16.12.2012 Views

Table of Contents - UCSD Continuing Medical Education

Table of Contents - UCSD Continuing Medical Education

Table of Contents - UCSD Continuing Medical Education

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

X V I I<br />

November 4-8, 2009<br />

World Congress <strong>of</strong><br />

PSYCHIATRIC<br />

GENETICS:<br />

Surfing the<br />

Wave <strong>of</strong><br />

Discovery<br />

Located at the Manchester Grand Hyatt in San Diego, CA


<strong>Table</strong> <strong>of</strong> <strong>Contents</strong><br />

PLENARY ABSTRACTS …………………………………………………………………………...3<br />

SYMPOSIA ABSTRACTS ………………………………………………………………………….12<br />

ORAL ABSTRACTS …………………………………………………………………………………..52<br />

POSTER ABSTRACTS<br />

ADHD …………………………………………………………………………………………..100<br />

EARLY CAREER INVESTIGATOR TRACK: ALTERNATIVE<br />

PHENOTYPES, GENE-GENE, AND GENE-ENVIRONMENT INTERACTION …...112<br />

ANIMAL MODELS ………………………………………………………………………...115<br />

ANXIETY DISORDERS ………………………………………………………………..118<br />

AUTISM …………………………………………………………………………………120<br />

EARLY CAREER INVESTIGATOR TRACK: CANDIDATE GENES …………………….124<br />

CANDIDATE GENES ………………………………………………………………………...127<br />

COGNITION …………………………………………………………………………………145<br />

DEMENTIA …………………………………………………………………………………147<br />

ENDOPHENOTYPES ………………………………………………………………………...148<br />

EPIGENETICS …………………………………………………………………………………155<br />

GENE X ENVIRONMENT INTERACTIONS ………………………………………………158<br />

GENETIC COUNSELING ………………………………………………………………..161<br />

GENETIC EPIDEMIOLOGY ………………………………………………………………..162<br />

EARLY CAREER INVESTIGATOR TRACK: GENOMEWIDE AND<br />

META-ANALYTIC APPROACHES ……………………………………………………….168<br />

GENOMICS …………………………………………………………………………………172<br />

HIGH THROUGHOUT SEQUENCING ……………………………………………………….180<br />

MOOD DISORDERS ………………………………………………………………………...180<br />

OTHER CHILDHOOD DISORDERS ……………………………………………………….199<br />

PERSONALITY AND TEMPERAMENT ………………………………………………200<br />

PHARMACOGENETICS ………………………………………………………………..205<br />

SCHIZOPHRENIA ………………………………………………………………………...214<br />

STATISTICS AND BIOINFORMATICS ………………………………………………239<br />

SUBSTANCE ABUSE ………………………………………………………………………...247<br />

LEGEND<br />

P – PLENARY<br />

S – SYMPOSIA<br />

O – ORAL<br />

ECI – EARLY CAREER INVESTIGATOR TRACK<br />

Page


PLENARY<br />

ABSTRACTS


P1.1 NEURONAL PLASTICITY AND<br />

NEURONAL DIVERSITY<br />

F. Gage* (1)<br />

1. The Salk Institute<br />

*gage@salk.edu<br />

The first part <strong>of</strong> the talk will focus on evidence supporting the<br />

birth and maturation <strong>of</strong> new neurons in the adult dentate gyrus<br />

<strong>of</strong> the hippocampus in the mammalian brain. The mechanism<br />

by which the cells integrate and become functional will be<br />

discussed. In addition, the potential functional significance for<br />

adult neurogenesis in the context <strong>of</strong> the normal function <strong>of</strong> the<br />

hippocampus will be discussed. In the second part <strong>of</strong> the talk I<br />

will focus on the recent finding that LINE-1 (Long<br />

Interspersed Nucleotide Elements-1 or L1) retroelements are<br />

active in somatic neuronal progenitor cells (NPCs) providing<br />

an additional mechanism for neuronal diversification.<br />

Together with their mutated relatives, retroelement sequences<br />

constitute 45% <strong>of</strong> the mammalian genome with L1 elements<br />

alone representing 20%. The fact that L1 can retrotranspose in<br />

a defined window <strong>of</strong> neuronal differentiation, changing the<br />

genetic information in single neurons in an arbitrary fashion,<br />

allows the brain to develop in distinctly different ways. This<br />

characteristic <strong>of</strong> variety and flexibility may contribute to the<br />

uniqueness <strong>of</strong> an individual brain. However, the molecular<br />

mechanism that regulates L1 expression in NPCs is not<br />

completely understood. L1s are likely silenced in neural stem<br />

cells due to Sox2-mediated transcription repression.<br />

Down-regulation <strong>of</strong> Sox2 accompanies chromatin<br />

modifications, such as DNA de-methylation and<br />

histone acetylation, which in turn may trigger neuronal<br />

differentiation. The characterization <strong>of</strong> somatic neuronal<br />

diversification will not only be relevant for the understanding<br />

<strong>of</strong> brain complexity and neuronal organization in mammals,<br />

but may also shed light on the differences in cognitive<br />

abilities.<br />

P1.2 GENETICS OF ALCOHOLISM<br />

G. Schumann* (1)<br />

1. Institute <strong>of</strong> Psychiatry<br />

*gunter.schumann@kcl.ac.uk<br />

Alcohol use disorders are common psychiatric disorders that<br />

exert a very high cost to the individual and to society. They<br />

are a result <strong>of</strong> the interplay <strong>of</strong> multiple behavioural, genetic<br />

and environmental factors. Addictive behaviour is<br />

characterized by phenotypic heterogeneity, epistasis and<br />

polygenicity, implying a contribution <strong>of</strong> different<br />

neurobiological mechanisms to the clinical diagnosis.<br />

Therefore, treatments for most addiction-related disorders are<br />

<strong>of</strong>ten only partially effective, with a substantial proportion <strong>of</strong><br />

patients failing to respond. To address heterogeneity and<br />

polygenicity, strategies have been developed to identify more<br />

homogeneous subgroups <strong>of</strong> patients and to characterize genes<br />

contributing to their (endo-) phenotype. In this presentation,<br />

gene identification strategies using whole genome approaches<br />

as well as translational candidate gene strategies to identify the<br />

genetic and neurobiological basis <strong>of</strong> addictive behaviour will<br />

be presented, their mechanistic function will be characterised<br />

and their association with endophenotypes relevant for<br />

addiction-related disorders will be described. Applying these<br />

strategies in a translational context aims at improving<br />

therapeutic response by the identification <strong>of</strong> subgroups <strong>of</strong><br />

addiction patients for individualized, targeted treatment<br />

strategies. Finally, a European Integrated Project "IMAGEN",<br />

which investigates reinforcement-related behaviour using<br />

multicentric gene × neuroimaging in 2000 adolescents will be<br />

presented. This project aims to integrate the methodical<br />

approaches discussed above in order to identify the genetic<br />

and neurobiological basis <strong>of</strong> behavioural traits relevant to the<br />

development <strong>of</strong> addictions.


P2 DIAGNOSTIC AND PREDICTIVE<br />

GENETIC TESTING IN PSYCHIATRY:<br />

POTENTIAL AND PITFALLS<br />

P2.1 HOW DO PATIENTS RESPOND TO RECEIPT OF<br />

RESULTS FOR A PSYCHIATRIC GENETIC TEST?<br />

P. Mitchell* (1), K. Wilhelm (1), B. Meiser (1), P. Sch<strong>of</strong>ield<br />

(2), G. Parker (1)<br />

1. University <strong>of</strong> New South Wales 2. Prince <strong>of</strong> Wales <strong>Medical</strong><br />

Research Institute<br />

*phil.mitchell@unsw.edu.au<br />

In 2006, we (Wilhelm et al) replicated the gene-environment<br />

interaction between the serotonin transporter gene and<br />

stressful life events in depression which had been reported by<br />

Caspi et al (2003). Although recent meta-analyses have failed<br />

to confirm such an interaction (Risch et al, 2009; Munafo et al,<br />

2009), our study <strong>of</strong> response <strong>of</strong> individuals to being informed<br />

<strong>of</strong> their “depression risk genotype” (Wilhelm et al, 2009), and<br />

that <strong>of</strong> Green et al (2009) in individuals at risk <strong>of</strong> Alzheimer<br />

Disease, nonetheless provide the only evidence hitherto <strong>of</strong> the<br />

response <strong>of</strong> individuals to an actual (i.e. non-hypothetical)<br />

situation <strong>of</strong> receiving results from psychiatric genetic<br />

testing. We assessed predictors <strong>of</strong> the impact <strong>of</strong> receiving<br />

individual genotype data in 128 participants in a study <strong>of</strong><br />

gene-environment interaction in depression onset. Two-thirds<br />

decided to learn their individual genotype results (receivers)<br />

and prior to disclosure this decision was associated with a<br />

perception <strong>of</strong> greater benefit from receipt <strong>of</strong> the information.<br />

Receivers completing the 2-week and 3-month follow-up<br />

generally reported feeling pleased with the information and<br />

having had a more positive experience than distress. However,<br />

distress was related to genotype, with those with the “high<br />

risk” s/s allele being most affected. Those who elected not to<br />

learn their results generally did so because <strong>of</strong> feared<br />

repercussions <strong>of</strong> being compelled to provide insurance<br />

companies or workplaces with this information. Overall,<br />

there was high interest in, and satisfaction with, learning about<br />

this putative depression risk genotype. The recent report <strong>of</strong><br />

Green et al in which children <strong>of</strong> patients with Alzheimer<br />

disease responded positively to receipt <strong>of</strong> their APOE<br />

genotype is in accordance with our findings with risk to<br />

depression.<br />

P2.2 COMMUNICATING WITH AFFECTED<br />

INDIVIDUALS AND THEIR FAMILIES<br />

ABOUT PSYCHIATRIC<br />

GENETIC TESTS<br />

J.<br />

Austin* (1)<br />

1.<br />

University <strong>of</strong> British Columbia<br />

* jehannine.austin@ubc.ca<br />

At present - and at least for the near future - the best method<br />

for predicting an individual’s risk to develop psychiatric<br />

illness is based on documenting a detailed family psychiatric<br />

history, and individualizing empiric recurrence risk data-based<br />

on the unique presenting situation. However, individuals with<br />

psychiatric disorders and their families have expressed interest<br />

in having genetic tests for psychiatric disorders as they<br />

become available. The vast majority <strong>of</strong> psychiatric genetic<br />

tests will (like the family history based method) yield<br />

probabilistic rather than definitive information about risk to<br />

develop psychiatric illness. Research shows that neither<br />

laypersons nor healthcare providers readily understand<br />

probabilistic genetic risk information. Further, in linking<br />

genetic testing with psychiatric disorders there is the potential<br />

for potentially significant negative consequences, such as<br />

increases in self-stigma, fatalism or risk taking. Facilitating<br />

accurate understanding <strong>of</strong> psychiatric genetic test results, their<br />

context and implications will be therefore, <strong>of</strong> paramount<br />

importance. Genetic counseling involves providing families<br />

affected by health conditions that have a genetic component<br />

(like psychiatric disorders) with education and support about<br />

the causes <strong>of</strong> the illness in the family, genetic testing,<br />

recurrence risks, and management. Although it has<br />

traditionally been thought <strong>of</strong> as primarily relevant to<br />

pregnancy, or to single gene disorders, the genetic counseling<br />

model is readily applied to individuals with psychiatric<br />

disorders and their families - whether or not they are actively<br />

engaged in family planning. In this presentation, the process <strong>of</strong><br />

discussing genetics and psychiatric disorders with affected<br />

families in order to promote perceptions <strong>of</strong> control over<br />

illness, decrease self-stigma, and facilitate accurate risk<br />

perception<br />

will be detailed from a practical perspective.


P2.3 ATTITUDES TOWARD GENETIC TESTING<br />

AMONG SCHIZOPHRENIA-SPECTRUM<br />

PATIENTS AND<br />

FIRST-DEGREE<br />

RELATIVES<br />

J.<br />

Hoop* (1)<br />

1.<br />

<strong>Medical</strong> College <strong>of</strong> Wisconsin<br />

* jhoop@mcw.edu<br />

Rationale: The development <strong>of</strong> clinically valid psychiatric<br />

genetic tests requires not only empirical data regarding tests’<br />

clinical utility, but also an awareness <strong>of</strong> and sensitivity to the<br />

likely psychosocial impact on those who are tested. The<br />

current study was designed to explore this issue by gathering<br />

data on the attitudes concerning genetic testing <strong>of</strong><br />

schizophrenia-spectrum patients and first-degree relatives.<br />

Methods: In-depth, face-to-face semi-structured interviews<br />

were conducted with 52 outpatients with schizophrenia or<br />

schizoaffective disorder and 50 first-degree relatives <strong>of</strong> people<br />

with these disorders. Patients also completed the Brief<br />

Symptom Inventory (BSI) measure <strong>of</strong> psychological distress.<br />

Results: Schizophrenia or schizoaffective disorder patients and<br />

family members expressed strongly positive attitudes toward<br />

pharmacogenetic and susceptibility genetic testing in a range<br />

<strong>of</strong> clinical scenarios, and expressed more interest in tests with<br />

greater physician involvement, accuracy, and predictive power<br />

(>67%). Patients with higher levels <strong>of</strong> psychological distress<br />

had significantly more positive attitudes toward testing<br />

(p


P3.1 EPIGENETIC APPROACHES TO<br />

COMMON<br />

DISEASE<br />

A.<br />

Feinberg* (1)<br />

1. Center for Epigenetics, Institute for Basic<br />

Biomedical<br />

Sciences<br />

and Department <strong>of</strong> Medicine<br />

* afeinberg@jhu.edu<br />

Epigenetics is the study <strong>of</strong> information, heritable during cell<br />

division, other than the DNA sequence itself, such as DNA<br />

methylation (DNAm), a covalent modification <strong>of</strong> cytosine.<br />

DNAm is an attractive target for study, because it is easily<br />

measured in archived samples from pre-existing large patient<br />

cohorts. It is now well established from gene-specific studies<br />

that epigenetic alterations are important in cancer, and linked<br />

to oncogene activation, tumor suppressor gene silencing, and<br />

chromosomal instability. We are taking an integrated approach<br />

to catalyze the generalization <strong>of</strong> gene-specific to genomic<br />

epigenetics, and to advance the focus in this field from cancer<br />

to neuropsychiatric disease. Doing this requires an integration<br />

<strong>of</strong> new conceptual, technological, epidemiological and<br />

statistical approaches. Population variation in epigenetic<br />

marks may be a predisposing factor for common disease,<br />

consistent with a “common disease genetic and epigenetic”<br />

(CDGE) model. A key element <strong>of</strong> our approach is the use <strong>of</strong><br />

Comprehensive High-throughput Arrays for Relative<br />

Methylation (CHARM), which examines ~4 million CpG sites<br />

throughout the genome without bias toward preconceived<br />

notions <strong>of</strong> where differential DNAm will be found. We made<br />

the surprising discovery <strong>of</strong> “CpG island shores,” regions <strong>of</strong><br />

intermediate CpG density near high-CpG “islands,” that are a<br />

major target for tissue- and disease-related epigenetic<br />

variation. We have also found population variation in the<br />

stringency <strong>of</strong> epigenetic marks that may be a predisposing<br />

factor for common disease. This work will likely provide an<br />

important added dimension to genetic studies <strong>of</strong> common<br />

disease, and may also provide insight into environmental<br />

and<br />

stochastic changes in the genome that<br />

contribute to<br />

neuropsychiatric<br />

disorders.<br />

P3.2<br />

TED REICH YOUNG INVESTIGATOR AWARD<br />

LECTURE<br />

RARE STRUCTURAL VARIANTS IN THE<br />

GENOME AND<br />

COMMON DISORDERS OF<br />

THE<br />

BRAIN<br />

J.<br />

Sebat* (1)<br />

1.<br />

Cold Spring Harbor Laboratory<br />

* sebat@cshl.edu<br />

Studies by our group and others have shown that variation in<br />

genome copy number (CNV) is a major source <strong>of</strong> genetic<br />

difference among humans and an important contributor to<br />

disease risk. Rare genomic deletions and duplications have<br />

been implicated in multiple neuropsychiatric disorders. In<br />

addition, several mutations have been identified that confer<br />

risk <strong>of</strong> more than one disorder. Thus, genetic heterogeneity is<br />

a characteristic <strong>of</strong> all neuropsychiatric disorders; conversely,<br />

phenotypic heterogeneity is a characteristic <strong>of</strong> all<br />

disease-associated mutations. Identifying and characterizing<br />

rare mutations will be essential to understanding<br />

the<br />

neurobiological<br />

pathways underlying disease.


P4 THE PSYCHIATRIC GWAS<br />

CONSORTIUM<br />

P4.1 CHALLENGES IN WORLD<br />

WIDE GENETIC<br />

ASSOCIATION<br />

STUDIES<br />

S.<br />

Ripke*<br />

* ripke@chgr.mgh.harvard.edu<br />

In this talk, we describe the development and refinement <strong>of</strong> a<br />

quality control pipeline for genome-wide association SNP<br />

data. This pipeline was developed at MGH in Boston for the<br />

Psychiatric GWAS Consortium Statistical Analysis Group.<br />

The aims <strong>of</strong> this pipeline are to conduct the quality control <strong>of</strong><br />

GWAS-SNP data, impute genotypes based on a reference<br />

sample, and combine the data for mega-analyses. The quality<br />

control steps include the capacity to identify relatives and<br />

overlapping samples in and across studies, as well as to<br />

produce principal components representing population<br />

variation to control for stratification in downstream-analysis<br />

and ancestry outlier exclusion. Through the use <strong>of</strong><br />

parallelization and massive computer clusters such as the GCC<br />

(Netherlands), preliminary results can be generated in as little<br />

as<br />

one week, even for study sizes <strong>of</strong> over 40,000 individuals.<br />

P4.2 PSYCHIATRIC GWAS CONSORTIUM:<br />

MEGA-ANALYSIS OF GWAS FOR MAJOR<br />

DEPRESSIVE DISORDER<br />

P.<br />

Sullivan* (1), Psychiatric GWAS Consortium<br />

1.<br />

UNC/Genetics<br />

* pfsulliv@med.unc.edu<br />

Major depressive disorder (MDD) is a common complex trait<br />

with enormous public health significance. As part <strong>of</strong> the<br />

Psychiatric GWAS Consortium, the MDD Working Group<br />

conducted a "mega-analysis" <strong>of</strong> individual level genotype and<br />

phenotype data from 9 genome-wide association studies <strong>of</strong><br />

MDD (approximately 12,000 cases and 12,000 controls). All<br />

cases were directly interviewed using a structured diagnostic<br />

instrument and met DSM-IV criteria for MDD. All controls<br />

were population-based and had never met criteria for MDD.<br />

Data files were uploaded to a cluster computer in the<br />

Netherlands and all samples run through a common quality<br />

control, imputation, and analysis pipeline. Careful attention<br />

was paid to investigating and addressing bias (particularly<br />

inflation <strong>of</strong> type 1 error due to population stratification). An<br />

interim mega-analysis <strong>of</strong> five samples has been completed,<br />

and the remaining four samples are scheduled for completion<br />

in September 2009. Final results are to be presented<br />

for the<br />

first<br />

time at the WCPG PGC plenary session.


P4.3 COMBINED "FREEZE 1"<br />

SCHIZOPHRENIA-GWAS ANALYSIS<br />

Psychiatric<br />

GWAS Consortium<br />

Due to the very small genetic effects in schizophrenia (SZ)<br />

and the large number <strong>of</strong> common variants in the human<br />

genome, the detection <strong>of</strong> susceptibility loci from genome-wide<br />

association studies (GWAS) requires large individual sample<br />

sizes. Moreover, combining datasets with available GWAS<br />

can substantially improve statistical power. Our goal is to<br />

perform a combined analysis <strong>of</strong> all available SZ GWAS data<br />

sets to detect new associations and to test for confirmation <strong>of</strong><br />

previously published ones. To be included in the analysis,<br />

each sample met stringent criteria for quality <strong>of</strong> phenotypic<br />

data and genotyping. The overall study information and<br />

quality, errors, missing data, population structure, and<br />

intra- and inter-study inflationary factors were assessed.<br />

After<br />

QC exclusions (including sample overlapping and<br />

population outliers), the combined dataset consisted <strong>of</strong> 10<br />

GWAS studies which included 17 independently collected<br />

clinical samples from Europe, North America, and Australia<br />

with a grand total <strong>of</strong> >12,200 cases and >9,300 controls<br />

<strong>of</strong><br />

European ancestry (Freeze 1). The primary affected<br />

phenotype was defined as either SZ or schizoaffective<br />

disorder. The association analysis is to be implemented<br />

according to a set <strong>of</strong> a priori decisions following the<br />

QC phase. A set <strong>of</strong> exploratory phenotypic analyses is also<br />

planned. The Freeze 1 sample includes the large majority <strong>of</strong><br />

the SZ case<br />

samples already studied by GWAS <strong>of</strong> the entire<br />

world.<br />

P4.4 META-ANALYSIS OF GENOME-WIDE<br />

ASSOCIATION STUDIES OF ATTENTION DEFICIT<br />

HYPERACTIVITY DISORDER<br />

B. Neale* (1), Psychiatric GWAS Consortium<br />

1. MGH<br />

*bmneale@gmail.com<br />

We present the first round <strong>of</strong> meta-analysis for Attention<br />

Deficit/Hyperactivity Disorder (ADHD) as conducted by the<br />

ADHD subgroup <strong>of</strong> the Psychiatric GWAS Consortium<br />

(PGC). The meta-analysis for ADHD is comprised <strong>of</strong> six<br />

different core genome-wide association studies: International<br />

ADHD Multisite Genetics (IMAGE) projects I, IMAGE II,<br />

PUMWa, CHOP, McGough, Eli Lilly for a total sample<br />

size <strong>of</strong> ~2,000 trios and 1,500 cases. These cases have been<br />

matched to shared control resources ensuring protection<br />

against population stratification through identity-by-state<br />

matching. For each <strong>of</strong> these datasets, we have applied a<br />

standardized quality control pipeline to ensure that the level <strong>of</strong><br />

cleaning. These studies have been completed on genome-wide<br />

arrays including Perlegen 600K platform, Affymetrix 5.0,<br />

Illumina 317K, and Illumina 610K, and so we performed<br />

imputation on each <strong>of</strong> these studies individually to facilitate<br />

the meta-analysis. We combined the test statistics for each <strong>of</strong><br />

these studies weighted by the effective sample size (as defined<br />

by power to detect a modest association) and the quality <strong>of</strong> the<br />

imputation. We present the top findings <strong>of</strong> this effort as well<br />

as describe the what sort <strong>of</strong> effects we are unlikely to be<br />

present for ADHD. In addition to the SNP results, we present<br />

the results from interrogating copy number variation across a<br />

subset <strong>of</strong> these datasets.


P4.5 THE PSYCHIATRIC GWAS CONSORTIUM FOR<br />

BIPOLAR DISORDER: SIGNIFICANT ASSOCIATION<br />

FOR MULTIPLE GENES<br />

J. Kelsoe* (1), The Psychiatric GWAS Consortium<br />

1. University <strong>of</strong> California, San Diego Dept. <strong>of</strong> Psychiatry<br />

*jkelsoe@ucsd.edu<br />

Early genomewide association studies <strong>of</strong> bipolar disorder were<br />

striking for their relative paucity <strong>of</strong> genomewide significant<br />

genes. This has suggested that common variants that<br />

predispose to bipolar disorder must be <strong>of</strong> small effect sizes<br />

and that very large samples would be necessary. The PGC-BD<br />

was formed in order to achieve such large sample sizes<br />

through collaboration. Eleven international sites have<br />

combined data from their GWAS studies to produce a<br />

combined sample <strong>of</strong> 6,806 Caucasian cases and 7,952<br />

controls. Analyses were conducted using PLINK and<br />

BEAGLE. SNPs were imputed to HapMap2, to integrate<br />

across the multiple genotyping platforms. After covarying for<br />

site, population stratification and R2, a genomic control<br />

lambda <strong>of</strong> 1.12 was obtained. Four regions achieved nominal<br />

genomewide significance (p


P4.7 CROSS-DISORDER WORKGROUP: TAKING<br />

DNA BEYOND DSM<br />

J. Smoller* (1), Cross Disorder Group<br />

1. Massachusetts General Hospital<br />

*jsmoller@hms.harvard.edu<br />

Over the past 25 years, genetic epidemiologic studies have<br />

provided compelling evidence for shared genetic influences on<br />

a range <strong>of</strong> psychiatric disorders. In addition, the common<br />

comorbidity <strong>of</strong> different diagnoses supports the hypothesis <strong>of</strong><br />

substantial overlap in the underlying biology <strong>of</strong><br />

clinically-defined syndromes. The vast scope <strong>of</strong> the PGC<br />

provides unprecedented power to examine and dissect the<br />

shared heritability underlying psychiatric disorders using<br />

genomewide data. The PGC’s Cross Disorder Group (CDG)<br />

was established to pursue this aim by coordinating and<br />

conducting meta-analyses to identify convincing<br />

genotype-phenotype associations that overlap or transcend<br />

traditional diagnostic phenotypes. In this presentation, we<br />

will discuss the scientific rationale for cross-disorder analyses,<br />

insights from genetic studies in other areas <strong>of</strong> medicine, and<br />

proposed analytic approaches. Cross-disorder analyses may<br />

answer fundamental questions about the genetic basis<br />

<strong>of</strong><br />

psychopathology and provide a crucial resource for<br />

developing an etiology-based nosology for psychiatry.


SYMPOSIA<br />

ABSTRACTS


S1 ANTIDEPRESSANT<br />

PHARMACOGENETICS: GWAS AND<br />

BEYOND<br />

S1.1 COMPARATIVE PHARMACOGENOMICS OF<br />

SEROTONERGIC AND NORADRENERGIC<br />

ANTIDEPRESSANTS: A GENOME-WIDE ANALYSIS<br />

OF THE GENDEP SAMPLE<br />

R. Uher* (1), N. Perroud (1), M. Ng (1), M. Rietschel (2), W.<br />

Maier (3), O. Mors (4), J. Hauser (5), N. Henigsberg (6), B.<br />

Jerman (7), D. Souery (8), A. Placentino (9), P. Muglia (10),<br />

A. Butler (1), S. Cohen-Woods (1), K. Aitchison (1), A.<br />

Farmer (1), I. Craig (1), C. Lewis (1), P. McGuffin (1)<br />

1. King’s College London 2. Central Institute <strong>of</strong> Mental<br />

Health, Mannheim 3. University <strong>of</strong> Bonn 4. Aarhus University<br />

Hospital 5. Poznan University <strong>of</strong> <strong>Medical</strong> Sciences 6.<br />

University <strong>of</strong> Zagreb 7. Jozef Stefan Institute, Ljubljana 8.<br />

Université Libre de Bruxelles and Psy Pluriel 9. Centro San<br />

Giovanni di Dio, FBF, Brescia 10. GlaxoSmithKline<br />

*rudolf.uher@kcl.ac.uk<br />

The substantial individual variability in response to<br />

antidepressants appears to be partially genetically determined,<br />

but functional candidate genes have little predictive power.<br />

We report a genome-wide association study with a continuous<br />

trait <strong>of</strong> antidepressant response, indexed as percentage<br />

improvement on a depression severity scale over 12 weeks <strong>of</strong><br />

treatment with either a serotonin-reuptake-blocking<br />

antidepressant escitalopram or a predominantly<br />

norepinephrine-reuptake-blocking antidepressant nortriptyline.<br />

High quality Illumina Human 610-quad chip genotyping was<br />

available for 706 unrelated participants <strong>of</strong> European ancestry<br />

treated with escitalopram (n=394) or nortriptyline (n=312) in<br />

the Genome-based Therapeutic Drugs for Depression<br />

(GENDEP) project. Two regions containing common copy<br />

number polymorphisms were associated with outcome <strong>of</strong><br />

treatment with either antidepressant at suggestive levels <strong>of</strong><br />

significance (p=3.8x10-7 and p=7.4x10-7). A marker in a<br />

neurogenesis-related gene was associated with response to<br />

nortriptyline at a genome-wide level <strong>of</strong> significance<br />

(p=3.6x10-8) and a predicted functional marker in a gene<br />

involved in immune response was the best predictor <strong>of</strong><br />

response to escitalopram (p=2.8x10-6). While limited<br />

statistical power means that a number <strong>of</strong> true associations may<br />

have been missed, these results suggest that efficacy <strong>of</strong><br />

antidepressants may be predicted by genetic markers other<br />

than traditional candidates. If replicated, these findings may be<br />

among the first steps on a genome-wide avenue towards<br />

personalized medicine for depression.<br />

S1.2 A GENOME-WIDE ASSOCIATION STUDY OF<br />

CITALOPRAM RESPONSE IN THE STAR*D SAMPLE<br />

S. Hamilton* (1), H. Garriock (1), J. Kraft (1), S. Shyn (1), E.<br />

Peters (1), J. Yokoyama (1), G. Jenkins (2), M. Reinalda (2),<br />

Slager (2), P. McGrath<br />

1. UCSF 2. Mayo Clinic<br />

*steve.hamilton@ucsf.edu<br />

Antidepressant response is likely influenced by genetic<br />

constitution, but the actual genes involved have yet to be<br />

determined. We have carried out a genome-wide association<br />

study to determine if common DNA variation influences<br />

antidepressant response. Our sample is derived from Level 1<br />

participants in the Sequenced Treatment Alternatives to<br />

Relieve Depression (STAR*D) study, all treated with<br />

citalopram. Association for the response phenotype included<br />

883 responders and 608 non- responders. For the remission<br />

phenotype, 743 subjects that achieved remission were<br />

compared to 608 non-responders. We used a subset <strong>of</strong> 430K<br />

SNPs from the Affymetrix 500K and 5.0 Human SNP Arrays,<br />

and association analysis was carried out after correcting for<br />

population stratification. We identified three SNPs associated<br />

with response with p-values less than 1 x 10-5 near the<br />

UBE3C gene, another 100kb away from BMP7, and a third<br />

that is intronic in the RORA gene. These same SNPs were<br />

also associated with remission. Thirty-nine additional SNPs<br />

are <strong>of</strong> interest with p-values ≤ 0.0001 for the response and<br />

remission phenotypes. Although the findings reported here do<br />

not meet a genome-wide threshold for significance, the<br />

regions identified from this study provide targets for<br />

independent replication and novel pathways to investigate<br />

mechanisms <strong>of</strong> antidepressant response. This study was not<br />

placebo controlled, making it possible that we are also<br />

observing associations to non-specific aspects <strong>of</strong> drug<br />

treatment <strong>of</strong> depression.


S1.3 FIVE YEAR FOLLOW-UP IN THE KAISER<br />

PERMANENTE GENES AND RESPONSE TO<br />

ANTIDEPRESSANTS (GRAD) STUDY<br />

C. Schaefer* (1), L. Shen (1), V. Reus (2), C. Quesenberry<br />

(1)<br />

1. Kaiser Permanente Division <strong>of</strong> Research 2. Department <strong>of</strong><br />

Psychiatry University <strong>of</strong> California San Francisco<br />

*cathy.schaefer@kp.org<br />

The GRAD study is a prospective cohort study <strong>of</strong> genetic and<br />

non-genetic factors associated with response to antidepressant<br />

treatment in an ethnically diverse cohort <strong>of</strong> 1,004 adults with<br />

major depression. Standard assessments <strong>of</strong> therapeutic<br />

response and side effects were completed at initiation, 4<br />

weeks, and 8 weeks after participants began pharmacotherapy<br />

with fluoxetine or paroxetine. After 8 weeks, 70% had<br />

responded with a 50% reduction in Hamilton Depression Scale<br />

scores. Approximately five years following the acute<br />

treatment phase <strong>of</strong> the study, 764 (76%) participants were<br />

re-interviewed and electronic medical records were abstracted<br />

to determine long term response to antidepressant treatment,<br />

including residual symptoms, episode recurrence, treatment<br />

resistance, and response to alternative medications. The<br />

follow-up study included the same proportion <strong>of</strong> responders<br />

(70%) and nonresponders (30%) as the acute treatment phase.<br />

Analyses <strong>of</strong> factors associated with therapeutic response after<br />

8 weeks, as well as with outcomes <strong>of</strong> treatment after five<br />

years, have been conducted for nongenetic factors and for<br />

variants in candidate genes related to current<br />

pharmacodynamic and pharmacokinetic hypotheses<br />

concerning response to SSRI antidepressants. A genome-wide<br />

association study is planned. This longitudinal investigation<br />

<strong>of</strong> the cohort’s subsequent syptomatology, episode recurrence,<br />

treatment adherence, and response to alternative drugs will<br />

serve to define a new set <strong>of</strong> phenotypic variables that more<br />

accurately reflect disease heterogeneity and may better relate<br />

to underlying genetic variation than variables derived from<br />

observations made during acute treatment alone.<br />

S1.4 A GENOMEWIDE ASSOCIATION STUDY<br />

POINTS TO MULTIPLE LOCI THAT PREDICT<br />

ANTIDEPRESSANT DRUG TREATMENT OUTCOME<br />

IN DEPRESSION<br />

S. Lucae* (1), M. Ising (1), E. Binder (1), T. Bettecken (1),<br />

M. Uhr (1), S. Ripke (1), M. Kohli (1), J. Hennings (1), S.<br />

Horstmann (1), S. Kloiber (1), A. Menke (1), B. Bondy (2), R.<br />

Rupprecht (2), K. Domschke (3), A. Rush (4), F. Holsboer (1),<br />

B. Mueller-Myhsok (1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry, Munich, Germany<br />

2. Department <strong>of</strong> Psychiatry, Ludwig Maximilians University,<br />

Munich, Germany 3. Department <strong>of</strong> Psychiatry, Westfalian<br />

Wilhelms University, Muenster, Germany 4. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> Texas, Southwestern <strong>Medical</strong><br />

Center, Dallas, USA<br />

*lucae@mpipsykl.mpg.de<br />

The efficacy <strong>of</strong> antidepressant drug treatment is<br />

unsatisfactory; 1 in 3 patients does not fully recover even after<br />

several treatment trials. It was the aim <strong>of</strong> this study to identify<br />

genetic and clinical determinants <strong>of</strong> antidepressant drug<br />

treatment. We performed a genomewide pharmacogenetic<br />

association study in 339 depressed inpatients from the Munich<br />

Antidepressant Response Signature (MARS) project and in<br />

361 pooled DNAs from an independent German replication<br />

study. A set <strong>of</strong> 328 SNPs highly related to outcome in both<br />

studies was genotyped in 832 samples <strong>of</strong> the STAR*D study.<br />

We generated a multilocus genetic variable that described the<br />

individual number <strong>of</strong> alleles <strong>of</strong> the selected SNPs with<br />

beneficial treatment outcome in the MARS sample<br />

(“response” alleles) to evaluate additive genetic effects on<br />

antidepressant drug treatment outcome. Multilocus analysis<br />

revealed a significant contribution <strong>of</strong> a binary variable that<br />

categorized patients as carriers <strong>of</strong> a high vs. low number <strong>of</strong><br />

response alleles in the prediction <strong>of</strong> antidepressant treatment<br />

outcome in both samples (MARS and STAR*D). In addition,<br />

we observed that patients with a comorbid anxiety disorder<br />

combined with a low number <strong>of</strong> response alleles showed the<br />

least favourable outcome. These results demonstrate the<br />

importance <strong>of</strong> multiple genetic factors combined with clinical<br />

features in the prediction <strong>of</strong> antidepressant drug treatment<br />

outcome, which underscores the multifactorial nature <strong>of</strong> this<br />

trait.


S1.5 TOWARDS POPULATION-BASED<br />

ANTIDEPRESSANT PHARMACOGENOMICS<br />

R. Perlis* (1)<br />

1. Massachusetts General Hospital<br />

*rperlis@partners.org<br />

Recent genome-wide association studies in psychiatric<br />

disorders strongly suggest that very large cohorts will be<br />

required to detect the many common variants <strong>of</strong> modest<br />

effects which are likely to underlie psychiatric phenotypes.<br />

Individual large clinical trials may be insufficient to detect or<br />

replicate such variants, much less determine their clinical<br />

validity. An alternative approach makes use <strong>of</strong> large electronic<br />

medical records databases to identify subjects with the<br />

phenotypes <strong>of</strong> interest and obtain blood for DNA collection<br />

from them. Strategies to validate the outcomes <strong>of</strong> interest in<br />

these data sets and address sources <strong>of</strong> heterogeneity have been<br />

developed, allowing a novel cohort <strong>of</strong> 3,000 subjects treated<br />

for MDD to be collected. Such population-based approaches<br />

<strong>of</strong>fer a feasible, low-cost means <strong>of</strong> identifying genetic and<br />

clinical predictors <strong>of</strong> treatment response for psychiatric<br />

disorders.<br />

S2 DNA METHYLATION IN HUMAN BRAIN<br />

AND NEUROPSYCHIATRIC DISEASES<br />

S2.1 COMPREHENSIVE DNA METHYLATION<br />

ANALYSIS IN NEURONS AND NON-NEURONS FROM<br />

HUMAN POSTMORTEM BRAINS AND ITS<br />

APPLICATION TO MENTAL DISORDERS<br />

T. Kato* (1), K. Iwamoto (1)<br />

1. RIKEN Brain Science Institute<br />

*kato@brain.riken.jp<br />

The role <strong>of</strong> DNA methylation in the pathophysiology <strong>of</strong><br />

mental disorders has not been well established yet, in spite <strong>of</strong><br />

recent studies using a candidate gene approach or a<br />

comprehensive DNA methylation analysis in the brain. The<br />

brain is a heterogeneous tissue, and there may be substantial<br />

differences <strong>of</strong> DNA methylation status between cell types.<br />

Thus, we should clarify the DNA methylation difference<br />

between neurons and glial cells to search for the genes<br />

differentially methylated in the neurons <strong>of</strong> patients with<br />

mental disorders. For this purpose, we are analyzing the DNA<br />

methylation differences between neuronal and non-neuronal<br />

nuclei. After separating neuronal nuclei from frozen human<br />

brains using anti-NeuN antibody and fluorescent cell sorter,<br />

we performed comprehensive DNA methylation analysis<br />

using promoter tiling arrays and site-specific methylation<br />

analysis <strong>of</strong> about 1500 CpG sites using Illumina Golden Gate<br />

assays. Global methylation level was also assessed by a<br />

LUMA assay. Neuronal nuclei tended to be hypomethylated<br />

compared with non-neuronal nuclei, and methylation patterns<br />

<strong>of</strong> bulk brain tissue were more similar to the patterns in<br />

non-neuronal nuclei compared with neuronal nuclei. Genes<br />

commonly methylated both in neuronal and non-neuronal<br />

nuclei included genes related to cell-cycle. Neuronal- and<br />

non-neuronal nuclei showed distinctive DNA methylation<br />

patterns, compatible with their functional differences.<br />

Application <strong>of</strong> this method to patients with mental disorders<br />

will provide a clue to understand the role <strong>of</strong> epigenetics in<br />

mental disorders.


