS of BIS-11 #### Attentional We identified one significant association (rs10196237, *p*=1.10E-08) on chromosome 2, near the genes *SPHKAP and PID1. SPHKAP* has been previously associated with educational attainment (9), but the association with impulsivity is novel. We also detected a nominal association (rs145225651, *p*=5.00E-07), which is near the gene *CADM2*. Also of note is a nominal association within *PABPC4* (rs1601647, *p*=6.30E-07), on chromosome 4, which was also nominally associated with Nonplanning, but novel in relation to impulsivity. #### Motor We detected one significant association near *CADM2* (rs35614735, *p*=3.20E-11). We also considered one suspect association (rs111502401, *p*=2.00E-08), on chromosome 19, near the genes *ZNF229* and *ZNF180*, but inspection of the regional association is not supportive of a strong association (**Figure S17**). #### Nonplanning We detected 2 previously associated variants: rs35614735 (*p*=4.70E-12) near *CADM2*, which was the same SNP identified for Motor impulsivity; and rs6872863 (*p*=1.20E-08) in the gene *ELOVL7*. Variants in strong LD with rs6872863 have been reported for a variety of traits including educational attainment, mathematical ability (9), household income (79), and brain morphology, such as cortical surface area (80) (**Table S9**). However, there is extensive LD in this region, making the association difficult to interpret. Again, we detected a nominal association within *PABPC4* (rs1601647, *p*=5.30E-07); with *AFF3* (rs72819158, *p*=1.10E-07), a robust candidate gene associated with externalizing (74); and within *FOXP2* (rs936146, *p*=5.50E-07), a robust candidate for cannabis use disorders (81), smoking initiation (66), general risk tolerance (82), cognitive ability, years of educational attainment and schizophrenia (83). ### GWAS of Drug Experimentation We previously reported (4) a suggestive association (rs2163971, *p*=3.00E-07) near the *CADM2* gene. In the present study, we identified a nearby SNP that was genome-wide significant (rs35614735, *p*=2.80E-15). We also report 4 novel hits (rs951740, *p*=9.70E-10, *PTPRF*; rs12713405, *p*=9.70E-09, *BLC11A;* rs67660520, *p*=7.60E-09, *CADPS2;* rs7128649, *p*=2.50E-09, *NCAM1*). Intriguingly, *PTPRF* has been recently associated with problematic prescription opioid use (25) and opioid use disorder (84), as well as smoking initiation/cessation (66), cognition (85), and educational attainment (9) (**Table S9**). Variants in strong LD with rs67660520 have been associated with ADHD (86), smoking initiation (66), number of sexual partners (82) and BMI (69) (**Table S9**). *NCAM1* variants have been previously associated with alcohol, cannabis and smoking behaviors (66,87), mathematical ability (9), and anxiety and depression (72), among other traits. We note a nominal association near the gene encoding the Glutamate Metabotropic Receptor 3 (*GRM3*; rs12673181, *p*=4.50E-07). Phenotypes associated with *GRM3* include educational attainment (9), schizophrenia (88), and neuroticism (89). We also identified a nominal association near *HTR3B* (rs6589400, p=2.80E-07), which encodes the serotonin receptor 3B and is implicated in various forms of impulsivity and the reward system (90). ### Gene-based analyses Similar to the GWAS results, gene-based analyses in MAGMA identified an association (Bonferroni *p*<2.53E-06; **Table S10**) between *CADM2* and 6 of the 9 traits examined in this paper: Premeditation, Sensation Seeking (UPPS-P); Attentional, Motor and Nonplanning (BIS-11); and Drug Experimentation. *TCF4*, which was significantly associated with Premeditation and Sensation Seeking in the GWAS, was significantly associated with these traits in the gene-based analysis. *MAPT*, which has been previously associated with many traits including multiple alcohol-related behaviors (13), was implicated in Negative Urgency. Lastly, *KDM4A*, which was recently related to problematic opioid use and interacts with selective serotonin reuptake inhibitors and dopaminergic agents (25), was significantly associated with Drug Experimentation. ### Phenotypic and genetic correlations A phenotypic and genetic correlation matrix of all 9 traits is shown in **Figure S22** and **Tables S11-12**. Consistent with the literature and our prior work (4,5,91,92), both phenotypic and genetic inter-correlations among the UPPS-P and BIS subscales were high and positive, with the exception of Sensation Seeking and Perseverance, suggesting that these traits may represent relatively different constructs (5,13,91). Drug experimentation was positively and significantly associated with all impulsive personality traits. All impulsivity traits were phenotypically associated (*r*=-0.34-0.11) with demographic variables (**Table S12**), impulsivity scores being greater in male and younger research participants, compared to female and older participants; and in participants with higher BMI, lower household income, and fewer years of education, as we previously reported (13). **Figure 2** shows a genetic correlation matrix of BIS, UPPS-P, Drug Experimentation and several other phenotypes (full results shown in **Table S13**). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/03/2022.01.29.22270095/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2022/05/03/2022.01.29.22270095/F2) Figure 2. Genetic correlations (rg) between UPPS-P, BIS, and Drug Experimentation, and other substance use, psychiatric, personality, cognitive, metabolic, health, pain, longevity and sleep traits (see **Table S9** for full results). All values survive 5% FDR correction. As anticipated, we found positive moderate to high genetic correlations between virtually all UPPS-P (except Perseverance and Sensation Seeking) and BIS subscales, and Drug Experimentation, and substance use disorders [e.g., alcohol dependence, rg=0.25-0.79; cannabis use disorder, rg=0.19-0.75; opioid use disorder from MVP, rg=0.28-0.75, and GENOA, rg=0.25-0.75]. The strongest associations were observed between Drug Experimentation and alcohol dependence (rg=0.79), smoking and cannabis initiation (rg=0.84 and 0.72, respectively), problematic opioid use (rg=0.76) and opioid use disorders (MVP, rg=0.70; GENOA, rg=0.75). We also observed moderate to strong associations between all impulsive subscales (except UPPS-P Perseverance) and other personality traits, such as risk-taking (rg=0.15-0.65), neuroticism (rg=-0.23-0.84), and loneliness (rg=0.17-0.54), particularly for Positive and Negative Urgency. As we anticipated, extraversion was positively associated with Sensation Seeking (rg=0.34). Externalizing psychopathology, which represents disorders and behaviors characterized by deficits in inhibition, was strongly associated with all impulsivity facets (rg=0.28-0.92), except Perseverance. We also identified positive associations with an array of psychiatric phenotypes, including ADHD (rg=0.20-0.47), depression (rg=-0.13-0.47) and anxiety (rg=-0.38-0.61) disorders, and cross-disorder (rg=0.12-0.44). The associations were again primarily significant for all except Perseverance and Sensation Seeking. Other disorders showed weaker associations (e.g., schizophrenia, rg=-0.09-0.15) or were only significantly associated with one impulsivity facet [e.g., anorexia nervosa (Perseverance, rg=-0.16); bipolar disorder (Motor, rg=0.22)]. Most impulsivity subscales were also genetically correlated with socioeconomic variables, including negative genetic associations with educational attainment (rg=-0.49 to -0.16) and income (rg=-0.38 to -0.16) and positive genetic associations with Townsend index (rg=0.18-0.58). Metabolic and medical phenotypes, such as BMI (rg=0.18-0.28), chronic pain (rg=0.22-0.46), insomnia (rg=0.20-0.42), and coronary artery disease (rg=0.18-0.30) were genetically correlated with all impulsive subscales (except Perseverance and Sensation Seeking). We also noted negative genetic associations with parental longevity (rg=-0.17 to -0.32). ### PheWAS To explore the impact of specific variants in and around *CADM2*, we performed a PheWAS using 1,291 traits and the 5 most implicated SNPs (**Figure 3**). The list of PheWAS association results using the 23andMe cohort after 5% FDR correction is available in **Tables S14** (summary), **S15** (Europeans), **S16** (Latin American) and **S17** (African Americans). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/03/2022.01.29.22270095/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2022/05/03/2022.01.29.22270095/F3) Figure 3. FDR-significant associations from *CADM2* PheWAS in individuals of European ancestry (**A**) and Latin American ancestry (**B**). No FDR-significant findings were detected in individuals of African American ancestry. The size of the dots represents the magnitude of the effect size for each trait. The effect sizes ranged from -0.14 to 0.13 in the European cohort and from -0.08 to 0.16 in the Latin American cohort. In European cohorts, *CADM2* variants had been previously identified to be significantly associated with numerous traits (**Table S18**). Most SNPs were highly correlated (R2>0.1) and tagged similar traits (**Figure S23**), but the overlap was incomplete (**Figure S24** and **Table S19**). rs993137, located in 85,449,885 bp on 3p12.1 (GRCh37/hg19), showed the highest number of associations (378), which we describe below. We replicated all previously known associations in 23andMe participants of European ancestry, identifying signals across all categories tested (**Table S15**). These included negative associations with risky behavior (e.g., lower risk for adventurousness [β=-0.05, *p*=1.33E-08], risk-taking tendencies [β=-0.02, *p*=1.13E-07]) and substance use behaviors (e.g., lower risk for alcohol consumption [β=-0.03, *p*=2.05E-09] and tobacco initiation (β=-0.02, *p*=3.66E-12; but see packs per day, β=0.01, *p*=1.05E-03), as well as negative associations with psychiatric disorders characterized by deficits in impulsivity, such as lower risk for ADHD (β=-0.05, *p*=2.17E-41). Furthermore, we found positive associations with educational outcomes (e.g., higher educational attainment (β=0.03, *p*=1.67E-12). Novel findings included positive associations with allergies (β=0.04, *p*=4.51E-03), anxiety (e.g., panic [β=0.02, *p*=6.82E-08]), and medical conditions (e.g., IBS [β=0.02, *p*=8.89E-07]), anemia (β=0.01, *p*=8.30E-74), hepatitis C (β=-0.06, p=8.36E-10). Intriguingly, we also detected positive associations with pain phenotypes (β=0.02, p=8.37E-12) and a need for a higher dose of pain medication (β=0.01, *p*=1.02E-06). For the overlapping phenotypes, UK Biobank PheWAS results (93) largely supported the 23andMe PheWAS findings (except for smoking behaviors). For example, we identified associations with dietary traits (e.g., daily fruit and vegetable intake (β=-0.01, *p*=4.23E-11), pastry frequency (β=0.01, *p*=7.36E-06), sleep quality (β=-0.01, *p*=2.53E-03), and number of pregnancies (β=-0.01, *p*=7.69E-04), among others [**Table S15**, (12)]. In the Latin American cohort, up to 47 traits were significantly associated with *CADM2* variants (**Table S16**). The highest number of associations were again observed for rs993137 (47), which are described below. Similarly, although some of the SNPs were correlated (R2>0.1; **Figure S24**), the overlap was incomplete (**Figure S26** and **Table S20**). The pattern of associations was consistent with those described in the European cohort. Again, the strongest associations were with risky behaviors, such as adventurousness (β=-0.04, *p*=1.76E-17), risk-taking (β=-0.02, *p*=5.90E-07), alcohol consumption (β=-0.03, *p*=1.41E-12), and disorders characterized by high levels of impulsivity, such as ADHD (β=-0.04, *p*=4.74E-10). The novel findings were, again, with multiple forms of allergies (e.g., seasonal allergies, β=0.03, *p*=3.0E-04), migraine (β=0.04, *p*=1.56E-04), sleep behaviors (e.g., sleep apnea, β=-0.03, *p*=6.76E-04), among others. All findings that were in common between the European and Latin American cohorts showed the same direction of effect and similar effect sizes. We did not identify FDR-significant associations in the African American cohort (**Table S17**). The effect sizes were generally extremely small (**Figures S27-28**), as is expected for a single gene and complex traits. Note that the statistical significance of the associations is, in part, a function of sample size, as evidenced by the correlation between sample size and observed *p*-values (r=-0.16, *p*=1.27E-09, European; r=-0.23, *p*=1.56E-02, Latin American). ### Mouse results **Figure 4** summarizes the mouse results across the five cohorts tested. Full statistics and additional secondary measures are described in the **Supplementary Material** and **Table S20**. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/03/2022.01.29.22270095/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2022/05/03/2022.01.29.22270095/F4) Figure 4. Complete and partial loss of *Cadm2* function and behavioral consequences in the Probability Breakpoint Ratio Task (PBRT, **A**), Probabilistic Reversal Learning Task (PRLT, **B**), Iowa gambling task (IGT, **C-D**), Behavioral Pattern Monitor task (BPM, **E-F**), Prepulse Inhibition (PPI, **G-H**), 5-Choice Serial Reaction Time Task (5CSRTT) performance (**I-M**), Locomotor (LCM) activity (**N**), Elevated Plus Maze (EPM, **O**), Light-Dark Box (LDB, **P**), and Drinking in the Dark (DID, **Q**); longitudinal body weight changes (**R**), and dendrite morphology (DSM) in the nucleus accumbens (**S**). Western blot (**WB**) analysis (**T**) of Cadm2 protein in whole brain, frontal cortex and striatum. WT, wildtype, HET, heterozygote, HOM, homozygote. * *p*<0.05, ** *p*<0.01, \***| *p*<0.001. #### Cohort 1 - Motivation, inhibition, and risk-taking behavior No differences in motivation were found between WT and HET mice during the Progressive Breakpoint task [F(1,51)=0.003, *p*=9.57E-01; **Figure 4A**]. However, we noted significant genotype differences in behavioral flexibility in the Probabilistic Reversal Learning Task, as indexed by the number of trials to first reversal [F(1,42)=4.27, *p*=4.50E-02; **Figure 4B**], and risky behavior in the IGT [F(1,51)=4.70, *p*=3.50E-02; **Figure 4C**], HET mice requiring fewer trials to reach criterion and choosing risky options less frequently than WT mice (*p*<0.05), respectively. The number of premature responses, on the contrary, were higher in HET mice [F(1,51)=5.78, *p*=2.00E-02] compared to WT mice (*p*<0.05; **Figure 4D**). In the Behavioral Pattern Monitor test, HET mice exhibited greater exploratory behavior, as shown by an increase in hole-pokes [F(1,53)= 4.88, *p*=3.20E-02; **Figure 4E**], compared to WT mice (*p*<0.05), but general levels of activity, such as distance traveled (F(1,53)= 0.42, *p*=5.21E-01; **Figure 4F**), were similar across the genotypes. Lastly, although the startle response was equal across the groups (**Figure 4G**), prepulse inhibition was larger in HET mice compared to WT mice (*p*<0.05; **Figure 4H**), particularly at ISI 25 and 100 in HET mice [F(1,53)=8.23, *p*=6.00E-03, F(1,53)=4.50, *p*=3.90E-02, respectively]. #### Cohort 2 - Motoric impulsivity The main outcome tested in cohort 2 were premature responses via the 5CSRTT (**Figure 4J-M**). Premature responses were lower in HOM (*p*<0.001) and WT (*p*<0.02) mice compared to HET mice under standard conditions (F(2,36)=8.74, *p*=8.06E-04; **Figure 4J**), and compared to both HET (*p*<0.001) and WT (*p*<0.01) mice during a long ITI session (H(2)=16.10, *p*=3.19E-04; **Figure 4L**). We also noted that HOM mice were faster at learning the 5CSRTT, requiring fewer days for adequate baseline performance (F(2,36)=7.42, *p*=2.00E-03; **Figure 4I**), compared to WT mice (*p*<0.01). #### Cohort 3 - General locomotion, anxiety-like behavior, and ethanol consumption We found a significant effect of genotype on the distance traveled in the OF [F(2,70)=7.525, *p*=1.00E-03; **Figure 4N**], with HOM mice showing higher levels of locomotor activity than WT mice (*p*=1.40E-02). As illustrated in **Figure 4O-P**, no differences in anxiety-like behavior were detected across WT, HET or HOM mice in the EPM or LDB tests (**Table S20**). The total amount of ethanol consumed during the DID paradigm did not differ between the groups ([F(2,78)=1.084, *p*=3.44E-01]; **Figure 4Q**). #### Cohort 4 - Body weight Relative to WT mice, there was a significant reduction in body weight in HOM mice from week 21 onwards (β=-3.74±1.27, *p*=4.00E-03; **Figure 4R**). The apparent trend towards a reduction in weight in HET mice was non-significant (β=-0.83±0.7, *p*=2.30E-01). Although there was a significant effect of sex (β=8.94±0.89, *p*<1.00E-03), we did not identify any sex by genotype interactions (**Supplementary Material**). #### Cohort 5 - Dendrite morphology Quantitative analyses of MSN in the NAc revealed no difference in dendritic spine density across the groups (**Figure 5S**). ## DISCUSSION In this study, we performed the largest