S2.2 DNA METHYLATION SIGNATURES WITHIN<br />

THE MAJOR DEPRESSIVE DISORDER BRAIN<br />

S. Sabunciyan (1), M. Aryee (1), R. Irizarry (1), M. Webster<br />

(4), R. Yolken (1), A. Feinberg (1), J. Potash* (1)<br />

1. Johns Hopkins School <strong>of</strong> Medicine 2. Uniformed Services<br />

University<br />

*jpotash@jhmi.edu<br />

Major depressive disorder (MDD) is influenced by genetic<br />

factors, but heritability has been estimated at 37%.<br />

Genome-wide association studies are ongoing to identify<br />

sequence variation that influences MDD susceptibility.<br />

Epigenetic marks, such as DNA methylation, which can be<br />

influenced by the environment, may also play a role in MDD<br />

etiopathogenesis. Here we present a genome-wide DNA<br />

methylation scan in MDD. We compared 39 postmortem<br />

MDD brain samples and 27 control brain samples, all<br />

prefrontal cortex, obtained from the Stanley <strong>Medical</strong> Research<br />

Institute. DNA from these samples was hybridized to our<br />

Comprehensive High-throughput Arrays for Relative<br />

Methylation (CHARM) platform. This 2.1 million probe<br />

platform assays 140,000 genomic GC-rich regions with an<br />

average <strong>of</strong> 15 probes per region. Smoothing is performed<br />

across each region to derive a single methylation value per<br />

region. Follow-up is being done using bisulfite<br />

pyrosequencing to validate results. Gene expression levels will<br />

be assayed by real-time RT-PCR for correlation with<br />

methylation patterns.<br />

S2.3 QTLS FOR GENE-SPECIFIC METHYLATION IN<br />

HUMAN BRAIN<br />

D. Zhang* (1), L. Cheng (1), J. Badner (1), C. Chen (1), D.<br />

Craig (2), M. Redman (2), E. Gershon (1), C. Liu (1)<br />

1. The University <strong>of</strong> Chicago 2. The Translational Genomic<br />

Research Institute<br />

*dandanz@uchicago.edu<br />

We have observed extensive inter-individual differences in<br />

DNA methylation at specific CpG sites in human adult<br />

cerebellum. To study genetic factors that might affect this<br />

variation we studied CpG sites <strong>of</strong> 14,495 genes, almost all<br />

from promoter regions. We performed a genome-wide single<br />

nucleotide polymorphism (SNP)-based association mapping<br />

for methylation Quantitative Trait Loci (mQTLs) in 153 adult<br />

cerebellum samples. Cis-association refers to methylation<br />

correlation with SNPs within 1 megabase <strong>of</strong> a CpG site. 1,923<br />

SNP-CpG pairs showed cis-association that was region-wide<br />

significant by permutation. Of these SNP-CpG pairs, 629 were<br />

also phenotype-wide significant (after additional permutation<br />

corrections for all the tested methylation phenotypes).<br />

Methylation level was negatively correlated with gene<br />

expression in the cerebellum for 14 <strong>of</strong> 15 genes with<br />

significant cis-association (FDR q value


S2.4 REGULATION AND FUNCTION OF CPG<br />

METHYLATION AND DNA METHYLTRANSFERASES<br />

IN POST-MITOTIC NEURONS<br />

R. Sharma* (1)<br />

1. University <strong>of</strong> Illinois at Chicago<br />

*rsharma@psych.uic.edu<br />

We know the domain structure, substrate preference,<br />

protein-protein interactions, and cellular expression pr<strong>of</strong>iles<br />

(including brain) <strong>of</strong> the major DNA methyltransferases. We<br />

also now know that DNA methyltransferases are demonstrably<br />

albeit variably expressed in the adult brain, and that global<br />

CpG methylation <strong>of</strong> brain DNA is abundant, dynamic,<br />

modular and possibly disease related. We also know that in<br />

individual gene studies, promoter CpG methylation can be<br />

modified in response to neuronal depolarization, behavioral<br />

and cognitive experimentation, and pharmacological<br />

treatments. And yet, remarkably little is known about the<br />

regulation/function <strong>of</strong> either CpG methylation or DNA<br />

methyltransferases in the post-mitotic neurons as in the<br />

developing or adult brain. We have approached this question<br />

by attempting to identify the cis-regulatory sequences<br />

upstream <strong>of</strong> the transcription start site (TSS) in postmitotic<br />

primary neuron cultures as a best approximation to their<br />

in-vivo regulation in brain neurons. In these studies, we are<br />

able to demonstrate a vigorous promoter activity with DNA<br />

protein interactions in the immediate region surrounding the<br />

TSS. Parallel studies have included a DNMT1 knockdown<br />

strategy in neurons, where we can demonstrate a role for DNA<br />

methytransferases in the regulation <strong>of</strong> candidate gene<br />

promoter methylation. Also, neuronal depolarization as a<br />

model <strong>of</strong> neuronal activity causes a decrease in DNMT1 and<br />

DNMT3a mRNA expression. The accumulating evidence<br />

suggests that the function and regulation <strong>of</strong> CpG methylation<br />

and DNA methyltransferases in neurons is partly unique to<br />

these post-mitotic cells.<br />

S2.5 CORRELATIONS BETWEEN GENE<br />

EXPRESSION AND DNA METHYLATION IN HUMAN<br />

BRAIN<br />

C. Chen* (1), D. Zhang (1), L. Cheng (1), J. Badner (1), E.<br />

Gershon (1, 2), C. Liu (1)<br />

1. Department <strong>of</strong> Psychiatry, The University <strong>of</strong> Chicago,<br />

Chicago, IL 60637 2. Department <strong>of</strong> Human Genetics, The<br />

University <strong>of</strong> Chicago, Chicago, IL 60637<br />

*chenchaor@gmail.com<br />

DNA methylation plays a critical role in the regulation <strong>of</strong> gene<br />

expression. Here, we studied the correlation <strong>of</strong> DNA<br />

methylation and gene expression at the genome level in the<br />

human brain. The methylation status <strong>of</strong> 27,578 CpG sites from<br />

14,495 genes in 153 cerebellum samples was measured in our<br />

lab using Illumina HumanMethylation27 BeadChips.<br />

Cerebellum gene expression data for 45 <strong>of</strong> these individuals<br />

were obtained from the Stanley <strong>Medical</strong> Research Institute<br />

Online Genomics Database, having been analyzed with<br />

Affymetrix Genechip Human Genome U95 Set. After<br />

eliminating covariate and batch effects, and excluding low<br />

quality data, we identified the CpG sites that were within 10kb<br />

<strong>of</strong> the transcription start site (TSS) <strong>of</strong> a gene measured on the<br />

expression array. Five hundred fifty-three CpG sites were<br />

located within 10 kb <strong>of</strong> one or more TSSs, resulting in 625<br />

CpG site-expression pairs for correlation analysis.<br />

Seventy-five nominally significant correlations were observed.<br />

Twenty-four <strong>of</strong> them remained significant after multiple test<br />

correction (FDR, q ≤ 0.05). Twenty <strong>of</strong> the 24 (83.3%)<br />

significant correlations were negative, as expected.<br />

Interestingly, at four CpG sites (16.7%), increased DNA<br />

methylation was associated with increased gene expression.<br />

We did not observe a significant correlation between gene<br />

expression and DNA methylation for imprinted genes.


S3 RISK VARIANTS THROUGH BIOLOGY<br />

TO TREATMENT: MAPPING THE<br />

CHALLENGES AND THE POTENTIAL<br />

S3.1 DELINEATING THE EFFECT ON SZ OF GWAS<br />

IDENTIFIED RISK ALLELES BASED ON<br />

NEUROPSYCHOLOGICAL PERFORMANCE.<br />

G. Donohoe* (1), J. Walters (2), D. Morris (1), L. Kaladjieva<br />

(3), A. Jablensky (3), M. Gill (1), M. O'Donovan (1), D.<br />

Rujescu (4), M. Owen (2), A. Corvin (1)<br />

1. Trinity College Dublin 2. Cardiff University 3. University<br />

<strong>of</strong> Western Australia 4. Munich University<br />

*donoghug@tcd.ie<br />

Recent SZ genome wide association studies have led to the<br />

identification <strong>of</strong> several new risk loci for the disorder. The<br />

biological role <strong>of</strong> these variants is largely unknown, making<br />

an understanding <strong>of</strong> the neural mechanisms they control a<br />

priority. One approach to this question is the cognitive<br />

intermediate or endophenotype approach. This was originally<br />

proposed as a method <strong>of</strong> gene discovery but increasingly is<br />

used as a means <strong>of</strong> identifying the neural mechanisms by<br />

which risk is conferred. The endophenotype strategy in<br />

psychiatric genetics is based on a number <strong>of</strong> critical (and<br />

criticised) assumptions, including the unproven assumption<br />

that it is located on the risk pathway between illness<br />

phenotype and genotype, thus acting to either mediate or<br />

modify risk. If this is unlikely for all risk variants, then<br />

distinguishing between those variants whose risk is<br />

cognitively mediated from variants that are not will be<br />

informative about the neural mechanism underlying this risk.<br />

Demonstrating its utility, we will present data on the<br />

application <strong>of</strong> this approach to first allele to have received<br />

genome wide support for psychosis: The Zinc Finger Protein<br />

804A gene (ZNF804A), which is brain-expressed but <strong>of</strong><br />

unknown function. We tested for association between<br />

ZNF804A rs1344706 and cognitive functions known to be<br />

impaired in schizophrenia (IQ, episodic memory, working<br />

memory, and attentional control) in an Irish discovery sample.<br />

We then tested significant results in a German replication<br />

sample. We observed an interaction between ZNF804A<br />

genotype and diagnosis for measures <strong>of</strong> episodic and working<br />

memory in the Irish patient sample but not controls. These<br />

findings replicated in the same direction in the German<br />

sample. Furthermore, in both samples the association between<br />

ZNF804A and schizophrenia strengthened when patients with<br />

lower general cognitive function were excluded. We conclude<br />

from these data that in a disorder characterized by<br />

heterogeneity, a risk variant at ZNF804A appears to delineate<br />

a patient subgroup characterized by relatively spared cognitive<br />

ability. Further work is required to establish if this represents a<br />

discrete molecular pathogenesis that differs from other patient<br />

groups and whether this also has consequences for nosology,<br />

illness course or treatment.<br />

S3.2 THE EFFECTS OF RISK VARIANTS ON BRAIN<br />

STRUCTURE, FUNCTION AND CONNECTIVITY<br />

A. Macintosh<br />

S3.3 MUTANT MOUSE MODELS:<br />

PSYCHOPATHOLOGY AND TRANSLATIONAL<br />

PSYCHOPHARMACOLOGY<br />

J. Waddington* (1), C. O'Tuathaigh (1)<br />

1. Royal College <strong>of</strong> Surgeons in Ireland<br />

*jwadding@rcsi.ie<br />

In the face <strong>of</strong> the complexity <strong>of</strong> psychiatric disorders, the<br />

etiology <strong>of</strong> which likely involves a combination <strong>of</strong> genetic and<br />

environmental factors, a number <strong>of</strong> key technologies are<br />

converging: for example, identification <strong>of</strong> putative<br />

susceptibility genes through GWAS and CNV studies leads to<br />

investigation <strong>of</strong> the behavioural roles <strong>of</strong> these genes by<br />

targeted manipulation in mice and their phenotypic<br />

characterisation (‘gene-driven’ approach); in a complementary<br />

manner, identification <strong>of</strong> putative pathophysiological<br />

processes and indicative therapeutic pathways leads to<br />

investigation <strong>of</strong> behavioural phenotype in mice mutant for<br />

genes regulating such processes and pathways<br />

(‘phenotype-driven’ approach). There are many challenges in<br />

mutant phenotyping as it relates to models <strong>of</strong><br />

psychopathology, underlying brain mechanisms and<br />

psychopharmacological pr<strong>of</strong>iling, and in translating findings<br />

in mutants to the identification <strong>of</strong> novel therapeutic targets.<br />

Recent studies relating to psychosis particularly illustrate these<br />

issues. Gene x environment and gene x gene interactions<br />

constitute additional problems. Despite rapid advances over<br />

the past several years, it is clear that we continue to face<br />

substantive challenges in applying mutant models to better<br />

understand the genetics <strong>of</strong> psychopathology. However, the<br />

breadth and depth <strong>of</strong> ongoing studies holds the prospect<br />

<strong>of</strong> progress. The authors' studies are supported by Science<br />

Foundation Ireland and the Health Research Board.


S3.4 SCHIZOPHRENIA PATHOGENESIS: INSIGHTS<br />

FROM ETIOLOGICALLY VALID MOUSE MODELS<br />

OF RARE DISEASE VARIANTS<br />

J. Gogos* (1)<br />

1. Columbia University<br />

*jag90@columbia.edu<br />

Genetic variation influences the risk for and manifestation <strong>of</strong><br />

mental illnesses. In light <strong>of</strong> recent genetic advances,<br />

psychiatric genetics is currently at a crossroads concerning<br />

how to efficiently translate genetic findings into meaningful<br />

insights for disease prevention and treatment. Teasing apart<br />

the contribution <strong>of</strong> genetic variants to neural development and<br />

function, and ultimately disease risk, necessitates the use <strong>of</strong><br />

model systems. In order for mutant animal models to uncover<br />

a genuine pathogenetic link between a candidate gene and<br />

disease risk, the association <strong>of</strong> the genetic variant must be<br />

unequivocal, its functional effect known and the risk allele<br />

modeled accurately. Studies assessing rare variants have<br />

identified specific, causative and highly penetrant<br />

schizophrenia-associated alleles. These rare alleles are far<br />

more likely to have deleterious functional consequences than<br />

common alleles due to clear effects on gene products. As a<br />

result they can be faithfully modeled in mice and have higher<br />

potential for revealing genuine insights into disease<br />

pathogenesis. In this talk we review results from the study <strong>of</strong><br />

mouse models <strong>of</strong> rare schizophrenia-associated structural<br />

variants generated and characterized in our lab. We found that<br />

rare disease-associated variants result in specific impairments<br />

in cognition and brain connectivity and identified some <strong>of</strong> the<br />

molecular and cellular/synaptic processes underlying these<br />

impairments. The latter can serve as substrates for novel drug<br />

development efforts.<br />

S3.5 ESTABLISHMENT OF A METHOD FOR<br />

HIGH-THROUGHPUT FUNCTIONAL SCREENING OF<br />

PATIENT REGULATORY & CODING VARIANTS IN<br />

MICE<br />

J. Schmouth (1, 2), R. Bonaguro (1), C. de Leeuw (1, 3), L.<br />

Dreolini (4), R. Holt (4, 5, 6), E. Simpson* (1, 3, 5)<br />

1. Centre for Molecular Medicine and Therapeutics at the<br />

Child & Family Research Institute, University <strong>of</strong> British<br />

Columbia, Vancouver, Canada 2. Genetics Graduate Program,<br />

University <strong>of</strong> British Columbia, Vancouver, Canada 3.<br />

Department <strong>of</strong> <strong>Medical</strong> Genetics, University <strong>of</strong> British<br />

Columbia, Vancouver, Canada 4. Canada's Michael Smith<br />

Genome Sciences Centre, British Columbia Cancer Agency,<br />

Vancouver, Canada 5. Department <strong>of</strong> Psychiatry, University<br />

<strong>of</strong> British Columbia, Vancouver, Canada 6. Department <strong>of</strong><br />

Molecular Biology and Biochemistry, Simon Fraser<br />

University, Burnaby, Canada<br />

*simpson@cmmt.ubc.ca<br />

Large scale association studies and deep sequencing are<br />

generating huge numbers <strong>of</strong> human variants for which the<br />

functional significance is unknown. Traditionally, the mouse<br />

is the in vivo model in which to assay candidate human<br />

mutations. However, standard mouse approaches do not<br />

effectively address the challenges <strong>of</strong> this dataset: many <strong>of</strong> the<br />

mutations may be regulatory, not coding; variant-phenotypic<br />

effect may be small; and most variants will not be mutations.<br />

We propose an approach that addresses these issues. A human<br />

BAC, >100 kb including the gene <strong>of</strong> interest and variant<br />

region, is “recombineered” to carry the candidate variant. The<br />

BAC is also retr<strong>of</strong>itted for homologous recombination at a<br />

locus 5' <strong>of</strong> the Hprt1 gene in ESCs, and mice derived. Since<br />

each variant will be studied without copy number or site <strong>of</strong><br />

insertion variation, direct functional comparisons can be made.<br />

Because the human allele is not located at the homologous<br />

mouse locus, the variant can be tested in the presence <strong>of</strong> two,<br />

one, or no mouse alleles. The latter, in which brain<br />

development and function are entirely dependent upon the<br />

human allele, may best reveal subtle Wt versus variant<br />

differences. Currently in testing are variants <strong>of</strong> NR2E1; a gene<br />

critical for neural stem cell function and “associated” with<br />

bipolar disorder. Germline animals carrying the Wt human<br />

allele have been obtained and variants are in chimeras and<br />

ESCs. Thus, we will test the functional significance <strong>of</strong> human<br />

NR2E1 variants and provide “pro<strong>of</strong> in principle” for this<br />

approach to test other gene variants.


S3.6 PROMOTION OF SYNAPTIC AND CIRCUIT<br />

MATURATION PARTIALLY REVERSES SYMPTOMS<br />

IN A MOUSE MODEL OF RETT SYNDROME<br />

D. Tropea* (1, 2), E. Giacometti (3), N. Wilson (2), R.<br />

Iaenisch (3), M. Sur (2)<br />

1. Trinity College Dublin 2. MIT 3. Whitehead Institute<br />

*tropea@mit.edu<br />

Rett Syndrome (RTT) is a neurodevelopmental disorder<br />

caused by mutations in the gene coding for methyl<br />

CpG-binding protein 2 (MECP2). We examined the<br />

hypothesis that MeCP2 deficiency leads to a deficit in the<br />

maturation <strong>of</strong> synapses and circuits in the cerebral cortex <strong>of</strong><br />

MeCP2 null mice. Consistent with the hypothesis, we found<br />

reductions in synaptic amplitudes measured by patch clamp<br />

recording from motor cortex neurons, reductions in the<br />

postsynaptic density protein PSD95 in motor cortex measured<br />

with immunohistochemistry, and reductions in the density <strong>of</strong><br />

spines on motor cortex neurons. An in vivo assay <strong>of</strong> synapse<br />

maturation and developmental plasticity in visual cortex<br />

revealed that synapses had abnormally prolonged plasticity in<br />

MeCP2 null mice. Microarray analyses have shown that the<br />

Insulin-like Growth Factor 1 (IGF-1) signaling pathway has a<br />

key role in the stabilization and maturation <strong>of</strong> cortical<br />

synapses. Thus, we reasoned that upregulation <strong>of</strong> the IGF-1<br />

pathway would reverse symptoms in MeCP2 null mice.<br />

Systemic treatment <strong>of</strong> young MeCP2 mutant mice with an<br />

active fragment <strong>of</strong> IGF-1 increased synaptic amplitudes,<br />

upregulated cortical PSD95 and stabilized cortical plasticity to<br />

wild-type levels. In addition, the treatment extended the life<br />

span <strong>of</strong> the mutant mice, partially restored regularity in heart<br />

rate and breathing, and improved locomotor function. Our<br />

results suggest that truncated or full-length IGF-1 are<br />

promising candidates for pharmacological treatment <strong>of</strong> Rett<br />

Syndrome, and potentially <strong>of</strong> other CNS disorders caused by<br />

delayed synaptic maturation. A similar approach can be used<br />

for studying the molecular mechanisms and potential therapies<br />

<strong>of</strong> other neurodevelopmental disorders, such as Autism<br />

Spectrum Disorders (ASD) and schizophrenia.<br />

S4 GENOME-WIDE ASSOCIATION STUDIES<br />

ON MAJOR DEPRESSION: WHAT’S NEXT?<br />

S4.1 GENOME-WIDE ASSOCIATION STUDIES IN<br />

MAJOR DEPRESSION. THE ROLE OF<br />

ENDOPHENOTYPES<br />

W. Hoogendijk* (1), P. Sullivan (2), Z. Bochdanovits (3), M.<br />

Bevova (3), S. Woudstra (4), B. Penninx (4), P. Heutink (3)<br />

1. Dept. Psychiatry, VU University <strong>Medical</strong> Center 2.<br />

Department <strong>of</strong> Genetics, Psychiatry, & Epidemiology, UNC<br />

Chapel Hill, Department <strong>of</strong> Genetics, USA 3. Section <strong>Medical</strong><br />

Genomics, Department <strong>of</strong> Clinical Genetics, VU <strong>Medical</strong><br />

Center, Amsterdam, the Netherlands 4. Department <strong>of</strong><br />

Psychiatry, Neuroscience Campus VU medical center<br />

Amsterdam, the Netherlands<br />

*w.hoogendijk@ggzingeest.nl<br />

According to the WHO, depression will be the second leading<br />

cause <strong>of</strong> disability worldwide by 2020. Diagnostic accuracy<br />

and prediction <strong>of</strong> treatment-outcome is limited by subjective<br />

phenotypic assessments and would strongly benefit from<br />

biological endophenotypes, together with<br />

genome-wide genotyping. Gene-expression <strong>of</strong> peripheral<br />

blood cells, measured as RNA concentrations, is the<br />

endophenotype that is most proximal to the genotype. The<br />

endophenotype that is most proximal to the brain disorder<br />

is brain structure and function, assessed<br />

using (functional) magnetic resonance imaging (fMRI).<br />

Recent data from our campus in twins concordant or<br />

discordant for depression and anxiety have indicated that<br />

abnormal responses to emotional stimuli are associated<br />

primarily with genetic factors, whereas hippocampal atrophy<br />

was observed only in discordant twins and is therefore likely<br />

to be due to environmental factors. Several studies have tried<br />

to associate individual candidate genes with neuroimaging<br />

data as endophenotype. However, results are inconclusive,<br />

especially concerning depression. Recently, a genome wide<br />

association study (GWAS) for MDD was performed on<br />

subjects from Netherlands study on depression and anxiety<br />

(NESDA) and the Netherlands twin register (N= 1738 cases<br />

and 1802 controls). Eleven SNPs within the top 200 smallest<br />

p-values appeared to be within to the presynaptic piccolo<br />

(PCLO) gene (Sullivan et al., Mol. Psychiatry 2009). Here, we<br />

report on preliminary results on the association <strong>of</strong> these PCLO<br />

SNPs with endophenotypes like brain morphology and<br />

activity in depressed subjects compared to healthy controls.


S4.2 GENOME-WIDE ANALYSIS IN MAJOR<br />

DEPRESSION - AND NOW?<br />

B. Müller-Myhsok* (1)<br />

1. MPI Psychiatry, Munich<br />

*bmm@mpipsykl.mpg.de<br />

Major Depression is a common disorder with a heritable<br />

component clearly established. However, conventional<br />

association studies, including GWAs, have shown<br />

comparatively little success. This apparent lack <strong>of</strong> success<br />

may be related to an overly simplistic underlying model. Thus<br />

a refined model or additional data might be necessary. These<br />

additional aspects include gene by gene, gene by environment<br />

interaction, rare variants, especially CNVs and animal models.<br />

In the second part <strong>of</strong> my talk I will focus on gene by<br />

environment interaction.<br />

S4.3 MICE THEN HUMANS: A COMPLEMENTARY<br />

APPROACH TO DISSECTING THE BIOLOGY OF<br />

MDD<br />

P. Sullivan* (1)<br />

1. UNC/Genetics<br />

*pfsulliv@med.unc.edu<br />

It is possible that the etiological underpinnings <strong>of</strong> major<br />

depressive disorder (MDD) are more intricate than for other<br />

complex traits. The phenotype may blend into normalcy,<br />

etiological heterogeneity could be marked, genetic effects may<br />

be very subtle, and gene-environment interactions may be<br />

particularly salient. If true, then sample size requirements in<br />

humans for unbiased, genome-wide studied are prohibitively<br />

large. In this talk, I will describe an alternative approach. With<br />

Drs Lisa Tarantino, Fernando Pardo-Manuel de Villena and<br />

UNC colleagues, we have recently received a NHGRI center<br />

<strong>of</strong> excellence award to attempt to dissect MDD-like behaviors<br />

in mouse. This work is made possible due to the development<br />

<strong>of</strong> a new systems biology platform, the Collaborative Cross,<br />

along with advances in mouse genomics (e,g. a new<br />

Affymetrix mouse GWAS array) and dense phenotyping.<br />

Central to our study is an experimental manipulation to assess<br />

the impact <strong>of</strong> stressful environments and gene-environment<br />

interactions on MDD-like behaviors. The fundamental idea is<br />

to screen the vast number <strong>of</strong> possible genetic and<br />

gene-environment effects to derive a far smaller number <strong>of</strong><br />

models. High probability models can then be tested in human<br />

samples. This approach will yield a detailed understanding <strong>of</strong><br />

MDD-like mouse behavior, and the applicability <strong>of</strong> these<br />

models can then be tested in humans.<br />

S4.4 GENOME-WIDE PREDICTION OF FUNCTIONAL<br />

GENE-GENE INTERACTIONS FOR DEPRESSION<br />

Z. Bochdanovits* (1)<br />

1. VU <strong>Medical</strong> Center<br />

*z.bochdanovits@vumc.nl<br />

The contribution <strong>of</strong> individual risk factors to complex<br />

disorders is modest and current strategies aimed at identifying<br />

such susceptibility genes in very large case-control studies are<br />

still thought to be underpowered. One biologically obvious<br />

reason why susceptibility genes would be difficult to identify<br />

based on searching for their main effects alone is that in reality<br />

they might work together in a non-additive fashion, i.e. the<br />

risk is actually conveyed by specific combinations <strong>of</strong> such<br />

factors. Although this possibility is widely acknowledged, a<br />

full screen for epistatic effects on a genome-wide scale is<br />

unfeasible because statistical power would be seriously<br />

compromised by correcting for multiple testing. One possible<br />

way to avoid this problem is to ascertain pairs <strong>of</strong> variants that<br />

a priori are more likely to be involved in functional gene-gene<br />

interactions and test only these for association with a complex<br />

phenotype. Classical population genetic theory predicts that<br />

linkage disequilibrium (LD) between interacting loci will<br />

emerge if the phenotype affected by the combination <strong>of</strong> loci is<br />

under selection. Consequently, ascertainment <strong>of</strong> gene-pairs<br />

based on deviation from two-locus equilibrium genotype<br />

frequency could predict functional interactions. We applied<br />

this approach to multiple genome-wide datasets and propose<br />

gene-pairs that are potentially involved in conveying<br />

susceptibility to depression.


S4.5 DISCOVERY AND TESTING OF DELETIONS<br />

FOR ASSOCIATION WITH MDD IN GAIN STUDY<br />

M. Bevova*(1), Z. Bochdanovits (1), E. de Geus (2), G.<br />

Willemsen (2), B. Penninx (2), D. Boomsma<br />

(2), W. Hoogendijk (2), P. Heutink (1)<br />

1. VUMC 2. VU<br />

*m.bevova@vumc.nl<br />

GWAS on major depressive disorder (MDD) had been<br />

performed in the frame <strong>of</strong> six initial Genetic Information<br />

Network studies. This study was conducted in GAIN-MDD<br />

cohort comprised <strong>of</strong> 1738 MDD cases and 1802 controls<br />

genotyped for around 435 000 SNPs using a Perlegen array.<br />

There are different methods <strong>of</strong> discovering CNVs in<br />

genome-wide data sets. Most methods based on intensity<br />

differences between SNP alleles allowing detection <strong>of</strong> CNV<br />

gains and losses. A chemistry <strong>of</strong> the Perlegen platform<br />

however, do not allow to distinguish intensity differences and<br />

therefore CNV gains, but we still have the opportunity to<br />

identify CNV losses (deletions). Several approaches based on<br />

violation <strong>of</strong> Hardy–Weinberg or Mendelian transmission as<br />

well as on non random distribution <strong>of</strong> null genotypes enable to<br />

identify deletions from genotype data only. We developed an<br />

algorithm there we first used 30 trios from GAIN-MDD cohort<br />

to discover deletions using MICRODEL s<strong>of</strong>tware and then we<br />

use runs <strong>of</strong> homozygosity and stretches <strong>of</strong> missing genotypes<br />

in the GAIN MDD unrelated cohort as a footprint for possible<br />

deletions in genotyping data as defined by adjusted settings <strong>of</strong><br />

PLINK s<strong>of</strong>tware. Detected runs <strong>of</strong> homozygosity and missing<br />

genotypes for each individual will be compared with the CNV<br />

dataset obtained from trio data. Individuals having either runs<br />

<strong>of</strong> homozygosity or missing genotypes overlapping with<br />

deletion identified by MICRODEL s<strong>of</strong>tware in trios are<br />

considered as having a deletion in this region. For the<br />

identified deletions with frequencies >1% association studies<br />

with MDD as a main outcome has been performed.<br />

S5 NEW INSIGHTS FROM STUDIES OF<br />

RARE STRUCTURAL VARIATION IN<br />

PSYCHIATRIC DISORDERS<br />

S5.1 SCHIZOPHRENIA: A COMMON DISEASE DUE<br />

TO MULTIPLE RARE ALLELES?<br />

M. King (1), J. McClellan (1)<br />

1. University <strong>of</strong> Washington<br />

Objectives: To review the role <strong>of</strong> rare structural variants in<br />

persons with schizophrenia. Methods: We screened<br />

genome-wide for genomic deletions and duplications in 150<br />

individuals with schizophrenia and 268 ancestry-matched<br />

healthy controls, using array CGH technology. We focused on<br />

rare mutations <strong>of</strong> size >100kb that disrupted genes, that were<br />

only detected in cases or in controls, and that were not<br />

reported in the Database <strong>of</strong> Genomic Variants (DGV). Results:<br />

Individuals with schizophrenia were significantly more likely<br />

to harbor rare deletions and duplications mutations than<br />

healthy controls (15% vs 5%, P = 0.0008). This risk was even<br />

higher in patients with onset <strong>of</strong> illness by age 18 years <strong>of</strong> age<br />

(20% vs 5%, P = 0.0001). Each affected individual harbored a<br />

different mutation that impacted different genes. Mutations in<br />

cases disproportionally disrupted genes involved in<br />

neurodevelopment, including neuregulin and glutamate<br />

pathways (Walsh et al., Science 2008). Independent groups<br />

have since confirmed and extended these findings (Xu et al.,<br />

2008; Stefansson et al., 2008; International Schizophrenia<br />

Consortium, 2008; Need et al., 2009). Our present work<br />

extends these experiments to rare small structural variants and<br />

to rare point mutations. Conclusions: Individuals with<br />

schizophrenia are more likely than healthy persons to harbor<br />

rare structural events that delete, duplicate, or disrupt genes.<br />

These results suggest that a substantial portion <strong>of</strong><br />

schizophrenia arises from rare mutations, either de novo or<br />

very recent in origin. This degree <strong>of</strong> genetic heterogeneity has<br />

important implications for genetic and treatment research.


S5.2 DELETIONS IN NRXN1 IN SCHIZOPHRENIA<br />

G. Kirov* (1), D. Rujescu (2), A. Ingason (3), D. Collier (4),<br />

M. O'Donovan (1), M. Owen (1)<br />

1. MRC Centre for Neuropsychiatric Genetics and Genomics,<br />

School <strong>of</strong> Medicine, Cardiff University, Heath Park, Cardiff,<br />

CF14 4XN, United Kingdom 2. Division <strong>of</strong> Molecular and<br />

Clinical Neurobiology, Dept. <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Munich, Germany 3. Research Institute <strong>of</strong> Biological<br />

Psychiatry, Mental Health Center Sct. Hans, Copenhagen<br />

University Hospital, Roskilde, Denmark 4. Social, Genetic and<br />

Developmental Psychiatry Centre, Institute <strong>of</strong> Psychiatry,<br />

King's College, London, UK<br />

*kirov@cardiff.ac.uk<br />

Several studies have implicated deletions in NRXN1 in the<br />

aetiology <strong>of</strong> schizophrenia. Similar findings are also available<br />

in the literature on autism. We have conducted a joint analysis<br />

on all available studies on CNVs in schizophrenia, in order to<br />

assess the significance <strong>of</strong> these reports. At the time <strong>of</strong> writing<br />

there are 7 studies that have published data on<br />

non-overlapping samples. The total number <strong>of</strong> subjects studied<br />

have been 7,935 patients with schizophrenia, and 41,234<br />

controls. We re-analysed all available data for CNVs >100kb,<br />

in order to make all studies comparable. We find 15 deletions<br />

in cases (0.19%) and 17 in controls (0.04%). This corresponds<br />

to a Fisher Exact Test p=0.000047; OR 4,59; 95% CI<br />

2,29-9.17. The results are very similar for those CNVs that<br />

disrupt exons (12 in cases and 9 in controls). We conclude that<br />

the association for NRXN1 deletions in schizophrenia is<br />

strongly supported by the evidence. However, more samples<br />

should be analysed, and higher-resolution platforms should be<br />

used in future studies, in order to confirm the findings and<br />

identify which deletions are pathogenic.<br />

S5.3 VERY RARE CNVS ASSOCIATED WITH<br />

BIPOLAR DISORDER<br />

D. Zhang (1), C. Liu (1), E. Gershon* (1)<br />

1. University <strong>of</strong> Chicago<br />

*egershon@yoda.bsd.uchicago.edu<br />

With the paucity <strong>of</strong> significant associations from GWAS<br />

analyses <strong>of</strong> Schizophrenia and Bipolar disorder, the<br />

underlying common-variant association model has been called<br />

into question. Rare variants such as Copy Number Variations<br />

(CNVs) could account for some <strong>of</strong> the missing variance<br />

(difference between heritability in twin studies and variance<br />

accounted for by GWAS). Comparative intensity <strong>of</strong><br />

hybridization (or similar) signal at each locus studied with<br />

GWAS chips can be used to detect CNVs. The size <strong>of</strong> CNVs<br />

that are reliably detectable is limited because <strong>of</strong> the average 3<br />

kb distance between markers. There are batch effects and<br />

other artifacts <strong>of</strong> microarray data, and the s<strong>of</strong>tware used for<br />

analyzing intensity data (CEL files in the Affymetrix system)<br />

and calling CNVs varies in accuracy. Bipolar disorder: GWAS<br />

data was analyzed to detect CNVs in 1001 cases and 1034<br />

controls from the Bipolar Genome Study (BiGS), using the<br />

Affymetrix single nucleotide polymorphism (SNP) 6.0<br />

platform. Plate-wise normalization was done by the<br />

Affymetrix Power Tool plug-in to Birdsuite 1.5.2. Very rare<br />

CNVs (singletons; occur once in a data set) were significantly<br />

more frequent in Bipolar patients than in controls (Zhang et<br />

al., 2009). Quantitative PCR (qPCR) was used to validate<br />

CNVs. These data are consistent with numerous rare variants<br />

contributing to a very heterogeneous disorder. Multiple<br />

s<strong>of</strong>tware packages exist for analyzing CEL files and for<br />

calling CNVs. These include Birdsuite, HelixTree (Golden<br />

Helix, Inc.), Partek, and PennCNV-Affymetrix. For singleton<br />

deletions, confirmation by qPCR was most consistent for<br />

Birdsuite and Partek, and least consistent for HelixTree.


S5.4 FROM CNV “HOT SPOTS” TO THE REST OF<br />

THE GENOME, LOOKING BEYOND THE LOW<br />

LYING FRUIT<br />

S. Jonathan* (1)<br />

1. Cold Spring Harbor Laboratory<br />

*sebat@cshl.edu<br />

Recent studies have established an important role for rare<br />

structural variants in the etiology <strong>of</strong> schizophrenia and other<br />

psychiatric disorders. Studies <strong>of</strong> rare variants have so far been<br />

limited to relatively large (>100 kb) copy number variants<br />

(CNVs). However, large CNVs account for a very small<br />

fraction <strong>of</strong> all variants and presumably account for only a<br />

subset <strong>of</strong> risk alleles. In order to further enhance the power <strong>of</strong><br />

the CNV-based approach, new computational and<br />

experimental strategies must be considered. Promising new<br />

computational approaches to the analysis <strong>of</strong> CNVs include<br />

gene-based and pathway-based association methods. We will<br />

present preliminary findings from these studies, and we will<br />

address the specific technical challenges that apply to CNV<br />

data. New experimental methods that are in development for<br />

highly-sensitive detection <strong>of</strong> CNVs include new microarray<br />

CGH and Next Generation sequencing platforms. We will<br />

address how these methods can be applied to genetic studies<br />

<strong>of</strong> psychiatric disease.<br />

S5.5 ANALYSIS OF RARE STRUCTURAL VARIANTS<br />

IN BIPOLAR DISORDER AND SCHIZOPHRENIA<br />

S. Purcell* (1)<br />

1. Mass General<br />

*shaun@pngu.mgh.harvard.edu<br />

We and others have previously reported evidence that<br />

implicates rare, large deletions and duplications as strong risk<br />

factors for schizophrenia, both at specific loci and in terms <strong>of</strong><br />

genome-wide burdens. In this talk, I consider a number <strong>of</strong><br />

related questions: 1) the role <strong>of</strong> structural variants in bipolar<br />

disorder, 2) an analysis <strong>of</strong> the intersection <strong>of</strong> regions emerging<br />

from association analysis <strong>of</strong> common single nucleotide<br />

polymorphism data with rare structural variant data, and 3)<br />

methodological issues in testing for enrichment <strong>of</strong> variants in<br />

affected individuals occurring in particular sets <strong>of</strong> genes.<br />

S6 PSYCHIATRIC DISORDER<br />

BIOMARKERS<br />

S6.1 THE QUEST FOR PSYCHIATRIC DISORDER<br />

BIOMARKERS: FROM DIFFERENTIAL EXPRESSION<br />

TO ISOFORMS TO PATHWAYS<br />

C. Turck* (1), R. Landgraf (1), M. Filiou (1), Y. Zhang (1),<br />

C. Webh<strong>of</strong>er (1), G. Maccarrone (1), C. Ditzen (1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry<br />

*turck@mpipsykl.mpg.de<br />

Proteomic technologies in combination with pathway analysis<br />

promise to be <strong>of</strong> great value in molecular medicine,<br />

particularly in the discovery and validation <strong>of</strong> disease markers.<br />

The research <strong>of</strong> the “Proteomics and Biomarkers” group is<br />

aimed at the identification <strong>of</strong> markers that can categorize<br />

subsets <strong>of</strong> psychiatric patients in a more consistent manner<br />

than is presently achievable. This will allow a more precise<br />

definition and categorization <strong>of</strong> affective disorders and in turn<br />

facilitate investigations <strong>of</strong> the pathogenesis <strong>of</strong> the diseases and<br />

enhance the ability for treatment. Biomarker detection efforts<br />

range from classical proteomics approaches such as<br />

quantitative mass spectrometry <strong>of</strong> brain tissue and body fluid<br />

proteins to phage display screens with cerebrospinal fluid<br />

antibodies. A particular focus is the use <strong>of</strong> animal models that<br />

represent selected endophenotypes characteristic for the<br />

respective clinical phenotype in humans. A comprehensive<br />

and sensitive proteomics platform that is based on metabolic<br />

labeling <strong>of</strong> mouse models with stable isotopes is used for mass<br />

spectrometry quantitation and disease relevant pathway<br />

discovery. In a complementary approach human specimens<br />

from patient groups that have been characterized according to<br />

their clinical as well as endophenotypes are analyzed. An<br />

antibody array platform serves to interrogate a great number <strong>of</strong><br />

proteins in human cerebrospinal fluids with the aim to extract<br />

patterns <strong>of</strong> biomarkers that can be used for clinical diagnosis.


S6.2 BIOMARKER DISCOVERY IN SCHIZOPHRENIA<br />

AND BIPOLAR DISORDER: TRANSCRIPTOMIC,<br />

SPLICEOMIC, AND PATHWAY ANALYSIS FINDINGS<br />

S. Glatt (1), C. Bousman (2), G. Chana (2), S. Chandler (2),<br />

W. Kremen (2), I. Everall (2)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Sciences, and<br />

<strong>Medical</strong> Genetics Research Center; SUNY Upstate <strong>Medical</strong><br />

University; 750 East Adams Street; Syracuse, NY, 13210;<br />

U.S.A. 2. Center for Behavioral Genomics; Department <strong>of</strong><br />

Psychiatry; University <strong>of</strong> California, San Diego; 9500 Gilman<br />

Drive; La Jolla, CA 92039; U.S.A.<br />

Recently, high-throughput biomarker discovery efforts have<br />

focused on pr<strong>of</strong>iling various “omes” to assist in elucidating<br />

dynamic molecular signatures in major psychiatric disorders.<br />

For the last four years, our group has focused largely on the<br />

transcriptome (i.e., the full compendium <strong>of</strong> mRNAs expressed<br />

in a given tissue) as a potential source <strong>of</strong> biomarkers for<br />

various mental disorders. Gene expression levels are<br />

heritable, yet are also influenced strongly by environmental<br />

and epigenomic influences; thus, the transcriptome may<br />

represent a final common pathway wherein the joint<br />

influences <strong>of</strong> genome, epigenome, and envirome may be<br />

represented. This ability to represent multiple influences may<br />

be an advantage for a biomarker discovery platform when<br />

considering complex multifactorial polygenic disorders, such<br />

as psychiatric disorders. In this presentation, we will provide<br />

a review <strong>of</strong> our recent efforts utilizing these approaches to<br />

identify viable biomarkers for schizophrenia (SCZ) and<br />

bipolar disorder (BPD). These efforts have included the use <strong>of</strong><br />

supervised and unsupervised hierarchical clustering <strong>of</strong><br />

individual transcripts; receiver-operating curve analyses;<br />

ANOVA and ANCOVA; ontology, pathway, and protein<br />

structure-similarity analyses; and home-grown biostatistical<br />

and bioinformatic approaches. The various strengths and<br />

weaknesses <strong>of</strong> these approaches will be discussed in the<br />

context <strong>of</strong> our empirical work that has implicated various<br />

genes and biological pathways as sources <strong>of</strong> potentially useful<br />

biomarkers for SCZ, BPD, and their sometimes-shared feature<br />

<strong>of</strong> psychosis. We will conclude with a discussion <strong>of</strong> how<br />

these findings may be interrelated and how they may guide<br />

future biomarker discovery efforts.<br />

S6.3 IDENTIFICATION OF BLOOD BIOMARKERS<br />

FOR PSYCHOSIS USING CONVERGENT<br />

FUNCTIONAL GENOMICS<br />

S. Kurian (1), H. Le-Niculescu (2), S. Patel (2), D. Bertram<br />

(3), C. Dike (4), N. Yehyawi (3), J. Davis (3), P. Lysaker (3),<br />

M. Caligiuri (5), J. Lohr (4, 5), D. Lahiri (2), J. Nurnberger<br />

(2), S. Faraone (6), M. Geyer (5), M. Tsuang (5), N. Schork<br />

(1), D. Salomon (1), A. Niculescu* (2, 3)<br />

1. Scripps Research Institute 2. Indiana University 3.<br />

Indianapolis VA <strong>Medical</strong> Center 4. San Diego VA <strong>Medical</strong><br />

Center 5. <strong>UCSD</strong> 6. SUNY Upstate<br />

* anicules@iupui.edu<br />

There are to date no objective clinical laboratory blood tests<br />

for psychotic disease states. We provide pro<strong>of</strong> <strong>of</strong> principle for<br />

a Convergent Functional Genomics (CFG) approach to help<br />

identify and prioritize blood biomarkers for two key psychotic<br />

symptoms, one sensory (hallucinations) and one cognitive<br />

(delusions). We used gene expression pr<strong>of</strong>iling in whole blood<br />

samples from patients with schizophrenia and related<br />

disorders, with phenotypic information collected at the time <strong>of</strong><br />

blood draw, then cross-matched the data with other human and<br />

animal model lines <strong>of</strong> evidence. Topping our list <strong>of</strong> candidate<br />

blood biomarkers for hallucinations, we have four genes<br />

decreased in expression in high hallucinations states (Fn1,<br />

Rhobtb3, Aldh1l1, Mpp3), and three genes increased in high<br />

hallucinations states (Arhgef9, Phlda1,S100a6). All <strong>of</strong> these<br />

genes have prior evidence <strong>of</strong> differential expression in<br />

schizophrenia patients. At the top <strong>of</strong> our list <strong>of</strong> candidate<br />

blood biomarkers for delusions, we have fifteen genes<br />

decreased in expression in high delusions states (such as Drd2,<br />

Apoe, Scamp1, Fn1, Idh1, Aldh1l1), and sixteen genes<br />

increased in high delusions states (such as Nrg1, Egr1, Pvalb,<br />

Dctn1, Nmt1, Tob2). Twenty five <strong>of</strong> these genes have prior<br />

evidence <strong>of</strong> differential expression in schizophrenia patients.<br />

Predictive scores, based on panels <strong>of</strong> top candidate<br />

biomarkers, show good sensitivity and negative predictive<br />

value for detecting high psychosis states in the original cohort<br />

as well as in three additional cohorts. These results have<br />

implications for the development <strong>of</strong> objective laboratory tests<br />

to measure illness severity and response to treatment in<br />

devastating disorders such as schizophrenia.


S6.4 LITHIUM MEDIATED EXPRESSION CHANGES<br />

IN LYMPHOBLASTOID CELL LINES<br />

H. Chen (1), M. Burmeister (1), M. McInnis* (1)<br />

1. University <strong>of</strong> Michigan<br />

* mmcinnis@umich.edu<br />

Lithium (Li) is an effective maintenance therapy <strong>of</strong> bipolar<br />

disorder (BPD). There are several hypotheses on the<br />

mechanism <strong>of</strong> lithium’s action, many derived from studies that<br />

have utilized post-mortem brain tissue from bipolar<br />

individuals. The changes <strong>of</strong> gene expression in postmortem<br />

tissue exposed to Li are confounded by agonal and other<br />

factors such as postmortem interval, tissue pH, medication<br />

status, age, and sex. Lymphoblastoid cell lines (LCL) derived<br />

from subjects with the illness <strong>of</strong> interest represent an<br />

accessible proxy cellular system to study gene expression<br />

patterns. We studied gene expression in 12 LCLs cultured<br />

with and without presence <strong>of</strong> 1 mM LiCl in the culture for 4,<br />

8, and 16 day using the Illumina RefSeq8_v2 BeadChip<br />

microarrays. We identified 218 transcripts that showed<br />

significant changes over the time course <strong>of</strong> Li treatment at<br />

false discovery rate (FDR) < 5%. Of the 218 significant<br />

transcripts, only C8orf33 showed a consistently positive slope<br />

change in expression pattern, and the rest showed negative<br />

slope changes in expression. C8orf33 is a novel gene mapped<br />

to the region <strong>of</strong> 8q24 linked to BPD. Molecular interaction<br />

analysis using the Michigan Molecular Interaction data base<br />

(MiMI) search algorithms identified that C8orf33 directly<br />

interacted with three other genes (GIT1, GPRASP1, and<br />

HAP1), and through indirect interactions forms a network <strong>of</strong><br />

total 144 nodes (genes) and 345 edges. Among the 144 nodes,<br />

21% <strong>of</strong> them were reported to be regulated by Li in mouse<br />

brain (McQuillin et al., 2007). Functional annotation analysis<br />

using the EASE algorithms suggests that the 144 genes in the<br />

network are enriched in biological pathways, including a<br />

G-protein coupled receptor protein signaling pathway<br />

(p=2.27E-8), neuroactive ligand-receptor interaction(P =<br />

7.38E-16), calcium signaling pathway (P = 7.90E-10), and<br />

regulation <strong>of</strong> actin cytoskeleton (P = 3.16E-05). Our data<br />

suggest that individual gene expression changes induced by Li<br />

treatment are modest. The results reported here suggest a<br />

model that includes the effect from networked genes<br />

contributes to the molecular basis <strong>of</strong> Li's therapeutic action.<br />

S6.5 EXON ARRAY FINDINGS IN LYMPHOBLASTIC<br />

CELL LINES IN SCHIZOPHRENIA<br />

M. Vawter<br />

S7 SMALL RNA IN NEUROPSYCHIATRY<br />

S7.1 BIOINFORMATIC ANALYSIS OF POTENTIAL<br />

MIRNA INVOLVEMENT IN PSYCHIATRIC DISEASE<br />

S. Jugurnauth*, G. Breen, D. Collier<br />

*sarah.jugurnauth@kcl.ac.uk<br />

The ability <strong>of</strong> microRNAs to regulate expression <strong>of</strong> genes via<br />

UTR regions opens up these relatively overlooked sequences<br />

as new candidates for disease susceptibility. In many cases<br />

associations map to "gene deserts", and association peaks<br />

<strong>of</strong>ten indicate areas where no causative change is obvious. A<br />

number <strong>of</strong> these fall within computationally predicted miRNA<br />

target sites as reviewed in Barnes 2007. MiRNAs, with their<br />

increasingly recognized roles in neurodevelopment and<br />

synaptic plasticity may contribute to the multiple genetic<br />

factors influencing the as yet unknown dysfunction and/or<br />

neurodevelopmental incidents believed to result in psychiatric<br />

disease. Evidence is emerging for aberrant miRNA expression<br />

or miRNA involvement in many psychiatric diseases including<br />

schizophrenia, autism spectrum disorder, alzheimers disease,<br />

neurodegenerative disorders and behavioral disorders. I intend<br />

to support evidence for disruptive SNPs in candidate miRNA<br />

target sites to highlight miRNAs that might impact on<br />

neurotransmission pathways and pathways involving<br />

candidate psychiatric genes. Identification <strong>of</strong> relevant<br />

computationally predicted target sites will take the following<br />

into account, multiple targets in a gene, strength and quality <strong>of</strong><br />

targets, target prediction by more than one s<strong>of</strong>tware, gene<br />

weighting and function, similar expression levels and location<br />

<strong>of</strong> miRNA and target previously observed in literature,<br />

previous associations with neuropsychiatric disease in<br />

candidate region, target accessibility (mRNA folding) and<br />

where possible epistasis and eQTL pQTL data.


S7.2 ALTERATION OF CORTICAL MICRORNA<br />

BIOGENESIS IN SCHIZOPHRENIA AND ITS<br />

INFLUENCE ON GENE EXPRESSION<br />

M. Cairns (1), N. Beveridge (1), A. Carroll (1), E. Gardiner<br />

(1), P. Tooney (1), D. Santarelli (1)<br />

1. Schizophrenia Research Institute, Sydney. School <strong>of</strong><br />

Biomedical Sciences, The University <strong>of</strong> Newcastle. Hunter<br />

<strong>Medical</strong> Research Institute<br />

* murray.cairns@newcastle.edu.au<br />

Gene expression is altered in multiple brain regions in<br />

schizophrenia. While some <strong>of</strong> these changes relate directly to<br />

changes at the DNA level, most are probably a function <strong>of</strong><br />

regulatory influences. Bioinformatic approaches are helping to<br />

make sense <strong>of</strong> these changes by identifying pathways and<br />

networks <strong>of</strong> related genes with plausible implications for this<br />

complex phenotype. Unfortunately, this analysis is yet to<br />

provide a cohesive and reproducible signature indicative <strong>of</strong><br />

systematic failure in gene regulation. In our laboratory, we<br />

have been investigating the hypothesis that<br />

post-transcriptional gene silencing is altered in schizophrenia,<br />

causing an underlying change in the regulatory matrix. In<br />

support <strong>of</strong> this we have identified widespread alteration <strong>of</strong><br />

microRNA expression in two cortical regions that appeared to<br />

be a consequence <strong>of</strong> elevated miRNA biogenesis. This<br />

corresponded with an increase in the expression <strong>of</strong> DGCR8, a<br />

component <strong>of</strong> the microprocessor complex involved in<br />

post-transcriptional modification and regulation <strong>of</strong> most<br />

primary microRNA transcripts. Interestingly, this<br />

up-regulation <strong>of</strong> miRNA expression is inversely correlated<br />

with gene expression in these tissues and could have important<br />

implications for the development and progress <strong>of</strong><br />

schizophrenia - perhaps through the regulation <strong>of</strong> genes<br />

involved in synaptic structure and function, which feature<br />

strongly amongst predicted target genes.<br />

S7.3 ALTERATION OF THE GENE REGULATORY<br />

NETWORK IN THE FRONTAL CORTEX OF<br />

HIV-INFECTED INDIVIDUALS<br />

E. Tatro* (1), I. Everall (1)<br />

1. <strong>UCSD</strong> School <strong>of</strong> Medicine Department <strong>of</strong> Psychiatry<br />

* etatro@ucsd.edu<br />

Background: MicroRNAs are small, non-coding RNAs that<br />

regulate gene networks, helping control cell function and<br />

phenotype. MicroRNAs are recognized as key players in CNS<br />

patterning, function, and disease. Past studies focused on<br />

expression levels <strong>of</strong> coding mRNAs in the brain <strong>of</strong><br />

HIV-infected individuals. Because microRNAs affect the<br />

abundance and downstream functions <strong>of</strong> mRNAs, it is<br />

important to understand both mRNA and miRNA changes<br />

concurrently. We report the first genome-wide microRNA<br />

pr<strong>of</strong>ile in the HIV-infected human frontal cortex. We present<br />

methods to integrate mRNA expression with microRNA<br />

expression data in a Target BiasAnalysis by determining the<br />

probability that the number <strong>of</strong> target-genes <strong>of</strong> dysregulated<br />

miRNAs would be dysregulated in HIV infection versus<br />

expected by chance. Methods: We used Affymetrix arrays for<br />

comparing gene expression in frontal cortex from<br />

6HIV-infected males (no cognitive impairment or<br />

encephalitis) and 6 age-matched controls at the mRNA level.<br />

We pooled equivalent RNA samples and utilized Applied<br />

Biosystems PCR-based array to assess a panel <strong>of</strong> 379<br />

microRNAs. Results: Target bias analysis indicated that<br />

microRNAs clustered into four types: A) Those with many<br />

dysregulated mRNA targets <strong>of</strong> less stringent significance, B)<br />

Fewer dysregulated target-genes <strong>of</strong> highly stringent<br />

significance, C) spectrum from non-bias to combinations <strong>of</strong> A<br />

and B. The dysregulated miRNAs clustered on Chromosomes<br />

14, 17, 19, and X. Those miRNAs that affect many genes may<br />

be important "circuits" in gene regulatory networks pertinent<br />

to the effects <strong>of</strong> HIV infection in CNS. These may be<br />

functional and diagnostic markers <strong>of</strong> neurologic disease in<br />

HIV-infected patients.


S7.4 MICRORNAS IN THE TRANSITION FROM<br />

CONTROLLED TO COMPULSIVE COCAINE-TAKING<br />

P. Kenny (1), J. Hollander (1), H. Im (1)<br />

1. The Scripps Research Institute<br />

* pjkenny@scripps.edu<br />

Rats with extended access to cocaine over prolonged time<br />

periods can develop compulsive-like cocaine seeking<br />

behaviors, including intake that progressively escalates. The<br />

dorsal striatumis considered an important brain region in the<br />

development <strong>of</strong> compulsive cocaine intake. MicroRNAs are<br />

small (21–23 nucleotides) noncoding RNA transcripts that<br />

regulate many basic aspects <strong>of</strong> cell biology, but little is known<br />

<strong>of</strong> their roles in addiction. We investigated the role for<br />

microRNAs in the dorsal striatum in regulating the<br />

development <strong>of</strong> escalated cocaine intake in rats with extended<br />

access to the drug. We found that microRNA-212 (miR-212)<br />

was significantly up regulated in the dorsal striatum <strong>of</strong> rats<br />

with extended (6-h) daily access to cocaine<br />

self-administration, demonstrating compulsive-like escalated<br />

intake, compared with rats that had restricted (1-h) access and<br />

cocaine-naïve rats. Furthermore, miR-212overexpression in<br />

dorsal striatum decreased, whereas its inhibition increased,<br />

cocaine intake only in rats with extended but not restricted<br />

access to thedrug. Finally, we found that the effects <strong>of</strong><br />

miR-212 on cocaine-taking were regulated in part through the<br />

novel CREB coactivator TORC. These findings identify<br />

miR-212 as a cocaine-responsive microRNA whose<br />

expression is increased in dorsal striatum in rats that<br />

over-consume cocaine. Moreover, miR-212 may be a novel<br />

protect factor against the development <strong>of</strong> compulsive<br />

cocaine-taking behaviors. Finally, we identify apreviously<br />

unknown role TORC in regulating drug-taking behavior.<br />

Noncoding RNAs such as the microRNAs may thus serve as a<br />

novel targets for the future development <strong>of</strong> anti-addiction<br />

therapeutics.<br />

S8 ATTENTION DEFICIT HYPERACTIVITY<br />

DISORDER: ENVIRONMENTAL<br />

INFLUENCES ON THE GENOME<br />

2009 RICHARD TODD AWARD LECTURE<br />

S8.1 WHOLE GENOME METHYLATION STUDY OF<br />

48 MONOZYTGOTIC PAIRS CONCORDANT OR<br />

DISCORDANT FOR ATTENTION PROBLEMS<br />

R. Alth<strong>of</strong>f* (1), D. Boomsma (2), C. van Beijesterveldt (2), P.<br />

Rizzu (2), E. de Geus (2), F. Middleton (3), Y. Zhang-James<br />

(3), S. Faraone (3), J. Hudziak (1)<br />

1. University <strong>of</strong> Vermont 2. VU University 3. SUNY Upstate<br />

University<br />

* ralth<strong>of</strong>f@uvm.edu<br />

Despite large sample sizes Genome Wide Association Studies<br />

for ADHD have produced relative few new genetic<br />

associations. We sought to determine if a whole genome<br />

methylation scan <strong>of</strong> monozygotic twin pairs, who mostly share<br />

their DNA sequence, would reveal new genetic areas for<br />

study. Method: 48 monozygotic (MZ) twin pairs from the<br />

Netherlands Twin Registry were selected for persistent<br />

longitudinal concordance or discordance on CBCL attention<br />

problems (AP). Extracted buccal DNA from 11 discordant<br />

pairs, 20 concordant affected, and 17 concordant unaffected<br />

was restriction digested and incubated with antibodies to<br />

5-methylcytosine. Antibody-bound DNA was separated from<br />

non-antibody bound DNA and the antibodies were removed to<br />

reveal methylation sites on the DNA. After PCR amplification<br />

methylated DNA was hybridized for Affymetrix Human<br />

Promoter Arrays tiled upstream and downstream <strong>of</strong> every<br />

known gene. Raw signal values for each <strong>of</strong> the 400 probes for<br />

each promoter were extracted, quantile normalized, and<br />

median polished within each promoter region. The end result<br />

was median methylation signals for each <strong>of</strong> 16,441 gene<br />

promoter regions. Empirical distributions for each twin type<br />

were created as a screening tool to identify those promoter<br />

regions where concordant affected, concordant unaffected and<br />

discordant twin pairs differ more from each other than from<br />

other individuals in the sample. MZ co-twin methylation<br />

falling outside <strong>of</strong> this empirical distribution interval for a<br />

particular individual was considered a possible positive result.<br />

Results revealed significant differences between discordant<br />

twins in at least one gene region previously associated with<br />

attention problems in animal models.


S8.2 THE DEVELOPMENTAL COURSE OF GENE<br />

EXPRESSION IN BRAINS OF RATS PRENATALLY<br />

EXPOSED TO POLYCHLORINATED BIPHENYLS<br />

T. DasBanerjee* (1), F. Middleton (1), S. Faraone (1)<br />

1. SUNY Upstate <strong>Medical</strong> University<br />

* dast@upstate.edu<br />

Although ADHD is a heritable neurobehavioral disorder,<br />

environmental influences or gene-environment interactions<br />

play an important role. Studies in humans and animals support<br />

the relationship between exposure to polychlorinated<br />

biphenyls (PCBs) and risk <strong>of</strong> developing ADHD. In this study,<br />

we investigated the developmental course <strong>of</strong> expression <strong>of</strong><br />

ADHD candidate genes, or IMAGE genes, in Sprague-Dawley<br />

(SD) rats exposed to PCBs prenatally and postnatally. We<br />

performed gene expression analysis <strong>of</strong> three brain regions (the<br />

prefrontal cortex, midbrain and vermis), which abundantly<br />

express the IMAGE genes. We analyzed three time points-<br />

PND23, PND35 and PND70. At each time point we have 36<br />

rats (18 males, 18 females; within each gender, 6 controls, 6<br />

exposed to PCBs only gestationally and 6 exposed to PCBs<br />

only during lactation).The brains were harvested at the above<br />

time points and used for isolation <strong>of</strong> RNA from each brain<br />

region. For microarray screen, we pooled equal amounts <strong>of</strong><br />

RNA from each animal in each group and each brain area to<br />

create a total <strong>of</strong> 36 RNA samples for analysis (18 samples in<br />

duplicate). RNA was processed using the recommended<br />

Affymetrix protocol and hybridized to the GeneChip Rat Gene<br />

1.0 ST array. Our results show that a small subset <strong>of</strong> IMAGE<br />

genes shows robust differences in expression in some or all<br />

brain regions <strong>of</strong> PCB-exposed SD rats compared to SD<br />

controls. A couple <strong>of</strong> these genes (COMT, Csnk1a1) have<br />

been confirmed by real time quantitative PCR (p< 0.005).<br />

Collectively, the data provide strong evidence that exposure to<br />

PCBs, either gestational or lactational, can lead to pr<strong>of</strong>ound<br />

effects on the brain circuits involved in ADHD.<br />

S8.3 A PILOT STUDY OF METHYLATION IN ADHD<br />

YOUTH PRENATALLY EXPOSED TO CIGARETTE<br />

SMOKE<br />

S. Faraone* (1), F. Middleton (1), T. DasBanerjee (1), Y.<br />

Zhang-James (1), Tourette GWAS Consortium<br />

1. SUNY Upstate <strong>Medical</strong> University<br />

* faraones@upstate.edu<br />

Attention deficit hyperactivity disorder (ADHD) is a complex<br />

disorder having both genetic and environmental causes. Both<br />

human and animal model studies show that prenatal exposure<br />

to cigarette smoke is a risk factor for ADHD symptoms and<br />

associated features. Despite this evidence, little is known<br />

about the mechanisms whereby smoke exposure increases risk<br />

for ADHD. We hypothesized that methylation <strong>of</strong> ADHD<br />

candidate genes by cigarette exposure could be one such<br />

mechanism. As a preliminary test <strong>of</strong> this hypothesis, we<br />

examined the methylation status <strong>of</strong> 51 ADHD candidate genes<br />

in 34 subjects from the International Multisite ADHD<br />

Genetics (IMAGE) project, using DNA extracted from blood:<br />

15 ADHD smoke exposed subjects, 4 non-ADHD smoke<br />

exposed siblings; 10 ADHD non-smoke exposed subjects and<br />

5 non-ADHD non-smoke exposed siblings. We assessed<br />

methylation using bisulfite treatment followed by analysis on<br />

the Human Promoter 1.0R Array (Affymetrix). Signal data<br />

from all annotated CpG islands in the genome were extracted<br />

and quantile normalized. Then, the probe and CpG island data<br />

for individual IMAGE candidate genes were compared<br />

between subject groups. After correction for multiple<br />

comparisons, we found no significant effect <strong>of</strong> either ADHD<br />

status or smoke exposure on methylation values. However,<br />

three genes showed nominally significant (p


S8.4 JOINT EFFECTS OF TOXIN EXPOSURES AND<br />

RISK GENOTYPES ON ADHD SYMPTOMS<br />

J. Nigg* (1)<br />

1. Oregon Health & Science University<br />

* niggj@ohsu.edu<br />

Two studies are presented here that illustrate both<br />

psychosocial environment and biological environment effects<br />

on gene correlation with ADHD in children. In study 1, blood<br />

lead is examined in 347 children carefully characterized with<br />

regard to ADHD or non-ADHD status. Blood lead is reliably<br />

associated with ADHD symptoms. However, when iron<br />

metabolism (HFE) and dopamine genes are considered, a<br />

composite genotype is created that is invulnerable to the lead<br />

effect (r=.00), versus a risk genotype that exhibits robust<br />

response to blood lead (r=.30, p


S9.2 USING NEUROCOGNITION AS AN<br />

ENDOPHENOTYPE IN A GWAS DATASET<br />

K. Burdick* (1), P. DeRosse (1), T. Lencz (1), A. Malhotra<br />

(1)<br />

1. The Zucker Hillside Hospital; Feinstein Institute for<br />

<strong>Medical</strong> Research; Albert Einstein College <strong>of</strong> Medicine<br />

* kburdick@lij.edu<br />

One promising strategy proposed as a way to simplify the<br />

genetics <strong>of</strong> complex disorders, such as schizophrenia, has been<br />

through the use <strong>of</strong> intermediate phenotypes or<br />

endophenotypes. An endophenotype is a measurable trait that<br />

is thought to be more closely linked to the underlying<br />

pathophysiology <strong>of</strong> the disease than is the categorical<br />

diagnosis itself. The use <strong>of</strong> such biomarkers in molecular<br />

genetic studies may enhance power to detect susceptibility loci<br />

first by reducing the complexity <strong>of</strong> the phenotype through<br />

dissecting it into its component parts and second by assaying a<br />

trait that is believed to be more proximal to gene action. A<br />

growing body <strong>of</strong> evidence suggests that measures <strong>of</strong><br />

neurocognitive function may have utility in genetic studies by<br />

enhancing power and/or elucidating gene function at the<br />

endophenotype level. However, while the originally proposed<br />

methodology was to first identify genetic variants associated<br />

with the endophenotype and then subsequently to test for their<br />

effects on risk for illness, most studies to date have used these<br />

traits as post-hoc to disease susceptibility analyses. In the<br />

current study, we attempted to utilize the endophenotype<br />

approach as it was initially intended. Specifically, we<br />

conducted a genome-wide association study (GWAS) in 200<br />

Caucasian healthy controls using general cognitive ability (g)<br />

as the primary outcome measure. We then tested the top five<br />

hits for case-control association in 249 healthy controls and<br />

280 patients with schizophrenia. This presentation will focus<br />

on the findings from these analyses, including the detection <strong>of</strong><br />

novel schizophrenia-associated loci.<br />

S9.3 PHARMACOGENETIC APPROACHES IN A<br />

GENOME-WIDE DATASET<br />

A. Malhotra* (1), T. Lencz (1), J. Zhang (1), J. Kane (1), D.<br />

Robinson (1)<br />

1. The Zucker Hillside Hospital; Feinstein <strong>Medical</strong> Research<br />

Institute; Albert Einstein College <strong>of</strong> Medicine<br />

* malhotra@lij.edu<br />

Genome-wide association studies (GWAS) are now being<br />

utilized across a number <strong>of</strong> phenotypes in psychiatric research,<br />

including studies that seek to identify molecular genetic<br />

factors influencing antipsychotic drug response. To date, these<br />

studies have been limited by phenotypic heterogeneity<br />

(including multiple drugs, dosages, and levels <strong>of</strong> compliance)<br />

and smaller sample sizes than usually recommended for<br />

GWAS; more innovative study designs may need to be<br />

considered. We have embarked on a large scale study that<br />

aims to combine the strengths <strong>of</strong> GWAS with the advantages<br />

<strong>of</strong> focused candidate gene work to identify variants than<br />

influence antipsychotic drug efficacy and adverse events<br />

associated with treatment. Initially, we will conduct a GWAS<br />

in a group <strong>of</strong> patients characterized by clinical assignment to<br />

treatment with clozapine, the atypical antipsychotic drug<br />

commonly reserved for treatment refractory patients, and<br />

compare the clozapine-treated patients to<br />

non-clozapine-treated patients as a proxy measure <strong>of</strong> treatment<br />

responsiveness. Top SNPs will be functionally characterized<br />

with in vivo and in vitro approaches; functionally relevant<br />

SNPs will then be assessed in large prospectively treated<br />

cohort <strong>of</strong> 1000 first episode schizophrenia patients collected in<br />

the U.S and Europe for relationship to drug efficacy and<br />

drug-induced weight gain. In this presentation, we will present<br />

early data with this approach including: GWAS data<br />

identifying SNPs within 5 genes that were differentially<br />

distributed between groups (p< 5 x 10-6); candidate gene work<br />

implicating specific promoter region polymorphisms in first<br />

episode schizophrenia patients’ clinical response to treatment<br />

as well as propensity for acute weight gain and finally; data<br />

from a recent meta analysis suggesting that the dopamine D2<br />

receptor (DRD2) gene plays a modest, yet significant, role in<br />

the variation in antipsychotic drug efficacy.


S9.4 ALTERNATIVE PHENOTYPES IN BIPOLAR<br />

DISORDER: DOES UNDERLYING GENETIC<br />

ARCHITECTURE MAP ONTO CLINICAL<br />

FEATURES?<br />

J. Kelsoe, T. Greenwood, K. Oedergaard, R. McKinney,<br />

Bipolar Genome Study (BiGS)<br />

It has long been suspected that different aspects <strong>of</strong> bipolar<br />

disorder might have different genetic bases and study <strong>of</strong> a<br />

specific subform might reduce the problem <strong>of</strong> genetic<br />

heterogeneity. Large scale GWAS studies have now given us<br />

both the genotyping density and sample sizes to begin to<br />

address this question. In support <strong>of</strong> the idea are studies that<br />

demonstrate the familiality <strong>of</strong> a variety <strong>of</strong> clinical<br />

presentations such as psychotic mania, irritable mania,<br />

comorbid anxiety disorders or substance abuse and many<br />

others. Our collaborative group has taken great pains to assess<br />

these clinical features and have been actively examining their<br />

value as alternative phenotypes. In general, it seems clear that<br />

many alternative phenotypes provide stronger evidence <strong>of</strong><br />

association than seen in the analysis <strong>of</strong> the entire sample<br />

despite much smaller samples sizes. Striking examples <strong>of</strong> this<br />

are irritable mania, comorbid migraine and disability. Though<br />

in an early stage these analyses suggest different genetic bases<br />

to these different forms. This elucidation <strong>of</strong> underlying<br />

architecture may guide biological hypotheses <strong>of</strong> disease.<br />

S10 OBSESSIVE COMPULSIVE DISORDER<br />

AND TOURETTE DISORDER: PHENOTYPE,<br />

GENOTYPE AND GENOME-WIDE<br />

ASSOCIATION STUDIES<br />

S10.1 OBSESSIVE COMPULSIVE DISORDER AND<br />

TOURETTE DISORDER: PHENOTYPE, GENOTYPE<br />

AND GENOME-WIDE ASSOCIATION STUDIES.<br />

C. Mathews (1), D. Cath (2), P. Arnold (3), J. Scharf (4), S.<br />

Stewart (4), D. Pauls * (4)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California <strong>Medical</strong><br />

School, San Francisco, CA; USA 2. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Utrecht, the Netherlands 3. Hospital for Sick<br />

Children, University <strong>of</strong> Toronto, Toronto, Ontario, Canada 4.<br />

Psychiatric and Neurodevelopmental Genetics Unit, Center for<br />

Human Genetic Research, Massachusetts General Hospital,<br />

Harvard <strong>Medical</strong> School, Boston, MA, USA<br />

* dpauls@pngu.mgh.harvard.edu<br />

Obsessive Compulsive Disorder (OCD) and Tourette Disorder<br />

(TD) are familial, phenotypically heterogeneous<br />

neuropsychiatric disorders that are likely to have a shared<br />

genetic etiology with additional independent genetic and<br />

non-genetic contributors. The first two presenters will discuss<br />

attempts to disaggregate these conditions into more<br />

homogeneous quantitative phenotypes. In the first<br />

presentation, factor and latent class analyses <strong>of</strong> 1,200 and<br />

1,600 individuals with OCD, respectively, will be described.<br />

Results suggest an underlying latent OCD susceptibility and<br />

five symptom-based factors (contamination, taboo, doubts,<br />

hoarding/perfectionism, superstitions/rituals), four <strong>of</strong> which<br />

are heritable. In the second presentation, latent class analyses<br />

<strong>of</strong> 500 TD-affected individuals will be summarized. Three<br />

classes <strong>of</strong> tics (socially inappropriate behaviors/tics, head tics,<br />

body tics) were observed, one <strong>of</strong> which (body tics) is<br />

heritable. The next speaker will summarize a family-based<br />

association study <strong>of</strong> 308 OCD families in which DLGAP3 (the<br />

human homologue <strong>of</strong> SAPAP3 a gene associated with<br />

compulsive grooming in mice) was examined. Several SNPs<br />

were nominally associated with OCD, although none survived<br />

correction for multiple testing. Finally, preliminary results <strong>of</strong><br />

genome-wide association studies for OCD and TD will be<br />

presented. Two-thousand-five-hundred OCD-affected<br />

individuals and 1,800 TD-affected individuals were genotyped<br />

using the Illumina Human610 Beadchip. Preliminary results<br />

suggest that at least two TD loci are associated with p-values<br />

less than 10-8-10-9. To date, no OCD loci have achieved<br />

genome-wide significance, but several were associated with<br />

p-values in the 10-6-10-7 range. Results presented in this<br />

symposium should help to advance our understanding <strong>of</strong> the<br />

genetic etiologies <strong>of</strong> OCD and TD.


S10.2 LATENT STRUCTURE IN OBSESSIVE<br />

COMPULSIVE DISORDER<br />

C. Mathews* (1), M. Andresen (2), D. Cath (3), K. Delucchi<br />

(1), D. Denys (5), H. Katerberg (6), C. Lochner (7), D. Stein<br />

(7), S. Stewart (8)<br />

1. UCSF 2. University <strong>of</strong> Minnesota 3. Utrecht 4. UCSF 5.<br />

University <strong>of</strong> Amsterdam 6. University <strong>of</strong> Groningen 7.<br />

University <strong>of</strong> Stellenbosch 8. Massachusetts General Hospital<br />

* cmathews@lppi.ucsf.edu<br />

Objective: Obsessive-compulsive disorder (OCD)<br />

isphenomenologically heterogeneous, and data reduction<br />

methods have been increasingly used to identify more<br />

homogeneous subgroups, with varying results. We present the<br />

results <strong>of</strong> factor and latent class analyses in a large<br />

multi-center sample <strong>of</strong> OCD patients and their relevance to<br />

genetics and treatment outcome. Method: Principal<br />

components factor (PCA) and latent class analyses (LCA)<br />

using obsessive-compulsive (OC) symptoms from the<br />

Yale-Brown Obsessive-Compulsive Scale were conducted in<br />

1200 and 1600 OCD-affected individuals, respectively.<br />

Heritability <strong>of</strong> the resulting factors and relationships between<br />

latent class membership and treatment response, gender,<br />

symptom severity and comorbid tics were tested. Results:<br />

PCA resulted in a five-factor model: 1) taboo, 2)<br />

contamination/cleaning, 3)doubts, 4) superstitions/rituals, and<br />

5) symmetry/hoarding. All factors except factor 4 were<br />

heritable. LCA best-fit models yielded three or five classes.<br />

Classes in the three-class solution differed only in frequency<br />

<strong>of</strong> endorsement <strong>of</strong> symptoms, irrespective <strong>of</strong> symptom type.<br />

The five-class solution was similar to the three-class, with the<br />

addition <strong>of</strong> a minimal endorsement class and a class with<br />

predominantly contamination/cleaning symptoms. Classes<br />

with higher symptom endorsement were associated with<br />

earlier age <strong>of</strong> onset, male gender, and comorbid tics. The<br />

contamination/cleaning class was associated with improved<br />

treatment outcome. Conclusions: These results provide<br />

support both for the validity <strong>of</strong> OCD as a single diagnostic<br />

entity with varying levels <strong>of</strong> severity/symptom endorsement,<br />

and for additional separate, genetically distinct OC symptom<br />

dimensions. The large size and heterogeneous nature <strong>of</strong> the<br />

sample has the advantage <strong>of</strong> increasing the generalizability <strong>of</strong><br />

the findings.<br />

S10.3 GENOME-WIDE ASSOCIATION STUDY OF<br />

TOURETTE DISORDER IN EUROPEAN ANCESTRY<br />

SAMPLES AND FOUR POPULATION ISOLATES<br />

J. Scharf* (1), B. Neale (1), S. Service (2), A. Tikhomorov<br />

(3), J. Fagerness (1), A. Pluzhnikov (3), D. Yu (1), J. Crane<br />

(1), M. State (4), J. Tischfield (5), D. Cath (6), G. Rouleau (7),<br />

A. Ruiz-Linares (8), C. Mathews (9), C. Sabatti (2), S. Purcell<br />

(1), N. Freimer (2), N. Cox (3), D. Pauls (1), GENEVA<br />

1. Massachusetts General Hospital 2. University <strong>of</strong> California,<br />

Los Angeles 3. University <strong>of</strong> Chicago 4. Yale University 5.<br />

Rutgers University 6. University <strong>of</strong> Utrecht, Netherlands 7.<br />

University <strong>of</strong> Montreal, Canada 8. University College London,<br />

England 9. University <strong>of</strong> California, San Francisco<br />

* jscharf@partners.org<br />

Tourette Disorder (TD) is highly heritable, though no<br />

definitive susceptibility genes have been identified. Here,<br />

results from the first genome-wide association study (GWAS)<br />

<strong>of</strong> TD in 1,749 cases and 4,410 ancestry-matched controls will<br />

be reported. Cases included 983 subjects <strong>of</strong> European ancestry<br />

(EA) as well as 766 subjects from four population isolates:<br />

French Canadian (FC), Ashkenazi Jewish (AJ), Antioquia<br />

Colombian (CO) and Central Valley Costa Rica (CR).<br />

Diagnoses were made using DSM-IV-TR criteria. Case<br />

samples and population isolate controls from FC and CO were<br />

genotyped on the Illumina Human610Quad Beadchip; EA and<br />

AJ controls were previously genotyped on the Illumina<br />

Human550K and Human317K Beadchips, respectively. QC<br />

and population-stratified analyses were performed in PLINK.<br />

Preliminary results are based on analysis <strong>of</strong> 95% <strong>of</strong> the data.<br />

After standard QC to remove poorly performing SNPs and<br />

individuals, 1552 cases and 4027 controls remained.<br />

Multi-dimensional scaling was performed to control for<br />

population substructure; population outliers were excluded.<br />

Data were analyzed using logistic regression under an additive<br />

model with significant MDS axes included as covariates.<br />

Preliminary association analysis identified 8 loci with genomic<br />

control-adjusted p-values in the 10-7-10-9 range (seven in the<br />

EA sample and one in the AJ sample). In the FC and CR/CO<br />

populations, no loci had GC-adjusted p


S10.4 A GENOME-WIDE ASSOCIATION STUDY OF<br />

OBSESSIVE-COMPULSIVE DISORDER<br />

S. Stewart* (1), B. Neale (1), J. Fagerness (1), C. Mathews<br />

(2), J. Knowles (3), P. Arnold (4), G. Hanna (5), D. Cath (6),<br />

D. Denys (7), A. Hounie (8), M. Wagner (9), C. Lochner (10),<br />

M. Cavallini (11), H. Nicolini (12), D. Murphy (13), G.<br />

Nestadt (14), S. Purcell (1), D. Pauls (1)<br />

1. Massachusetts General Hospital 2. University <strong>of</strong> California,<br />

San Francisco 3. University <strong>of</strong> Southern California 4.<br />

University <strong>of</strong> Toronto 5. University <strong>of</strong> Michigan 6. Utrecht<br />

University 7. University <strong>of</strong> Amsterdam, Institute for<br />

Neuroscience 8. University <strong>of</strong> São Paulo 9. University <strong>of</strong><br />

Bonn 10. Stellenbosch University 11. Fondazione Centro San<br />

Raffaele del Monte Tabor 12. Universidad Autónoma de la<br />

Ciudad de México 13. National Institute <strong>of</strong> Mental Health 14.<br />

Johns Hopkins University<br />

* stewart@pngu.mgh.harvard.edu<br />

Introduction: Obsessive-Compulsive Disorder (OCD) is a<br />

complex genetic illness. This first genome-wide association<br />

study (GWAS) <strong>of</strong> OCD included European ancestry cases &<br />

family trios from the OCF Genetics Collaborative. Methods:<br />

1558 OCD cases <strong>of</strong> European ancestry and 477 OCD trios<br />

were included, as diagnosed via DSM-IV criteria. OCD<br />

samples were genotyped on the Illumina Human610Quad<br />

Beadchip; 3, 212 European ‘iControl’ samples were<br />

previously genotyped on the Illumina Human550K Beadchip.<br />

QC & stratified analyses were performed in PLINK. TDT &<br />

case-control samples were weighted by power. GPC based<br />

non-centrality parameters were employed. A weighted Z<br />

approach transformed P into Z-values for combining the<br />

samples. Results/Conclusions: Standard QC removed poorly<br />

performing SNPs & individuals, leaving 1558 cases. Checks<br />

for familial relationship and Mendel errors left 477 trios.<br />

Multi-dimensional scaling (MDS) was performed to exclude<br />

population outliers and to be included as covariates in logistic<br />

regression analyses under an additive model. Combining these<br />

two datasets yielded a genomic control ? <strong>of</strong> 1.09. Preliminary<br />

analyses identified two loci with p-values in the 10-7 range &<br />

12 loci with p-values in the 10-6 range. Four <strong>of</strong> the top 14<br />

associated genes are involved in glutamate neurotransmission<br />

(GRK4, GRID2, GRIN2B & DLGAP1), consistent with<br />

current etiologic theories <strong>of</strong> OCD and previous candidate gene<br />

studies. Although results <strong>of</strong> this first GWAS study <strong>of</strong> OCD<br />

were promising, with multiple biologically plausible<br />

associated loci, results fell short <strong>of</strong> genome-wide significance.<br />

Additional genotyping in independent samples will be<br />

necessary to confirm these findings.<br />

S10.5 DLGAP3 AND OBSESSIVE-COMPULSIVE<br />

DISORDER<br />

P. Arnold* (1), S. Stewart (2), S. Shaheen (1), J. Fagerness<br />

(2), S. Taillefer (1), B. Doan (1), A. Hounie (3), J. Kennedy<br />

(4), E. Cook (5), D. Pauls (1), M. Richter (6), C. Mathews (7),<br />

G. Hanna (8)<br />

1. Hospital for Sick Children 2. Massachusetts General<br />

Hospital 3. University <strong>of</strong> Sao Paulo 4. Centre for Addiction<br />

and Mental Health 5. University <strong>of</strong> Illinois at Chicago 6.<br />

Sunnybrook Health Sciences Centre 7. University <strong>of</strong><br />

California at San Francisco 8. University <strong>of</strong> Michigan<br />

* paul.arnold@sickkids.ca<br />

A recently described putative animal model <strong>of</strong><br />

obsessive-compulsive disorder (OCD) is the SAPAP3 (human<br />

homologue DLGAP3) knockout mouse, which exhibits<br />

compulsive grooming behaviour and corticostriatal glutamate<br />

transmission dysregulation. We hypothesized that a significant<br />

association would be identified between DLGAP3 and OCD<br />

using a family-based association approach. Tag single<br />

nucleotide polymorphisms (SNPs) were selected using<br />

publicly available databases (dbSNP, HapMap) and the<br />

Haploview program. The sample comprised OCD-affected<br />

probands and their first degree relatives from a combined<br />

sample <strong>of</strong> 1575 individuals (308 families), recruited from<br />

specialized OCD clinics by investigators at six sites (Toronto,<br />

Michigan, Boston, Paris, San Francisco, and Brazil).<br />

Genotyping was performed on the Sequenom iPlex platform.<br />

Markers with genotyping success rates < 90%, with minor<br />

allele frequency < 0.05, with greater than 10 Mendelian errors<br />

were excluded, leaving a set <strong>of</strong> 26 markers for analysis. Single<br />

marker and haplotype analyses were conducted using FBAT<br />

(Family Based Association Test) and Haploview s<strong>of</strong>tware. In<br />

the whole sample, several SNPs were nominally associated<br />

with OCD, although none remained significant after<br />

permutation testing. Exploratory analysis stratified by gender<br />

revealed that one SNP lying 5’ <strong>of</strong> the gene, rs16837122 was<br />

significantly associated (p = 0.003, not corrected for multiple<br />

testing) when examining transmission to male rather than<br />

female probands. Consistent with findings reported from other<br />

glutamate system genes (e.g. the glutamate transporter<br />

SLC1A1), these preliminary findings suggest a possible<br />

gender-specific association between sequence variation in or<br />

near the DLGAP3 gene and OCD in males.