GWAS of impulsive personality traits to date, we conducted the first multi-ancestral exploration of the role of *CADM2* on a diverse array of traits, and we created a mouse model to functionally explore the role of *Cadm2* in impulsivity and other behaviors. We identified positive genetic correlations across multiple domains, particularly substance use disorders, confirming that NIMH RDoC transdiagnostic domains (6), or endophenotypes, such as impulsive personality traits, can be used to dissect the genetic basis of psychiatric illness and normal functioning. Using mouse and human correlates, we provide further evidence that *CADM2* is a robust candidate gene for impulsivity and an important modulator of numerous other psychiatric and somatic traits. We increased the sample size of our prior GWAS of impulsivity by almost 6-fold and identified 21 genomic loci implicated in impulsive personality and Drug Experimentation. For instance, SNPs located in the gene *TCF4* were implicated in 3 subscales; this gene is also highly pleiotropic for other psychiatric conditions. Furthermore, we identified associations with *NCAM1*, which, intriguingly, is a critical member of the NTAD (*NCAM1-TTC12-ANKK1-DRD2*) gene cluster (94) and variants correlated with *NCAM1* in that cluster have been associated with differences in D2 receptor density (95). We also detected associations near *XKR6* and *AFF3*, which have been recently implicated in externalizing psychopathology (74), and *PTPRF* and *KDM4A*, recently implicated in problematic opioid use (25) and opioid use disorder (84). Although in this report we focused on *CADM2*, functional studies of those genes are warranted. Furthermore, we nominally replicate prior candidate gene studies implicating monoamine neurotransmitters in impulsivity and Drug Experimentation (*DRD2, HTR3B*). High impulsivity depends on a neural network that includes the ventral striatum (subsuming the NAc) with top-down control from prefrontal cortical regions, and is modulated by monoamine neurotransmitters including dopamine and serotonin (94); this is the first GWAS to implicate genes modulating these systems as robust candidate genes for impulsivity. We extend on prior findings (4,5) showing that the genetic architecture of impulsivity facets is distinct. Although impulsivity has been proposed as an endophenotype for ADHD and substance use disorders, individual differences across the domains. These results suggest that impulsivity facets only partially overlap, providing further support to the idea of impulsivity being a multifaceted construct even at the genetic level (5). Recent studies have implicated the *CADM2* gene in impulsivity and traits associated with reward sensitivity and multiple domains of human health. We confirmed numerous previously reported associations and extended our findings of variants related to *CADM2*. Here, *CADM2* was significantly associated with 4 out of the 9 traits that we measured in GWAS and 6 out of the 9 traits that we measured in gene-based analyses. Our PheWAS provided a comprehensive profile of human traits associated with each variant we evaluated, including hundreds of traits that had not been well studied thus far. *CADM2* variants were associated with decreased risk for externalizing psychopathology, but also increased risk for internalizing psychopathology (anxiety, depression, OCD). We also observed novel associations with migraines and various allergies. Others using a similar approach with UK Biobank data have found that this enrichment of associations is higher than expected (12) compared to other genes. Taken together, our results provide evidence that *CADM2* variants are associated with broad health outcomes, but whether this gene affects human health via disruptions in inhibition control or reward systems, or whether it acts via multiple pathways (94), is still not fully understood. A relatively unique feature of our study is that, to follow up on the *CADM2* loci implicated in human studies, we generated and phenotyped a *Cadm2* mutant mouse line. Unlike GWAS analyses, functional experiments can provide information about the causality and directionality of effects (e.g., whether higher or lower *CADM2* expression is associated with risky behavior) and whether *CADM2*, and not other nearby genes in the locus, are responsible for the reported associations. Using this approach, we found evidence that loss of *Cadm2* resulted in *less* risky behavior and *improved* information processing, consistent with prior human work (4,10,74,82,95). Mice performing the IGT generally work toward minimizing punishment as much as maximizing reward (96). Here, HET mice exhibited a greater preference for selecting the safe option vs. their WT littermates, indicative of avoidance of risk and high punishment. In contrast, HET mice showed *elevated* premature responses, which represents another form of impulsivity (motoric impulsivity). However, premature responses have also been linked to temporal discrimination, wherein mice and humans overestimating the passage of time exhibit higher premature responses (97,98). Interestingly, in the 5CSRTT, premature responding was *reduced* in HOM mice, suggesting that the type of impulsivity measured can modulate the influence of the *Cadm2* genotype. Therefore, the preference for less risky options of HET mice in the IGT could reflect their misjudgment of time – resulting in higher premature responses – and thus avoidance of higher temporal punishment in the IGT. Furthermore, hole-poking in the BPM test is thought to reflect specific exploratory behavior, whereby animals gain information about their environment. The source of the small increase in nose-poking of the HET mice is unclear, but their increased information gathering could drive their better risk preference in the IGT. In addition, HET mice exhibited better reversal learning performance in the PRL task relative to their WT littermates. This enhanced performance could be driven by their *higher* information gathering, possibly processing information. The fact that the HET mice may exhibit heightened sensitivity to temporal punishment, used in conjunction with rewards for training in the PRL task, could drive their enhanced learning in the IGT. Another important finding supporting enhanced information and speed of processing is the better performance of HET mice in the ISI (PPI) challenge, especially at the shorter time-point. PPI reflects sensory information processing over short temporal windows. HET mice exhibiting better PPI at the shortest temporal window (25 ms) supports the premise that these mice have faster temporal processing that underlies behavior. Finally, findings from the 5CSRTT provide consistent evidence that deletion of *Cadm2* may improve some facets of information processing and impulsivity while being detrimental to others. We observed that HOM mice acquired baseline task criteria faster than WT littermates, supporting that learning may be enhanced by *Cadm2* reduction. Consistent with IGT results suggesting an association between *Cadm2* and motoric impulsivity, HET mice were the most likely to commit premature responses, although HOM mice were surprisingly the least likely to make premature responses. Interestingly, although not significant, there was a consistent elevation in the number of premature responses committed by the HOM mice as the stimulus duration was reduced in the RSD (5CSRT) task, which could suggest that HOM mice, like HET mice, may show motoric impulsivity deficits when performing tasks that require greater attentional demand; this requires further investigation. Compared with WT mice, HOM mice also showed impaired accuracy performance under RSD conditions, in line with our human findings of *CADM2* association with BIS-11 Attentional subscore, and cognitive function by others (10). The heterogeneity of performance outcomes in the HOM mice (i.e., better motoric impulsivity but impaired attentional performance) further supports a unique but overlapping contribution of genetics across impulsivity domains. Although most human traits are difficult to model in mice, we have several examples where the mouse and human data are concordant. For example, we show that *CADM2* was implicated both in human impulsive personality and impulsive behavior in mice. Furthermore, our mouse data indicate that *Cadm2* was specific for impulsivity but did not have more general effects on behavior; for instance, we did not observe deficits