S10.6 GENOME-WIDE LINKAGE ANALYSES OF<br />

COSTA RICAN FAMILIES WITH OBSESSIVE<br />

COMPULSIVE DISORDER<br />

J. Ross* (1), A. Azzam (1), D. Chavira (2), C. Mathews (1),<br />

H. Garrido (3)<br />

1. Department <strong>of</strong> Psychiatry, Langley Porter Psychiatric<br />

Institute, UCSF, San Francisco, CA 2. Department <strong>of</strong><br />

Psychiatry, <strong>UCSD</strong>, La Jolla, CA 3. Department <strong>of</strong> Psychiatry<br />

and Pediatrics, University <strong>of</strong> Costa Rica School <strong>of</strong> Medicine,<br />

San Jose, Costa Rica<br />

* jross@lppi.ucsf.edu<br />

Introductions: Obsessive Compulsive Disorder(OCD) is a<br />

complex neuropsychiatric disorder with a prevalence <strong>of</strong> 1-2%<br />

in most populations, and a component <strong>of</strong> heritability<br />

demonstrated through , twin, and segregation studies.<br />

Published OCD linkage studies have identified several<br />

possible susceptibility loci, pointing to both a complex genetic<br />

architecture and genetic heterogeneity. Methods: Extensive<br />

clinical information and DNA samples were ascertained from<br />

28 families from both Central Valley <strong>of</strong> Costa Rica and the<br />

US, but as initial linkage analysis revealed that these were two<br />

distinct genetic populations with regards genetic etiology for<br />

OCD, they were analyzed separately. These are the results <strong>of</strong><br />

parametric non-parametric(NPL) linkage <strong>of</strong> 3<br />

multi-generational families from Cost Rica with multiple<br />

members with OCD. Individuals were genotyped using the<br />

Illumina Linkage Panel IV(approximately 5800 markers).<br />

Linkage analyses were done using MERLIN and<br />

SIMWALK2snp. Significance analyses were also completed.<br />

Results: Initial linkage analysis revealed that NPL scores over<br />

1.5 on chromosomes several chromosomes, with a maximum<br />

NPL <strong>of</strong> 2.8. Several loci identified, including the one with the<br />

maximum NPL score, were in close proximity to loci<br />

identified in prior published OCD genome wide linkage<br />

studies. Bioinformatic pathway analysis <strong>of</strong> genes with max lod<br />

scores over 1.5 identified 11 genes at these loci that were<br />

involved in neurodevelopmental pathways that had less than 2<br />

degrees <strong>of</strong> separation. Discussion: The results <strong>of</strong> this study<br />

provide additional evidence <strong>of</strong> several genetic loci conferring<br />

susceptibility to OCD, and suggest neurodevelopmental<br />

pathways where several loci may interact in the pathology <strong>of</strong><br />

this disease.<br />

S10.7 TIC SYMPTOM DIMENSIONS AND THEIR<br />

HERITABILITY IN TOURETTE’S SYNDROME<br />

M. de Haan (1), K. Delucchi (2), D. Cath* (3), C. Mathews<br />

(2)<br />

1. Department <strong>of</strong> Psychiatry, VU University 2. Department <strong>of</strong><br />

Psychiatry, UCSF 3. Department <strong>of</strong> clinical psychology,<br />

Utrecht University<br />

* cath@xs4all.nl<br />

Objective: Gilles de la Tourette’s disorder (GTS) is both<br />

genotypically and phenotypically heterogeneous. Factor<br />

analytic strategies to reduce symptom heterogeneity are useful<br />

for unravelling heritability patterns and genetic susceptibility<br />

genes for GTS. Method: A self-report questionnaire for<br />

multiple tic symptoms was collected in two large Caucasian<br />

cohorts (UCSF San Francisco; VU University Amsterdam),<br />

totalling 494 patients. Item-level factor analysis using a<br />

tetrachoric correlation matrix in Mplus version 5.1 was<br />

conducted on twenty-six tic symptom variables that<br />

overlapped between the two samples. Heritability analysis was<br />

conducted for the resulting symptom dimensions using<br />

Sequential Oligogenic Linkage Analysis Routine (SOLAR) in<br />

a subset <strong>of</strong> subjects and their family members. Results: Three<br />

factors were identified: 1) Complex vocal tics & obscene<br />

behaviour; 2) Motor body tics; and 3) Motor head tics. The<br />

heritability analysis showed moderate heritability (h2= 10%),<br />

for factor 3 only. Conclusion/Discussion: The factor analytic<br />

results <strong>of</strong> this study are in line with a previous category-based<br />

FA study (Robertson et al., 2008) and a Latent Class Analytic<br />

study (Mathews et al., 2007). The heritability analyses suggest<br />

that the tic symptom dimension encompassing simple facial<br />

motor tics is heritable. These findings have consequences for<br />

future genetic studies in GTS, although the findings need<br />

replication in independent samples.


S11 PHARMACOGENETICS OF<br />

PSYCHIATRIC TREATMENTS – ARE WE<br />

READY FOR THE CLINIC?<br />

S11.1 PHARMACOGENETICS OF ATYPICAL<br />

ANTIPSYCHOTIC EXPOSURE, FOLATE<br />

METABOLISM, CARDIOVASCULAR DISEASE, AND<br />

ENDOTHELIAL FUNCTIONING<br />

V. Ellingrod* (1), T. Grove (1), S. Taylor (1), J. Moline (2),<br />

D. Miller (3)<br />

1. University <strong>of</strong> Michigan College <strong>of</strong> Pharmacy and School <strong>of</strong><br />

Medicine, Department <strong>of</strong> Psychiatry 2. Cleveland Clinics 3.<br />

University <strong>of</strong> Iowa Carver College <strong>of</strong> Medicine, Department<br />

<strong>of</strong> Psychiatry<br />

* vellingr@umich.edu<br />

Purpose: To examine the relationship between the<br />

methylenetetrahydr<strong>of</strong>olate reductase (MTHFR) 677C/T<br />

variant, metabolic syndrome, and endothelial functioning in<br />

schizophrenia and bipolar subjects receiving antipsychotics for<br />

≥ 6 months. Methods: 157 subjects were recruited from the<br />

Universities <strong>of</strong> Iowa and Michigan and screened<br />

cross-sectionally for the metabolic syndrome (NCEP ATP-III<br />

criteria), MTHFR genotype, plasma folate, vitamin B12,<br />

homocysteine, and endothelial functioning. Results: 60<br />

subjects (37%) met metabolic syndrome criteria. The group’s<br />

mean age (± s.d.) was 41.75 ± 11.65 years, 81% were<br />

Caucasian, 52% were male, mean BMI was 31.04 ± 7.53<br />

kg/m2 and 68% were receiving an atypical antipsychotic<br />

(AAP). There were no differences in age, gender, race, AAP<br />

exposure, or BMI between the genotype groups. Overall, a<br />

significant relationship was found between the TT genotype,<br />

AAP use, and metabolic syndrome (c2 = 6.44, p = 0.011). TT<br />

subjects receiving an AAP had a relative risk <strong>of</strong> 2.27 (95% CI:<br />

1.52-3.37) for metabolic syndrome. This was not found for<br />

subjects not receiving AAPs (c2 = 0.617, p = 0.43). Higher<br />

plasma folate concentrations resulted in lower homocysteine<br />

levels for the CC genotype group, whereas no relationship was<br />

found for T allele carriers (F = 6.971, df = 3,88, p = 0.0003).<br />

Data analysis for the endothelial functioning suggests higher<br />

folate plasma concentrations positively correlate with better<br />

endothelial functioning. Conclusion: Overall, results suggest<br />

that MTHFR genotype, AAPs exposure and folate, may be<br />

associated with metabolic complications and CVD in<br />

schizophrenia and bipolar subjects. However, due to the small<br />

sample size results should be taken cautiously.<br />

Acknowledgements: This project was supported by the NIMH<br />

(K08 MH64158), the NIH-NCRR, General Clinical Research<br />

Centers Program (M01-RR-59 and UL1RR024986), a<br />

University <strong>of</strong> Michigan College <strong>of</strong> Pharmacy Vahlteich<br />

Award, National Alliance on Research in Schizophrenia and<br />

Depression (NARSAD), and Washtenaw Community Health<br />

Organization (WCHO).<br />

S11.2 VARIANTS OF ABC TRANSPORTERS:<br />

CURRENT RELEVANCE IN PSYCHIATRY<br />

L. DeVane (1)<br />

1. <strong>Medical</strong> University <strong>of</strong> South Carolina<br />

* devaneL@musc.edu<br />

In addition to their role in gastrointestinal drug absorption,<br />

hepatic, and renal elimination, several ABC transporter<br />

proteins are expressed in tissues where their activity<br />

can influence the efficacy and safety <strong>of</strong> psychoactive drugs.<br />

These include P-glycoprotein (P-gp), breast cancer resistance<br />

protein (BCRP), multidrug resistance proteins 1-3 (MRP1-3),<br />

and others localized in endothelial cells <strong>of</strong> cerebral capillaries<br />

comprising the blood brain barrier (BBB). A similar cadre <strong>of</strong><br />

transporters in trophoblastic cells <strong>of</strong> the placenta modify fetal<br />

drug exposure during pregnancy, a period <strong>of</strong> great concern for<br />

the pharmacologic treatment <strong>of</strong> mental illness. About 35% <strong>of</strong><br />

women take at least one psychoactive drug during their<br />

pregnancy. The major antipsychotic and antidepressant drugs,<br />

and opioid analgesics, are substrates for one or more <strong>of</strong> these<br />

transporters. In rodents, both induction and inhibition <strong>of</strong><br />

transporter activity can be shown to affect brain and fetal<br />

exposure to psychoactive drugs as well as producing<br />

alterations in relevant pharmacologic activity. Emerging data<br />

support the choice <strong>of</strong> specific psychoactive drugs for clinical<br />

use should consider both their ABC transporter substrate<br />

status as well as patient genotype for transporter expression.


S11.3 PHARMACOGENETICS OF SSRI ASSOCIATED<br />

SEXUAL DYSFUNCTION AND MEDICATION<br />

INTOLERANCE<br />

J. Bishop* (1)<br />

1. Departments <strong>of</strong> Pharmacy Practice and Psychiatry,<br />

University <strong>of</strong> Illinois at Chicago<br />

* jbishop@uic.edu<br />

Sexual dysfunction is a common and disconcerting side effect<br />

<strong>of</strong> selective serotonin reuptake inhibitors (SSRIs) that<br />

influences a patient’s desire to continue long-term<br />

antidepressant treatment. Studies specifically assessing<br />

changes in sexual well-being over time illustrate that the<br />

incidence <strong>of</strong> sexual side effects from SSRIs ranges from 20 to<br />

70% depending on the characteristics <strong>of</strong> the study sample<br />

assessed. Developing strategies to predict who may be at the<br />

highest risk for adverse changes in their sexual well-being is<br />

an important step in improving the quality <strong>of</strong> life and<br />

treatment <strong>of</strong> patients who require antidepressant agents.<br />

Although initial studies investigating antidepressant<br />

pharmacogenetics focused on predicting treatment response,<br />

an increasing number <strong>of</strong> studies have recently focused on<br />

identifying genetic markers <strong>of</strong> medication intolerance. Studies<br />

investigating medication intolerance in general, as well as<br />

more specific lasting side-effects such as sexual dysfunction<br />

have identified common genetic variations in serotonin and<br />

glutamate system genes along with clinical variables that may<br />

predict risk for these outcomes in patients taking<br />

antidepressants. The results <strong>of</strong> studies investigating genetic<br />

variations in drug metabolism enzymes and their relationships<br />

to antidepressant-associated adverse effects have been mixed.<br />

Continued efforts to identify the relationships between genetic<br />

markers and antidepressant outcomes and to translate this<br />

knowledge to patient care has the potential to significantly<br />

improve the empiric selection <strong>of</strong> antidepressant agents and to<br />

minimize the risk for intolerable side effects that may<br />

compromise long term treatment.<br />

S11.4 VARIANTS OF THE SEROTONIN SYSTEM AND<br />

BONE MINERALIZATION IN BOYS TREATED WITH<br />

SELECTIVE SEROTONIN REUPTAKE INHIBITORS<br />

C. Calarge* (1), V. Ellingrod (2), M. Bliziotes (3), W.<br />

Coryell (1)<br />

1. University <strong>of</strong> Iowa 2. University <strong>of</strong> Michigan 3. Oregon<br />

Health and Science University<br />

* chadi-calarge@uiowa.edu<br />

Objective: To investigate whether genetic variants <strong>of</strong> the<br />

serotonin (5HT) system moderate the effect <strong>of</strong> selective<br />

serotonin reuptake inhibitors (SSRIs) on bone mineral density<br />

(BMD) in boys. Methods: Healthy, 7-17 year-old<br />

non-Hispanic Caucasian boys, treated with risperidone for ≥ 6<br />

months, were included in this analysis. We measured BMD at<br />

the lumbar spine using dual x-ray absorptiometry and total and<br />

trabecular BMD at the distal radius using peripheral<br />

quantitative computerized tomography. Genotyping <strong>of</strong><br />

selected polymorphisms <strong>of</strong> the genes transcribing the 5HT<br />

transporter (5HTTLPR and Stin2 VNTR) and 5HT receptors<br />

1A (A(-1019)C), 1B (G861C), 1D (T1350C), and 2A<br />

(A(-1438)G) was done following standard procedures.<br />

Results: The mean age <strong>of</strong> the sample was 12.1 years<br />

(SD=2.7). Of 71 boys enrolled, 41 (58%) received an SSRI<br />

for a median <strong>of</strong> 3.1 years (IQR=2.9). SSRI-treated boys were<br />

less physically active and more likely to have a depressive, an<br />

anxiety, or a pervasive developmental disorder compared to<br />

those not taking SSRIs. Using multiple linear regression<br />

analysis, after controlling for the relevant covariates, we found<br />

a trend for the 5-HTTLPR genotype x SSRI treatment<br />

interaction effect to be significant in predicting lumbar spine<br />

BMD Z-score (p=0.055). In fact, the largest detrimental effect<br />

<strong>of</strong> SSRIs on the lumbar BMD Z-score was noted among boys<br />

with the ‘ls’ genotype (Cohen’s d= 1.04, p


S11.5 CYP450 GENOTYPING IN THE CLINICAL USE<br />

OF ANTIPSYCHOTIC AND ANTIDEPRESSANT DRUG<br />

TREATMENT<br />

D. Müller* (1), R. Hwang (2), A. Tiwari (2), J. Sturgess (1),<br />

C. Zai (2), J. Lieberman (3), P. Richter (4), J. Kennedy (2)<br />

1. Pharmacogenetics Research Clinic, Centre for Addiction<br />

and Mental Health, Toronto, Canada 2. Neurogenetics Section,<br />

Centre for Addiction and Mental Health, Toronto, Canada 3.<br />

Dept. <strong>of</strong> Psychiatry, Columbia University, NY 4. Sunnybrook<br />

Health Sciences Center, Toronto<br />

* daniel_mueller@camh.net<br />

Introduction: A substantial number <strong>of</strong> antidepressant and<br />

antipsychotic medications are metabolized by the liver<br />

cytochrome P450 enzymes (CYP450) CYP2D6 and<br />

CYP2C19. An individual’s genetic makeup allows for<br />

classification by phenotype as poor (PM), intermediate (IM),<br />

extensive or normal (EM), or ultra rapid drug metabolizers<br />

(UM). The AmpliChip CYP450 Test provides genotyping<br />

<strong>of</strong> CYP2D6 and CYP2C19 enzymes using microarray<br />

technology which combines DNA hybridization with<br />

fluorescent labeling. Methods: In our study we used 74<br />

AmpliChips to analyze for an association<br />

between metabolizer with response pattern to antipsychotics (n<br />

= 23) and occurrence <strong>of</strong> tardive dyskinesia (TD; n = 12) in<br />

patients with schizophrenia as well as response to<br />

antidepressants in a sample <strong>of</strong> patients affected with OCD (n =<br />

39). <strong>Medical</strong> records were also reviewed in cases who were<br />

non-extensive metabolizers to assess the individual course <strong>of</strong><br />

treatment response and side effects. Response was assessed<br />

with clinical assessment scales such as CGI and<br />

PANSS whereas severity <strong>of</strong> TD was assessed by using AIMS<br />

scores. Results: Most patients were extensive metabolizers<br />

limiting statistical power. Overall, no association could be<br />

detected between patients' metabolizer status and response to<br />

antipsychotics, antidepressants or TD occurrence. However, a<br />

significant association was observed in patients who<br />

were CYP2D6 rapid metabolizers and non-response in<br />

treatment trials with various CYP2D6 metabolized<br />

antidepressant (p = .006). Conclusion: Our results suggest that<br />

genotyping <strong>of</strong> CYP450 genes is a useful tool to optimize<br />

clinical drug treatment for particular individuals.<br />

S12 UNDERSTANDING NORMAL<br />

VARIATION IN BRAIN STRUCTURE AND<br />

FUNCTION WILL<br />

INFORM PSYCHIATRIC GENETICS<br />

S12.1 HERITABILITY OF FMRI RESPONSE IN<br />

YOUNG ADULT TWINS<br />

N. Martin* (1)<br />

1. Queensland Inst <strong>Medical</strong> Research<br />

* nick.martin@qimr.edu.au<br />

While structural MRI studies consistently show a moderate to<br />

strong influence <strong>of</strong> genes on brain structure, it is largely<br />

unknown to what extent individual differences in neural<br />

activity, as captured by fMRI, are influenced by genetic and<br />

environmental factors. Working memory related brain<br />

activation has been widely studied using the N-back working<br />

memory task, and inefficient or abnormal function is evident<br />

in several neurodegenerative and neuropsychiatric disorders,<br />

and in the healthy siblings <strong>of</strong> patients for some disorders.<br />

Deficits in physiological function may be therefore not only<br />

associated with a disease, but also may reflect familial<br />

(possibly genetic) factors predisposing to the disorder. Using a<br />

regions <strong>of</strong> interest analysis we investigated the heritability <strong>of</strong><br />

brain activation during the N-back working memory task in a<br />

sample <strong>of</strong> 120 young adult twins and provided the first support<br />

that individual variation in working memory related brain<br />

activation is to some extent influenced by genes, with<br />

(non-significant) heritability estimates (14-30%) in the<br />

low-moderate range. Most recently we have extended this to a<br />

voxel-by-voxel analysis in a much larger sample (315 twins).<br />

Overall the MZ twin correlations are significantly larger than<br />

the DZ correlations, and there is good test-retest reliability for<br />

a sub-sample (N=40) <strong>of</strong> twins that were scanned twice. Our<br />

work establishes that there are significant genetic influences<br />

on working memory brain activation, and that there are also<br />

sizeable environmental effects on brain activation, which are<br />

largely due to unique environmental factors rather than<br />

measurement error.


S12.2 GENETIC ANALYSIS FOR NORMAL<br />

VARIATION IN COGNITION, SLEEP AND SEASONAL<br />

RHYTHMS: EXPERIENCES FROM TWIN AND<br />

POPULATION-BASED STUDIES FROM FINLAND<br />

T. Paunio* (1)<br />

1. Public Health Genomics Unit, National Institute for Health<br />

and Welfar<br />

* tiina.paunio@thl.fi<br />

Understanding genetic background for normal variation in<br />

brain structure and function is likely to provide insight into<br />

illnesses associated with related processes, such as impaired<br />

cognition in schizophrenia or disturbed sleep in mood<br />

disorders. We evaluated the contribution <strong>of</strong> AKT1, encoding<br />

the protein kinase B and previously associated with the genetic<br />

etiology <strong>of</strong> schizophrenia and bipolar disorder, to cognition<br />

and brain structure in a Finnish twin sample comprising both<br />

control pairs as well as pairs affected with schizophrenia or<br />

bipolar disorder. We found association <strong>of</strong> an AKT1 allele<br />

defined by a variation in the UTR with verbal learning and<br />

memory (P = 0.0005) and, moreover, a higher degree <strong>of</strong><br />

resemblance in pairs sharing the genotype. The same allele<br />

also associated with decreased gray matter density in medial<br />

and dorsolateral prefrontal cortex, suggesting that and AKT1<br />

may exert its effect on verbal cognitive processes via neural<br />

networks involving prefrontal cortex (Pietiläinen et al,<br />

2009).In an approach ultimately aiming to reveal genetic<br />

background for disturbed mood, we searched for genetic<br />

influences determining variation in sleep, known to be<br />

affected in most patients with a mood disorder. In our previous<br />

study in Finnish twins we found that depressed mood, as<br />

measured by life dissatisfaction, predicted a consistent pattern<br />

<strong>of</strong> life dissatisfaction (OR = 2.1), whereas life dissatisfaction<br />

did not consistently predict poor sleep. The shared genetic<br />

component between sleep quality and life dissatisfaction was<br />

relatively weak, consistent with the hypothesis that poor sleep<br />

may have direct effects on the brain, emotions, and mood<br />

(Paunio et al. 2009). We then searched for genetic background<br />

for variation in normal sleep length and seasonal rhythms in<br />

2200 Finnish individuals from a population-based Health 2000<br />

project by a genome-wide DNA association analysis. We<br />

found several new genes and variants that associated to total<br />

sleep length. Interestingly, the gene ontology analysis showed<br />

that sleep length and seasonal changes in mood share common<br />

genes and molecular pathways. Altogether, the data evidences<br />

for a strong genetic background for variation in normal sleep<br />

and seasonal rhythms and the findings will have implication in<br />

our understanding patophysiological processes <strong>of</strong> common<br />

diseases related to disturbed sleep, such as mood disorders.<br />

Acknowledgments: Pietiläinen OP, Loukola A,<br />

Tuulio-Henriksson A, Kieseppä T, Thompson P, Toga AW,<br />

van Erp TG, Silventoinen K, Soronen P, Hennah W, Turunen<br />

JA, Wedenoja J, Palo OM, Silander K, Lönnqvist J, Kaprio J,<br />

Cannon TD, and Peltonen L for the study on AKT1 in twins;<br />

Korhonen T, Hublin C, Partinen M, Kivimäki M, Koskenvuo<br />

M, and Kaprio J. for the analysis <strong>of</strong> life dissatisfaction and<br />

sleep quality in twins; Ollila HM, Partonen T, Kronholm E,<br />

Männistö S, Lönnqvist J, Salomaa V, Peltonen L, Perola M,<br />

and Porkka-Heiskanen T for the study on sleep length and<br />

seasonal rhythms in Health 2000; Academy <strong>of</strong> Finland (grant<br />

124404 to Paunio T) References: Pietiläinen OP, Paunio T,<br />

Loukola A, et al. (2009) Association <strong>of</strong> AKT1 with verbal<br />

learning, verbal memory, and regional cortical gray matter<br />

density in twins. Am J Med Genet B Neuropsychiatr Genet.<br />

2009 Jul 5;150B(5):683-92. Paunio T, Korhonen T, Hublin C,<br />

et al. (2009) Longitudinal study on causality and genetic risk<br />

factors for the association between poor sleep and life<br />

dissatisfaction in a nationwide cohort <strong>of</strong> twins. Am J<br />

Epidemiol. 169(2):206-13.<br />

S12.3 A GENETIC INVESTIGATION OF<br />

SCHIZOPHRENIA ENDOPHENOTYPES IN A<br />

NEPALESE POPULATION GENETIC ISOLATE<br />

T. Sitnikova (1), M. Hall (2), J. Subedi (3), S. Ojha (4), S.<br />

Park (5), J. VandeBerg (6), J. Blangero (6), S.<br />

Williams-Blangero (6), S. Santangelo* (1, 7)<br />

1. Harvard <strong>Medical</strong> School, Dept Psychiatry, Boston, MA,<br />

USA 2. Psychology Research Laboratory, McLean Hospital,<br />

Belmont, MA, USA 3. Miami University, Dept. Sociology,<br />

Gerontology, Oxford, OH, USA 4. Tribhuvan University<br />

Institute <strong>of</strong> Medicine, Dept. Psychiatry, Kathmandu, Nepal 5.<br />

Dept. Psychology Vanderbilt University, Nashville, TN, USA<br />

6. Southwest Foundation for Biomedical Research, San<br />

Antonio, TX, USA 7. Center for Human Genetic Research,<br />

Massachusetts General Hospital, Boston, MA, USA<br />

* ssantangelo@pngu.mgh.harvard.edu<br />

Schizophrenia endophenotypes were measured in a population<br />

genetic isolate in Jiri, Nepal, encompassing over 2500<br />

surveyed individuals in one extended pedigree. This is both an<br />

epidemiological sample (complete ascertainment <strong>of</strong> 4 <strong>of</strong> 7<br />

villages) and a pedigree sample. The amplitude and latency <strong>of</strong><br />

the N100, N200, and P300 electrophysiological evoked<br />

potentials (ERPs), as well as the power <strong>of</strong> evoked and induced<br />

oscillatory brain activity in the gamma-band range (35-45Hz),<br />

were measured during an auditory "oddball" paradigm. The<br />

P50 ERP amplitude was measured in a double-click paradigm.<br />

In addition, performance was tested on cognitive tasks,<br />

including go-no-go, set shifting, strategic target detection, and<br />

spatial working memory. Several other tasks in the original<br />

battery were discontinued when they were found to be<br />

excessively language- or culture-bound. Data collection just<br />

ended, and data are being analyzed for the 960 individuals<br />

who participated in these studies. Each <strong>of</strong> the heritable<br />

measures is subjected to a genome-wide linkage analysis,<br />

using a 10 cM microsatellite marker set. A new genome scan,<br />

using the Illumina v. 610 SNP array, is in progress. Analysis<br />

<strong>of</strong> the cognitive test data is ongoing. However, we found<br />

significant linkage to at least one <strong>of</strong> the ERPs (P300 amplitude<br />

measured at the Pz electrode, at chromosome 2p15, reported<br />

elsewhere at this meeting) and suggestive linkage to others<br />

(e.g. induced gamma measured at the P4 electrode at 2p13).<br />

Genes in this region include sepiapterin reductase, an enzyme<br />

required forsynthesis <strong>of</strong> tetrahydrobiopterin (BH4), an<br />

essential c<strong>of</strong>actor for synthesis <strong>of</strong> many neurotransmitters,<br />

including serotonin.


S12.4 GENOME-WIDE COMBINED<br />

LINKAGE/ASSOCIATION SCAN LOCALIZES TWO<br />

QTLS INFLUENCING HUMAN CAUDATE NUCLEUS<br />

VOLUME<br />

D. Glahn* (1), A. Winkler (1), J. Curran (2), M. Carless (2), J.<br />

Charlesworth (2), M. Johnson (2), H. Göring (2), T. Dyer (2),<br />

E. Moses (2), L. Almasy (2), P. Fox (3), P. Kochunov (3), R.<br />

Duggirala (2), J. Blangero (2)<br />

1. Yale University & Olin Neuropsychiatric Research Center,<br />

Institute <strong>of</strong> Living 2. Department <strong>of</strong> Genetics, Southwest<br />

Foundation for Biomedical Research 3. Research Imaging<br />

Center, UT Health Science Center at San Antonio<br />

* david.glahn@yale.edu<br />

The caudate nucleus (CN), a brain region located within the<br />

basal ganglia, is associated with higher-order motor control,<br />

learning and memory, feedback processing, language and<br />

other executive functions. The CN, which is innervated by<br />

dopaminergic neurons, is thought to be a locus <strong>of</strong> pathology<br />

for several neurological and psychiatric disorders (e.g.<br />

Schizophrenia and Bipolar Disorder). While the size and<br />

function <strong>of</strong> the CN are thought to be under genetic control, the<br />

genes influencing the volume <strong>of</strong> this brain region are<br />

unknown. To better understand the genetic basis <strong>of</strong> CN<br />

volume variation in the normal population, we undertook the<br />

first large-scale genome-wide search for genes influencing<br />

human brain structure. We collected high-resolution<br />

T1-weighted MRI scans from approximately 400<br />

Mexican-American individuals from 30 extended pedigrees<br />

who participated in the Genetics <strong>of</strong> Brain Structure and<br />

Function study and who have Illumina HumanHap 550K SNP<br />

genotyping available. Imaging data was analyzed with<br />

FreeSurfer and genetic analyses were performed with SOLAR.<br />

The heritability <strong>of</strong> CN volume was estimated to be 0.685<br />

(p=2.3×10-10). Combined linkage/association analysis<br />

identified two regions exhibiting genome-wide significant<br />

evidence for harboring QTLs influencing caudate volume.<br />

One QTL was localized on chromosome 7p22 with a strongly<br />

associated SNP in the PRKAR1B gene (nominal p-value =<br />

2.3×10-8, genome-wide p-value = 0.02). This gene codes for<br />

the protein kinase, cAMP-dependent, regulatory, type I, beta,<br />

which is involved in brain development and linked to poor<br />

memory. Examination <strong>of</strong> the potential pleiotropic associations<br />

<strong>of</strong> this SNP with neurocognitive function revealed significant<br />

associations with processing speed (p = 0.005), episodic<br />

memory (p = 0.04) and working memory (p=0.05). Thus, this<br />

QTL appears to influence both brain structure and function. A<br />

second QTL influencing caudate volume was localized to<br />

chromosomal region 2p12 with strong association to a SNP<br />

near the LRRTM4 gene (nominal p-value = 1.3×10-7,<br />

genome-wide p-value = 0.049) which codes for a neuronal<br />

protein <strong>of</strong> unknown function. Given the strength <strong>of</strong> these<br />

signals, we are currently deeply sequencing these genes to<br />

identify potential functional variants. Dissection <strong>of</strong> the genetic<br />

basis <strong>of</strong> normal human brain structural variation should lead to<br />

the identification <strong>of</strong> genes likely to be involved in disorders <strong>of</strong><br />

brain structure/function.<br />

S12.5 GENOMIC AND TRANSCRIPTOMIC<br />

APPROACHES TO IDENTIFYING GENES<br />

INFLUENCING COGNITION AND NEUROANATOMY<br />

L. Almasy* (1), M. Carless (1), J. Curran (1), M. Zlojutro (1),<br />

H. Goring (1), T. Dyer (1), R. Duggirala (1), D. Glahn (2), J.<br />

Blangero (1)<br />

1. Southwest Foundation for Biomedical Research, San<br />

Antonio, TX, USA 2. Yale University, New Haven, CT, USA<br />

* almasy@sfbrgenetics.org<br />

This presentation will discuss the rationale for studying<br />

normal variation in brain structure and function and how such<br />

studies provide insight into genetics <strong>of</strong> psychiatric disorders.<br />

Examples will be provided from the Genetics <strong>of</strong> Brain<br />

Structure and Function Study, which makes use <strong>of</strong> an existing<br />

resource <strong>of</strong> large Mexican American families in San Antonio,<br />

Texas, for which a GWAS and genome-wide transcription<br />

pr<strong>of</strong>iling in lymphocytes have already been completed.<br />

Individuals from these families are currently being recalled for<br />

cognitive testing, structural MRI, and psychiatric screening.<br />

Currently this data is available for 1000 individuals. This<br />

resource is being used to 1) document heritability <strong>of</strong> potential<br />

psychiatric endophenotypes; 2) assess pleiotropic influences<br />

among these quantitiative risk factors and between<br />

endophenotypes and high prevalence disorders such as<br />

depression and substance abuse; 3) localize genes influencing<br />

normal variation in brain structure and function in which<br />

other, more extreme, genetic variants might influence risk <strong>of</strong><br />

psychiatric disorder; 4) identify upstream genes regulating<br />

expression <strong>of</strong> previously identified genes important for<br />

psychiatric phenotypes; 5) identify downstream genes whose<br />

expression is regulated by genes <strong>of</strong> interest for psychiatric<br />

disorders.


S13 ANOREXIA NERVOSA – GENES OR<br />

JEANS<br />

S13.1 A GENOME-WIDE ASSOCIATION STUDY IN<br />

PATIENTS WITH ANOREXIA NERVOSA<br />

K. Wang* (1), H. Zhang (1), C. Bloss (2), N. Schork (2), W.<br />

Berrettini (3), H. Hakonaron (1), P. Collaborative Group (4)<br />

1. Children's Hospital <strong>of</strong> Philadelphia 2. Scripps Translational<br />

Science Institute 3. University <strong>of</strong> Pennsylvania 4. Price<br />

Foundation<br />

* tumorim@gmail.com<br />

Anorexia nervosa (AN) is a serious psychiatric illness<br />

characterized by restricted eating and obsessive fears <strong>of</strong> being<br />

fat, with an inability to maintain a normal healthy body<br />

weight. The current prevalence estimates ranged from 0.1% to<br />

5.7% in female subjects in Western countries. The heritability<br />

<strong>of</strong> anorexia is estimated to be 56%, with sibling relative risk <strong>of</strong><br />

11.3, implicating a strong genetic component in anorexia<br />

susceptibility, most <strong>of</strong> which remains uncovered. To identify<br />

genetic risk factors for anorexia, we carried out a<br />

genome-wide association study (GWAS) on 1,016 female<br />

cases and 1,796 female control subjects, all <strong>of</strong> whom were <strong>of</strong><br />

European ancestry and were genotyped on the Illumina<br />

HumanHap610 platform. The top association results were<br />

examined in a separate cohort <strong>of</strong> 24 cases and 648 control<br />

subjects <strong>of</strong> European ancestry genotyped on the<br />

HumanHap550 platform. Several SNPs residing between<br />

CDH10 (cadherin 10) and CDH9 (cadherin 9) – a locus<br />

previously implicated in autism spectrum disorders -<br />

demonstrated genome-wide significance (most significant<br />

P=1.5x10-8). Analysis <strong>of</strong> previously reported AN-associated<br />

SNPs replicated association with OPRD1 (rs1042114,<br />

P=0.02), but failed to detect association with HTR1D<br />

(rs7532266, P=0.83). Furthermore, nominal significance was<br />

observed for SNPs near DRD2 (rs10891556, P=0.0023), near<br />

HTR2A (rs7331274, P=0.0025), near HTR1B (rs6454038,<br />

P=0.0048), within HTR7 (rs7920627, P=0.0048), near DRD3<br />

(rs9825563 , P=0.0087), near CNR1 (rs9344757, P=0.0058),<br />

but they do not survive corrections for multiple testing <strong>of</strong><br />

SNPs within or near each gene. Taken together, we have<br />

identified a genome-wide significant locus in AN and<br />

replicated OPRD1, demonstrating the utility <strong>of</strong><br />

whole-genome, unbiased survey in identifying novel genomic<br />

regions and confirming previously reported candidate genes.<br />

S13.2 GENOME-WIDE ASSOCIATION OF<br />

PERSONALITY TRAITS IN INDIVIDUALS WITH<br />

ANOREXIA NERVOSA<br />

C. Bloss* (1), K. Wang (2), W. Berrettini (3), A. Bergen (4),<br />

P. Magistretti (5), W. Kaye (6), H. Hakonarson (2), N. Schork<br />

(1)<br />

1. Scripps Translational Science Institute 2. Children's<br />

Hospital <strong>of</strong> Philadelphia 3. University <strong>of</strong> Pennsylvania 4. SRI<br />

International 5. University <strong>of</strong> Lausanne 6. University <strong>of</strong><br />

California, San Diego<br />

* cbloss@scripps.edu<br />

Several studies have shown that certain facets <strong>of</strong> personality<br />

differ markedly between women with anorexia nervosa (AN)<br />

and controls, as well as between women with different<br />

subtypes <strong>of</strong> AN. As such, certain temperament traits have<br />

been hypothesized to both predispose to AN, as well as predict<br />

illness course and response to treatment. Given these<br />

observations, coupled with the known moderate heritability <strong>of</strong><br />

temperament, we investigated the extent to which common<br />

genetic polymorphisms may underlie aspects <strong>of</strong> personality in<br />

this disorder by conducting a genome-wide association study<br />

<strong>of</strong> the Temperament and Character Inventory (TCI) in women<br />

with AN. Participants were n = 1,001 women with AN<br />

enrolled in the Price Foundation Collaborative Study <strong>of</strong><br />

Anorexia and Bulimia Nervosa. Participants were all <strong>of</strong><br />

European ancestry and were administered the TCI and<br />

genotyped on the Illumina HumanHap610 platform. In<br />

addition to standard univariate quantitative trait association<br />

analyses conducted with each temperament scale <strong>of</strong> the TCI,<br />

several multivariate analytic approaches were also leveraged<br />

to assess genetic association with TCI pr<strong>of</strong>iles. Standard<br />

univariate analyses revealed different associations (p < 5 x<br />

10-6) for each scale <strong>of</strong> the TCI, including SNPs near RNF165<br />

for harm avoidance, ZNF645 for novelty seeking, BTG3 for<br />

reward dependence, and RBAK for persistence. Furthermore,<br />

analyses that included relevant covariates, as well as cluster<br />

and other multivariate approaches identified additional regions<br />

<strong>of</strong> interest. Findings suggest novel genes that may underlie<br />

single dimensions <strong>of</strong> personality, as well as personality<br />

pr<strong>of</strong>iles in anorexia nervosa.


S13.3 GENETICALLY MEDIATED, PRE-MORBID<br />

VULNERABILITIES OF TEMPERAMENT AND<br />

PERSONALITY IN ANOREXIA AND BULIMIA<br />

NERVOSA<br />

W. Kaye* (1)<br />

1. University <strong>of</strong> California San Diego<br />

* whkaye@gmail.com<br />

Many individuals desire to diet and lose weight, but relatively<br />

few develop an eating disorder. Recent studies show that<br />

certain childhood temperament and personality traits such as<br />

negative emotionality, harm avoidance, perfectionism,<br />

inhibition, drive for thinness, altered interoceptive awareness,<br />

and obsessive-compulsive personality, create a vulnerability<br />

for developing anorexia nervosa (AN) and bulimia nervosa<br />

(BN). Moreover, studies done on 3 continents have shown that<br />

for AN and BN individuals with a lifetime history <strong>of</strong> an<br />

anxiety disorder diagnosis, the anxiety disorder most <strong>of</strong>ten<br />

began in childhood before the onset <strong>of</strong> the ED. Such<br />

symptoms may be susceptibility factors that make people<br />

vulnerable to developing an ED. Malnutrition tends to<br />

exaggerate these premorbid behavioral traits after the onset <strong>of</strong><br />

the illness. The process <strong>of</strong> recovery in AN is poorly<br />

understood and, in most cases, protracted. Still,<br />

approximately 50% to 70% <strong>of</strong> affected individuals will<br />

eventually have complete or moderate resolution <strong>of</strong> the illness,<br />

<strong>of</strong>ten in the early to mid 20’s. It is important to emphasize<br />

that temperament and personality traits such as negative<br />

emotionality, harm avoidance and perfectionism, obsessional<br />

behaviors (particularly symmetry, exactness, and order) persist<br />

after recovery from both AN and BN and are similar to the<br />

symptoms described premorbidly in childhood. Compared to<br />

the ill state, symptoms in REC AN and BN tend to be mild to<br />

moderate. Interestingly, REC AN and BN tend to be more<br />

alike than different on many <strong>of</strong> these measures, although there<br />

are some differences on factors related to impulse control or<br />

stimuli seeking, such as novelty seeking. In summary,<br />

individuals with restricting type AN are more likely to have<br />

restricted eating, constricted affect and emotional mood<br />

expression, and impulse over control, as well as personality<br />

traits <strong>of</strong> marked rigidity, conformity, and reduced social<br />

spontaneity. Individuals with BN may show similar traits, but<br />

in addition, may exhibit histories <strong>of</strong> episodic overeating,<br />

extremes <strong>of</strong> intense affect, and impulse dysregulation. AN<br />

and BN most commonly develop during adolescence or young<br />

adulthood 19 in proximity to puberty. Adolescence is a time<br />

<strong>of</strong> pr<strong>of</strong>ound biological, psychological and sociocultural<br />

change, and it demands a considerable degree <strong>of</strong> flexibility to<br />

successfully manage the transition into adulthood.<br />

Psychologically, change may challenge the rigidity <strong>of</strong> those at<br />

risk for AN and BN, and thus open a window <strong>of</strong> vulnerability.<br />

Thus these vulnerabilities, which are biologically mediated,<br />

may significantly enhance the risk <strong>of</strong> onset <strong>of</strong> an ED,<br />

particularly in women.<br />

S13.4 THE PRICE FOUNDATION STUDIES – A<br />

RESOURCE FOR ANOREXIA NERVOSA RESEARCH<br />

A. Bergen* (1), The Price Foundation Collaborative Group<br />

1. SRI International<br />

* andrew.bergen@sri.com<br />

Over a period <strong>of</strong> ten years, a multinational collaboration<br />

ascertained over 1000 unrelated individuals with a DSM-IV<br />

diagnosis <strong>of</strong> anorexia nervosa in three separate studies<br />

encompassing family (sib-pair and trio) and unrelated<br />

collection methods, and nearly 700 unrelated individuals<br />

screened for DSM-IV diagnoses. A series <strong>of</strong> linkage studies in<br />

pedigrees and association analyses in both family based and<br />

unrelated individuals have been published. In particular, a<br />

series <strong>of</strong> linkage and association studies have identified<br />

significant linkage in the chr1p34-36 region and association at<br />

the genes coding for the delta opioid and 1D serotonin<br />

receptors in samples ascertained by this collaboration and in<br />

an independent sample. Despite this early example <strong>of</strong> a<br />

replicated association, candidate gene association studies in<br />

the world anorexia nervosa literature have been characterized<br />

by less than optimal sample sizes and incomplete investigation<br />

<strong>of</strong> gene variation at candidates. These limitations are being<br />

addressed using candidate gene and genome wide association<br />

approaches focusing on common variation, and candidate gene<br />

resequencing studies focusing on both common and rare<br />

variation, though limitations with respect to sample size and<br />

power continue to influence this field in common with all<br />

fields <strong>of</strong> genetic research, suggesting that additional<br />

technologies and collaborations will be necessary to promote<br />

further discovery. For example, biospecimens (DNA,<br />

lymphoblastoid cell lines, viably frozen cells) from both<br />

affected and unaffected individuals recruited in the Price<br />

Foundation studies are available and <strong>of</strong>fer the potential for<br />

genetical genomics studies, with appropriate and extensive<br />

control assessments to avoid confounding due to participant,<br />

biospecimen processing, cell line and technical characteristics.


S13.5 LARGE-SCALE CANDIDATE GENE<br />

RESEQUENCING IN ANOREXIA NERVOSA<br />

A. Scott* (1), C. Bloss (1), S. Murray (2), R. Tewhey (3), V.<br />

Bansal (1), O. Libiger (1), A. Torkamani (1), M. Shaw (2), J.<br />

Bishop (4), T. Price Foundation Collaborative Group, P.<br />

Magistretti (5), A. Bergen (6), W. Berrettini (7), A. Harris (8),<br />

W. Kaye (9), E. Topol (2), N. Schork (2)<br />

1. Scripps Translational Science Institute and The Scripps<br />

Research Institute 2. Scripps Genomic Medicine, Scripps<br />

Translational Science Institute and The Scripps Research<br />

Institute 3. Divison <strong>of</strong> Biological Sciences, University <strong>of</strong><br />

California, San Diego and The Scripps Research Institute 4.<br />

Life Technologies 5. Ecole Polytechnique Federale De<br />

Lausanne, Switzerland6. Molecular Genetics, SRI<br />

International 7. Center for Neurobiology and Behavior,<br />

University <strong>of</strong> Pennsylvania 8. Life Technologies 9. Eating<br />

Disorder Treatment & Research Program, Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> California, San Diego<br />

* ashleys@scripps.edu<br />

Anorexia nervosa (AN) is a complex psychiatric illness<br />

characterized by restricted eating patterns and an intense fear<br />

<strong>of</strong> weight gain. Current estimates place the incidence <strong>of</strong> AN<br />

between 3 and 8 per 100,000 persons per year, affecting<br />

women significantly more <strong>of</strong>ten than men. Although there is<br />

evidence <strong>of</strong> significant heritability (0.58-0.76) and genetic<br />

liability for anorexia, previous candidate gene and current<br />

genome-wide association (GWA) studies have elucidated only<br />

a small fraction <strong>of</strong> the genetic vulnerability for the disorder.<br />

Therefore, we are undertaking a large-scale sequence-based<br />

association study <strong>of</strong> anorexia to uncover highly penetrant rare<br />

variants associated with the disorder. Over the past 15 years,<br />

the Price Foundation has supported the collection <strong>of</strong> a large<br />

cohort <strong>of</strong> individuals with AN. To discover additional genetic<br />

variants that contribute to AN, we are resequencing<br />

approximately 1 Mb <strong>of</strong> exomic DNA in 400 women with<br />

restricting-subtype AN (RAN) and 100 control women using<br />

the Life Technologies SOLiD next-generation sequencing<br />

platform. Over 100 genes and genomic regions were selected<br />

based on results from previous GWA and candidate gene<br />

studies, as well as a priori neurobiologically relevant candidate<br />

genes. Targeted capture and amplification <strong>of</strong> conserved and<br />

exonic regions within these genes was performed using<br />

RainDance microdroplet-based emulsion PCR. In addition to<br />

case-control comparisons to identify sequence-based<br />

associations with RAN, we are also investigating<br />

neurobehavioral subclinical phenotypes within the RAN cases.<br />

This study has the potential to provide new insights into the<br />

spectrum <strong>of</strong> genetic variation in anorexia nervosa.<br />

S14 SUB-PHENOTYPES AND MACRO-<br />

PHENOTYPES IN MOOD DISORDERS<br />

S14.1 APPROACHES TO ANALYZING GWAS DATA<br />

IN BIPOLAR DISORDER USING INFORMATION<br />

ABOUT ITS CLINICAL HETEROGENEITY<br />

P. Zandi* (1), M. Pirooznia (2), P. Belmonte (2), F. Seiffudin<br />

(2), J. Judy (2), T. Phenome Group, J. Potash (2)<br />

1. Johns Hopkins University Bloomberg School <strong>of</strong> Public<br />

Health 2. Johns Hopkins University School <strong>of</strong> Medicine<br />

* pzandi@jhsph.edu<br />

Family, twin and adoption studies show that genetic factors<br />

play a significant role in the etiology <strong>of</strong> bipolar disorder.<br />

However, recent genome-wide association studies (GWAS)<br />

have yet to convincingly identify individual susceptibility<br />

genes with large effects on risk. This may be due to the fact<br />

that bipolar disorder is likely caused by multiple different<br />

genes with small effect sizes acting in complex pathways to<br />

illness. It is notable that bipolar disorder is clinically very<br />

heterogeneous. For example, data suggests the age at onset<br />

ranges anywhere from adolescence to old age with a median<br />

between 18-21 years <strong>of</strong> age, and that up to 58% <strong>of</strong> bipolar<br />

patients present with psychotic symptoms, 61% have a<br />

substance abuse disorder, 53% suffer with a co-morbid anxiety<br />

disorder, and 15% die from suicide. Some investigators have<br />

proposed that this clinical heterogeneity may reflect the<br />

underlying genetic heterogeneity <strong>of</strong> the disorder. Therefore,<br />

analytic methods that utilize information about the clinical<br />

heterogeneity <strong>of</strong> the disorder may facilitate the identification<br />

<strong>of</strong> relevant susceptibility genes. We explore three different<br />

genetic models that may explain how multiple genes<br />

contribute to the apparent clinical heterogeneity <strong>of</strong> bipolar<br />

disorder, including a heterogeneity model, a modifier model,<br />

and an epistatic model. We examine approaches for analyzing<br />

GWAS data to identify susceptibility genes and draw<br />

inferences about the model for how they contribute to risk.<br />

The approaches are compared through simulation and by<br />

application to real data.


S14.2 A GENOME- WIDE ASSOCIATION STUDY OF<br />

BIPOLAR DISORDER AND CO-MORBID MIGRAINE:<br />

IDENTIFICATION AND REPLICATION OF GENETIC<br />

ASSOCIATIONS IN THE 13Q14.1 REGION<br />

HARBOURING THE GENE KIAA0564<br />

K. Oedegaard* (1), T. Greenwood (2), S. Johansson (3), K.<br />

Jacobsen (3), A. Halmoy (3), O. Fasmer (4), H. Akiskal (2, 5),<br />

Bipolar Genome Study (BiGS), J. Haavik (9), J. Kelsoe (2, 5)<br />

1. University <strong>of</strong> Bergen, Moodnet Research Group, Haukeland<br />

University Hospital, Norway 2. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> California San Diego, La Jolla, CA, USA 3.<br />

Department <strong>of</strong> Biomedicine, University <strong>of</strong> Bergen, Norway 4.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Bergen, Norway 5.<br />

Department <strong>of</strong> Psychiatry, VA San Diego Healthcare System,<br />

La Jolla, CA, USA<br />

* keti@haukeland.no<br />

Both migraine and Bipolar Disorder (BPAD) are complex<br />

phenotypes with significant genetic and non-genetic<br />

components. Epidemiological and clinical studies have<br />

demonstrated a high degree <strong>of</strong> co-morbidity between migraine<br />

and BPAD. Numerous genome- wide linkage studies in BPAD<br />

and migraine have shown overlapping regions <strong>of</strong> linkage on<br />

chromosomes, and two functionally similar voltage-dependent<br />

calcium channels, CACNA1A and CACNA1C, have been<br />

identified in familial hemiplegic migraine and recently in three<br />

BPAD genome-wide association studies (GWAS),<br />

respectively. To identify susceptibility factors for the<br />

BPAD/migraine phenotype, we conducted a GWAS in 1001<br />

cases with bipolar disorder collected through the Foundation<br />

for the National Institutes <strong>of</strong> Health Genetic Information<br />

Association Network (GAIN) initiative. We compared BPAD<br />

patients without any headache (n= 699) to BPAD patients with<br />

doctor diagnosed migraine (n=56). The strongest evidence for<br />

association was found for several SNPs in a 317 kb region<br />

encompassing the uncharacterized gene KIAA0564 (e.g.<br />

rs9566845 (OR=4.98 (95%CI: 2.6-9.48), p= 7.7 x10-8 ) and<br />

rs9566867 (p= 8,2 x 10-8 )). Furthermore, marker rs9566845<br />

was genotyped and found associated with migraine in an<br />

independent Norwegian sample <strong>of</strong> adult ADHD patients with<br />

and without co-morbid migraine (n=131 and n=324<br />

respectively), OR=2.42 (1.18-4.97), p=0.013. In conclusion,<br />

this is the first GWAS examining patients with bipolar<br />

disorder and co-morbid migraine, and the first migraine<br />

GWAS in any sample. Together these data suggest that<br />

genetic variants in the KIAA0564 gene region may predispose<br />

to migraine headaches in subgroups <strong>of</strong> patients with both<br />

BPAD and ADHD.<br />

S14.3 A GENOME-WIDE ASSOCIATION STUDY OF<br />

AGE AT ONSET IN A COMBINED BIGS/GERMAN<br />

BIPOLAR DISORDER SAMPLE<br />

P. Belmonte (1), D. Jancic (1), T. Schulze (2), Bipolar<br />

Genome Study (BiGS), S. Cichon (3), M. Nothen (3), R.<br />

Breuer (4), F. McMahon (2), M. Rietschel (4), J. Potash* (1),<br />

P. Zandi (5)<br />

1. Johns Hopkins School <strong>of</strong> Medicine 2. National Institute <strong>of</strong><br />

Mental Health 3. Life and Brain Center, University <strong>of</strong> Bonn 4.<br />

Central Institute <strong>of</strong> Mental Health, Mannheim 5. Johns<br />

Hopkins Bloomberg School <strong>of</strong> Public Health<br />

* jpotash@jhmi.edu<br />

Age at onset <strong>of</strong> bipolar disorder (BP) aggregates in families,<br />

with affected relatives <strong>of</strong> early onset probands being 4.5 times<br />

more likely than others to have early onset themselves.<br />

Linkage regions related to age at onset in BP have been<br />

identified on chromosomes 21q22 and 18p11. We initially<br />

examined age at onset data from a genome-wide association<br />

study <strong>of</strong> BP in the Genetic Association Information Network<br />

(GAIN) BP sub-sample <strong>of</strong> the Bipolar Genetics Study<br />

(BiGS). This sample consisted <strong>of</strong> 1,034 controls and 1,001 BP<br />

cases, <strong>of</strong> whom 681 have early onset (age at onset ≤ 21) and<br />

285 have late onset (age at onset > 21). Genotyping was<br />

performed using Affymetrix Genome-Wide Human SNP<br />

Array 6.0 and 729,779 SNPs passing QC were examined for<br />

association with age at onset in BP. We analyzed the data<br />

using polytomous logistic regression with three outcome<br />

groups: early onset BP, late onset BP and controls. Our initial<br />

focus was on the early vs. late onset BP comparison, which<br />

yielded 375 loci at p


S14.4 GENOME-WIDE ASSOCIATION STUDIES<br />

IDENTIFY A LOCUS ON CHROMOSOME 3P THAT<br />

INFLUENCES BOTH BIPOLAR AND UNIPOLAR<br />

DISORDER<br />

F. McMahon* (1), N. Akula (1), T. Schulze (1), S.<br />

Detera-Wadleigh (1), C. Steele (1), R. Breuer (2), J.<br />

Strohmaier (2), J. Wendland (1), T. Mühleisen (3), W. Maier<br />

(4), M. Nöthen (5), S. Cichon (5), M. Rietschel (2)<br />

1. NIMH 2. Central Institute <strong>of</strong> Mental Health, Mannheim<br />

3. Dept <strong>of</strong> Genomics, Life & Brain Center, Univ <strong>of</strong> Bonn 4.<br />

Dept <strong>of</strong> Psychiatry, Univ <strong>of</strong> Bonn 5. Dept <strong>of</strong> Genomics, Life<br />

& Brain Center, Univ <strong>of</strong> Bonn<br />

* mcmahonf@mail.nih.gov<br />

The major mood disorders, which include bipolar disorder and<br />

major depressive disorder, are substantially heritable, but few<br />

risk loci have been identified. We performed a meta-analysis<br />

<strong>of</strong> five case-control samples assessed for the presence <strong>of</strong> a<br />

major mood disorder, totaling 13,743 unique individuals<br />

genotyped with approximately 500,000 to 1 million single<br />

nucleotide polymorphism (SNP) markers on high-density<br />

arrays. Imputation was used to compare SNPs across<br />

platforms, and test statistics were weighted for sample size.<br />

We found genome-wide significant evidence that SNPs in a<br />

region <strong>of</strong> chromosome 3p21.1—containing the known genes<br />

STAB1, NT5DC2, PBRM1, NEK4, SPCS1, GNL3,<br />

GLT8D1,and ITIH1, 3, and 4—were associated with major<br />

mood disorders. The SNP rs2251219 returned the smallest<br />

meta-analysis p-value, 3.63 x 10-8, with a pooled odds ratio <strong>of</strong><br />

0.87. These results imply that one or more genes in 3p21.1<br />

play an etiologic role in major mood disorders and suggest<br />

that bipolar disorder and major depressive disorder share<br />

genetic risk factors.<br />

S14.5 GWAS AND META-ANALYSIS RESULTS FOR<br />

PSYCHOSIS<br />

N. Craddock* (1)<br />

1. Cardiff University<br />

* craddockn@cardiff.ac.uk<br />

Even before the advent <strong>of</strong> GWAS, the pattern <strong>of</strong> molecular<br />

genetic findings demonstrated evidence for an overlap in<br />

genetic susceptibility across the traditional classification<br />

categories - including association findings at DISC1 and<br />

NRG1. Genome-wide association studies (GWAS) now<br />

provide greater power to explore the relationship between<br />

mood and psychotic illness. Several large-scale studies have<br />

been undertaken, including those by our collaborators at the<br />

Broad Institute <strong>of</strong> MIT and Harvard and the GAIN<br />

Collaboration. Within the context <strong>of</strong> the Wellcome Trust Case<br />

Control Consortium (WTCCC) we studied 2700<br />

mood-psychosis cases and 3000 controls. In our data the<br />

genome-wide significant associations at CACNA1C in bipolar<br />

disorder and ZNF804A in schizophrenia show evidence for a<br />

contribution to susceptibility across the traditional diagnostic<br />

boundaries. Using both specific single risk alleles and also<br />

composite “polygenic scores”, susceptibility to broadly<br />

defined psychotic bipolar disorder does not appear to be<br />

related in a simple way to susceptibility to schizophrenia.<br />

However, there is evidence for the existence <strong>of</strong> some<br />

relatively specific relationships between genotype and<br />

psychopathology. For example, in our dataset variation at<br />

GABAA receptor genes is associated with susceptibility to a<br />

form <strong>of</strong> illness with mixed features <strong>of</strong> schizophrenia and<br />

bipolar disorder. Recent genetic analyses <strong>of</strong> the WTCCC and<br />

the larger collaborative datasets will be considered and the<br />

implications for understanding psychotic bipolar disorder<br />

discussed.


S14.6 CLINICAL HETEROGENEITY IN MOOD<br />

DISORDERS<br />

J. Smoller* (1)<br />

1. Massachusetts General Hospital<br />

* jsmoller@hms.harvard.edu<br />

The modern distinctions between psychotic and mood<br />

disorders date to Kraepelin’s foundational descriptions <strong>of</strong><br />

dementia praecox and manic depressive insanity more than a<br />

century ago and Leonhard’s later division <strong>of</strong> bipolar and<br />

unipolar disorders. These distinctions continue to be<br />

fundamental organizing principles for psychiatric nosology,<br />

but the diagnoses enumerated in DSM-IV are descriptive<br />

syndromes that may not optimally capture the heritable basis<br />

<strong>of</strong> psychiatric illness. Empirical evidence from clinical,<br />

epidemiologic and genetic research has led to a growing<br />

recognition that these diagnostic categories comprise<br />

substantial heterogeneity. For example, several features <strong>of</strong><br />

mood disorders, including psychosis and early age at onset,<br />

may index familial and genetic subtypes. The emergence <strong>of</strong><br />

large-scale, genomewide analyses have provided new<br />

opportunities to dissect this heterogeneity. This presentation<br />

will provide an overview <strong>of</strong> the evidence for clinical and<br />

genetic heterogeneity in mood disorder syndromes and the<br />

prospects for identifying genetic influences that transcend<br />

diagnostic categories. The exploration <strong>of</strong> alternative<br />

phenotype definitions present methodologic challenges but<br />

may have important implications for enhancing the power <strong>of</strong><br />

genetic studies and informing an etiology-based nosology.<br />

S15 THE WNT SIGNALING PATHWAY AND<br />

SCHIZOPHRENIA<br />

S15.1 GENETIC ASSOCIATION ANALYSIS OF THE<br />

WNT SIGNALING PATHWAY IN SCHIZOPHRENIA<br />

D. Collier* (1), P. Priotsi (1), The SGENE Consortium , S.<br />

Lovestone (1)<br />

1. Institute <strong>of</strong> Psychiatry, King's College, London<br />

* david.collier@kcl.ac.uk<br />

There is evidence from multiple sources that Wnt signalling is<br />

involved in the aetiopathology <strong>of</strong> schizophrenia, suggesting<br />

the hypothesis that genes in the wnt pathway are genetic<br />

susceptibility factors for this illness. The accumulation <strong>of</strong><br />

genome wide association data in schizophrenia has made it<br />

possible to test this hypothesis. In order to do so we performed<br />

pathway association analysis using all known wnt signalling<br />

genes their modifiers and targets in case-control GWAs data.<br />

This includes the Dickkopf 4 (DKK4), which has previously<br />

been found to show altered expression in schizophrenia brain<br />

and to bind to neuregulin, and shows evidence <strong>of</strong> association<br />

with schizophrenia.<br />

S15.2 THE DISC1 PATHWAY MODULATES<br />

EXPRESSION OF NEURODEVELOPMENTAL,<br />

SYNAPTOGENIC AND SENSORY PERCEPTION<br />

GENES<br />

W. Hennah* (1), D. Porteous (2)<br />

1. Institute for Molecular Medicine Finland FIMM, Nordic<br />

EMBL Partnership for Molecular Medicine, Helsinki, Finland<br />

2. <strong>Medical</strong> Genetics Section, University <strong>of</strong> Edinburgh, EH4<br />

2XU, Edinburgh, UK<br />

* william.hennah@thl.fi<br />

Background: Genetic and biological evidence supports a role<br />

for DISC1 across a spectrum <strong>of</strong> major mental illnesses,<br />

including schizophrenia and bipolar disorder. There is<br />

evidence for genetic interplay between variants in DISC1 and<br />

in biologically interacting loci in psychiatric illness. DISC1<br />

also associates with normal variance in behavioral and brain<br />

imaging phenotypes. With recent studies demonstrating that<br />

DISC1 binding partners also play a role in mental illness.<br />

Methodology: Here, we analyze public domain datasets and<br />

demonstrate correlations between variants in the DISC1<br />

pathway genes and levels <strong>of</strong> gene expression. Genetic variants<br />

<strong>of</strong> DISC1, NDE1, PDE4B and PDE4D regulate the expression<br />

<strong>of</strong> cytoskeletal, synaptogenic, neurodevelopmental and<br />

sensory perception proteins. Interestingly, these regulated<br />

genes include existing targets for drug development in<br />

depression and psychosis. Conclusions: Our systematic<br />

analysis provides further evidence for the relevance <strong>of</strong> the<br />

DISC1 pathway to major mental illness, identifies additional<br />

potential targets for therapeutic intervention and establishes a<br />

general strategy to mine public datasets for insights into<br />

disease pathways.


S15.3 COPY NUMBER VARATION IN<br />

SCHIZOPHRENIA<br />

D. St. Clair (1)<br />

1. Department <strong>of</strong> Mental Health, University <strong>of</strong> Aberdeen<br />

A series <strong>of</strong> major articles have reported associations with<br />

schizophrenia <strong>of</strong> copy number variants at 1q21, 15q11.2,<br />

15q13.3, 16p11.2, 22q12, and Neurexin 1 loci. These are rare<br />

high-penetrant mutations that increase risk not only <strong>of</strong><br />

schizophrenia but also <strong>of</strong> a range <strong>of</strong> other psychiatric disorders<br />

including autism and mental retardation. Several copy number<br />

variant loci include genes which impact on the wnt signalling<br />

pathway. In some cases, the same phenotype can occur<br />

irrespective <strong>of</strong> whether the copy number variant causes a<br />

deletion or duplication. Although the loci identified so far<br />

account for only a small proportion <strong>of</strong> cases, many more are<br />

likely to be discovered over the next few years. A major focus<br />

<strong>of</strong> research will be to identify the key, the genetic and<br />

environmental determinants <strong>of</strong> schizophrenia risk in carriers <strong>of</strong><br />

these copy number variants, and to discover whether their<br />

rates <strong>of</strong> mutation are unstable or fixed.<br />

S15.4 HDAC-WNT/TCF SIGNALING CONTROLS<br />

OLIGODENDROCYTE DIFFERENTIATION<br />

Q. Lu* (1)<br />

1. UT Southwestern <strong>Medical</strong> Center<br />

* qrichard.lu@utsouthwestern.edu<br />

Oligodendrocyte development is regulated by the interplay <strong>of</strong><br />

repressors and activators in a complex transcriptional network.<br />

Here we report that two histone-modifying enzymes, HDAC1<br />

and HDAC2, are required for oligodendrocyte formation.<br />

Genetic deletion <strong>of</strong> both HDAC1 andHDAC2 in<br />

oligodendrocyte lineage cells resulted in stabilization and<br />

nuclear translocation <strong>of</strong> -catenin, which negatively regulates<br />

oligodendrocyte development by repressing Olig2 expression.<br />

We further identified an oligodendrocyte-restricted<br />

transcription factor TCF7L2/TCF4 as a bipartite co-effector <strong>of</strong><br />

-catenin for regulating oligodendrocyte differentiation.<br />

Targeted disruption <strong>of</strong> TCF7L2 in mice leads to severe defects<br />

in oligodendrocyte maturation, while expression <strong>of</strong> its<br />

dominant repressive form promotes precocious<br />

oligodendrocyte specification in developing chick neural tube.<br />

Transcriptional co-repressors HDAC1 and HDAC2 compete<br />

with -catenin for TCF7L2 interaction to regulate downstream<br />

genes involved in oligodendrocyte differentiation. Hence,<br />

crosstalk between HDAC1/2 and the canonical Wnt signaling<br />

pathway mediated by TCF7L2 serves as a regulatory<br />

mechanism for oligodendrocyte differentiation.<br />

S15.5 DISC1 REGULATES NEURAL PROGENITOR<br />

PROLIFERATION VIA MODULATION OF<br />

GSK3BETA/BETA-CATENIN SIGNALING<br />

Y. Mao, X. Ge, C. Frank, J. Madison, A. Koehler, M. Doud,<br />

C. Tassa, E. Berry, T. Soda, K. Singh, T. Biechele, T.<br />

Petryshen , R. Moon , S. Haggarty , L. Tsai (1)<br />

1. Howard Hughes <strong>Medical</strong> Institute; Picower Institute for<br />

Learning and Memory, Department <strong>of</strong> Brain and Cognitive<br />

Sciences, Massachusetts Institute <strong>of</strong> Technology, Cambridge,<br />

MA 02139<br />

* lhtsai@mit.edu<br />

Schizophrenia is a severe brain illness that affects 0.5-1% <strong>of</strong><br />

the world population. While the etiology is poorly understood,<br />

accumulating evidence suggest that neurodevelopmental<br />

defects are involved. Recent studies have identified many risk<br />

genes associated with schizophrenia. Among these genetic<br />

factors, the (1; 11) (q42; q14.3) translocation allele <strong>of</strong> the<br />

DISC1 gene closely segregates with the manifestation <strong>of</strong><br />

psychiatric disorders in a large Scottish pedigree. Functional<br />

studies revealed that DISC1 is involved in neurite outgrowth,<br />

neuronal migration, integration <strong>of</strong> newborn neurons, and<br />

cAMP signaling. In addition, mouse models created by either<br />

overexpressing a dominant negative form, deleting certain<br />

exons, or ENU mutagenesis <strong>of</strong> DISC1 resulted in the<br />

manifestation <strong>of</strong> schizophrenia- or depression-like behavioral<br />

phenotypes in these mice. We found a novel role for DISC1 in<br />

the proliferation <strong>of</strong> neural progenitors during embryonic and<br />

adult neurogenesis. In searching for mechanisms to explain<br />

how DISC1 regulates neurogenesis, we discovered that<br />

downregulating levels <strong>of</strong> DISC1 abolished the ability <strong>of</strong><br />

Wnt3a to stimulate proliferation <strong>of</strong> neural progenitor cells,<br />

suggesting DISC1 functions in the Wnt signaling pathway.<br />

Moreover, the N-terminal region <strong>of</strong> DISC1 was found to<br />

directly bind and inhibit GSK3b, thereby stabilizing b-catenin<br />

levels. Most striking was the observation that the in vitro and<br />

in vivo phenotypes due to DISC1 knockdown (both in the<br />

embryonic or adult brain) could be rescued by either<br />

overexpression <strong>of</strong> a degradation-resistant b-catenin, or by<br />

pharmacological inhibition <strong>of</strong> GSK3b. This suggests the<br />

cellular and behavioral results due to DISC1 knockdown can<br />

be alleviated by hyperactivation <strong>of</strong> the canonical Wnt<br />

signaling pathway. Together, these data strongly argue that<br />

DISC1 is an important regulator <strong>of</strong> neural progenitor<br />

proliferation that acts by positively modulating Wnt signaling<br />

via inhibition <strong>of</strong> GSK3b.


S16 GENES, ENVIRONMENT AND<br />

SUBSTANCE USE: EMERGING RESULTS<br />

FROM SAGE<br />

S16.1 ANALYSIS OF GWAS DATA FOR NICOTINE<br />

DEPENDENCE: THE IMPORTANCE OF PHENOTYPE<br />

DEFINITION AND CO-MORBID DIAGNOSES<br />

J. Rice* (1), S. Saccone (1), N. Saccone (1), W. Howells (1),<br />

K. Bucholz (1), D. Dick (2), K. Doheny (3), H. Edenberg (4),<br />

V. Hesselbrock (5), J. Kramer (6), R. Krueger (1), S.<br />

Kuperman (6), C. Laurie (7), R. Neuman (1), J. Nurnberger,<br />

Jr. (4), B. Porjesz (8), E. Pugh (3), J. Tischfield (9), J. Wang<br />

(1), L. Bierut (1), COGA, COGEND, GENEVA<br />

1. Washington University, St. Louis, MO 2. Commonwealth<br />

University, Richmond VA 3. CIDR Genotyping Lab and JHU<br />

Center, Baltimore, MD 4. Indiana University, Indianapolis, IN<br />

5. University <strong>of</strong> Connecticut, Farmington CT 6. University <strong>of</strong><br />

Iowa, Iowa City, IA 7. University <strong>of</strong> Washington, Seattle, WA<br />

8. SUNY, Brooklyn, NY 9. Rutgers University, Piscataway,<br />

NJ<br />

* john@zork.wustl.edu<br />

The SAGE addiction project, one <strong>of</strong> the GENEVA<br />

(Gene-Environment Association) GWAS studies, consists <strong>of</strong><br />

data from three studies (alcohol dependence, cocaine<br />

dependence and nicotine dependence). Cases had to have had<br />

a diagnosis <strong>of</strong> alcohol dependence, and controls had neither a<br />

diagnosis <strong>of</strong> alcohol dependence nor cocaine dependence.<br />

Genotyping was performed on the Illumina 1M chip at CIDR<br />

and data were cleaned following stringent GENEVA<br />

standards. We analyzed the phenotype <strong>of</strong> nicotine dependence<br />

using 1294 nicotine dependent cases and 2071 controls that<br />

had smoked cigarettes but were not dependent. We first<br />

considered issues related to ascertainment, co-morbidity and<br />

phenotype definition, and then performed logistic regression<br />

analysis for the phenotype <strong>of</strong> nicotine dependence. Covariates<br />

consisted <strong>of</strong> gender, age (coded in four quartiles), ethnicity,<br />

co-morbid alcohol dependence with and without cocaine<br />

dependence, and SNP genotype coded additively with one<br />

degree <strong>of</strong> freedom. We analyzed 1,341 SNPs present on the<br />

Illumina chip in a set <strong>of</strong> 56 candidate genes previously<br />

identified by a panel <strong>of</strong> experts and found significance in the<br />

beta nicotinic receptor (CHRNB3) on chromosome 8 (p=2.4 x<br />

10-8, OR = 1.54). These SNPs in CHRNB3 were the most<br />

significant under GWAS analysis. Odds ratios were similar<br />

for African Americans and European Americans as well as for<br />

those with and without a co-morbid diagnosis. We emphasize<br />

the importance <strong>of</strong> addressing co-morbidity in the analysis <strong>of</strong><br />

GWAS data for psychiatric phenotypes.<br />

S16.2 A GENOMEWIDE ASSOCIATION STUDY<br />

(GWAS) OF THE FIVE FACTOR MODEL (FFM) OF<br />

PERSONALITY<br />

R. Krueger* (1), A. Agrawal (1), J. Derringer (1), N. Wray<br />

(2), R. Grucza (1), H. Edenberg (3), V. Hesselbrock (4), J.<br />

Kramer (5), S. Kuperman (5), M. Lynskey (1), J. Nurnberger<br />

Jr. (3), J. Tischfield (6), GENEVA, L. Bierut (1)<br />

1. Washington University 2. Queensland Institute <strong>of</strong> <strong>Medical</strong><br />

Research 3. Indiana University 4. University <strong>of</strong> Connecticut 5.<br />

University <strong>of</strong> Iowa 6. Rutgers University<br />

* rkrueger@artsci.wustl.edu<br />

Twin studies are consistent in attributing a substantial<br />

proportion <strong>of</strong> the variation in human personality to genetic<br />

factors. Nevertheless, the identities <strong>of</strong> the molecular genetic<br />

variants that contribute to personality variation remain<br />

elusive. We therefore pursued a Genomewide Association<br />

Study (GWAS) <strong>of</strong> the Five Factor Model (FFM) <strong>of</strong><br />

personality, as assessed by the NEO-Five Factor Inventory<br />

(NEO-FFI) in 2,223 unrelated research participants.<br />

Genotyping was based on the 1M Illumina Platform,<br />

with 948,142 single nucleotide polymorphisms (SNPs)<br />

available for analysis after initial data cleaning. Three types<br />

<strong>of</strong> results will be presented. First, we present standard GWAS<br />

findings for each <strong>of</strong> the five personality traits (i.e., SNPs with<br />

the lowest p-values).Second, we present p-values for SNPs<br />

found to be associated with FFM traits in previous GWAS<br />

research. Third, we present the results <strong>of</strong> a novel approach<br />

to developing “genotypic scores.” Multiple SNPs found to be<br />

predictive <strong>of</strong> personality variation in a development sample (a<br />

random half <strong>of</strong> the participants) were used to develop five<br />

scores characterizing genotypic standing on the five FFM<br />

traits. Persons with higher genotypic scores have a greater<br />

number <strong>of</strong> alleles predictive <strong>of</strong> higher FFM scores in the<br />

development sample. The validity <strong>of</strong> these genotypic scores<br />

for predicting personality was then ascertained in the<br />

remaining half <strong>of</strong> the sample, i.e., in those participants not<br />

used to develop the genotypic scores.