in anxiety-like behavior or general motivation, as some of the PheWAS findings revealed. A few other measures were also inconsistent across species, particularly measures of alcohol consumption, where *CADM2* showed a role in humans (7,13,99,100) but not mice. Future studies should use different assays to determine whether *Cadm2* may be associated with other aspects of alcohol use than the one we studied via the drinking-in-the-dark paradigm. Furthermore, *CADM2* has been shown to be implicated in BMI in humans (24,101) and energy homeostasis in mice (24). We further replicate these findings by showing body weight reductions in adult mutant mice. Although changes in BMI in humans may be secondary to impulsive decision-making, it is harder to imagine that the influences are attributable to deficits in impulsivity in mice. Lastly, some measures, such as allergies and other medical conditions, do not have direct correlates and will be harder to model in mice. *CADM2* encodes the immunoglobulin adhesion protein SynCAM 2, which is part of the family of synaptic adhesion molecules known as SynCAMs. Studies have shown the large influence of SynCAMs on synaptogenesis (58,102–105), axon guidance (106), and neuron myelination (107–109), processes that have direct effects on the pathology of neurodevelopmental diseases, such as autism spectrum disorders, intellectual disability, and schizophrenia (79). *CADM2* appears to be most strongly expressed in the striatum and frontal cortex, which are core regions that regulate impulsivity; for these reasons, we measured synapse morphology in striatal regions of the brain. The lack of phenotype in terms of MSN spine density suggests that *Cadm2* may not have a role as a postsynaptic organizer of spines in MSN in the NAc, or may have redundant functions that are compensated in the mutant mice by other molecules. However, we tested mice during adulthood and may have missed a critical window of change. Based on *in-silico* analyses in humans, *CADM2* expression seems to be greater at earlier stages of development (**Figure S29**); whether *Cadm2* may affect earlier stages of development (prenatal and early postnatal) that are compensated in adulthood has not been investigated in this study. Several limitations of this study are worth noting. The discovery GWAS only includes participants of European ancestry; future studies examining these traits in other ancestries are urgently needed. Our results are also biased by potential ascertainment and characteristics of the sample; the 23andMe participant has higher socioeconomic status and lower levels of drug use and impulsivity than the general US population (110). Moreover, although the traits we studied are extracted via well-established questionnaires, they are self-reported measures, which are different from behavioral phenotypes (110,111). Similarly, PheWAS uses minimal self-reported phenotyping; however, the tradeoff is the power to survey multiple phenotypes within a single study. PheWAS analyses also have additional challenges. The first challenge of the PheWAS approach is to reliably distinguish true pleiotropic associations of a SNP (or SNPs in strong LD with the lead SNP) from unrelated associations driven by independent SNPs at a locus. A second challenge to PheWAS is the existence of common comorbidities among endpoints, or an insufficient distinction between phenotypes. Although we tested multiple variants in the *CADM2* loci, further conditional analyses are required to determine if this signal and previously reported associations implicating *CADM2* loci, including a large non-coding rare deletion in the first intron of *CADM2* (94), may tag the same underlying genetic effect. We are also unaware of the sequence of events, and whether there is true pleiotropy or mediation effects has not been examined. The analyses were well powered for moderate and large effect sizes. Still, for unclear reasons, despite similar minor allele frequencies and imputation quality of the SNPs we tested across all ancestries, we identified no significant associations in the African American cohort. Finally, although we detected some discordant cross-species effects of *Cadm2* on behavior (anxiety, acute doses of ethanol), background strain effects (104) or subtle allelic variations (vs whole KO) may explain task differences. In conclusion, we show that impulsivity facets are extremely polygenic, but of very high transdiagnostic significance. Further, we propose that genetic studies using research participants not ascertained for neuropsychiatric disorders may represent an efficient and cost-effective strategy for elucidating the genetic basis and etiology of genetically complex psychiatric diseases. Using homologous measures of impulsivity in mice and humans across three ancestral backgrounds, we provide evidence of the overarching role of *CADM2* on impulsivity, and a much broader impact on human health. Future studies are urged to further examine the role of *CADM2* on behavior at the molecular, cellular, and circuit levels. ## Supporting information Supplementary Material [[supplements/270095_file03.docx]](pending:yes) Supplementary Tables [[supplements/270095_file04.xlsx]](pending:yes) ## Data Availability We will provide summary statistics for the top 10,000 SNPs upon publication (Tables S22-30). Full GWAS summary statistics will be made available through 23andMe to qualified researchers under an agreement with 23andMe that protects the privacy of the 23andMe participants. Please visit [https://research.23andme.com/collaborate/#dataset-access/](https://research.23andme.com/collaborate/#dataset-access/) for more information and to apply to access the data. ## Data availability We will provide summary statistics for the top 10,000 SNPs upon publication (**Tables S22-30**). Full GWAS summary statistics will be made available through 23andMe to qualified researchers under an agreement with 23andMe that protects the privacy of the 23andMe participants. Please visit [https://research.23andme.com/collaborate/#dataset-access/](https://research.23andme.com/collaborate/#dataset-access/) for more information and to apply to access the data. ## Disclosures MVJ, SBB, SSR and AAP were supported by funds from the California Tobacco-Related Disease Research Program (TRDRP; Grant Number 28IR-0070 and T29KT0526). SBB was also supported by P50DA037844, SSR was also supported by NIDA DP1DA054394. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. AMBL was supported by NIH-NIAAA grant K99AA027835. TB acknowledges support by NIH/NIDA R01 DA018928. JM is supported by the Peter Boris Chair in Addictions Research. HHAT is funded through a Natural Science and Engineering Research Council PGS-D scholarship, and studies in Cohort 2 were supported by the Canadian Institutes of Health Research Project Grant (PJT-173442 to JYK). PF and SLE are employees of 23andMe, Inc., and hold stock or stock options in 23andMe. JY reports having received grant support funding from Sunovion, Heptares, and Gilgamesh, as well as honoraria from Marvel Biotech, none of which were involved in the current project. The other authors report no conflict of interest. ## Acknowledgements We would like to thank the research participants and employees of 23andMe for making this work possible. The following members of the 23andMe Research Team contributed to this study: Stella Aslibekyan, Adam Auton, Elizabeth Babalola, Robert K. Bell, Jessica Bielenberg, Katarzyna Bryc, Emily Bullis, Daniella Coker, Gabriel Cuellar Partida, Devika Dhamija, Sayantan Das, Teresa Filshtein, Kipper Fletez-Brant, Will Freyman, Karl Heilbron, Pooja M. Gandhi, Karl Heilbron, Barry Hicks, David A. Hinds, Ethan M. Jewett, Yunxuan Jiang, Katelyn Kukar, Keng-Han Lin, Maya Lowe, Jey C. McCreight, Matthew H. McIntyre, Steven J. Micheletti, Meghan E. Moreno, Joanna L. Mountain, Priyanka Nandakumar, Elizabeth S. Noblin, Jared O’Connell, Aaron A. Petrakovitz, G. David Poznik, Morgan Schumacher, Anjali J. Shastri, Janie F. Shelton, Jingchunzi Shi, Suyash Shringarpure, Vinh Tran, Joyce Y. Tung, Xin Wang, Wei Wang, Catherine H. Weldon, Peter Wilton, Alejandro Hernandez, Corinna Wong, Christophe Toukam Tchakouté. We would also like to thank The Externalizing Consortium for sharing the GWAS summary statistics of externalizing. The Externalizing Consortium: Principal Investigators: Danielle M. Dick, Philipp Koellinger, K. Paige Harden, Abraham A. Palmer. Lead Analysts: Richard Karlsson Linnér, Travis T. Mallard, Peter B. Barr, Sandra Sanchez-Roige. Significant Contributors: Irwin D. Waldman. The Externalizing Consortium has been supported by the National Institute on Alcohol Abuse and Alcoholism (R01AA015416 -administrative supplement), and the National Institute on Drug Abuse (R01DA050721). Additional funding for investigator effort has been provided by K02AA018755, U10AA008401, P50AA022537, as well as a European Research Council Consolidator Grant (647648 EdGe to Koellinger). The content is solely the responsibility of the authors and does not necessarily represent the official views of the above funding bodies. The Externalizing Consortium would like to thank the following groups for making the research possible: 23andMe, Add Health, Vanderbilt University Medical Center’s BioVU, Collaborative Study on the Genetics of Alcoholism (COGA), the Psychiatric Genomics Consortium’s Substance Use Disorders working group, UK10K Consortium, UK Biobank, and Philadelphia Neurodevelopmental Cohort. * Received January 29, 2022. * Revision received May 3, 2022. * Accepted May 3, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## REFERENCES 1. 