S16.3 GENOMEWIDE ASSOCIATION STUDIES OF<br />

CANNABIS AND COCAINE DEPENDENCE<br />

A. Agrawal* (1), M. Lynskey (2), A. Hinrichs (2), R. Grucza<br />

(2), N. Saccone (2), S. Saccone (2), R. Krueger (3), R.<br />

Neuman (2), W. Howells (2), S. Fisher (2), L. Fox (2), R.<br />

Cloninger (2), D. Dick (4), K. Doheny (5), H. Edenberg (6),<br />

A. Goate (2), V. Hesselbrock (7), E. Johnson (8), J. Kramer<br />

(9), S. Kuperman (10), J. Nurnberger (6), E. Pugh (5), J.<br />

Tischfield (11), COGA, COGEND, GENEVA, J. Rice (2), K.<br />

Bucholz (2), L. Bierut (2)<br />

1. Washington University School <strong>of</strong> Medicine Dept. <strong>of</strong><br />

Psychiatry, Email:arpana@wustl.edu 2. Washington Univ Sch<br />

Med 3. Washington Univ St. Louis 4. Virginia<br />

Commonwealth Univ 5. CIDR 6. Indiana Univ Sch Med 7.<br />

Univ Connecticut Sch Med 8. RTI 9. Univ <strong>of</strong> Iowa College <strong>of</strong><br />

Med 10. Univ <strong>of</strong> Iowa Hospitals 11. Rutgers Univ<br />

* arpana@wustl.edu<br />

Cannabis is the most commonly used illicit psychoactive<br />

substance in developed nations. After cannabis, cocaine<br />

misuse contributed to 13% <strong>of</strong> admissions to treatment settings<br />

in 2007. Prior twin studies have demonstrated considerable<br />

genetic influences on both cannabis and cocaine dependence,<br />

some <strong>of</strong> which are overlapping. Linkage and candidate gene<br />

association studies have identified some variants associated<br />

with dependence on either drug but results have been<br />

equivocal. We conduct one <strong>of</strong> the first genomewide<br />

association studies <strong>of</strong> DSM-IV cannabis dependence and<br />

cocaine dependence using data from the Study <strong>of</strong> Addiction:<br />

Genes and Environment (SAGE) which genotyped, using the<br />

Illumina 1M, 4121 (54% female; 32.5% African-American)<br />

individuals including 1944 cases (1965 exposed controls and<br />

212 other/drug misuse) ascertained for alcohol dependence.<br />

For our analyses, 753 and 1130 cases were available for<br />

DSM-IV cannabis and cocaine dependence respectively. Our<br />

top findings for cannabis dependence included polymorphisms<br />

on chromosome 3 (p


S16.5 INTERPLAY OF GENETIC RISK FACTORS AND<br />

PARENTS MONITORING IN RISK FOR NICOTINE<br />

DEPENDENCE<br />

L. Chen* (1), E. Johnson (2), N. Breslau (3), D. Hatsukami<br />

(4), N. Saccone (5), R. Grucza (1), J. Wang (1), A. Hinrichs<br />

(1), L. Fox (1), A. Goate (1), J. Rice (1), L. Bierut (1)<br />

1. Washington University School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Psychiatry 2. Research Triangle Institute International 3.<br />

Michigan State University, Department <strong>of</strong> Epidemiology 4.<br />

University <strong>of</strong> Minnesota, Department <strong>of</strong> Psychiatry 5.<br />

Washington University School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Genetics<br />

* chenli@psychiatry.wustl.edu<br />

Background: Several studies have found replicable<br />

associations between nicotine dependence and specific<br />

variants in the nicotinic receptor genes CHRNA5(rs16969968)<br />

and CHRNA3(rs3743078). How these newly identified<br />

genetic risks combine with known environmental risks is<br />

unknown. This study examined whether the level <strong>of</strong> parent<br />

monitoring during early adolescence modified the risk <strong>of</strong><br />

nicotine dependence associated with these genetic variants.<br />

Methods: In a cross-sectional case control study <strong>of</strong> US-based<br />

community sample <strong>of</strong> 2027 subjects, we use a systematic<br />

series <strong>of</strong> regression models to examine the effect <strong>of</strong> parent<br />

monitoring on risk associated with two distinct variants in the<br />

nicotinic receptor genes CHRNA5(rs16969968) and<br />

CHRNA3(rs3743078). Results: Low parent monitoring as well<br />

as the previously identified genetic variants were associated<br />

with an increased risk <strong>of</strong> nicotine dependence. An interaction<br />

was found between the SNP(rs16969968) and parent<br />

monitoring (p=0.034). The risk for nicotine dependence<br />

increased significantly with the risk genotype <strong>of</strong><br />

SNP(rs16969968) when combined with lowest quartile parent<br />

monitoring. In contrast, there was no evidence <strong>of</strong> an<br />

interaction between SNP(rs3743078) and parent monitoring<br />

(p=0.80). Conclusions: The genetic risk <strong>of</strong> nicotine dependent<br />

associated with rs16969968 was modified by level <strong>of</strong> parent<br />

monitoring, while the genetic risk associated with rs3743078<br />

was not, suggesting that the increased risk due to some genes<br />

may be mitigated by environmental factors such as parent<br />

monitoring.<br />

S16.6 GENETIC INFLUENCES ON SENSATION<br />

SEEKING: MODERATION BY RELIGIOUS SERVICE<br />

ATTENDANCE<br />

J. Derringer* (1), R. Krueger (1, 2), D. Dick (3), R. Grucza<br />

(2), A. Agrawal (2), L. Almasy (4), H. Edenberg (5), T.<br />

Foroud (6), V. Hesselbrock (7), J. Kramer (8), S. Kuperman<br />

(9), J. Nurnberger Jr. (10), B. Porjesz (11), M. Schuckit (12),<br />

COGA, GENEVA, L. Bierut (2)<br />

1. Department <strong>of</strong> Psychology, Washington University 2.<br />

Department <strong>of</strong> Psychiatry, Washington University 3. Virginia<br />

Institute for Psychiatric and Behavioral Genetics, Virginia<br />

Commonwealth University 4. Genetics, Southwest Foundation<br />

for Biomedical Research 5. Center for <strong>Medical</strong> Genomics,<br />

Indiana University 6. Department <strong>of</strong> <strong>Medical</strong> and Molecular<br />

Genetics, Indiana University 7. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Connecticut 8. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Iowa 9. Division <strong>of</strong> Child Psychiatry, University<br />

<strong>of</strong> Iowa 10. Institute <strong>of</strong> Psychiatric Research, Indiana<br />

University 11. Henri Begleiter Neurodynamics Laboratory,<br />

SUNY Downstate <strong>Medical</strong> Center 12. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> California – San Diego<br />

* derringer@wustl.edu<br />

The trait <strong>of</strong> sensation seeking (Zuckerman 1971) has been<br />

associated with a range <strong>of</strong> psychiatric disorders, including<br />

addiction, mania, and psychopathy. Twin studies indicate that<br />

genetic factors account for significant variance in sensation<br />

seeking, and sensation seeking has been associated with<br />

biological markers, including MAO activity and sex hormone<br />

levels (see Zuckerman, Buchsbaum, & Murphy 1980).<br />

Religiosity moderates the influence <strong>of</strong> genetic factors on the<br />

disinhibition scale <strong>of</strong> sensation seeking, where genetic factors<br />

account for a greater proportion <strong>of</strong> variance in disinhibition for<br />

individuals without a religious upbringing than for those<br />

raised in religious environments (Boomsma et al. 1999). As<br />

part <strong>of</strong> the Study <strong>of</strong> Addiction: Genes and Environment<br />

(SAGE), we have been exploring the moderating role <strong>of</strong><br />

religious service attendance, one index <strong>of</strong> religiosity, on<br />

genetic influences on substance dependence and related<br />

phenotypes (Dick et al; accompanying presentation). We<br />

examined moderation <strong>of</strong> genetic influences on sensation<br />

seeking, a trait associated with substance dependence, by<br />

religious service attendance. Our sample consisted <strong>of</strong><br />

644individuals from SAGE. More frequent religious service<br />

attendance was associated with decreased overall sensation<br />

seeking (r=-0.155, p


S16.7 INCORPORATING ENVIRONMENTAL<br />

INFORMATION INTO GWAS: TESTING FOR<br />

GENE-ENVIRONMENT INTERACTION AND<br />

CORRELATION BETWEEN ALCOHOL<br />

DEPENDENCE AND CHURCH ATTENDANCE<br />

D. Dick* (1), J. Yan (2), F. Aliev (2), R. Krueger (3), A.<br />

Agrawal (4), H. Edenberg (5), J. Kramer (6), M. Schuckit (7),<br />

J. Tishcfield (8), B. Porjesz (9), V. Hesselbrock (10), S.<br />

Kuperman (11), L. Almasy (12), J. Nurnberger (5), K. Kendler<br />

(2), S. Aggen (2), L. Bierut (4), COGA, COGEND, GENEVA<br />

1. Virginia Institute for Psychiatric and Behavioral Genetics<br />

Virginia Commonwealth University Department <strong>of</strong> Psychiatry<br />

2. Virginia Commonwealth University 3. Washington<br />

University in St. Louis 4. Washington University School <strong>of</strong><br />

Medicine 5. Indiana University School <strong>of</strong> Medicine 6.<br />

University <strong>of</strong> Iowa 7. University <strong>of</strong> California San Diego 8.<br />

Rutgers University 9. State University <strong>of</strong> New York 10.<br />

University <strong>of</strong> Connecticut School <strong>of</strong> Medicine 11. University<br />

<strong>of</strong> Iowa Hospitals 12. Southwest Foundation for <strong>Medical</strong><br />

Research<br />

* ddick@vcu.edu<br />

Although most psychiatric disorders show considerable<br />

evidence for genetic influence, the environment also plays an<br />

important role. Furthermore, genetic and environmental<br />

influences do not act in isolation, with twin studies providing<br />

compelling evidence for both gene-environment interaction,<br />

whereby the importance <strong>of</strong> genetic effects varies as a function<br />

<strong>of</strong> the environment, and gene-environment correlation,<br />

whereby the selection and experience <strong>of</strong> environmental events<br />

is impacted by genetically-influenced characteristics. Despite<br />

widespread recognition <strong>of</strong> the complex interplay between<br />

genetic and environmental risk factors on psychiatric outcome,<br />

most large-scale gene identification efforts to date have<br />

largely ignored environmental effects. We have conducted a<br />

series <strong>of</strong> analyses aimed at exploring the relationship between<br />

alcohol dependence (AD) and church attendance. Religiosity<br />

is known to play an important role in the development <strong>of</strong> AD,<br />

with protective main effects (Kendler et al. 2003), and<br />

moderating effects on genetic influences on alcohol use and<br />

related phenotypes (Koopmans et al., 1999; Boomsma et al.,<br />

1999). Analyses in the Virginia Adult Twin Study <strong>of</strong><br />

Psychiatric and Substance Use Disorders demonstrate genetic<br />

covariance contributing to the correlation between church<br />

attendance and AD. In the Study <strong>of</strong> Addiction: Genes and<br />

Environment, a sample with GWAS data on 1944 AD cases<br />

and 1965 controls, we find evidence for several loci that may<br />

be involved in this gene-environment correlation between<br />

religiosity and AD, as well as other loci where the association<br />

with AD is moderated by religiosity. This study represents a<br />

first step toward incorporating environmental information into<br />

GWAS, using twin data to guide these efforts.


ORAL ABSTRACTS


O1 MOOD DISORDERS1<br />

O1.1 SUPPORT FOR A BIPOLAR AFFECTIVE<br />

DISORDER SUSCEPTIBILITY LOCUS ON<br />

CHROMOSOME 12Q24.3<br />

L. Foldager* (1), H. Buttenschøn (1), T. Flint (1), I. Olsen<br />

(1), T. Deleuran (1), M. Nyegaard (2), M. Hansen (3), P.<br />

Kallunki (4), K. Christensen (4), D. Blackwood (5), W. Muir<br />

(5), S. Straarup (3), T. Als (3), M. Nordent<strong>of</strong>t (6), A. Børglum<br />

(2), O. Mors (3)<br />

1. Centre for Psychiatric Research, Aarhus University<br />

Hospital, Risskov, Denmark 2. Institute <strong>of</strong> Human Genetics,<br />

Aarhus University, Denmark 3. Centre for Psychiatric<br />

Research, Aarhus University Hospital, Risskov, Denmark 4.<br />

H. Lundbeck A/S, Valby, Denmark 5. Division <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Edinburgh, UK 6. Psychiatric Centre Bispebjerg,<br />

University <strong>of</strong> Copenhagen, Denmark<br />

* Leslie.Foldager@ps.rm.dk<br />

Linkage and association studies <strong>of</strong> bipolar affective disorder<br />

(BP) point out chromosome 12q24 as a region <strong>of</strong> interest. In<br />

order to investigate this region further, we conducted an<br />

association study <strong>of</strong> 22 DNA markers within a 1.14 Mb region<br />

in a Danish sample <strong>of</strong> 166 patients with BP and 311 control<br />

individuals. Two hundred and four Danish patients with<br />

schizophrenia (SZ) were also included in the study. We<br />

observed highly significant allelic and genotypic association<br />

between BP and two markers. The risk allele <strong>of</strong> both markers<br />

considered separately conferred an odds ratio (OR) <strong>of</strong> two to<br />

an individual carrying one risk allele and an OR <strong>of</strong> four for<br />

individuals carrying both risk alleles assuming an additive<br />

genetic model. These findings were supported by the<br />

haplotype analysis. In addition we obtained a replication <strong>of</strong><br />

four markers associated with BP in a previous U.K. study.<br />

The most significantly associated marker was also analyzed in<br />

a Scottish case-control sample and was previously associated<br />

with BP in the U.K. cohort. That particular marker was<br />

strongly associated with BP in a meta-analysis <strong>of</strong> the Danish,<br />

Scottish and U.K. sample (P=0.0003).The chromosome region<br />

confined by our most distant markers is gene-poor and<br />

harbours only a few predicted genes. The present study<br />

implicates the Slynar locus. We confirmed one annotated<br />

Slynar transcript and identified a novel transcript in human<br />

brain cDNA. This study confirms 12q24.3 as a region <strong>of</strong><br />

functional importance in the pathogenesis <strong>of</strong> BP and<br />

highlights the importance <strong>of</strong> focused genotyping.<br />

O1.2 PER2 VARIATION IS ASSOCIATED WITH<br />

DEPRESSION VULNERABILITY<br />

C. Lavebratt (1), L. Sjöholm (1), T. Partonen (2), M.<br />

Schalling (1), Y. Forsell (1)<br />

1. Karolinska Institutet 2. Helsinki National Institute <strong>of</strong> Health<br />

and Welfare<br />

The circadian clock is driven by transcription-translation<br />

feedback loops and regulates rhythms that approximate the<br />

24-h day-night cycle or light-dark transitions. Disruptions <strong>of</strong><br />

the circadian rhythms are common in depressed patients,<br />

expressed for example as sleep disturbances. Genetic<br />

variations in core circadian genes may in part explain these<br />

abnormalities. To investigate whether genetic variation in core<br />

circadian genes associates with vulnerability to depression, we<br />

genotyped 18 genes in a Swedish population based sample.<br />

Genetic variations indicative <strong>of</strong> association with depression, or<br />

with winter depression in our previous study, were tested for<br />

association to depression in a second Swedish<br />

depression-control sample set. PER2 genetic variation was<br />

associated with depression vulnerability, and this genetic risk<br />

did not seem to require exposure to potential sleep disturbance<br />

factors such as negative life event or financial strain that are<br />

known to increase the risk for depression. Polymorphisms<br />

in the circadian genes NPAS2, ARNTL and RORA were also<br />

suggested to contribute to depression vulnerability. The<br />

findings we report for PER2, ARNTL and RORA are<br />

supported by at least two <strong>of</strong> the three sample sets. In<br />

conclusion, genetic variation in PER2 is associated with<br />

depression vulnerability a Swedish population-based sample.<br />

More studies are needed to determine if this is the case also<br />

for NPAS2, ARNTL and RORA.


O1.3 GENOME WIDE ASSOCIATION SCAN OF<br />

SUICIDALITY IN MAJOR DEPRESSION.<br />

A. Schosser* (1), A. Butler (2), M. Ng (2), R. Uher (2), S.<br />

Cohen-Woods (2), K. Pirlo (2), A. Elkin (2), N. Craddock (3),<br />

M. Owen (2), K. Aitchison (2), A. Korszun (4), M. Barnes (5),<br />

P. Muglia (5), M. Lathrop (6), S. Heath (6), G. Breen (2), A.<br />

Farmer (2), I. Craig (2), P. McGuffin (2), C. Lewis (2)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London, UK; Dep. <strong>of</strong> Psychiatry and Psychotherapy, <strong>Medical</strong><br />

Univ. Vienna, Austria 2. MRC SGDP Centre, Institute <strong>of</strong><br />

Psychiatry, King's College London, UK 3. Department <strong>of</strong><br />

Psychological Medicine, Cardiff University, UK 4. Centre for<br />

Psychiatry, Wolfson Institute <strong>of</strong> Preventive Medicine, Barts<br />

and The London <strong>Medical</strong> School, Queen Mary University <strong>of</strong><br />

London, UK 5. GlaxoSmithKline Research & Development<br />

6. Centre National de Genotypage, 91057 Evry, France<br />

* alexandra.schosser@iop.kcl.ac.uk<br />

Suicidal behaviour seems to cluster in families, and eritability<br />

<strong>of</strong> completed suicide has been estimated as about 43%. Genes<br />

predisposing to suicide may overlap with those predisposing<br />

to affective disorder, since approximately half <strong>of</strong> those<br />

committing suicide have a diagnosis <strong>of</strong> depression. We carried<br />

out a genome-wide association (GWA) study in 2947 subjects<br />

with DSM-IV and/or ICD-10 major depression, derived from<br />

three studies: DeCC (Depression Case Control, N=1145),<br />

GENDEP (Genome-based Therapeutic Drugs for Depression,<br />

N=729), DeNt (Depression Network, N=918), and a sample<br />

collected by GSK (Glaxo Smith Kline, N=155). Depression<br />

and suicidality were assessed using SCAN, and a new variable<br />

summarising suicidal behaviour (SSB), composed <strong>of</strong> SCAN<br />

items 6.011 (suicide or self-harm, score 0-4) and 6.012 edium<br />

vitae, score 0-3), was created. All subjects therefore had a<br />

score between 0 and 7 for the worst and 2nd worst episode, or<br />

current episode in GENDEP. This quantitative trait SSB was<br />

correlated with number <strong>of</strong> episodes <strong>of</strong> depression and with<br />

early age <strong>of</strong> onset, but not gender. We performed enome-wide<br />

analysis <strong>of</strong> the quantitative SSB phenotype, and a discrete trait<br />

(SSB6, with N=275 cases), correcting for the<br />

Europe-wide ascertainment <strong>of</strong> cases. Four SNPs attained<br />

p-values <strong>of</strong>


O1.5 SYSTEMATIC ANALYSIS OF CIRCADIAN<br />

GENES IN A POPULATION-BASED SAMPLE<br />

REVEALS ASSOCIATION OF TIMELESS TO<br />

DEPRESSION AND SLEEP DISTURBANCE<br />

S. Utge (1), P. Soronen (1), A. Loukola (1), E. Kronholm (2),<br />

V. Leppä (3), S. Pirkola (4), T. Porkka-Heiskanen (5), T.<br />

Partonen (6), T. Paunio* (7)<br />

1. Public Health Genomics Unit, National Institute for Health<br />

and Welfare, Helsinki 2. Department <strong>of</strong> Health and Functional<br />

Capacity, Laboratory for Population Research 3. Public Health<br />

Genomics Unit, National Institute for Health and Welfare,<br />

Helsinki and Institute for Molecular Medicine Finland FIMM,<br />

University <strong>of</strong> Helsinki 4. Department <strong>of</strong> Psychiatry, Helsinki<br />

University Central Hospital, Helsinki<br />

5. Department <strong>of</strong> Physiology, University <strong>of</strong> Helsinki, Helsinki,<br />

Finland 6. Department <strong>of</strong> Mental Health and Substance Abuse<br />

Services, National Institute for Health and Welfare, Helsinki<br />

7. Public Health Genomics Unit, National Institute for Health<br />

and Welfare, Helsinki and Department <strong>of</strong> Psychiatry, Helsinki<br />

University Central Hospital, Helsinki<br />

* tiina.paunio@thl.fi<br />

Background: Circadian rhythm abnormalities and disturbed<br />

sleep is a prevalent sign <strong>of</strong> depression. We have recently<br />

showed that poor sleep quality is a risk factor for incidence <strong>of</strong><br />

depressed mood (Paunio et al. 2009) and that there are<br />

sex-dependent and symptom specific differences in the genetic<br />

back ground <strong>of</strong> depression at population level (Utge et al. in<br />

press). We hypothesized that some <strong>of</strong> the genetic variants<br />

from the circadian system might be a contributing causal<br />

factor for depression with signs for disturbed sleep, early<br />

morning awakenings, or day-time fatigue. Materials and<br />

methods: We tested this hypothesis in a population-based<br />

sample <strong>of</strong> Health 2000 from Finland, including 384 depressed<br />

individuals and 1270 controls, all with detailed information on<br />

sleep and day-time vigilance, and analyzed this set <strong>of</strong><br />

individuals against 113 single nucleotide variants from 18<br />

genes <strong>of</strong> the circadian system. Association <strong>of</strong> genetic variants<br />

to depression with signs <strong>of</strong> disturbed sleep was investigated<br />

using allelic permutation tests, followed by haplotype<br />

analyses, and finally gene-gene interaction analyses for the<br />

most significant findings. Results: We found significant<br />

association <strong>of</strong> variants from TIMELESS to depression with<br />

fatigue (P = 0.0001 for single variant and P = 0.000008 for a<br />

haplotype across the gene) in females and suggestive<br />

association to depression with early morning awakenings (P =<br />

0.0009 for single variant and P = 0.0001 for 2-SNP haplotype)<br />

in males. There was some evidence <strong>of</strong> interaction between<br />

TIMELESS and other circadian genes both in females and<br />

males. Conclusions: The current findings support the<br />

involvement <strong>of</strong> circadian system in the regulation <strong>of</strong><br />

depression and sleep in a sex dependent manner.<br />

References: Paunio T, Korhonen T, Hublin C, Partinen M,<br />

Kivimäki M, Koskenvuo M, Kaprio J. (2009) Longitudinal<br />

study on causality and genetic risk factors for the association<br />

between poor sleep and life dissatisfaction in a nationwide<br />

cohort <strong>of</strong> twins. Am J Epidemiol. 169(2):206-13; Utge S,<br />

Soronen P, Partonen T, Loukola A, Kronholm E, Pirkola S,<br />

Nyman E, Porkka-Heiskanen T, Paunio T. A population-based<br />

association study <strong>of</strong> candidate genes for depression and sleep<br />

disturbance. Am J Med Genet B Neuropsychiatr Genet. in<br />

press Acknowledgments: Funding from STREP, Call:<br />

FP6-2004-Lifescihealth-5 ; Enough sleep (to T. Paunio)<br />

O1.6 PI3K/AKT SIGNALING IN DOPAMINERGIC<br />

ACTION<br />

K. Celestrin (1), K. Carr (1), T. Franke (1)<br />

1. NYU School <strong>of</strong> Medicine<br />

One biochemical change acquired following induction <strong>of</strong><br />

depression-like behavior and exposure to drugs <strong>of</strong> abuse is<br />

altered phosphorylation <strong>of</strong> the Aktserine-threonine kinase in<br />

dopaminergic neurons. Here, we have asked if altered Akt<br />

signal transduction affects behavioral responses in<br />

experimental paradigms related to mesolimbic/nigrostriatal<br />

dopamine signaling. To answer the question, we have used<br />

Akt1-mutant mice. Akt1 is a major is<strong>of</strong>orm <strong>of</strong> the Akt kinase<br />

family and expressed at high levels in dopaminergic neurons.<br />

Loss-<strong>of</strong>-function Akt1-mutantmice exhibited a phenotype in<br />

the learned helplessness paradigm. In this experimental model<br />

for acquired behavioral depression, the exposure to<br />

inescapable electric foot shocks (IES) disrupts the escape to<br />

subsequent noxious stimuli. While no significant differences<br />

were observed between sham-treated control mice depending<br />

on genotypes, IE-shocked Akt1-mutant mice showed<br />

increased latencies <strong>of</strong> crossing and a higher number <strong>of</strong> escape<br />

failures when compared to IES-treated wild-type mice. In<br />

further support <strong>of</strong> the importance <strong>of</strong> intact Akt signaling in<br />

dopaminergic signaling, Akt1-mutantmice displayed an<br />

enhanced response to acute cocaine and a diminished<br />

sensitization effect to chronic cocaine exposure. Western<br />

blotting and immunohistochemical analyses after IES training<br />

revealed genotype-dependent changes in Akt phosphorylation<br />

and regulation <strong>of</strong> its downstream substrates <strong>of</strong> Akt and its<br />

substrates following IES training. These biochemical state<br />

changes phenocopied differences in behavioral outputs when<br />

comparing wild-type and Akt1-mutantmice. This work is<br />

supported in part by the National Science Foundation (NSF<br />

award IOS-0757780) and the G. Harold & Leila Y. Mathers<br />

Foundation (toTFF), and by the National Institute on Drug<br />

Addiction/NIH (DA003956) (to KDC). Dr. Franke is<br />

supported in part by a 2008 NARSAD Independent<br />

Investigator Award.


O1.7 TPH2 RISK HAPLOTYPE ASSOCIATED WITH<br />

IMPULSIVE AGGRESSION IN BORDERLINE<br />

PERSONALITY DISORDER<br />

L. Siever* (1), A. New (1), M. Perez-Rodriguez (2), S.<br />

Weinstein (2), L. Bevilacqua (3), Q. Yuan (3), Z. Zhou (3), C.<br />

Hodgkinson (3), M. Goodman (1), H. Koenigsberg (1), D.<br />

Goldman (3)<br />

1. Mount Sinai School <strong>of</strong> Medicine/Bronx VA <strong>Medical</strong> Center<br />

2. Mount Sinai School <strong>of</strong> Medicine 3. NIAAA/NIH<br />

* Larry.Siever@va.gov<br />

The serotonin system has been implicated in the regulation <strong>of</strong><br />

impulsive aggression and in the pathogenesis <strong>of</strong> borderline<br />

personality disorder on the basis <strong>of</strong> neuropharmacologic,<br />

neuroimaging, and treatment studies. Reduced serotonergic<br />

activity is associated with reduced prefrontal control <strong>of</strong> the<br />

emergence <strong>of</strong> aggression upon emotional provocation.<br />

Responses to fenfluramine agent and imaging <strong>of</strong> serotonin<br />

synthesis with methyl-L-tryptophan trapping suggest reduced<br />

presynaptic serotonergic activity. TPH2 is the rate limiting<br />

step for serotonin synthesis in the brain and this study was<br />

designed to evaluate the genetics <strong>of</strong> TPH2 in relation to<br />

impulsive aggression in borderline personality and a cohort <strong>of</strong><br />

251 personality disorder patients and 103 healthy controls.<br />

All subjects were free <strong>of</strong> medication, screened for medical<br />

problems, and met criteria for a personality disorder and were<br />

evaluated diagnostically and using a composite measure <strong>of</strong><br />

aggression based on the Buss Perry Aggression Inventory.<br />

123 Caucasian subjects were evaluated for a risk haplotype<br />

associated with reduced serotonergic activity and suicide in a<br />

Finnish cohort (Zhou et al, 2005). In our study subjects<br />

carrying the risk haplotype demonstrated higher aggression<br />

scores and were more likely to be diagnosed with borderline<br />

personality disorder (p


O2.2 MOLECULAR NETWORKS OF LITHIUM<br />

RESPONSE: CONVERGENCE ACROSS PHYLOGENY<br />

A. Gupta (1), T. Schulze (2), N. Akula (1), F. McMahon (1),<br />

S. Detera-Wadleigh (1)<br />

1. NIMH 2. NIMH PI3K/AKT Signaling In Dopaminergic<br />

Action<br />

How lithium elicits mood stabilization in bipolar disorder<br />

patients has not been fully explained. Perturbations by lithium<br />

in key components <strong>of</strong> the phosphoinositide and Wnt<br />

signaling pathways have been demonstrated consistently,<br />

recommending their prominent role in lithium response. To<br />

evaluate the global effect <strong>of</strong> lithium, we assembled expression<br />

studies performed in various organisms and identified<br />

differentially expressed genes and proteins. We sought to<br />

capture, through pathway analysis, the biological relationships<br />

<strong>of</strong> these genetic factors. This generated a molecular network<br />

that may unify the disparate mechanisms recruited by the drug<br />

and exposes potential markers for lithium response. Genes up-<br />

and down-regulated by lithium were initially identified from<br />

>20 gene-based and 5 protein-based studies, with a focus on<br />

large microarray studies. Homologs significantly regulated in<br />

the same direction across two or more species/studies were<br />

given preference and fed into the pathway analysis programs<br />

Ingenuity and Cytoscape. Enzymes classically shown to alter<br />

lithium response were also included. Networks resulting from<br />

both were compared for overlaps and hubs <strong>of</strong> importance<br />

verified. Two pathway analysis tools were trained on 19<br />

lithium-responsive genes and proteins/enzymes. Major gene<br />

hubs centered at AKT1, BCL2, CALM, EZR and GSK3B<br />

and minor ones localized at ATF4, PTGS2, BDNF, CRYAB,<br />

SYN1. Where these cores converged could be considered key<br />

features <strong>of</strong> the network. Although multiple genes populate the<br />

networks, coalescence into a more unified molecular<br />

repertoire may underlie the overall effect <strong>of</strong><br />

lithium. Conceivably, some genes may be used as new drug<br />

targets and diagnostic tools as well.<br />

O2.3 BEHAVIORAL ANALYSES OF TRANSGENIC<br />

MICE HARBORING BIPOLAR DISORDER<br />

CANDIDATE GENES, IMPA1 AND IMPA2<br />

T. Yoshikawa* (1), T. Ohnishi (1)<br />

1. RIKEN Brain Science Institute<br />

* takeo@brain.riken.jp<br />

IMPase (myo-inositol monophosphatase) generates free<br />

inositol from inositol monophosphate. The inositol depletion<br />

hypothesis proposes the inhibition <strong>of</strong> IMPase by lithium, a<br />

mood stabilizer, as a mechanism <strong>of</strong> lithium’s efficacy. This<br />

hypothesis predicts that the upregulation <strong>of</strong> this biochemical<br />

pathway may underlie the pathophysiology <strong>of</strong> bipolar<br />

disorder. In favor <strong>of</strong> this idea, we have recently shown that<br />

IMPA2 encoding IMPase is a candidate susceptibility gene for<br />

bipolar disorder and that the risk-conferring single nucleotide<br />

polymorphisms (SNPs) enhance the promoter activity <strong>of</strong><br />

IMPA2. Furthermore, some studies have suggested an<br />

increase in the myo-inositol content in the brains <strong>of</strong><br />

bipolar patients. However, it is yet unknown whether such<br />

upregulation has a biological role in bipolar disorder. To<br />

address this issue, we generated Tg (transgenic) mice for the<br />

two human IMPase genes (IMPA1 and IMPA2) under the<br />

control <strong>of</strong> brain-specific promoters, and examined them using<br />

multiple behavioral test batteries. The Tg mice behaved<br />

normally except for subtle changes in some paradigms<br />

under durg-naïve conditions, and did not exhibit signs for<br />

manic change when an antidepressant amitriptyline was<br />

chronically administrated. Interestingly, the male Tg mice for<br />

IMPA2 exhibited a lithium-resistant phenotype in the forced<br />

swim test. The current study did not support a substantial<br />

role <strong>of</strong> the upregulation <strong>of</strong> IMPase in the pathogenesis <strong>of</strong><br />

bipolar disorder, at least from animal experiments, although<br />

the lithium-insensitivity trait seen in IMPA2 Tg mice might<br />

represent some aspect relevant to the inositol depletion<br />

hypothesis.


O2.4 CANDIDATE GENE PHARMACOGENETICS<br />

STUDY OF MGLU 2/3 AGONIST LY2140023<br />

A. Downing (1), W. Liu (1), L. Shen (1), L. Munsie (1), P.<br />

Chen (1), B. Han (1), J. Brandt (1), R. Njau (1), B. Kinon (1),<br />

L. Nisenbaum (1)<br />

1. Eli Lilly and Company<br />

LY2140023 is the oral prodrug <strong>of</strong> LY404039, a selective<br />

agonist for metabotropic-glutamate 2/3 (mGlu) receptors.<br />

LY2140023 is currently under development for the treatment<br />

<strong>of</strong> schizophrenia and has the potential to introduce a novel<br />

mechanism <strong>of</strong> action for antipsychotics. The primary goal <strong>of</strong><br />

the pharmacogenetics effort is to identify genetic markers that<br />

are predictive <strong>of</strong> patients’ response to LY2140023 and could<br />

be used to target specific subpopulations for treatment.<br />

Genetic variants in eight candidate genes related to the<br />

mechanism <strong>of</strong> action <strong>of</strong> LY2140023 or the atypical<br />

antipsychotic olanzapine were investigated in DNA samples<br />

collected as part <strong>of</strong> the Phase II pro<strong>of</strong> <strong>of</strong> concept trial,<br />

H8Y-MC-HBBD. Primary statistical analysis was conducted<br />

on 193 Caucasian patients treated with either placebo,<br />

LY2140023 or olanzapine using an ANCOVA model<br />

assuming LOCF and additive inheritance. Associations<br />

between LY2140023 response and genotypes were identified<br />

for SNPs within the HTR2A and NRG1 genes. Three SNPs (1<br />

in NRG1, 2 in HTR2A) were statistically significantly<br />

associated with LY2140023 response at 28 days as measured<br />

by Positive and Negative Syndrome scale (PANSS) Total<br />

change from baseline. All three SNPs had unadjusted p-values<br />

less than 0.001 (FDR=0.114) at 28 days. These SNPs were<br />

investigated in a second cohort collected from trial<br />

H8Y-MC-HBBI, which had an inconclusive primary clinical<br />

endpoint. While the genetic results did not reach statistical<br />

significance in HBBI, the HTR2A minor homozygous group<br />

showed the greatest treatment response in both HBBD and<br />

HBBI. In conclusion, these data suggest that a genetic<br />

association may impact response to LY2140023 treatment.<br />

O2.5 CONVERGENT ANIMAL AND HUMAN<br />

EVIDENCE FOR A ROLE OF PPM1A GENE IN<br />

RESPONSE TO ANTIDEPRESSANTS<br />

K. Malki* (1), R. Uher (1), E. Binder (1), J. Payo-Cano (1),<br />

C. Lewis (1), J. Rietschel (1), W. Maier (1), O. Mors (1), J.<br />

Hauser (1), N. Henigsberg (1), B. Jerman (1), D. Souery (1),<br />

A. Placentino (1), K. Aitchison (1), A. Farmer (1), I. Craig (1),<br />

L. Schalkwyk (1)<br />

1. IOP<br />

* karim.malki@iop.kcl.ac.uk<br />

Anti-depressant drugs are used as first line treatment in<br />

depression but response has been shown to be highly<br />

heterogeneous with drugs <strong>of</strong>ten failing to have the desired<br />

therapeutic effect. We report on an integrative analysis from<br />

the Genome-based Therapeutic Drugs for Depression<br />

(GENDEP) study using gene expression from mice to inform<br />

prioritisation in human pharmacogenetic GWAS. The same<br />

two antidepressants were used in mice and humans:<br />

escitalopram (a serotonin-reuptake inhibitor) and nortriptyline<br />

(a norepinephrine-reuptake inhibitor). The animal study used<br />

four inbred strains <strong>of</strong> mice (129S1/SvlmJ, C57LB/6J, DBA/2J<br />

and FVB/NJ), and two stress protocols (maternal separation<br />

and chronic mild stress). Hippocampus mRNA data was<br />

measured in 144 animals using the Affymetrix 430 chip.<br />

Behavioural data informed gene-expression analysis looking<br />

for Strain(G)xDrug(E) interactions. Twenty-six genes were<br />

identified as being differentially expressed according to strain<br />

and drug, including the phosphatase 1A gene (Ppm1a)<br />

differentially expressed with nortriptyline vs. saline. Single<br />

nucleotide polymorphisms tagging common sequence<br />

variation in human homologues <strong>of</strong> these genes were tested for<br />

association with response to antidepressants in 706<br />

participants <strong>of</strong> the GENDEP human pharmacogenetic study,<br />

treated with escitalopram or nortriptyline for 12 weeks, with<br />

available high-quality Illumina 610quad chip genotyping.<br />

Several polymorphisms in the protein phosphatase 1A gene<br />

(PPM1A) were significantly associated with response to<br />

nortriptyline in humans after correction for multiple testing.<br />

PPM1 encodes a phosphatase involved in MAP kinase<br />

signalling and cell stress response. The convergent evidence<br />

from mice and humans points to a role <strong>of</strong> the PPM1A in<br />

response to noradrenergic but not serotonergic antidepressants.


O2.6 INDIVIDUALIZED ANTIPSYCHOTIC THERAPY<br />

FOR SCHIZOPHRENIA<br />

K. Aberg* (1), J. McClay (1), D. Adkins (1), J. Bukszar (1),<br />

P. Jia (2), Z. Zhao (2), V. Vladimirov (3), J. Lieberman (4), P.<br />

Beardsley (5), P. Sullivan (6), E. van den Oord (7)<br />

1. Center for Biomarker Research and Personalized Medicine,<br />

Virginia Commonwealth University 2. Department <strong>of</strong><br />

Biomedical Informatics, Vanderbilt University <strong>Medical</strong> Center<br />

3. Virginia Institute for Psychiatric and Behavioral Genetics,<br />

Virginia Commonwealth University 4. Department <strong>of</strong><br />

Psychiatry, Columbia University 5. Department <strong>of</strong><br />

Pharmacology and Toxicology, Virginia Commonwealth<br />

University 6. Department <strong>of</strong> Genetics, University <strong>of</strong> North<br />

Carolina at Chapel Hill 7. Center for Biomarker Research and<br />

Personalized Medicine, Virginia Commonwealth University;<br />

Virginia Institute for Psychiatric and Behavioral Genetics,<br />

Virginia Commonwealth University<br />

* kaaberg@vcu.edu<br />

Understanding individual variation causing differences in<br />

efficacy and development <strong>of</strong> adverse effects, as a response to<br />

antipsychotic therapy is essential to optimize and individualize<br />

the treatment for schizophrenia. Genetic variations may<br />

potentially affect the response to antipsychotic treatment.<br />

We are conducting a series <strong>of</strong> genomewide association studies<br />

(GWAS) in the Clinical Antipsychotic Trials <strong>of</strong> Intervention<br />

Effectiveness (CATIE) for a wide variety <strong>of</strong> efficacy (e.g.<br />

disease symptoms, neurocognition) and toxicity (e.g. QT<br />

prolongation, movement and metabolic side effects) measures.<br />

To study these measures, we developed a systematic method<br />

to estimate treatment effects in CATIE that combines<br />

information from all assessments in an empirical fashion.<br />

After quality control, 492K successfully genotyped SNPs were<br />

available for 738 schizophrenia patients. Several signals<br />

reached genomewide significance at our pre-specified<br />

threshold ensuring that no more than 10% <strong>of</strong> our significant<br />

findings are expected to be false discoveries. A SNP in MEIS2<br />

mediated the effect <strong>of</strong> risperidone on hip and waist<br />

circumference and showed secondary associations with BMI,<br />

and blood pressure. Another SNP, located in SLC22A23,<br />

mediated the effect <strong>of</strong> quetiapine on QT prolongation. Many<br />

other signals were found for genes that are good candidates for<br />

further studies. We are developing a user-friendly databases to<br />

store all GWAS results. This database allow other researchers<br />

to design replication studies, search the CATIE results to<br />

confirm their findings, perform cross-outcome analyses, and<br />

combine new data with the CATIE GWAS results.<br />

Furthermore, we are currently developing methods to<br />

incorporate pathway information in the analyses, which will<br />

improve the statistical power.<br />

O3 ANXIETY<br />

O3.1 THE SYMPTOMATIC PROFILE OF PANIC<br />

DISORDER IS SHAPED BY THE 5-HTTLPR<br />

POLYMORPHISM<br />

T. Lonsdorf (1), M. Schalling* (1), C. Ruck (1), J. Bergstrom<br />

(1), G. Andersson (2), A. Ohman (1), N. Lindefors (1)<br />

1. Karolinska Institutet 2. Uppsala University<br />

* martin.schalling@ki.se<br />

The short allele <strong>of</strong> a functional polymorphism (5-HTTLPR) in<br />

the serotonin transporter is associated with reduced serotonin<br />

transporter (5-HTT) expression and also with neuroticism,<br />

amygdalare activity and facilitated fear conditioning. Thus the<br />

5-HTTLPR is a candidate variant for panic disorder. However,<br />

case-control studies have consistently failed to show an<br />

association between 5-HTTLPR and panic disorder. As<br />

psychiatric disorders are broadly defined phenotypes merging<br />

different symptoms to syndromes, they may not be very well<br />

suited for genetic association studies. An alternative approach<br />

is to measure symptoms along continuous psychological<br />

dimensions which may more appropriately reflect their<br />

biological underpinnings and may reveal subpopulations<br />

within clinical diagnostic groups. We recorded the<br />

symptomatic pr<strong>of</strong>ile in 73 mainly Caucasian panic disorder<br />

patients using observer-rated instruments (Panic Disorder<br />

Severity Scale, PDSS; Montgomery-Åsberg Depression<br />

Rating Scale, MADRS) prior to treatment, after10-weeks <strong>of</strong><br />

psychological treatment and six months after treatment. We<br />

observed a strong and time-stable association between bi-and<br />

triallelic 5-HTTLPR polymorphisms and the symptomatic<br />

pr<strong>of</strong>ile. Carriers <strong>of</strong> the 5-HTTLPR s-allele suffered from more<br />

severe panic and depressive symptoms as compared to those<br />

homozygous forthe l-allele. Our data highlight the importance<br />

<strong>of</strong> defining appropriate phenotypes for psychiatric genetic<br />

studies and demonstrate that the 5-HTTLPR genotype may be<br />

related to the symptomatic pr<strong>of</strong>ile rather than to the<br />

vulnerability to develop panic disorder.


O3.2 POLYMORPHISMS IN THE GENE ENCODING<br />

THE NEUROPEPTIDE GALANIN ARE ASSOCIATED<br />

WITH HPA-AXIS DYSREGULATION AND<br />

SYMPTOME SEVERITY IN MAJOR-DEPRESSIVE-<br />

AND ANXIETY-DISORDER PATIENTS.<br />

P. Unschuld* (1), M. Ising (1), D. Roeske (1), A. Erhardt (1),<br />

M. Specht (1), S. Ripke (1), M. Uhr (1), S. Kloiber (1), B.<br />

Müller-Myhsok (1), F. Holsboer (1), E. Binder (1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry, Kraepelinstr. 2-10,<br />

80804 München, Germany<br />

* unschuld@mpipsykl.mpg.de<br />

Galanin (GAL) is an estrogen inducible neuropeptide, highly<br />

expressed in brain-regions processing mood and behavior.<br />

GAL possibly has a direct modulatory effect on<br />

hypothalamic-pituitary-adrenal axis (HPA) regulation, as<br />

recent pharmacological and genetic studies indicate a<br />

significant role <strong>of</strong> GAL in stress-related disorders.<br />

By using a tag SNP approach covering the GAL-gene, we<br />

expanded an earlier finding <strong>of</strong> a genetic association with<br />

panic-disorder in female subjects to a larger sample <strong>of</strong> 268<br />

outpatients with anxiety disorders (AD) and 541 inpatients<br />

with major depressive disorder (MDD). As intermediate<br />

phenotype for treatment response, HPA-axis dysregulation<br />

using the combined dexamethasone suppression / CRH<br />

stimulation test both at admission and discharge was evaluated<br />

in 298 MDD-inpatients. To test for case-control associations,<br />

a healthy control sample (n=541) was used. Genotyping<br />

revealed high LD in the promotor area <strong>of</strong> the GAL-gene,<br />

including predescribed estrogen binding element sites. We<br />

found associations <strong>of</strong> the rare allele <strong>of</strong> rs948854 in female<br />

AD-patients with more severe anxious pathology and in<br />

premenopausal MDD-patients with higher HPA-axis activity<br />

at admission, more severe anxious pathology at discharge and<br />

non-remission. No significant case-control associations could<br />

be observed. The here presented data suggest a gender-specific<br />

role <strong>of</strong> GAL SNPs in liability for anxious and depressive<br />

disorders, characterized by a particularly dysregulated<br />

HPA-axis.<br />

O3.3 VARIANTS IN SLITRK1 ARE ASSOCIATED<br />

WITH THE OBSESSIVE-COMPULSIVE DISORDER<br />

SPECTRUM<br />

U. Ozomaro* (1), R. Moessner (2), M. Cuccaro (1), M.<br />

Pericak-Vance (1), M. Wagner (2), G. Feng (3), S. Zuchner<br />

(1)<br />

1. Miami Institute for Human Genomics, University <strong>of</strong> Miami,<br />

Miami, USA 2. Psychiatrische Klinik der Universitaet Bonn,<br />

Bonn, Germany 3. Department <strong>of</strong> Neurobiology, Duke<br />

University <strong>Medical</strong> Center, Durham, USA<br />

*uozomaro@med.miami.edu<br />

Obsessive-Compulsive Disorder (OCD) and the spectrum <strong>of</strong><br />

closely related disorders are characterized by persistent<br />

intrusive thoughts (obsessions), repetitive actions<br />

(compulsions), and excessive anxiety. Although heritability<br />

studies in OCD have shown an increased risk for first degree<br />

relatives, and twin studies revealed higher concordance<br />

amongst monozygotic twins (compared to dizygotic twins),<br />

the identification <strong>of</strong> the underlying risk-conferring genetic<br />

variation by means <strong>of</strong> classic genetic association studies has<br />

proven to be difficult. Recently, the possibility <strong>of</strong> a larger<br />

contribution <strong>of</strong> rare genetic variants to the risk <strong>of</strong> psychiatric<br />

disorder has been suggested by several successful studies. In<br />

accordance with this hypothesis we applied a rare variant<br />

association study design to explore SLITRK1, a gene<br />

previously linked to disorders <strong>of</strong> the OCD spectrum. We<br />

directly resequenced SLITRK1 in 279 OCD spectrum cases<br />

and in 185 psychiatric controls. We identified two novel<br />

nonsynonymous variants in five cases, N400I and T418S. The<br />

N400I variant was found in one case, while the T418S change<br />

was identified in four cases. Neither variant, nor any other<br />

nonsynonymous changes were detected in the controls. In a<br />

previous study, we identified two rare variants in SLITRK1,<br />

R584K and S593G, whose sequence changes were<br />

significantly associated with trichotillomania, an OCD<br />

spectrum disorder. Taking into consideration the combined<br />

mutation load <strong>of</strong> our two studies, OCD spectrum cases are<br />

significantly associated with variants in SLITRK1 (complete<br />

gene sequencing; p=0.043, Fisher’s exact test). SLITRK1<br />

codes for a neurite modulating protein and is found in the<br />

striatum. Additional support for a role <strong>of</strong> SLITRK1 in OCD<br />

spectrum disorders comes from a recent SLITRK1 knockout<br />

mouse characterized by anxiety and mirroring aspects <strong>of</strong> the<br />

human disorder. We are currently performing functional<br />

in-vitro studies to further clarify the molecular consequences<br />

<strong>of</strong> these variants.


O3.4 ACCELERATED TELOMERE SHORTENING IN<br />

A POPULATION-BASED NESTED CASE-CONTROL<br />

STUDY OF ANXIETY DISORDER PATIENTS<br />

L. Kananen (1), I. Surakka (2), S. Pirkola (3), J. Suvisaari (4),<br />

J. Lönnqvist (4), L. Peltonen (5), S. Ripatti (2), I. Hovatta*<br />

(6)<br />

1. Mol Neurol Res Program, Univ Helsinki, Finland 2. Public<br />

Health Genomics Unit, Natl Inst for Health and Welfare;<br />

FIMM, Inst for Mol Med Finland, Univ Helsinki, Finland 3.<br />

Dept <strong>of</strong> Psychiatry, Helsinki University Central Hospital<br />

Finland 4. Mental Health and Substance Abuse Services, Natl<br />

Inst for Health and Welfare, Helsinki, Finland 5. FIMM, Inst<br />

<strong>of</strong> Mol Med Finland, Univ Helsinki; Wellcome Trust Sanger<br />

Inst, Hinxton, UK 6. Mol Neurol Res Program, Univ Helsinki;<br />

Mental Health and Substance Abuse Services, Natl Inst for<br />

Health and Welfare; Dept <strong>of</strong> Med Genet, Univ Helsinki,<br />

Finland<br />

* iiris.hovatta@helsinki.fi<br />

Accelerated telomere shortening has been previously<br />

associated to self-perceived stress and psychiatric disorders,<br />

including schizophrenia and mood disorders. We hypothesized<br />

that accelerated telomere shortening might be involved in the<br />

etiology <strong>of</strong> anxiety disorders, in which stress and stressful life<br />

events are major environmental predisposing factors. We set<br />

out to test this hypothesis in a randomly selected<br />

population-based Health 2000 cohort with 974 samples<br />

including 321 anxiety disorder patients and 653 matched<br />

controls aged 30-87 years, who all underwent Composite<br />

International Diagnostic Interview. Relative telomere length <strong>of</strong><br />

peripheral blood cells was measured by quantitative real-time<br />

PCR. Anxiety disorder patients in the older age group (48-87<br />

years) exhibited significantly shorter telomeres than healthy<br />

controls. The association was highly stable and robust across<br />

adjustments for previously associated factors and potential<br />

confounders. In the whole sample and the younger age group<br />

(30-47 years), shorter telomere length was associated with<br />

greater number <strong>of</strong> childhood adverse life events, both in the<br />

anxiety disorder cases and controls. Childhood chronic illness<br />

was the most significantly associated single event affecting<br />

telomere length at the adult age. Self-reported current<br />

psychological distress, as measured by GHQ-12, did not have<br />

an effect on the telomere length. Our results suggest that<br />

accelerated telomere shortening in anxiety disorder patients is<br />

likely due to a long-term stress related to anxiety disorders.<br />

This finding strengthens the hypothesis that oxidative stress,<br />

which is known to affect telomere length, is involved in the<br />

pathogenesis <strong>of</strong> anxiety disorders.<br />

O3.5 PRIORITIZATION AND ASSOCIATION<br />

ANALYSIS OF MURINE-DERIVED CANDIDATE<br />

GENES IN ANXIETY-SPECTRUM DISORDERS<br />

J. Hettema (1), B. Webb (1), A. Guo (2), Z. Zhao (2), B.<br />

Maher (1), X. Chen (1), S. An (1), C. Sun (1), K. Kendler (1),<br />

P. Kuo (3), J. Flint (4), E. van den Oord (1)<br />

1. Virginia Commonwealth University 2. Vanderbilt<br />

University 3. National Taiwan University 4. Oxford<br />

University<br />

* jhettema@vcu.edu<br />

Introduction: Anxiety disorders are common psychiatric<br />

conditions that are highly comorbid with each other and<br />

related phenotypes such as depression and personality,<br />

possibly due to a shared genetic basis. Fear-related behaviors<br />

in mice have long been investigated as potential models <strong>of</strong><br />

anxiety-spectrum disorders, making integration <strong>of</strong> information<br />

from both murine and human genetic data a powerful strategy<br />

for identifying potential susceptibility genes for these<br />

conditions. Methods: A genome-wide association analysis<br />

(GWAS) <strong>of</strong> fear-related behaviours in heterogeneous<br />

stock mice identified a preliminary list <strong>of</strong> 54 novel candidate<br />

genes. We ranked these according to three complementary<br />

sources <strong>of</strong> anxiety-related genetic data: (1) human linkage<br />

scans, (2) a human GWAS study, and (3) linkage and<br />

knock-out studies in mice. We genotyped tagging SNPs<br />

covering the nine top-ranked genic regions in a two-stage<br />

association study <strong>of</strong> subjects genetically informative for<br />

anxiety-spectrum disorders. Results: Stage 1 screening<br />

identified SNPs in several <strong>of</strong> the regions that demonstrated<br />

suggestive association that were carried forward for<br />

genotyping in stage 2. While most <strong>of</strong> these did not retain their<br />

association in stage 2, multiple SNPs and related haplotypes in<br />

the PPARGC1A gene did. Conclusions: Integration <strong>of</strong> genetic<br />

data across human and murine studies suggests PPARGC1A<br />

as a potential susceptibility gene for anxiety-spectrum<br />

disorders.


O4 ADHD<br />

O4.1 HAPLOTYPE SEGREGATION IN MULTIPLEX<br />

ADHD FAMILIES AT DIFFERENT CHROMOSOMAL<br />

LOCI<br />

M. Lin* (1), T. Nguyen (2), C. Freitag (3), H. Palmason (1),<br />

C. Seitz (4), T. Renner (5), M. Romanos (5), S. Walitza (5), C.<br />

Jacob (6), K. Lesch (6), J. Meyer (1)<br />

1. Institute <strong>of</strong> Psychobiology, Department <strong>of</strong> Neurobehavioral<br />

Genetics, Trier, Germany 2. Institute <strong>of</strong> <strong>Medical</strong> Biometry and<br />

Epidemiology, Philipps-University, Marburg, Germany 3.<br />

Goethe-University Frankfurt am Main, Department <strong>of</strong> Child<br />

and Adolescent Psychiatry, Frankfurt, Germany 4. Saarland<br />

University Hospital, Department <strong>of</strong> Child and Adolescent<br />

Psychiatry, Homburg, Germany 5. University<strong>of</strong> Wuerzburg,<br />

Department <strong>of</strong> Child and Adolescent Psychiatry and<br />

Psychotherapy, Wuerzburg, Germany Wuerzburg, Germany 6.<br />

University <strong>of</strong> Wuerzburg, Department <strong>of</strong> Psychiatry and<br />

Psychotherapy, Wuerzburg, Germany<br />

* s1kalinn@uni-trier.de<br />

Attention-deficit hyperactivity disorder (ADHD) is a<br />

neuropsychiatric disorder characterized by symptoms <strong>of</strong><br />

inattention, hyperactivity and increased impulsiveness.<br />

Despite many studies over the last decades, the etiology <strong>of</strong><br />

ADHD remains unknown. Eight large ADHD-affected<br />

families were recruited in a multicenter collaboration study<br />

conducted by three clinical units <strong>of</strong> child and adult psychiatry<br />

(Universities <strong>of</strong> Trier, Homburg/Saar, and Wuerzburg). They<br />

comprised <strong>of</strong> 191 individuals, <strong>of</strong> which 95 were afflicted with<br />

ADHD. Fine-mapping with microsatellite markers <strong>of</strong> the<br />

significant loci reported in our recent paper (Romanos et al,<br />

2008) <strong>of</strong> a genome-wide linkage study <strong>of</strong> the eight large<br />

families was carried out. Using genetic data derived from<br />

related individuals within each <strong>of</strong> the large families, an<br />

attempt is made to identify predisposing chromosomal regions<br />

in these families by verifying which <strong>of</strong> these significant<br />

regions actually segregate among the ADHD-affected<br />

individuals in the pedigrees. Several <strong>of</strong> the large ADHD<br />

families show evidence <strong>of</strong> haplotype segregation at 1q25<br />

(HLOD = 2.99, p = 0.00021, alpha = 0), 9q22 (LOD = 2.61, p<br />

= 0.00053), 12p13 (LOD = 3.07, p = 0.00017), p = 0.0000065)<br />

and 18q11 (LOD = 2.90, p = 0.00026). Evidence <strong>of</strong> multiple<br />

segregating loci suggests genetic heterogeneity. To better<br />

discern the complexly inherited trait <strong>of</strong> ADHD, we will assess<br />

the segregating loci in these families in order to identify<br />

plausible candidate genes and variants contributing to the<br />

disorder. Further studies on these segregating regio regions <strong>of</strong><br />

chromosomes will be executed with next-generation<br />

sequencing techniques.<br />

O4.2 A NEURODEVELOPMENTAL PATHWAY FOR<br />

ATTENTION DEFICIT HYPERACTIVITY DISORDER<br />

(ADHD) : INTEGRATING THE GENOME-WIDE<br />

ASSOCIATION FINDINGS<br />

G. Poelmans (1), D. Pauls (2), J. Buitelaar (1), B. Franke* (3)<br />

1. Department <strong>of</strong> Psychiatry, Donders Institute for Brain,<br />

Cognition and Behaviour, Radboud University Nijmegen<br />

<strong>Medical</strong> Centre, Nijmegen, The Netherlands 2. Psychiatric and<br />

Neurodevelopmental Genetics Unit, Center for Human<br />

Genetic Research, Massachusetts General Hospital, Harvard<br />

<strong>Medical</strong> School, Boston, USA 3. Department <strong>of</strong> Psychiatry,<br />

Donders Institute for Brain, Cognition and Behaviour,<br />

Radboud University Nijmegen <strong>Medical</strong> Centre, Nijmegen,<br />

The Netherlands and Department <strong>of</strong> Human Genetics,<br />

Radboud University Nijmegen <strong>Medical</strong> Centre, Nijmegen,<br />

The Netherlands<br />

* g.poelmans@psy.umcn.nl<br />

Attention Deficit Hyperactivity Disorder (ADHD) is a<br />

common neuropsychiatric disorder observed in children and<br />

adults. The disorder is characterised by inappropriate levels <strong>of</strong><br />

hyperactive, impulsive and inattentive behaviours that lead to<br />

significant clinical and psychosocial impairments. ADHD<br />

belongs to the multifactorial disorders and is highly heritable.<br />

Recently, the first genome-wide association studies (GWAS)<br />

for ADHD have been published. Firstly, we compiled a list <strong>of</strong><br />

100 top single nucleotide polymorphisms (SNPs) in gene<br />

regions from the three published GWAS for ADHD, taking<br />

into account the reported SNPs with association at p <<br />

5.00E-04. Secondly, we searched the literature for the<br />

(proposed) function <strong>of</strong> these 100 candidate genes and tried to<br />

functionally integrate them into one pathway. We found that<br />

approximately 1 in 4 <strong>of</strong> the ADHD candidate genes emerging<br />

from the GWAS for ADHD (i.e. 26 out <strong>of</strong> the 100 putative<br />

ADHD candidate genes) fitted into a neurodevelopmental<br />

pathway that links axonal guidance genes/proteins at the<br />

synapse (such as the ROBO1, UNC5B, UNC5C, LPL,<br />

CDH13, CTNNA2, NTRK3, ADCYAP1R1 and NUCB1<br />

genes) with several downstream-acting adaptor and (neuronal)<br />

cytoskeleton associated genes/proteins (such as the FHIT,<br />

NEDD4L, SEC14L4, VWC2L, RAB1B, MBOAT1, EREG,<br />

PPM1F, RBM35A, MAP1B, MOBP, DNM1, ITGA11,<br />

MMP24 and SPOCK3 genes) and eventually results in<br />

neuronal migration, i.e. directed neurite outgrowth.<br />

In summary, we have identified a common eurodevelopmental<br />

pathway that may provide an important contribution to the<br />

understanding <strong>of</strong> ADHD etiology.


O4.3 RISK PATHWAYS OR PLEIOTROPIC EFFECTS?<br />

AN INVESTIGATION INTO COGNITIVE<br />

INTERMEDIATE PHENOTYPES CONTRIBUTING TO<br />

THE ASSOCIATION BETWEEN COMT VAL158MET<br />

GENOTYPE AND ANTISOCIAL BEHAVIOUR IN<br />

CHILDREN WITH ADHD<br />

K. Langley* (1), J. Heron (2), M. O'Donovan (1), M. Owen<br />

(1), A. Thapar (1)<br />

1. MRC Centre for Neuropsychiatric Genetics & Genomics,<br />

Cardiff University 2. ALSPAC, Dept <strong>of</strong> Social Medicine,<br />

University <strong>of</strong> Bristol<br />

* langleyk@cardiff.ac.uk<br />

As replicated genetic susceptibility loci for psychiatric<br />

disorders are identified, it becomes necessary to investigate<br />

the mechanisms through which such genotypes exert their<br />

effect. One proposal is to identify intermediate phenotypes<br />

lying on the risk pathway between genotype and outcome.<br />

An association between the COMT Val158Met polymorphism<br />

in the presence <strong>of</strong> ADHD and antisocial behaviour has been<br />

independently replicated in three separate studies and pooled<br />

analysis. We replicated this association in a further sample,<br />

then investigated the role <strong>of</strong> two potential intermediate<br />

phenotypes; executive functioning and “Social cognitive<br />

dysfunction” (lack <strong>of</strong> empathy and awareness <strong>of</strong> others’<br />

emotions). Utilising data from the prospective longitudinal<br />

ALSPAC sample <strong>of</strong> children aged eight years old, (n=4365)<br />

we replicated previous findings that Val/Val genotype in the<br />

presence <strong>of</strong> ADHD is associated with increased antisocial<br />

behaviour (beta=0.07, t=4.47, p=3x10-6). Measures <strong>of</strong><br />

executive function were associated with both the COMT<br />

Val/Val genotype (p=0.02 to p


O5 GENETIC OVERLAP<br />

O5.1 COPY NUMBER VARIATION AND GENE<br />

DOSAGE CHANGES IN AUTISM: OVERLAP WITH<br />

REGIONS INVOLVED IN SCHIZOPHRENIA AND<br />

BIPOLAR DISEASE<br />

M. Smith (1), P. Flodman (1), J. Gargus (1), M. Simon (1), J.<br />

Heck (1), D. Wallace (1)<br />

1. University <strong>of</strong> California, Irvine<br />

We utilized AFFYMETRIX SNP 6.0 microarrays to search for<br />

genome copy number variants (CNV) in autism. We<br />

discovered rare large CNV and smaller structural variants.<br />

Microarray studies revealed the presence <strong>of</strong> deletion <strong>of</strong> 4M<strong>of</strong><br />

chromosome 12p and duplication <strong>of</strong> 4M <strong>of</strong> 2p in siblings with<br />

autism. Follow-up FISH studies revealed an unbalanced<br />

chromosome translocation between2p and 12p in each sib.<br />

Adjacent genes hemizygously deleted in this sib pair include<br />

the CACNA1C voltage gated ion channel gene, ion channel<br />

CACNA2D andERC2 neurotransmitter release gene.<br />

CACNA1C has been implicated in other cases <strong>of</strong> autism and<br />

in association studies <strong>of</strong> bipolar disorder. In a subject with<br />

autism and aggression we identified <strong>of</strong> a 2q37.3 chromosome<br />

deletion and duplication on5q33-q34 that included 5 GABA<br />

receptor loci, GABRA1, GABRG2, GABRA6, GABRB2,and<br />

GABRP. This chromosome region has been implicated in the<br />

etiology <strong>of</strong> schizophrenia in a number <strong>of</strong> different studies. In a<br />

subject with autism and seizures we confirmed presence <strong>of</strong> a<br />

deletion <strong>of</strong> 15q13.3 that leads to loss <strong>of</strong> a number <strong>of</strong> genes<br />

including CHRNA7 neurotransmitter receptor gene. Deletions<br />

in this region have been implicated in seizure disorder, mental<br />

retardation and in schizophrenia. Smaller structural variants in<br />

our autistic subjects occur predominantly in genes active at the<br />

synapse, in signal transduction and mitochondrial function.<br />

O5.2 ABCA13: A CANDIDATE GENE FOR<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER<br />

D. Blackwood* (1), H. Knight (2), B. Pickard (3), A. Maclean<br />

(2), M. Malloy (2), D. Soares (3), A. McRae (4), A. Condie<br />

(5), A. White (5), W. Hawkin (5), K. McGhee (2), M. van<br />

Beck (1), D. Macintyre (1), J. Starr (6), I. Deary (7), P.<br />

Visscher (4), D. Porteous (3), R. Cannon (8), D. St. Clair (9),<br />

W. Muir (2)<br />

1. Division <strong>of</strong> Psychiatry, University <strong>of</strong> Edinburgh, Royal<br />

Edinburgh Hospital, Edinburgh, EH10 5HF, UK<br />

2. Division <strong>of</strong> Psychiatry, University <strong>of</strong> Edinburgh, Royal<br />

Edinburgh Hospital, Edinburgh, EH10 5HF, UK; <strong>Medical</strong><br />

Genetics, Institute <strong>of</strong> Genetics and Molecular Medicine,<br />

University <strong>of</strong> Edinburgh, Molecular Medicine Centre, Western<br />

General Hospital, Crewe Road 3. <strong>Medical</strong> Genetics, Institute<br />

<strong>of</strong> Genetics and Molecular Medicine, University <strong>of</strong> Edinburgh,<br />

Molecular Medicine Centre, Western General Hospital, Crewe<br />

Road, Edinburgh, EH4 2XU, UK 4. Queensland Institute <strong>of</strong><br />

<strong>Medical</strong> Research, 300 Herston Road, Herston 4006, QLD,<br />

Australia 5. Wellcome Trust Clinical Research Facility,<br />

Western General Hospital, Crewe Road, Edinburgh,<br />

EH4 2XU, UK 6. Centre for Cognitive Ageing and Cognitive<br />

Epidemiology, Geriatric Medicine unit, University <strong>of</strong><br />

Edinburgh, Royal Victoria Hospital, Craigleith Road,<br />

Edinburgh, EH4 2DN, UK 7. Centre for Cognitive Ageing and<br />

Cognitive Epidemiology, Department <strong>of</strong> Psychology,<br />

University <strong>of</strong> Edinburgh, 7 George Square, Edinburgh, EH8<br />

9JZ, UK 8. Cancer Biology Group, National Center for<br />

Toxicogenomics, NIEHS, Research Triangle Park,<br />

North Carolina 27709, USA 9. Institute <strong>of</strong> <strong>Medical</strong> Sciences,<br />

University <strong>of</strong> Aberdeen, Foresterhill, Aberdeen, AB25 2ZD,<br />

UK<br />

* d.blackwood@ed.ac.uk<br />

We investigated the brain expressed lipid transporter gene<br />

ABCA13 for a role in mental illness after showing it was<br />

directly disrupted by a breakpoint in a complex chromosome<br />

abnormality involving chromosomes 7 and 8 in a person with<br />

chronic schizophrenia. Sequencing ABCA13 exons in 100<br />

cases with schizophrenia and 100 controls revealed rare<br />

coding variants including one nonsense and nine missense<br />

mutations specific to or more common in schizophrenia. These<br />

10 variants were assayed in additional cohorts <strong>of</strong> patients with<br />

schizophrenia (~1000) bipolar disorder (~600) and controls<br />

(~2000) and their frequency was greater than controls in<br />

schizophrenia (OR=1.93, p=0.0057) and bipolar disorder<br />

(OR=2.71, p=0.00007). In the next stage we examined for the<br />

presence <strong>of</strong> each <strong>of</strong> the 10 variants in the families <strong>of</strong> carriers.<br />

106 members <strong>of</strong> 21 families took part and family size ranged<br />

from 3 to 16. Non-parametric single point linkage analysis<br />

using MERLIN to estimate identity by descent (IBD)<br />

between all pairs <strong>of</strong> affected relatives showed significant<br />

linkage <strong>of</strong> ABCA13 mutations to a phenotype including<br />

schizophrenia, bipolar disorder and major depression<br />

(LOD=4.38; p=3.5 x 10-6). These findings establish a role for<br />

ABCA13 and lipid biology in schizophrenia and bipolar<br />

disorder and also demonstrate that different rare penetrant<br />

variants in one gene may cause either schizophrenia, bipolar<br />

disorder or major depression in different families.


O5.3 RARE COPY NUMBER VARIANTS: A POINT OF<br />

RARITY IN GENETIC RISK FOR BIPOLAR<br />

DISORDER AND SCHIZOPHRENIA?<br />

D. Grozeva* (1), G. Kirov (1), D. Ivanov (2), I. Jones (1), L.<br />

Jones (3), E. Green (1), D. StClair (4), A. Young (5), N.<br />

Ferrier (5), A. Farmer (6), P. McGuffin (6), W. Wellcome<br />

Trust Case Control Consortium (7), P. Holmans (2), M. Owen<br />

(1), M. O'Donovan (1), N. Craddock (1)<br />

1. MRC Centre for Neuropsychiatric Genetics and Genomics,<br />

Department <strong>of</strong> Psychological Medicine, School <strong>of</strong> Medicine,<br />

Cardiff University, Heath Park, Cardiff, CF14 4XN, United<br />

Kingdom 2. MRC Centre for Neuropsychiatric Genetics and<br />

Genomics, Biostatistics and Bioinformatics Unit, Department<br />

<strong>of</strong> Psychological Medicine, School <strong>of</strong> Medicine, Cardiff<br />

University, Heath Park, Cardiff, CF14 4XN, United Kingdom<br />

3. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Birmingham,<br />

National Centre for Mental Health, 25 Vincent Drive,<br />

Birmingham, B15 2FG, United Kingdom 4. University <strong>of</strong><br />

Aberdeen, Institute <strong>of</strong> <strong>Medical</strong> Sciences, Foresterhill,<br />

Aberdeen AB25 2ZD, United Kingdom 5. School <strong>of</strong><br />

Neurology, Neurobiology and Psychiatry, Royal Victoria<br />

Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1<br />

4LP, United Kingdom 6. SGDP, The Institute <strong>of</strong> Psychiatry,<br />

King's College London, De Crespigny Park, Denmark Hill,<br />

London SE5 8AF, United Kingdom 7. Wellcome Trust Case<br />

Control Consortium<br />

* GrozevaDV@cf.ac.uk<br />

Recent studies suggest that copy number variation in the<br />

human genome is extensive and may play an important role in<br />

susceptibility to disease, including neuropsychiatric disorders<br />

such as schizophrenia and autism. The possible involvement<br />

<strong>of</strong> copy number variants (CNVs) in Bipolar disorder (BD) has<br />

received little attention to date. We sought to determine<br />

whether large (>100kb) and rare (found in > 1% <strong>of</strong> the<br />

population) CNVs are associated with susceptibility to BD and<br />

to make comparisons with findings in schizophrenia.<br />

We performed a genome-wide survey for CNVs in 1697 BD<br />

patients and 2806 healthy controls from the Wellcome Trust<br />

Case Control Consortium, using the Affymetrix 500K array.<br />

The burden <strong>of</strong> large CNVs (≥1Mb) in BD was not increased<br />

compared with controls, and was significantly less than in<br />

schizophrenia cases. CNVs previously implicated in the<br />

aetiology <strong>of</strong> schizophrenia were not more common in cases.<br />

Schizophrenia and bipolar disorder differ with respect to CNV<br />

burden in general, and association with specific CNVs in<br />

particular. Our data are consistent with the possibility that<br />

possession <strong>of</strong> large rare deletions may modify the phenotype<br />

in those at risk <strong>of</strong> psychosis: those possessing such events are<br />

more likely to be diagnosed with schizophrenia; those without<br />

such events are more likely to be diagnosed with bipolar<br />

disorder.<br />

O5.4 BETA-CATENIN PROMOTER CHIP-CHIP<br />

REVEALS POTENTIAL SCHIZOPHRENIA AND<br />

BIPOLAR DISORDER GENE NETWORK<br />

E. Pedrosa (1), A. Shah (1), C. Villa (1), C. Tenore (2), M.<br />

Capogna (3), H. Lachman* (1)<br />

1. AECOM 2. SUNY, Binghamton 3. Boston College<br />

*lachman@aecom.yu.edu<br />

Therapeutic concentrations <strong>of</strong> lithium salts inhibit GSK3β and<br />

phosphoinositide (PI) signaling suggesting that abnormal<br />

activation <strong>of</strong> these pathways could be a factor in bipolar<br />

disorder (BD) pathogenesis. Involvement <strong>of</strong> these pathways is<br />

also supported by recent genome wide association (GWA)<br />

studies. One way investigators have tried to understand the<br />

molecular basis <strong>of</strong> BD and the therapeutic action <strong>of</strong> lithium is<br />

by microarray expression studies, since the signal transduction<br />

pathways affected by the drug are coupled to transcriptional<br />

activation and inhibition. However, expression pr<strong>of</strong>iling has<br />

some limitations and investigators cannot use the approach to<br />

carry out analyses on fetal brain tissue, arguably the most<br />

relevant biological structure related to BD pathogenesis. In<br />

addition, the relative contribution <strong>of</strong> PI and GSK3β signaling<br />

in mediating the effects <strong>of</strong> lithium on gene expression cannot<br />

be assessed by simply analyzing lithium-treated cells. To<br />

address these shortcomings, we have taken a novel approach<br />

to help understand lithium’s effect on gene expression using<br />

chromatin immunoprecipiation-enriched material annealed to<br />

microarrays (ChIP-chip) targeting genes in fetal brain tissue<br />

bound by β-catenin, a transcription factor that is directly<br />

regulated by GSK3β. The promoters for several hundred genes<br />

were found to bind β-catenin, many <strong>of</strong> which are<br />

schizophrenia (SZ) and BD candidate genes, including four<br />

(PLXNA2, CACNA1B, NRNG and NOTCH4 CACNA1B)<br />

that were identified as candidates in recently published GWA<br />

studies. The findings suggest that seemingly disparate<br />

candidate genes for SZ and BD can be incorporated into a<br />

common molecular network revolving around SK3β/β-catenin<br />

signaling. In addition, the finding that a subgroup <strong>of</strong> SZ<br />

candidate genes may be influenced by a lithium-responsive<br />

pathway could have therapeutic implications.


O5.5 WHOLE-GENOME ANALYSIS REVEALS AN<br />

OVERLAPPING GENETIC BASISFOR BIPOLAR<br />

DISORDER, MAJOR DEPRESSION, AND<br />

SCHIZOPHRENIA<br />

T. Schulze* (1), N. Akula (1), R. Breuer (2), M. Nalls (3), M.<br />

Nöthen (4), S. Cichon (4), A. Singleton (3), M. Rietschel (2),<br />

Bipolar Genome Study (BiGS) Consortium (1), F. McMahon<br />

(1)<br />

1. NIMH 2. CIMH (Mannheim, Germany) 3. NIA 4. Life &<br />

Brain, University <strong>of</strong> Bonn (Germany)<br />

* schulzet@mail.nih.gov<br />

Family, linkage, and association studies have long suggested<br />

some shared genetic basis for major psychiatric disorders. We<br />

used genome-wide association study (GWAS) data from the<br />

Genetic Association Information Network (GAIN) bipolar<br />

disorder study (n=2064), the Wellcome Trust Case<br />

Control Consortium (WTCCC) bipolar disorder sample<br />

(n=4840), a German bipolar GWAS data set (n=2066), the<br />

GAIN major depression study (n=3573), and the GAIN<br />

schizophrenia data set (n=2729) in order to test for genetic<br />

relationships among these disorders. A Parkinson Disease<br />

GWAS (n=537) dataset was included as a negative control.<br />

We weighted each SNP by the association results in a<br />

specified discovery sample and then assigned a score to each<br />

individual on the basis <strong>of</strong> the weighted burden <strong>of</strong> risk alleles<br />

(WBRA) across all SNPs in a specified test sample. Using the<br />

WBRA derived from the GAIN bipolar sample, we could<br />

discriminate cases from controls in the WTCCC sample (p =<br />

2.5 x 10-10) with a predictive accuracy (as expressed by the<br />

area under the curve; AUC) <strong>of</strong> 0.55. The same WBRA<br />

discriminated cases from controls in the German bipolar<br />

(p=4.6 x 10-8; AUC=0.57) and the GAIN major depression<br />

sample (p=7.32 x 10-7; AUC=0.57). Conversely, the WBRA<br />

derived from the WTCCC bipolar sample predicted<br />

schizophrenia affection status with a similar accuracy (p=2.9 x<br />

10-9; AUC=0.56). In contrast, the WBRA derived from the<br />

GAIN bipolar sample, had no predictive value in the<br />

Parkinson Disease dataset (AUC= 0.50, p=ns). These results<br />

confirm formal genetic evidence that major psychiatric<br />

disorders share largely overlapping sets <strong>of</strong> risk alleles, which<br />

are distinct from those that predispose to a neurological<br />

disorder such as Parkinson Disease.<br />

O6 SCHIZOPHRENIA 1<br />

O6.1 DUPLICATION AND DELETION AT 15Q11.2 ARE<br />

ASSOCIATED WITH SCHIZOPHRENIA<br />

Q. Zhao (1), G. Feng (2), K. Huang (1), Z. Zhang (1), X. Zhao<br />

(3), T. Li (1), G. He (1), T. Wang (1), Z. Zeng (1), D. St Clair<br />

(4), L. He (1), Y. Shi (1)<br />

1. Bio-X Center and Bio-X Center Affiliated Changning<br />

Mental Health Center, Shanghai Jiao Tong<br />

University, Shanghai, PR China; 2. Shanghai Institute <strong>of</strong><br />

Mental Health, Shanghai, PR China; 3. Institutes <strong>of</strong><br />

Biomedical Sciences, Fudan University Shanghai, PR China;<br />

4. Department <strong>of</strong> Mental Health, University <strong>of</strong> Aberdeen;<br />

Schizophrenia is a severe mental disorder marked by<br />

hallucinations, delusions, cognitive deficits and apathy, with a<br />

heritability estimated at 73–90%. However genetic variants<br />

involved are proved difficult to understand. In recent years,<br />

SNP chips led to the quick identification <strong>of</strong> copy number<br />

variatons (CNVs). As a result, rare copy number variations<br />

have been found to be important risk factors <strong>of</strong> schizophrenia.<br />

One <strong>of</strong> the those promissing loci is the CNV at 15q11.2,<br />

including four genes, TUBGCP5, CYFIP1, NIPA2 and<br />

NIPA1. TUBGCP5 is specific expressed in the subthalamic<br />

nuclei while the latter three are widely expressed in the central<br />

nervous system. Deletion <strong>of</strong> the four genes was implicated in<br />

delayed motor and speech development, behavioural problems<br />

and lower intellectual ability in PWS/AS patients. A recent<br />

study showed that microdeletion at this loci was significant<br />

associated with schizophrenia. In the present work, we<br />

screened this CNV in 1568 unrelated paranoid schizophrenia<br />

patients and 1728 randomly-chosen normal controls in total<br />

by affymetrix 500K/6.0 SNP arrays or RT-PCR then validated<br />

by MLPA and Taqman assays in the Chinese Han population.<br />

We detected 8 deletions in cases and 1 in controls in total<br />

(OR=8.95, p=0.012). We found that the CNV at 15q11.2 were<br />

significantly associated with schizophrenia (p=8.51×10-5).<br />

Our data provided further evidence that deletion at 15q11.2 is<br />

a risk factor and suggested that duplication at this loci was<br />

also implicated in the pathogenesis <strong>of</strong> schizophrenia in<br />

Chinese Han population. Our findings also supported the<br />

incomplete penetrance <strong>of</strong> duplication and deletion at this loci.


O6.2 STRUCTURAL IMAGING REVEALS NOVEL<br />

GENETIC INFLUENCES IN SCHIZOPHRENIA<br />

J. Turner* (1), J. Segall (2), G. Guffanti (1), V. Calhoun (2),<br />

H. Bockholt (2), S. Potkin (1), F. Macciardi (1), F. BIRN (1)<br />

1. University <strong>of</strong> California, Irvine 2. MIND Research Network<br />

* turnerj@uci.edu<br />

Purpose. We combined grey matter volumetric measures with<br />

GWS data to identify genetic influences on grey matter loss<br />

which are different in schizophrenia than in age- and<br />

gender-matched healthy subjects. Methods. One hundred<br />

fifty-four subjects either with schizophrenia (SZ) or healthy<br />

volunteers (HV) were part <strong>of</strong> a larger study for the Functional<br />

Imaging Biomedical Informatics Research. They were scanned<br />

with an MP-RAGE or equivalent protocol, using a mixture <strong>of</strong><br />

1.5T and 3T scanners. Mean grey matter density measures<br />

over standard anatomical regions which were significantly<br />

different between groups were combined into a set <strong>of</strong> 14<br />

quantitative traits (QT). Blood samples were genotyped using<br />

the Illumina HumanHap300 BeadArray. Each QT and single<br />

nucleotide polymorphism was analyzed in a general linear<br />

model to identify significant interactions (p < 10-6) between<br />

genotype and diagnosis on the phenotype. Results. The<br />

anterior cingulate and frontal medial areas both identified<br />

interactions with ADAMTS18 (16q23.1); frontal medial areas<br />

also showed a significant interaction with the MPRIP gene<br />

(17p11.2). The frontal inferior and middle regions identified<br />

areas in strong linkage disequilibrium with PRKCB1 and<br />

CHP2 (16p12.1). The superior temporal gyrus identified the<br />

CDKAL1 gene (6p22.3). Conclusions. Imaging genetics<br />

analyses in schizophrenia have identified neurodevelopmental<br />

and stress-related genetic effects on a functional phenotype;<br />

using structural phenotypes identifies genes known to be<br />

involved in inflammatory disorders (ADAMTS18, CDKAL1),<br />

metabolic disorder (PRKCB1, CHP2), regulation <strong>of</strong> synaptic<br />

transmission (PRKCB1), and stress actin fibers (MPRIP).<br />

These provide a picture <strong>of</strong> multiple interacting genetic<br />

influences on regional grey matter loss found in the disorder.<br />

O6.3 CLINICAL AND COGNITIVE ASSOCIATIONS<br />

AND DATA MINING EVIDENCE OF EPISTASIS IN<br />

THE NIMH/GCAP SCHIZOPHRENIA GWA STUDY<br />

R. Straub* (1), Y. Iizuka (1), N. Feng (1), C. Li (1), B.<br />

Kolachana (1), K. Vakkalanka (1), M. Egan (2), D. Dickinson<br />

(1), D. Weinberger (1)<br />

1. GCAP, NIMH, NIH 2. Merck Pharmaceuticals<br />

* straubr@intra.nimh.nih.gov<br />

We genotyped part <strong>of</strong> our GCAP sample - 270 controls, 197<br />

schizophrenia trios (115 with an unaffected sibling), and 55<br />

additional schizophrenics - with ILLUMINA 550k/610k chips,<br />

and downloaded 3 Illumina iControl genotype datasets and<br />

GAIN Schizophrenia genotypes from dbGAP. We ran TDT<br />

and case-control tests <strong>of</strong> association to the clinical phenotype<br />

and QTDT and regression analyses <strong>of</strong> a range <strong>of</strong> quantitative<br />

phenotypes in patients and controls, including cognition, EEG,<br />

fMRI, and TPQ. Genome wide analyses <strong>of</strong> the effects <strong>of</strong> risk<br />

associated SNPs on expression and expression correlation<br />

patterns were performed using our own expression database<br />

and publicly available databases, and potential biochemical<br />

interactions between genes investigated by pathway analysis.<br />

Two statistical approaches were used to examine genetic<br />

interactions. First we ran genome wide SNP-SNP case-control<br />

epistasis analysis. The most significant GCAP result (PLINK<br />

Epistasis p=4.53e-10) was between NOS1 and LRP2, each <strong>of</strong><br />

which physically bind to the known susceptibility gene<br />

NOS1AP (CAPON). Numerous other interactions involving<br />

known susceptibility genes were also observed. Second, we<br />

used the data mining programs RANDOM FOREST and<br />

TREENET from Salford Systems (San Diego, CA) to identify<br />

SNPs potentially involved in higher order interactions<br />

influencing clinical risk. After imputation <strong>of</strong> a random subset<br />

(522 schiz., 600 controls) <strong>of</strong> the GAIN (“GRU” <strong>of</strong> the<br />

MGS-EA) sample, we mined 332,236 SNPs in parallel in the<br />

GCAP (252 schiz, 270 controls) and GAIN samples. In<br />

GCAP, a set <strong>of</strong> 4430 SNPs (median PLINK Genotypic<br />

case-control p=0.1028) within or closest to 2868 genes were<br />

strongly predictive <strong>of</strong> group (TREENET 10-fold cross<br />

validation test ROC 0.987, schiz. prediction error 0.06, control<br />

prediction error 0.07). In GAIN, a set <strong>of</strong> 4371 SNPs SNPs<br />

within or closest to 2865 genes were strongly predictive <strong>of</strong><br />

GAIN group (TREENET test ROC 0.956, schiz prediction<br />

error 0.11, control prediction error 0.12). By chance,<br />

prediction error and prediction success are each 0.50, and only<br />

a fraction <strong>of</strong> these SNPs are likely to be tagging schizophrenia<br />

susceptibility genes. Nevertheless, we found that at both the<br />

SNP and gene level there was overlap between the two<br />

independent samples: 60 SNPs were in common, 182 genes<br />

had multiple SNPs from both samples closest to them, and 139<br />

<strong>of</strong> these had SNPs within them, and within 100kb in both<br />

studies. The initial pathway analysis (INGENUITY<br />

SYSTEMS, Redwood CA) <strong>of</strong> 131 <strong>of</strong> these showed 22<br />

potentially involved in protein-protein (PP) interactions, in 7<br />

groups: [CACNA1D, RYR2, HOMER, PARK2, PACRG,<br />

GRIA1], [ATXN1, A2BP1, FAT4, EIF4ENIF1], [PDZD2,


CTNND2, MAGI2], [ERC2, PCLO], [MAGI1, RPS6KA2],<br />

[PLXNA2, SEMA3A], and [GABRG3, GABRG2, GABRB3].<br />

Addition <strong>of</strong> just 4 PP “bridges” (CACNA1C, GRID2, DLG4,<br />

and NOVA1) connect the first 4 groups, which then include<br />

additional genes (eg. CDH13, ODZ2, NRG3, DPP6, KCNH4)<br />

from the 131, totalling 26. In contrast, genes from the top 131<br />

genome wide TDT SNPs, and the top and bottom 131 case<br />

control SNPs contained only 5, 4 and 6 PP interactors<br />

respectively, and these were not merged by adding single<br />

bridges. Our integrated database <strong>of</strong> clinical, cognitive, and<br />

data mining results also contains genes from the literature,<br />

tracking about 400 specific genes/regions. We have confirmed<br />

many prior gene findings, and some <strong>of</strong> the more interesting<br />

functional pathways we are filling out are indicated by the<br />

following selections, many <strong>of</strong> which are novel with regard to<br />

SNP association to schizophrenia : ASTN2, AUTS2,<br />

CAMTA1, CDH8, CMYA5, CNTN4, CNTNAP2, CSMD1,<br />

CSMD3, CSNK1A1L, CTNNA2, CTNNA3, DMD, DOCK2,<br />

ESRRG, FHIT, FSHR, GUCY1A2, GUCY1A3,<br />

HLA-DRB1/DRB5, HS3ST1, HS3ST3A1, MAD1L1, NBAS,<br />

ODZ4, OFCC1, OSBPL3, PINK1, PKHD1, PSMD13,<br />

PTPRD, PTPRG, RTP4, S100A7/S100A6, SGCZ,<br />

SLC10A2, SLC24A3, SLIT3, ST8SIA3, TLE3, UNC5C.<br />

Ongoing work includes typing the remainder <strong>of</strong> our sample<br />

(503 schiz total, 600 controls) with the ILLUMINA 660W<br />

chip, epistasis analysis and data mining <strong>of</strong> additional<br />

independent samples, and detection <strong>of</strong> CNVs with the<br />

AGILENT 1x1M chip. A multilevel, integrative approach<br />

fundamentally acknowledging the genotypic and phenotypic<br />

heterogeneity and epistasis inherent in the condition, and<br />

building on prior findings is essential to properly evaluate and<br />

utilize GWA results, and we think that this has produced many<br />

new candidates, each supported by multiple pieces <strong>of</strong><br />

evidence.<br />

O6.4 ASSOCIATION OF THE DISOCIDIN DOMIAN<br />

RECEPTOR 1 WITH POSITIVE YMPTOMATOLOGY<br />

IN SCHIZOPHRENIC PATIENTS<br />

S. de la Portilla (1), J. Valero (1), M. Cortes (1), B. Roig (1),<br />

E. Vilella* (1)<br />

1. HPUIPM, IISPV, URV<br />

* elisabet.vilella@urv.cat<br />

We recently published a genetic association between the<br />

discoidin domain receptor 1 (DDR1) gene and schizophrenia<br />

(Roig et al. 2007). DDR1 is a tyrosine kinase receptor present<br />

in the myelin sheath <strong>of</strong> CNS axons (Franco-Pons et al, 2006).<br />

Moreover, we showed that the rs1049623 expression in<br />

lymphocytes is genotype dependent (Roig et al. 2007). In the<br />

present work we explore possible influence <strong>of</strong> SNP<br />

rs1049623 on clinical variables. A total <strong>of</strong> 125 patients with a<br />

DSMIV schizophrenia diagnosis were evaluated with the<br />

positive and negative syndrome scale (PANSS). Patients with<br />

DDR1 SNP rs1049623 GG genotype have higher scores in<br />

delusions (P=0.004), excitement (P=0.03), grandiosity<br />

(P=0.001) and positive subscale (P=0.041) and lower scores in<br />

emotional and social withdrawal (P=0.26 and 0.01<br />

respectively) and total negative subscale (P=0.044). In the<br />

psychopathological subscale GG genotype was also associated<br />

with lower scores in disorientation (P=0.002), and disturbance<br />

<strong>of</strong> volition (P=0.03) and higher scores in lack <strong>of</strong> judgment and<br />

insight (P=0.03). No statistical differences were obtained for<br />

the total items <strong>of</strong> the PANSS. These data demonstrates that<br />

patients with the GG genotype punctuated differently in the<br />

PANSS scale and therefore a possible causal relationship<br />

between DDR1 and schizophrenia may exist.