1.Daruna JH, Barnes PA (1993): A neurodevelopmental view of impulsivity. The Impulsive Client: Theory, Research, and Treatment. Washington, D.C.: American Psychological Association, p 23. 2. 2.Coskunpinar A, Dir AL, Cyders MA (2013): Multidimensionality in Impulsivity and Alcohol Use: A Meta-Analysis Using the UPPS Model of Impulsivity. Alcohol Clin Exp Res 37: 1441–1450. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/acer.12131&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23578176&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 3. 3.Jackson JNS, MacKillop J (2016): Attention-Deficit/Hyperactivity Disorder and Monetary Delay Discounting: A Meta-Analysis of Case-Control Studies. Biol Psychiatry Cogn Neurosci Neuroimaging 1: 316–325. 4. 4.Sanchez-Roige S, Fontanillas P, Elson SL, Gray JC, de Wit H, MacKillop J, Palmer AA (2019): Genome-wide association studies of impulsive personality traits (BIS-11 and UPPSP) and drug experimentation in up to 22,861 adult research participants identify loci in the CACNA1I and CADM2 genes. J Neurosci 2662–18. 5. 5.Gustavson DE, Friedman NP, Fontanillas P, Elson SL (2020): The Latent Genetic Structure of Impulsivity and Its Relation to Internalizing Psychopathology. 11. 6. 6.Insel TR (2014): The NIMH Research Domain Criteria (RDoC) Project: Precision Medicine for Psychiatry. Am J Psychiatry 171: 395–397. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2014.14020138&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24687194&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000333501000005&link_type=ISI) 7. 7.Arends RM, Pasman JA, Verweij KJH, Derks EM, Gordon SD, Hickie I, et al. (2021): Associations between the CADM2 gene, substance use, risky sexual behavior, and self-control: A phenome-wide association study. Addict Biol 13. 8. 8.Boutwell B, Hinds D, Tielbeek J, Ong KK, Day FR, Perry JRB, et al. (2017): Replication and characterization of CADM2 and MSRA genes on human behavior. Heliyon 3: e00349. 9. 9.Lee JJ, Wedow R, Okbay A, Kong E, Maghzian O, Zacher M, et al. (2018): Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat Genet 50: 1112–1121. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0147-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30038396&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 10. 10.Ibrahim-Verbaas CA, Bressler J, Debette S, Schuur M, Smith AV, Bis JC, et al. (2016): GWAS for executive function and processing speed suggests involvement of the CADM2 gene. Mol Psychiatry 21: 189–197. 11. 11.Strawbridge RJ, Ward J, Cullen B, Tunbridge EM, Hartz S, Bierut L, et al. (2018): Genome-wide analysis of self-reported risk-taking behaviour and cross-disorder genetic correlations in the UK Biobank cohort. Transl Psychiatry 8: 39. 12. 12.Pasman J, Chen Z, Vink JM, Van Den Oever MC, Pattij T, Vries TJD, et al. (2021): The CADM2 Gene and Behavior: A Phenome-Wide Scan in UK-Biobank. medRxiv. [https://doi.org/10.1101/2021.04.16.21255141](https://doi.org/10.1101/2021.04.16.21255141) 13. 13.Sanchez-Roige S, Palmer AA, Fontanillas P, Elson SL, the 23andMe Research Team, the Substance Use Disorder Working Group of the Psychiatric Genomics Consortium, Adams MJ, et al. (2019): Genome-Wide Association Study Meta-Analysis of the Alcohol Use Disorders Identification Test (AUDIT) in Two Population-Based Cohorts. Am J Psychiatry 176: 107–118. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2018.18040369&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30336701&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 14. 14.Pasman JA, Verweij KJH, Gerring Z, Stringer S, Sanchez-Roige S, Treur JL, et al. (2018): GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal effect of schizophrenia liability. Nat Neurosci 21: 1161–1170. 15. 15.Karlsson Linnér R, Biroli P, Kong E, Meddens SFW, Wedow R, Fontana MA, et al. (2019): Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences. Nat Genet 51: 245–257. 16. 16.Karlsson Linnér R, (2021): Multivariate analysis of 1.5 million people identifies genetic associations with traits related to self-regulation and addiction. Nat Neurosci 24: 27. 17. 17.Casey JP, Magalhaes T, Conroy JM, Regan R, Shah N, Anney R, et al. (2012): A novel approach of homozygous haplotype sharing identifies candidate genes in autism spectrum disorder. Hum Genet 131: 565–579. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00439-011-1094-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21996756&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 18. 18.Demontis D, Walters RK, Martin J, Mattheisen M, Als TD, Agerbo E, et al. (2019): Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat Genet 51: 63–75. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0269-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30478444&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 19. 19.et al. (2015): Genetic studies of body mass index yield new insights for obesity biology. Nature 518: 197–206. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14177&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25673413&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 20. 20.Day FR, Helgason H, Chasman DI, Rose LM, Loh P-R, Scott RA, et al. (2016): Physical and neurobehavioral determinants of reproductive onset and success. Nat Genet 48: 617–623. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3551&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27089180&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 21. 21.Mills MC, Tropf FC, Brazel DM, van Zuydam N, Vaez A, eQTLGen Consortium, et al. (2021): Identification of 371 genetic variants for age at first sex and birth linked to externalising behaviour. Nat Hum Behav 5: 1717–1730. 22. 22.Justice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, et al. (2017): Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits. Nat Commun 8: 14977. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ncomms14977&link_type=DOI) 23. 23.Klimentidis YC, Raichlen DA, Bea J, Garcia DO, Wineinger NE, Mandarino LJ, et al. (2018): Genome-wide association study of habitual physical activity in over 377,000 UK Biobank participants identifies multiple variants including CADM2 and APOE. Int J Obes 42: 1161–1176. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41366-018-0120-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 24. 24.Yan X, Wang Z, Schmidt V, Gauert A, Willnow TE, Heinig M, Poy MN (2018): Cadm2 regulates body weight and energy homeostasis in mice. Mol Metab 8: 180–188. 25. 25.Sanchez-Roige S, Fontanillas P, Jennings MV, Bianchi S, Huang Y, Hatoum A, et al. (2021): Genome-Wide Association Study of Problematic Opioid Prescription Use in 132,113 23andMe Research Participants of European Ancestry. Mol Psych. [https://doi.org/10.1101/2021.06.03.21258260](https://doi.org/10.1101/2021.06.03.21258260) 26. 26.Sanchez-Roige S, Fontanillas P, Elson SL, Pandit A, Schmidt EM, et al. (2018): Genome-wide association study of delay discounting in 23,217 adult research participants of European ancestry. Nat Neurosci 21: 16–18. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41593-017-0032-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29230059&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 27. 27.Cyders MA, Littlefield AK, Coffey S, Karyadi KA (2014): Examination of a short English version of the UPPS-P Impulsive Behavior Scale. Addict Behav 39: 1372–1376. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.addbeh.2014.02.013&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24636739&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 28. 28.Whiteside SP, Lynam DR (2001): The Five Factor Model and impulsivity: using a structural model of personality to understand impulsivity. Personal Individ Differ 30: 669–689. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0191-8869(00)00064-7&link_type=DOI) 29. 29.Patton JH, Stanford MS, Barratt ES (1995): Factor structure of the barratt impulsiveness scale. Wiley Period Inc 51: 768–774. 30. 30.VanderBroek L, Acker J, Palmer AA, de Wit H, MacKillop J (2016): Interrelationships among parental family history of substance misuse, delay discounting, and personal substance use. Psychopharmacology (Berl) 233: 39–48. 31. 31.Durand EY, Do CB, Mountain JL, Macpherson JM (n.d.): Ancestry Composition: A Novel, Efficient Pipeline for Ancestry Deconvolution. 16. 32. 32.Henn BM, Hon L, Macpherson JM, Eriksson N, Saxonov S, Pe’er I, Mountain JL (2012): Cryptic Distant Relatives Are Common in Both Isolated and Cosmopolitan Genetic Samples. PLoS ONE 7: e34267. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0034267&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22509285&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 33. 33.Watanabe K, Taskesen E, van Bochoven A, Posthuma D (2017): Functional mapping and annotation of genetic associations with FUMA. Nat Commun 8: 1826. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-017-01261-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 34. 34.Leeuw CA de, Mooij JM, Heskes T, Posthuma D (2015): MAGMA: Generalized Gene-Set Analysis of GWAS Data. PLOS Comput Biol 11: e1004219. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pcbi.1004219&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25885710&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 35. 35.Bulik-Sullivan BK, Loh P-R, Finucane HK, Ripke S, Yang J, Patterson N, et al. (2015): LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet 47: 291–295. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3211&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25642630&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 36. 36.Verma A, Bradford Y, Dudek S, Lucas AM, Verma SS, Pendergrass SA, Ritchie MD (2018): A simulation study investigating power estimates in phenome-wide association studies. BMC Bioinformatics 19: 120. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12859-018-2135-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29618318&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 37. 37.Ehm MG, Aponte JL, Chiano MN, Yerges-Armstrong LM, Johnson T, Barker JN, et al. (2017): Phenome-wide association study using research participants’ self-reported data provides insight into the Th17 and IL-17 pathway. PLOS ONE 12: e0186405. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0186405&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29091937&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 38. 38.Lloyd KCK (2011): A knockout mouse resource for the biomedical research community: The KOMP Repository. Ann N Y Acad Sci 1245: 24–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1749-6632.2011.06311.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22211970&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000301289400009&link_type=ISI) 39. 39.de Visser L (2011): Rodent versions of the Iowa gambling task: opportunities and challenges for the understanding of decision-making. Front Neurosci 5. [https://doi.org/10.3389/fnins.2011.00109](https://doi.org/10.3389/fnins.2011.00109) 40. 40.Peña-Oliver Y, Sanchez-Roige S, Stephens DN, Ripley TL (2014): Alpha-synuclein deletion decreases motor impulsivity but does not affect risky decision making in a mouse Gambling Task. Psychopharmacology (Berl) 231: 2493–2506. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00213-013-3416-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24402137&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 41. 41.Sanchez-Roige S, Baro V, Trick L, Peña-Oliver Y, Stephens DN, Duka T (2014): Exaggerated Waiting Impulsivity Associated with Human Binge Drinking, and High Alcohol Consumption in Mice. Neuropsychopharmacology 39: 2919–2927. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npp.2014.151&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24947901&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 42. 42.Young JW, Markou A (2015): Translational Rodent Paradigms to Investigate Neuromechanisms Underlying Behaviors Relevant to Amotivation and Altered Reward Processing in Schizophrenia. Schizophr Bull 41: 1024–1034. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/schbul/sbv093&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26194891&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 43. 43.Bensadoun J-C, Brooks Simon P, Dunnett Stephen B (2004): Free operant and discrete trial performance of mice in the nine-hole box apparatus: validation using amphetamine and scopolamine. Psychopharmacology (Berl) 174. [https://doi.org/10.1007/s00213-003-1751-0](https://doi.org/10.1007/s00213-003-1751-0) 44. 44.Metha JA, Brian ML, Oberrauch S, Barnes SA, Featherby TJ, Bossaerts P, et al. (2020): Separating Probability and Reversal Learning in a Novel Probabilistic Reversal Learning Task for Mice. Front Behav Neurosci 13: 270. 45. 45.Minassian A, Henry BL, Geyer MA, Paulus MP, Young JW, Perry W (2010): The quantitative assessment of motor activity in mania and schizophrenia. J Affect Disord 120: 200–206. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jad.2009.04.018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19435640&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 46. 46.Geyer MA, Russo PV, Masten VL (1986): Multivariate assessment of locomotor behavior: Pharmacological and behavioral analyses. Pharmacol Biochem Behav 25: 277–288. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0091-3057(86)90266-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2875472&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1986D517800046&link_type=ISI) 47. 47.Young JW, van Enkhuizen J, Winstanley CA, Geyer MA (2011): Increased risk-taking behavior in dopamine transporter knockdown mice: further support for a mouse model of mania. J Psychopharmacol (Oxf) 25: 934–943. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0269881111400646&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21421642&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000292758800009&link_type=ISI) 48. 48.Varty GB, Powell SB, Lehmann-Masten V, Buell MR, Geyer MA (2006): Isolation rearing of mice induces deficits in prepulse inhibition of the startle response. Behav Brain Res 169: 162–167. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bbr.2005.11.025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16406103&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000236136800019&link_type=ISI) 49. 49.Beraldo FH, Palmer D, Memar S, Wasserman DI, Lee W-JV, Liang S, et al. (2019): MouseBytes, an open-access high-throughput pipeline and database for rodent touchscreen-based cognitive assessment. eLife 8: e49630. 50. 50.Williams R, Lim JE, Harr B, Wing C, Walters R, Distler MG, et al. (2009): A Common and Unstable Copy Number Variant Is Associated with Differences in Glo1 Expression and Anxiety-Like Behavior. PLoS ONE 4: e4649. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0004649&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19266052&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 51. 51.Distler MG, Plant LD, Sokoloff G, Hawk AJ, Aneas I, Wuenschell GE, et al. (2012): Glyoxalase 1 increases anxiety by reducing GABAA receptor agonist methylglyoxal. J Clin Invest 122: 2306–2315. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI61319&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22585572&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000304736300037&link_type=ISI) 52. 52.McMurray KMJ, D. X, Brownlee M, Palmer AA (2016): Neuronal overexpression of Glo1 or amygdalar microinjection of methylglyoxal is sufficient to regulate anxiety-like behavior in mice. Behav Brain Res 301: 119–123. 53. 53.Barkley-Levenson AM, Lagarda FA, Palmer AA (2018): Glyoxalase 1 (GLO1) Inhibition or Genetic Overexpression Does Not Alter Ethanol’s Locomotor Effects: Implications for GLO1 as a Therapeutic Target in Alcohol Use Disorders. Alcohol Clin Exp Res 42: 869–878. 54. 54.Dawson GR, Tricklebank MD (1995): Use of the elevated plus maze in the search for novel anxiolytic agents. Trends Pharmacol Sci 16: 33–36. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0165-6147(00)88973-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7762079&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QP89200001&link_type=ISI) 55. 55.Bourin M, Hascoët M (2003): The mouse light/dark box test. Eur J Pharmacol 463: 55–65. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0014-2999(03)01274-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12600702&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000181295800004&link_type=ISI) 56. 56.Kamdar NK, Miller SA, Syed YM, Bhayana R, Gupta T, Rhodes JS (2007): Acute effects of Naltrexone and GBR 12909 on ethanol drinking-in-the-dark in C57BL/6J mice. Psychopharmacology (Berl) 192: 207–217. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00213-007-0711-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17273875&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 57. 57.Dalley JW, Everitt BJ, Robbins TW (2011): Impulsivity, Compulsivity, and Top-Down Cognitive Control. Neuron 69: 680–694. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2011.01.020&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21338879&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000288413200011&link_type=ISI) 58. 58.Giza JI, Jung Y, Jeffrey RA, Neugebauer NM, Picciotto MR, Biederer T (2013): The Synaptic Adhesion Molecule SynCAM 1 Contributes to Cocaine Effects on Synapse Structure and Psychostimulant Behavior. Neuropsychopharmacology 38: 628–638. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npp.2012.226&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23169347&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 59. 59.Evangelou E, Gao H, Chu C, Ntritsos G, Blakeley P, Butts AR, et al. (2019): New alcohol-related genes suggest shared genetic mechanisms with neuropsychiatric disorders. Nat Hum Behav 3: 950–961. 60. 60.Ikeda M, Takahashi A, Kamatani Y, Momozawa Y, Saito T, Kondo K, et al. (2019): Genome-Wide Association Study Detected Novel Susceptibility Genes for Schizophrenia and Shared Trans-Populations/Diseases Genetic Effect. Schizophr Bull 45: 824–834. 61. 61.Howard DM, Adams MJ, Clarke T-K, Hafferty JD, et al. (2019): Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci 22: 343–352. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 62. 62.Yang H, Liu D, Zhao C, Feng B, Lu W, Yang X, et al. (2021): Mendelian randomization integrating GWAS and eQTL data revealed genes pleiotropically associated with major depressive disorder. Transl Psychiatry 11: 225. 63. 63.Baselmans BML, Jansen R, Ip HF, van Dongen J, et al. (2019): Multivariate genome-wide analyses of the well-being spectrum. Nat Genet 51: 445–451. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0320-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30643256&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 64. 64.Hill WD, Weiss A, Liewald DC, Davies G, Porteous DJ, Hayward C, et al. (2020): Genetic contributions to two special factors of neuroticism are associated with affluence, higher intelligence, better health, and longer life. Mol Psychiatry 25: 3034–3052. 65. 65.Teixeira JR, Szeto RA, Carvalho VMA, Muotri AR, Papes F (2021): Transcription factor 4 and its association with psychiatric disorders. Transl Psychiatry 11: 19. 66. 66.Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, et al. (2019): Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat Genet 51: 237–244. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0307-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 67. 67.Van Esch H, Colnaghi R, Freson K, Starokadomskyy P, Zankl A, Backx L, et al. (2019): Defective DNA Polymerase α-Primase Leads to X-Linked Intellectual Disability Associated with Severe Growth Retardation, Microcephaly, and Hypogonadism. Am J Hum Genet 104: 957–967. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2019.03.006&link_type=DOI) 68. 68.Christakoudi S, Evangelou E, Riboli E, Tsilidis KK (2021): GWAS of allometric body-shape indices in UK Biobank identifies loci suggesting associations with morphogenesis, organogenesis, adrenal cell renewal and cancer. Sci Rep 11: 10688. 69. 69.Pulit SL, Stoneman C, Morris AP, Wood AR, Glastonbury CA, Tyrrell J, et al. (2019): Meta-analysis of genome-wide association studies for body fat distribution in 694 649 individuals of European ancestry. Hum Mol Genet 28: 166–174. 70. 70.Nagel M, Watanabe K, Stringer S, Posthuma D, van der Sluis S (2018): Item-level analyses reveal genetic heterogeneity in neuroticism. Nat Commun 9: 905. 71. 71.Lo M-T, Hinds DA, Tung JY, Franz C, Fan C-C, Wang Y, et al. (2017): Genome-wide analyses for personality traits identify six genomic loci and show correlations with psychiatric disorders. Nat Genet 49: 152–156. 72. 72.Thorp JG, Campos AI, Grotzinger AD, Gerring ZF, An J, et al. (2021): Symptom-level modelling unravels the shared genetic architecture of anxiety and depression. Nat Hum Behav. [https://doi.org/10.1038/s41562-021-01094-9](https://doi.org/10.1038/s41562-021-01094-9) 73. 73.Feitosa MF, Kraja AT, Chasman DI, Sung YJ, Winkler TW, Ntalla I, et al. (2018): Novel genetic associations for blood pressure identified via gene-alcohol interaction in up to 570K individuals across multiple ancestries. PLOS ONE 13: e0198166. 74. 74.Karlsson Linnér R, Mallard TT, Barr PB, Sanchez-Roige S, Madole JW, Driver MN, et al. (2021): Multivariate analysis of 1.5 million people identifies genetic associations with traits related to self-regulation and addiction. Nat Neurosci. [https://doi.org/10.1038/s41593-021-00908-3](https://doi.org/10.1038/s41593-021-00908-3) 75. 75.Uhl GR, Drgon T, Liu Q-R, Johnson C, Walther D, Komiyama T, et al. (2008): Genome-Wide Association for Methamphetamine Dependence: Convergent Results From 2 Samples. Arch Gen Psychiatry 65: 345. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.65.3.345&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18316681&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253672200012&link_type=ISI) 76. 76.King CP, Militello L, Hart A, St. Pierre CL, Leung E, Versaggi CL, et al. (2017): Cdh13 and AdipoQ gene knockout alter instrumental and Pavlovian drug conditioning. Genes Brain Behav 16: 686–698. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/gbb.12382&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28387990&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 77. 77.Ruth KS, Day FR, Tyrrell J, Thompson DJ, Wood AR, et al. (2020): Using human genetics to understand the disease impacts of testosterone in men and women. Nat Med 26: 252–258. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32042192&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 78. 78.Rask-Andersen M, Karlsson T, Ek WE, Johansson Å (2019): Genome-wide association study of body fat distribution identifies adiposity loci and sex-specific genetic effects. Nat Commun 10: 339. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-018-08000-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 79. 79.Hill WD, Davies NM, Ritchie SJ, Skene NG, Bryois J, Bell S, et al. (2019): Genome-wide analysis identifies molecular systems and 149 genetic loci associated with income. Nat Commun 10: 5741. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-13585-5&link_type=DOI) 80. 80.Zhao B, Luo T, Li T, Li Y, et al. (2019): Genome-wide association analysis of 19,629 individuals identifies variants influencing regional brain volumes and refines their genetic co-architecture with cognitive and mental health traits. Nat Genet 51: 1637–1644. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-019-0516-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31676860&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 81. 81.Johnson EC, Demontis D, Thorgeirsson TE, Walters RK, Polimanti R, Hatoum AS, et al. (2020): A large-scale genome-wide association study meta-analysis of cannabis use disorder. Lancet Psychiatry 7: 1032–1045. 82. 82.Karlsson Linnér R, Biroli P, Kong E, Meddens SFW, Wedow R, Fontana MA, et al. (2019): Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences. Nat Genet 51: 245–257. 83. 83.Lam M, Hill WD, Trampush JW, Yu J, Knowles E, Davies G, et al. (2019): Pleiotropic Meta-Analysis of Cognition, Education, and Schizophrenia Differentiates Roles of Early Neurodevelopmental and Adult Synaptic Pathways. Am J Hum Genet 105: 334–350. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31374203&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 84. 84.Gaddis N, Mathur R, Marks J, Zhou L, Quach B, Waldrop A, et al. (2021): Multi-Trait Genome-Wide Association Study of Opioid Addiction: OPRM1 and Beyond. medRxiv. [https://doi.org/10.1101/2021.09.13.21263503](https://doi.org/10.1101/2021.09.13.21263503) 85. 85.Davies G, Lam M, Harris SE, Trampush JW, Luciano M, Hill WD, et al. (2018): Study of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive function. Nat Commun 9: 2098. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-018-04362-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29844566&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 86. 86.Rovira P, Demontis D, Sánchez-Mora C, Zayats T, et al. (2020): Shared genetic background between children and adults with attention deficit/hyperactivity disorder. Neuropsychopharmacology 45: 1617–1626. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41386-020-0664-5&link_type=DOI) 87. 87.Pritikin JN, Neale MC, Prom-Wormley EC, Clark SL, Verhulst B (2021): GW-SEM 2.0: Efficient, Flexible, and Accessible Multivariate GWAS. Behav Genet 51: 343–357. 88. 88.Pardiñas AF, Holmans P, Pocklington AJ, Escott-Price V, et al. (2018): Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat Genet 50: 381–389. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0059-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29483656&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 89. 89.Kichaev G, Bhatia G, Loh P-R, Gazal S, Burch K, Freund MK, et al. (2019): Leveraging Polygenic Functional Enrichment to Improve GWAS Power. Am J Hum Genet 104: 65–75. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2018.11.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 90. 90.Liu Z, Lin R, Luo M (2020): Reward Contributions to Serotonergic Functions. 24. 91. 91.MacKillop J, Weafer J, C. Gray J, Oshri A, Palmer A, de Wit H (2016): The latent structure of impulsivity: impulsive choice, impulsive action, and impulsive personality traits. Psychopharmacology (Berl) 233: 3361–3370. 92. 92.Gray JC, MacKillop J, Weafer J, Hernandez KM, Gao J, Palmer AA, de Wit H (2018): Genetic analysis of impulsive personality traits: Examination of a priori candidates and genome-wide variation. Psychiatry Res 259: 398–404. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.psychres.2017.10.047&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29120849&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 93. 93.1. X. Zhu Leppert B, Millard LAC, Riglin L, Davey Smith G, Thapar A, Tilling K, et al. (2020): A cross-disorder PRS-pheWAS of 5 major psychiatric disorders in UK Biobank (( X. Zhu, editor)). PLOS Genet 16: e1008185. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 94. 94.Collins RL, Glessner JT, Porcu E, Niestroj L-M, Ulirsch J, Kellaris G, et al. (2021): A Cross-Disorder Dosage Sensitivity Map of the Human Genome. medRxiv. [https://doi.org/10.1101/2021.01.26.21250098](https://doi.org/10.1101/2021.01.26.21250098) 95. 95.Greenbaum L, Ravona-Springer R, Livny A, Shelly S, Ganmore I, Alkelai A, et al. (2017): The CADM2 gene is associated with processing speed performance – evidence among elderly with type 2 diabetes. World J Biol Psychiatry 577–583. 96. 96.van Enkhuizen J, Henry BL, Minassian A, Perry W, Milienne-Petiot M, Higa KK, et al. (2014): Reduced Dopamine Transporter Functioning Induces High-Reward Risk-Preference Consistent with Bipolar Disorder. Neuropsychopharmacology 39: 3112–3122. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npp.2014.170&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25005251&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 97. 97.Cope ZA, Halberstadt AL, van Enkhuizen J, Flynn AD, Breier M, Swerdlow NR, et al. (2016): Premature responses in the five-choice serial reaction time task reflect rodents’ temporal strategies: evidence from no-light and pharmacological challenges. Psychopharmacology (Berl) 233: 3513–3525. 98. 98.Hsu T-Y, Lee H-C, Lane TJ, Missal M (2019): Temporal Preparation, Impulsivity and Short-Term Memory in Depression. Front Behav Neurosci 13: 258. 99. 99.Clarke T-K, Adams M, Davies G, Howard D, Hall L, Padmanabhan S, et al. (2017): Genome-wide association study of alcohol consumption and genetic overlap with other health-related traits in UK Biobank. Mol Psychiatry 9. 100.100.Kranzler HR et al. (2017): Genome-wide association study of alcohol consumption and use disorder in 274,424 individuals from multiple populations. Nat Commun 10(1):1499. 101.101.Morris J et al. (2019): Genetic variation in CADM2 as a link between psychological traits and obesity. Sci Rep 9: 7339. 102.102.Biederer T, Sara Y, Mozhayeva M, Atasoy D, Liu X, Kavalali ET, Südhof TC (2002): SynCAM, a Synaptic Adhesion Molecule That Drives Synapse Assembly. Science 297: 1525–1531. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIyOTcvNTU4Ni8xNTI1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMDMvMjAyMi4wMS4yOS4yMjI3MDA5NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 103.103.Fogel AI, Akins MR, Krupp AJ, Stagi M, Stein V, Biederer T (2007): SynCAMs Organize Synapses through Heterophilic Adhesion. J Neurosci 27: 12516–12530. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjExOiIyNy80Ni8xMjUxNiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzA1LzAzLzIwMjIuMDEuMjkuMjIyNzAwOTUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 104.104.Robbins EM, Krupp AJ, Perez de Arce K, Ghosh AK, Fogel AI, Boucard A, et al. (2010): SynCAM 1 Adhesion Dynamically Regulates Synapse Number and Impacts Plasticity and Learning. Neuron 68: 894–906. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2010.11.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21145003&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000285664900011&link_type=ISI) 105.105.Yamada A, Inoue E, Deguchi-Tawarada M, Matsui C, Togawa A, Nakatani T, et al. (2013): Necl-2/CADM1 interacts with ErbB4 and regulates its activity in GABAergic neurons. Mol Cell Neurosci 56: 234–243. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.mcn.2013.06.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23769722&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) 106.106.Niederkofler V, Baeriswyl T, Ott R, Stoeckli ET (2010): Nectin-like molecules/SynCAMs are required for post-crossing commissural axon guidance. Development 137: 427–435. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGV2ZWxvcCI7czo1OiJyZXNpZCI7czo5OiIxMzcvMy80MjciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wNS8wMy8yMDIyLjAxLjI5LjIyMjcwMDk1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 107.107.Maurel P, Einheber S, Galinska J, Thaker P, Lam I, Rubin MB, et al. (2007): Nectin-like proteins mediate axon–Schwann cell interactions along the internode and are essential for myelination. J Cell Biol 178: 861–874. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNiIjtzOjU6InJlc2lkIjtzOjk6IjE3OC81Lzg2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzA1LzAzLzIwMjIuMDEuMjkuMjIyNzAwOTUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 108.108.Spiegel I, Adamsky K, Eshed Y, Milo R, Sabanay H, Sarig-Nadir O, et al. (2007): A central role for Necl4 (SynCAM4) in Schwann cell–axon interaction and myelination. Nat Neurosci 10: 861–869. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn1915&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17558405&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F03%2F2022.01.29.22270095.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000247560200016&link_type=ISI) 109.109.Park J, Liu B, Chen T, Li H, Hu X, Gao J, et al. (2008): Disruption of Nectin-Like 1 Cell Adhesion Molecule Leads to Delayed Axonal Myelination in the CNS. J Neurosci 28: 12815–12819. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjExOiIyOC80OC8xMjgxNSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzA1LzAzLzIwMjIuMDEuMjkuMjIyNzAwOTUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 110.110.Enticott PG, Ogloff JRP, Bradshaw JL (2006): Associations between laboratory measures of executive inhibitory control and self-reported impulsivity. Personal Individ Differences 10. 111.111.Lane SD, Cherek DR, Rhoades HM, Pietras CJ, Tcheremissine OV (2003): Relationships Among Laboratory and Psychometric Measures of Impulsivity: Implications in Substance Abuse and Dependence. Addict Disord their Treat 2(2): 33–40. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00132576-200302020-00001&link_type=DOI)