O6.5 ASSOCIATION OF 5HT2C RECEPTOR<br />

POLYMORPHISMS WITHPSYCHOPATHOLOGY<br />

AND TREATMENT RESPONSE IN SCHIZOPHRENIA<br />

H. Meltzer* (1), K. Jayathilake (1), H. Hashimoto (2)<br />

1. Vanderbilt University School <strong>of</strong> Medicine 2. Graduate<br />

School <strong>of</strong> Pharmaceutical Sciences, Osaka University<br />

* herbert.meltzer@vanderbilt.edu<br />

The serotonin (5-HT) 2C receptor has important tonic<br />

inhibitory effects on dopamine release in brain areas critical<br />

to schizophrenia, including the frontal cortex, nucleus<br />

accumbens and ventral tegmentum. We will report an<br />

association between the 5-HT2C receptor polymorphisms<br />

rs3813929 (-759C/T), rs518147 (-697G/C), both <strong>of</strong> which are<br />

promoter polymorphisms, and rs6318 (Cys23Ser), a disputed<br />

functional SNP, and baseline psychopathology and response to<br />

treatment in Caucasian and Afro-American patients with<br />

schizophrenia (or schizoaffective disorder). The Ser23 allele<br />

carriage was strongly associated with higher baseline scores <strong>of</strong><br />

the BPRS psychosis subscale (p = 0.007) and total (p<br />

= 0.01) scores. The SNP -759C/T was also significantly<br />

associated with the psychosis subscale (p = 0.04). The<br />

haplotype (-759C)–(-697C)–(23Ser) (p = 0.00091) and<br />

(-759C)–(23Ser) (P = 0.00055) were strongly associated with<br />

BPRS psychosis subscale. In addition, the Ser23 allele<br />

carriage was significantly associated with improvement in<br />

negative symptoms following 6 weeks and 6 month APD<br />

treatment. The relationship <strong>of</strong> these SNPs to cognitive<br />

dysfunction and its amelioration will also be reported.<br />

O6.6 COPY NUMBER VARIATIONS OF<br />

SCHIZOPHRENIA SUSCEPTIBILITY LOCI<br />

ARE ASSOCIATED WITH A SPECTRUM OF SPEECH<br />

AND DEVELOPMENTAL DELAYS AND BEHAVIOR<br />

PROBLEMS<br />

T. Sahoo* (1), J. Rosenfeld (1), L. Shaffer (1)<br />

1. Signature Genomic Laboratories<br />

* sahoo@signaturegenomics.com<br />

Although the etiology <strong>of</strong> schizophrenia is not well understood,<br />

a genetic predisposition is presumed to be a determinant <strong>of</strong><br />

susceptibility to the disease. Recently, microarray-based<br />

comparative genomic hybridization (aCGH) and other<br />

molecular techniques have identified novel copy number<br />

variants (CNVs) in individuals with schizophrenia. We<br />

undertook a “genotype-first” analysis <strong>of</strong> individuals with<br />

microdeletions and microduplications overlapping ~30<br />

putative schizophrenia susceptibility loci among individuals<br />

referred to our laboratory with developmental or growth delay,<br />

birth defects, and/or behavior problems. After excluding<br />

individuals with CNVs commonly associated with<br />

schizophrenia (e.g., microdeletion <strong>of</strong> 22q11.21) and those with<br />

CNVs encompassing but substantially larger than those<br />

reported in the literature, we identified CNVs involving 10<br />

susceptibility loci among 52 individuals. In 18 individuals the<br />

CNVs encompassed or disrupted developmental pathway<br />

genes. None <strong>of</strong> the individuals had schizophrenia as an<br />

indication for study. The phenotypes included developmental<br />

and speech delay and behavioral problems, including<br />

obsessive behavior in a few. Of the 21 cases where parental<br />

studies were possible, the CNV was inherited in 14 and de<br />

novo in 7. Additionally, a search for significant aCGH<br />

findings in individuals referred for testing for schizophrenia<br />

identified six with CNVs, including one with a<br />

microduplication <strong>of</strong> the 22q11.21 DiGeorge syndrome region<br />

and one with a microdeletion at 16p12.2, which has been<br />

identified in individuals referred for autistic features. Our<br />

results suggest CNVs across schizophrenia susceptibility loci<br />

are <strong>of</strong>ten associated with phenotypes that minimally overlap<br />

with schizophrenia, and additional factors are likely required<br />

to lead to the development <strong>of</strong> schizophrenia.


O7 MOOD DISORDERS 2<br />

O7.1 COPY NUMBER VARIATION AT 15Q11.2 ARE<br />

ASSOCIATED WITH BIPOLAR DISORDER AND<br />

MAJOR DEPRESSION IN CHINESE HAN<br />

POPULATION<br />

Y. Shi* (1), Q. Zhao (1), L. He (1)<br />

1. Bio-X Center, Shanghai Jiao Tong University<br />

* shiyongyong@gmail.com<br />

Bipolar disorder and major depressive disorder are related to a<br />

distinct change <strong>of</strong> mood. Bipolar disorder is a relatively<br />

common psychiatric disease which is characterized by mood<br />

symptoms <strong>of</strong> mania and depression with a heritability<br />

estimated at 62~80%. Major depressive disorder is<br />

characterized by sadness or irritability and accompanied by at<br />

least several psychophysiological changes. Early-onset,<br />

severe and recurrent depression may have a higher heritability<br />

than other forms <strong>of</strong> depression. Up till now, Rrare copy<br />

number variations emerged as important risk factors <strong>of</strong> several<br />

major psychiatric disorders, and some <strong>of</strong> them could be risk<br />

factors for different diseases. One <strong>of</strong> the most promissing loci<br />

is the CNV at 15q11.2. 15q11.2 spans, including four genes,<br />

namely TUBGCP5, CYFIP1, NIPA2 and NIPA1. Deletions in<br />

this loci locus was commonly found in Prader-Willi<br />

Syndrome/Agenleman Syndrome patients. And, Aa recent<br />

study showed that schizophrenia was associated with<br />

microdeletion at this loci. was significant associated with<br />

schizophrenia. In the present work, we screened this CNV in<br />

1448 unrelated bipolar disorder patients, 1317 unrelated major<br />

depressive disorder patients (no sign <strong>of</strong> bipolar disorder<br />

symptoms during two-year track after onset) and 1728 normal<br />

controls in total by affymetrix 500K/6.0 SNP arrays or<br />

RT-PCR then validated by MLPA or Taqman assays. We<br />

found that the CNV at 15q11.2 were significantly associated<br />

with bipolar disorder ( p=7.4×10-5 ) and major depressive<br />

disorder ( p=1.95×10-4 ). Our data suggested that this CNV,<br />

including both microdeletions and duplications at this locus, is<br />

awere shared risk factor <strong>of</strong> bipolar disorder and major<br />

depressive disorder in Chinese Han population. Our findings<br />

also supported the incomplete penetrance and phenotypic<br />

diversity <strong>of</strong> duplication and deletioncopy number variations at<br />

this locilocus.<br />

O7.2 GENETICS OF DIURNAL CORTISOL<br />

SECRETION: A GENOME WIDE ASSOCIATION<br />

STUDY.<br />

F. Velders* (1), M. Kumari (2), C. Kirschbaum (3), A.<br />

H<strong>of</strong>man (4), F. Verhulst (1), C. van Duijn (4), H. Tiemeier (1)<br />

1. Department <strong>of</strong> Child and Adolescent Psychiatry, Erasmus<br />

MC – Sophia Children’s Hospital, Rotterdam, The<br />

Netherlands 2. Department <strong>of</strong> Epidemiology and Public<br />

Health, London, United Kingdom 3. Department <strong>of</strong><br />

Psychology, Technical University <strong>of</strong> Dresden, Dresden,<br />

Germany 4. Department <strong>of</strong> Epidemiology, Erasmus MC<br />

University <strong>Medical</strong> Center, Rotterdam, The Netherlands<br />

* f.velders@erasmusmc.nl<br />

Introduction: Genes involved in the Hypothalamic Pituitary<br />

Adrenal (HPA) axis regulation are potential candidate genes<br />

for psychiatric disorders. Although several genes are known to<br />

be involved in the regulation <strong>of</strong> the HPA axis, the relationship<br />

<strong>of</strong> variations in these genes with diurnal cortisol secretion is<br />

not very robust. In search <strong>of</strong> new candidate genes for<br />

psychiatric disorders, we performed a Genome Wide<br />

Association Study (GWAS) <strong>of</strong> diurnal cortisol secretion.<br />

Methods: The GWAS study was performed in an ongoing<br />

population-based cohort <strong>of</strong> 1711 elderly persons, using the<br />

Illumina Platform 550k v.3.0. Cortisol secretion was measured<br />

in 4 saliva samples obtained on one day and was expressed as<br />

the area under the curve. To replicate our results, we repeated<br />

analyses <strong>of</strong> top hits in 3000 participants <strong>of</strong> the Whitehall II<br />

Study. Findings: Top hits were found on chromosome 15<br />

(rs8026512, beta -0.57, p-value 6*10e-06), chromosome 3<br />

(rs2252459, beta 0.53, p-value 8.8*10e-06) and in the FKBP5<br />

gene region on chromosome 6 (rs9470080, beta -0.55, p-value<br />

1*10e-05), a known candidate gene for depression. FKBP5<br />

SNPs were associated with anxiety in participants <strong>of</strong> the<br />

Rotterdam Study. We did not replicate any cortisol top hits in<br />

the Whitehall II Study. Interpretation: The present<br />

population-based study shows that the glucocorticoid receptor<br />

co-chaperone FKBP5 is related to diurnal cortisol secretion<br />

and anxiety in the elderly. This extends previous FKBP5<br />

studies on treatment response in depressed patients. However,<br />

our approach to utilize cortisol data to identify new candidate<br />

genes for psychiatric disorders was less successful.


O7.3 CRY2 IS A KEY TO DEPRESSION<br />

C. Lavebratt (1), L. Sjöholm* (1), P. Soronen (2), T. Paunio<br />

(2), M. Vawter (3), W. Bunney (4), R. Adolfsson (5), . Forsell<br />

(6), J. Wu (4), J. Kelsoe (7), T. Partonen (8), M. Schalling (1)<br />

1. Karolinska Institutet at Karolinska University Hospital<br />

Solna, Department <strong>of</strong> Molecular Medicine and Surgery,<br />

Stockholm, Sweden 2. National Institute for Health and<br />

Welfare; Department <strong>of</strong> Chronic Disease Prevention; Public<br />

Health Genomics Unit, Helsinki, Finland 3. University <strong>of</strong><br />

California Irvine School <strong>of</strong> Medicine, Functional Genomics<br />

Laboratory, Irvine, California, USA 4. University <strong>of</strong><br />

California Irvine School <strong>of</strong> Medicine Department <strong>of</strong><br />

Psychiatry & Human Behavior, Irvine, California, USA<br />

5. Division <strong>of</strong> Psychiatry, Department <strong>of</strong> Clinical Sciences,<br />

University <strong>of</strong> Umeå, Umeå, Sweden 6. Department <strong>of</strong> Public<br />

Health Science, Karolinska Institutet at Karolinska University<br />

Hospital Solna, Stockholm, Sweden 7. University <strong>of</strong><br />

California San Diego School <strong>of</strong> Medicine, Department <strong>of</strong><br />

Psychiatry, La Jolla, California, USA 8. National Institute for<br />

Health and Welfare; Department <strong>of</strong> Mental Health and<br />

Substance Abuse Services; Mood, Depression and Suicidal<br />

Behaviour Unit, Helsinki, Finland<br />

* louise.sjoholm@ki.se<br />

Abnormalities in the circadian clockwork characterize major<br />

depressive and bipolar disorders. Circadian clock genes are<br />

targets <strong>of</strong> interest in these patients. CRY2 is a circadian gene<br />

that participates in regulation <strong>of</strong> the evening oscillator. This is<br />

<strong>of</strong> interest in depressive disorders where a lack <strong>of</strong> switch from<br />

evening to morning oscillators has been postulated. We<br />

observed a marked diurnal variation in human CRY2 mRNA<br />

levels from peripheral blood mononuclear cells and a<br />

significant upregulation following one-night total sleep<br />

deprivation. In depressed bipolar patients, levels <strong>of</strong> CRY2<br />

mRNA were decreased and a complete lack <strong>of</strong> increase was<br />

observed following sleep deprivation, a known antidepressant.<br />

To investigate a possible genetic contribution we undertook<br />

SNP genotyping <strong>of</strong> the CRY2 gene in two independent<br />

population-based samples from Sweden and Finland. The<br />

CRY2 gene was significantly associated with winter<br />

depression in both samples. A difference in risk haplotype<br />

identity between the two samples narrowed the suggestive<br />

region <strong>of</strong> the vulnerability locus. Taken together we propose<br />

that CRY2 harbors a vulnerability locus for depression, and<br />

that mechanisms <strong>of</strong> action involve dysregulation <strong>of</strong> CRY2<br />

expression.<br />

O7.4 GENOME-WIDE ASSOCIATION STUDY OF<br />

UNIPOLAR DEPRESSION IN THE UK POPULATION<br />

C. Lewis* (1), M. Ng (1), A. Butler (1), S. Cohen-Woods (1),<br />

K. Pirlo (1), R. Uher (1), K. Aitchison (1), S. Heath (2), M.<br />

Lathrop (2), N. Craddock (3), M. Owen (3), A. Korszun (4), L.<br />

Jones (5), I. Jones (3), M. Barnes (6), P. Muglia (6), G. Breen<br />

(1), I. Craig (1), A. Farmer (1), P. McGuffin (1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King’s College<br />

London, De Crespigny Park, London, SE5 8AF, U.K.<br />

2. Centre National de Génotypage, 91057 Evry, France<br />

3. Department <strong>of</strong> Psychological Medicine, Cardiff University,<br />

School <strong>of</strong> Medicine, Heath Park, Cardiff, CF14 4XN, U.K.<br />

4. Centre for Psychiatry, Wolfson Institute <strong>of</strong> Preventive<br />

Medicine, Barts and The London, Charterhouse Square,<br />

London, EC1M 6BQ, U.K. 5. Division <strong>of</strong> Neuroscience,<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Birmingham, The<br />

Barberry, 25 Vincent Drive, Edgbaston, Birmingham, B15<br />

2FG, U.K. 6. GlaxoSmithKline, R&D<br />

* cathryn.lewis@kcl.ac.uk<br />

Unipolar depression has substantial heritability with a<br />

postulated genetic contribution, but genome-wide association<br />

studies (GWAS) and candidate gene studies published to date<br />

have failed to convincingly identify susceptibility genes. We<br />

performed a GWAS using 1636 cases <strong>of</strong> depression<br />

ascertained in the UK, and 1594 controls, screened negative<br />

for psychiatric disorders. Cases were collected as part <strong>of</strong> an<br />

affected sibling pair linkage study for recurrent depression<br />

(DeNt; N=332), a case-control study for recurrent depression<br />

(DeCC; N= 1346) and a pharmacogenetic study (GENDEP;<br />

N=88). Cases and controls were genotyped on the<br />

Illumina Human610-Quad BeadChip. After applying stringent<br />

QC criteria for missing genotypes, departure from<br />

Hardy-Weinberg equilibrium, and low minor allele frequency,<br />

we tested for association to depression using logistic<br />

regression, correcting for population ancestry. Two SNPs<br />

(separated by 7 kb) attained p-values <strong>of</strong> < 5 x 10^-7, and two<br />

further SNPs (in different regions) had p-values <strong>of</strong> < 5 x<br />

10^-6. In the region with the most strongly associated SNPs,<br />

evidence <strong>of</strong> association strengthened to genome-wide<br />

significance when genotypes at HapMap markers were<br />

imputed using MACH (p = 6x10^-9, taking into account<br />

imputation uncertainty). At the most significant genotyped<br />

SNP, the minor allele conferred a protective effect (OR 0.72;<br />

case frequency 0.185, control frequency 0.235). This GWAS<br />

<strong>of</strong> recurrent depression cases and screened controls has<br />

identified strong evidence for a previously unknown gene and<br />

several suggestive associations for further exploration in other<br />

samples.


O7.5 GWAS ASSOCIATION STUDY OF CIRCADIAN<br />

GENES SUPPORTS ARNTL AS A<br />

MORNINGNESS-EVENINGNESS LOCUS<br />

C. Nievergelt (1), R. McKinney (1), J. Kelsoe (1), D. Kripke<br />

(1), Bipolar Genome Study (BiGS)<br />

1. Department <strong>of</strong> Psychiatyr, University <strong>of</strong> California, San<br />

Diego<br />

Objective: Prior studies have reported associations <strong>of</strong> various<br />

circadian clock genes with psychiatric and sleep disorder<br />

phenotypes. We conducted a comprehensive search <strong>of</strong><br />

genomic loci influencing circadian morningness-eveningness<br />

preference, using a large European American GWAS sample<br />

<strong>of</strong> bipolar disorder cases assembled by the Bipolar Disorder<br />

Genetics (BiGs) Consortium. Methods: Circadian<br />

morningness-eveningness preference as determined by the<br />

13-item Basic Language Morningness scale (BALM) were<br />

available for a subset <strong>of</strong> 948 participants previously genotyped<br />

on the Affymetrix 6.0 SNP array. Primary analysis focused on<br />

333 SNPs in 32 candidate circadian-rhythm-related genes.<br />

Results: A significant association was found between the<br />

BALM and rs1026070 at 11p15 in ARNTL (p < 0.016,<br />

Bonferroni corrected for 333 comparisons), which was<br />

supported by two other SNPs in this gene being nominally<br />

associated with this trait. Subjects homozygous for the rare<br />

CC genotype had a mean (±SD) morningness score <strong>of</strong> 28<br />

(11.31) compared to 29.79 (9.72) for CG and 33.11 (9.33) for<br />

the GG genotypes. A genome-wide analysis including over<br />

700K SNPs found the strongest statistical evidence for BALM<br />

association with rs1000078 in CDH13 (p = 7.09 x 10-6) at<br />

16q23 and rs8089008 in an intergenic region at 18q22 (p =<br />

1.34 x 10-5). The best circadian marker, rs1026070 in<br />

ARNTL, ranked 21st among the 700K SNP markers, though<br />

none were significant at the genome-wide level. Discussion:<br />

Several lines <strong>of</strong> evidence have repeatedly associated ARNTL<br />

with psychiatric and sleep disorder phenotypes and suggest<br />

that ARNTL polymorphisms influence behavioral circadian<br />

preference.<br />

O7.6 GENOME-WIDE ASSOCIATION STUDY OF<br />

TEMPERAMENT AS AN INTERMEDIATE<br />

PHENOTYPE FOR BIPOLAR DISORDER<br />

T. Greenwood* (1), H. Akiskal (1), Bipolar Genome Study<br />

(BiGS), J. Kelsoe (1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> San Diego and the<br />

San Diego VA Healthcare System, San Diego, CA<br />

* tgreenwood@ucsd.edu<br />

Despite the many attempts that have been made to identify<br />

genes for bipolar disorder (BD) there has been limited success,<br />

which has generally been attributed to genetic heterogeneity<br />

And small gene effects. However, it is also possible that the<br />

categorical phenotypes currently used in genetic studies <strong>of</strong> BD<br />

are not the most informative, efficient, or biologically<br />

relevant. Although quantitative phenotypes provide an<br />

alternative to categorical phenotypes based on DSM diagnosis,<br />

they have not been fully exploited in BD genetics, largely due<br />

to a lack <strong>of</strong> easily accessible biological measures. As a<br />

quantitative phenotype for BD we have explored the use <strong>of</strong><br />

temperament, a heritable personality factor that establishes a<br />

person’s baseline level <strong>of</strong> reactivity, mood, and energy. We<br />

hypothesize that the fundamental trait that is inherited is not<br />

BD but rather variations in temperament with more extreme<br />

temperamental variation conferring greater susceptibility to<br />

developing BD. To investigate this theory, we have performed<br />

a genome-wide association study using genotype data from the<br />

NIMH Bipolar Genome Study (BiGS) and 1191 bipolar<br />

subjects that completed the Akiskal Temperment Scale<br />

(TEMPS-A), which is designed to access lifelong, milder<br />

aspects <strong>of</strong> bipolar symptomatology. Five temperaments are<br />

defined by this scale: hyperthymic, dysthymic, cyclothymic,<br />

irritable, and anxious. Of these, the hyperthymic and<br />

dysthymic temperaments produced the most interesting<br />

genome-wide results with peak pvalues <strong>of</strong> 6.2X10-8 on<br />

chromosome 12 and 2.4X10-7 on chromosome 4,<br />

respectively. These results suggest that aspects <strong>of</strong><br />

temperament may have utility as intermediate phenotypes for<br />

BD.


O8 SCHIZOPHRENIA 2<br />

O8.1 MATERNAL HSV2 INFECTION INTERACTS<br />

WITH NMDA GENE POLYMORPHISMS IN THE<br />

OFFSPRING INFLUENCING THE RISK OF<br />

SCHIZOPHRENIA<br />

D. Demontis* (1), H. Buttenschön (2), M. Nyegaard (1), A.<br />

Hedemand (1), C. Pedersen (3), T. Flint (2), K. Sørensen (4),<br />

M. Nordent<strong>of</strong>t (5), B. Nørgaard-Pedersen (4), T. Werge (6), P.<br />

Andersen (4), D. Hougaard (4), P. Mortensen (3), O. Mors (2),<br />

A. Børglum (1)<br />

1. Institute <strong>of</strong> Human Genetics, Aarhus University, Aarhus,<br />

Denmark 2. Centre for Psychiatric Research, Aarhus<br />

University Hospital, Risskov, Aarhus, Denmark 3. National<br />

Centre for Register-based Research, University <strong>of</strong> Aarhus,<br />

Aarhus, Denmark 4. Department <strong>of</strong> Clinical Biochemistry,<br />

State Serum Institute, Copenhagen, Denmark 5. Psychiatric<br />

Centre Bispebjerg, Copenhagen NV, Denmark 6. Research<br />

Institute <strong>of</strong> Biological Psychiatry, Sct. Hans Hospital,<br />

Roskilde, Denmark<br />

* ditte@humgen.au.dk<br />

Schizophrenia (SZ) is a complex disorder, caused by genetic<br />

and environmental factors. The genes (GRIN1, GRIN2A,<br />

GRIN2B, GRIN2C and GRIN2D) encoding the<br />

N-methyl-D-aspartate receptor (NMDAR) have been found to<br />

associate with SZ in some studies. Glutamate receptors are<br />

expressed early in human brain development, and<br />

environmental factors such as maternal infection could impact<br />

on NMDA function in the fetus. We used 85 tag-SNPs in the<br />

five genes to test for association with SZ, in three Danish<br />

samples in total consisting <strong>of</strong> 986 individuals with SZ and<br />

1501 control persons. We investigated if maternal herpes<br />

simplex type 2 (HSV2) infection increases <strong>of</strong>fspring risk <strong>of</strong> SZ<br />

later in life depending on GRIN-gene variation, by testing for<br />

tag-SNPs interaction in the <strong>of</strong>fspring with maternal HSV2<br />

antibody level during pregnancy.<br />

Significant allelic association with SZ was found for 11 SNPs<br />

out <strong>of</strong> 32 SNPs in GRIN2B. One SNP remained significant<br />

after Bonferroni correction (rs1806194, p=0.00094). When<br />

testing for interaction between genetic variation and maternal<br />

HSV2 infection eight GRIN2B and two GRIN2A SNPs<br />

showed significant interaction. Three SNPs in GRIN2B<br />

remained significant after Bonferroni correction<br />

(rs1805539, rs1806201, rs1806205, p=0.00142-0.00024).<br />

Our results suggest that GRIN2B may be a risk gene for SZ<br />

and that maternal HSV2 infection interacts with GRIN2B in<br />

the <strong>of</strong>fspring, affecting the risk <strong>of</strong> SZ. The NR2B subunit,<br />

encoded by the GRIN2B gene is mainly present in immature<br />

neurons in the early postnatal brain. It is possible that early<br />

life distortion <strong>of</strong> NMDAR function caused by dysregulation <strong>of</strong><br />

GRIN2B due to maternal HSV2 infection could be important<br />

for the preposition to SZ.<br />

O8.2 EXON-BASED RE-SEQUENCING ANALYSIS OF<br />

THE SCHIZOPHRENIA SUSCEPTIBILITY GENE<br />

NRXN1<br />

T. Mühleisen* (1), A. Forstner (1), F. Basmanav (1), V.<br />

Nieratschker (2), R. Breuer (2), S. Moebus (3), M. Rietschel<br />

(2), M. Nöthen (1), S. Cichon (1)<br />

1. Institute <strong>of</strong> Human Genetics, Department <strong>of</strong> Genomics, Life<br />

& Brain Center, University <strong>of</strong> Bonn, Bonn, Germany<br />

2. Department <strong>of</strong> Genetic Epidemiology, Central Institute <strong>of</strong><br />

Mental Health, Mannheim, Germany 3. Institute for <strong>Medical</strong><br />

Informatics, Biometry and Epidemiology, University Hospital<br />

<strong>of</strong> Essen, University Duisburg-Essen, Essen, Germany<br />

* thomas.muehleisen@uni-bonn.de<br />

Exon-disrupting copy number variants (CNVs) in the neurexin<br />

1(NRXN1; 2p16.3) gene were recently found to be<br />

significantly over-represented in schizophrenia patients<br />

(Rujescu, Ingason et al. 2009), supporting previous findings <strong>of</strong><br />

two microdeletions in schizophrenia patients involving this<br />

gene (Kirov et al. 2008; Walsh et al. 2008). Deletions and<br />

duplications <strong>of</strong> variable sizes accumulated in the promoter<br />

region and the first exons. The gene product NRXN1 is a<br />

presynaptic scaffolding molecule which plays an important<br />

role in neurotransmission by interacting with neurexophilins<br />

and neuroligins. In the present study, we performed<br />

re-sequencing at the NRXN1locus to identify rare genetic<br />

variations - single base exchanges (SBEs) or small<br />

deletions/duplications - which might confer susceptibility to<br />

schizophrenia and are likely to be missed using common<br />

single nucleotide polymorphisms in genome-wide association<br />

studies. We investigated 96 patients (51% males, parents<br />

available) with a DSM-IV diagnosis <strong>of</strong> schizophrenia and 96<br />

sex-matched controls, all <strong>of</strong> German descent. Using the<br />

Sanger method, we re-sequenced all coding sequences and<br />

splice sites <strong>of</strong> the NRXN1is<strong>of</strong>orms alpha1, alpha2 and beta,<br />

totaling ~10kb per individual. We have finished re-sequencing<br />

in ~50% <strong>of</strong> the sample and observe an over-representation <strong>of</strong><br />

rare SBEs in patients (n=4) versus controls (n=1). We are<br />

currently analyzing the second half <strong>of</strong> the sample. All<br />

identified rare variants are being investigated by<br />

bioinformatical tools and databases. They will be tested for<br />

inheritance from their parents, too, and followed-up in<br />

extended samples <strong>of</strong> schizophrenia patients and controls.


O8.3 GENOME-WIDE ASSOCIATION STUDY OF<br />

REGIONAL BRAIN VOLUME IN 600 INDIVIDUALS<br />

SUGGESTS INVOLVEMENT OF KNOWN<br />

PSYCHIATRY CANDIDATE GENES, IDENTIFIES<br />

NEW CANDIDATES FOR PSYCHIATRIC DISORDERS<br />

AND POINTS TO POTENTIAL MODES OF THEIR<br />

ACTION<br />

B. Franke* (1), M. Rijpkema (2), A. Arias Vasquez (1), J.<br />

Veltman (3), H. Brunner (3), P. Hagoort (4), G. Fernandez (4)<br />

1. Department <strong>of</strong> Human Genetics and Department <strong>of</strong><br />

Psychiatry, Donders Institute for Brain, Cognition and<br />

Behavior, Radboud University Nijmegen <strong>Medical</strong> Centre,<br />

Nijmegen, The Netherlands 2. Donders Centre for Cognitive<br />

Neuroimaging, Donders Institute for Brain, Cognition and<br />

Behavior, Radboud University Nijmegen, Nijmegen, The<br />

Netherlands 3. Department <strong>of</strong> Human Genetics, Radboud<br />

University Nijmegen <strong>Medical</strong> Centre, Nijmegen, The<br />

Netherlands 4. Donders Centre for Cognitive Neuroimaging,<br />

Donders Institute for Brain, Cognition and Behavior, Radboud<br />

University Nijmegen, Nijmegen, The Netherlands<br />

*b.franke@antrg.umcn.nl<br />

Though most psychiatric disorders are highly heritable, it has<br />

been hard to identify genetic risk factors involved, which are<br />

most likely <strong>of</strong> small individual effect size. A possible way to<br />

aid identification <strong>of</strong> risk genes is the use <strong>of</strong> intermediate<br />

phenotypes. These are supposed to be closer to the biological<br />

substrate(s) <strong>of</strong> the disorder than psychiatric diagnoses, and<br />

therefore less genetically complex. Intermediate phenotypes<br />

can be defined e.g. at the level <strong>of</strong> brain function and <strong>of</strong><br />

regional brain structure. Both are highly heritable, and<br />

regional brain structure is linked to brain function. Within the<br />

Brain Imaging Genetics (BIG) study at the Radboud<br />

University Nijmegen (<strong>Medical</strong> Centre) we performed a<br />

genome-wide association study (GWAS) in 600 <strong>of</strong> the<br />

currently 1100 healthy study participants. For all BIG<br />

participants, structural MRI brain images were available. Gray<br />

and white matter volumes were determined by brain<br />

segmentation using SPM s<strong>of</strong>tware. FSL-FIRST was used to<br />

assess volumes <strong>of</strong> specific brain structures. Genotyping was<br />

performed on Affymetrix 6.0 arrays. Results implicate known<br />

candidates from earlier GWAS and candidate gene studies in<br />

mental disorders in the regulation <strong>of</strong> regional brain structure.<br />

E.g. polymorphisms in CDH13, featuring among the<br />

top-findings <strong>of</strong> GWAS in disorders including ADHD,<br />

addiction and schizophrenia, were found associated with<br />

amygdala volume. The ADHD candidate gene SNAP25 was<br />

found associated with caudate nucleus volume.<br />

In conclusion, the use <strong>of</strong> intermediate phenotypes based on<br />

(subcortical) brain volumes may shed more light on pathways<br />

from gene to disease, but can also be expected to facilitate<br />

gene identification in psychiatric disorders.<br />

O8.4 ASSOCIATION OF A FUNCTIONAL<br />

HAPLOTYPE OF THE POST-SYNAPTIC DENSITY 95<br />

(PSD-95) GENE WITH SCHIZOPHRENIA<br />

M. Cheng (1), D. Liao (2), J. Chen (3), Y. Wang (3), I. Lai (3),<br />

Y. Liou (3), C. Chen (2)<br />

1. Institute <strong>of</strong> <strong>Medical</strong> Sciences, Tzu-Chi University, Hualien,<br />

Taiwan 2. Division <strong>of</strong> Mental Health and Addiction<br />

Medicine, Institute <strong>of</strong> Population Health Sciences,<br />

National Health Research Institutes, Zhunan, Taiwan<br />

3. Department <strong>of</strong> Psychiatry, Yuli Veterans Hospital, Hualien,<br />

Taiwan<br />

There is compelling evidence supporting the association <strong>of</strong><br />

hyp<strong>of</strong>unction <strong>of</strong> the N-methyl-D-aspartate (NMDA)<br />

receptor with the pathophysiology <strong>of</strong> schizophrenia.<br />

Post-synaptic density protein 95 (PSD-95) binds to NMDA<br />

receptors and plays an essential role in regulating the activity<br />

<strong>of</strong> the NMDA receptor. Altered expression <strong>of</strong> PSD-95 in<br />

various brain regions in patients with schizophrenia has been<br />

reported in several postmortem studies. This study aimed to<br />

investigate whether there are variants <strong>of</strong> the PSD-95 gene that<br />

may confer an increased risk <strong>of</strong> schizophrenia. We<br />

re-sequenced the promoter region, all the exons (including 5’<br />

and 3’ UTR) and their flanking intronic sequences <strong>of</strong> the<br />

PSD-95 gene in a sample <strong>of</strong> Han Taiwanese schizophrenic<br />

patients (n=434) and control subjects (n=438), and conducted<br />

an association study. We also characterized the function <strong>of</strong><br />

several genetic polymorphisms <strong>of</strong> PSD-95 using a reporter<br />

gene assay. We did not detect any mutations at the<br />

protein-coding regions associated with schizophrenia <strong>of</strong> the<br />

PSD-95 gene in this sample. Nevertheless, we identified a<br />

specific haplotype at the promoter region <strong>of</strong> the PSD-95 gene<br />

that was nominally associated with schizophrenia<br />

(odd ratio: 1.24, 95% confidence interval: 1.01-1.52). The<br />

reporter gene assay showed that this haplotype had<br />

significantly greater activity than the other haplotypes. Our<br />

study provides genetic evidence to indicate that the PSD-95<br />

gene is associated with schizophrenia, and the increased gene<br />

expression <strong>of</strong> PSD-95 may be involved in the pathophysiology<br />

<strong>of</strong> schizophrenia.


O8.5 GENOME-WIDE ASSOCIATION STUDY (GWAS)<br />

OF SCHIZOPHRENIA IN A LARGE GENETICALLY<br />

HOMOGENEOUS IRISH POPULATION<br />

The Schizophrenia Genomics Ireland Consortium*, The<br />

Wellcome Trust Case Control Consortium<br />

* acorvin@tcd.ie<br />

Schizophrenia (SZ; OMIM 181500) is a complex genetic<br />

disorder <strong>of</strong> substantial heritability. Three recent genome-wide<br />

association studies (GWAS) provide support for several<br />

common susceptibility loci, including the HLA-region (The<br />

International Schizophrenia Consortium (ISC), 2009; Shi et<br />

al, 2009; Stefansson et al, 2009). Additionally, the ISC study<br />

reported evidence that ~30% <strong>of</strong> susceptibility can be explained<br />

by common risk variants, many <strong>of</strong> which are likely to be <strong>of</strong><br />

small effect. Combining datasets may increase power to<br />

discover small effects, but may be compromised by individual<br />

differences in study design, ascertainment, phenotyping,<br />

population stratification or genotyping platforms.<br />

We report a large SZ GWAS performed as part <strong>of</strong> WTCCC2<br />

<strong>of</strong> 2,357 cases and 2,000 controls from the ethnically<br />

homogeneous Irish population. The study also accesses 6,000<br />

additional controls from the UK. All cases were collected<br />

using similar study design and ascertainment procedures<br />

including a structured clinical interview (SCID) and consensus<br />

diagnosis procedure. This dataset has ≥80% power to detect<br />

allelic odds ratios (ORs) 1.2-1.3 with minor allele frequency<br />

(MAF) 0.2-0.3. Combining data with existing GWAS studies<br />

may also allow sequential discovery <strong>of</strong> additional risk<br />

variants. GWAS data was analysed using the Affymetrix 6.0<br />

array. For all samples passing Affymetrix’s laboratory QC,<br />

raw intensities (from the .CEL files) were renormalized within<br />

collections using CelQuantileNorm (see<br />

http://outmodedbonsai.sourceforge.net/). These normalized<br />

intensities were used to call genotypes with an updated version<br />

<strong>of</strong> the Chiamo s<strong>of</strong>tware (see<br />

www.stats.ox.ac.uk/~marchini/s<strong>of</strong>tware/gwas/chiamo.html),<br />

adapted for Affymetrix 6.0 SNP data. Primary analysis <strong>of</strong> the<br />

GWAS data will be presented with p-values from 1-df<br />

Cochran-Armitage tests for trend. REFERENCES 1. The<br />

International Schizophrenia Consortium. Common polygenic<br />

variation contributes to risk <strong>of</strong> schizophrenia and bipolar<br />

disorder. Nature. 2009 Jul 1. PMID: 19571811. 2: Shi J, et al.<br />

Common variants on chromosome 6p22.1 are associated with<br />

schizophrenia. Nature. 2009 Jul 1. PMID: 19571809. 3.<br />

Stefansson H, et al. Common variants conferring risk <strong>of</strong><br />

schizophrenia. Nature. 2009 Jul 1. PMID:19571808.<br />

O8.6 ZNF804A: SCHIZOPHRENIA RISK ALLELE<br />

ASSOCIATED WITH NEUROANATOMIC,<br />

COGNITIVE, AND CLINICAL PHENOTYPES<br />

T. Lencz* (1), P. Szeszko (1), P. DeRosse (1), K. Burdick (1),<br />

A. Malhotra (1)<br />

1. The Zucker Hillside Hospital<br />

* lencz@lij.edu<br />

A recent genomewide association study identified a novel<br />

SNP in ZNF804A (rs1344706) as a risk factor for<br />

schizophrenia (O’Donovan et al. 2008; Nat Gen, 40:1053-5).<br />

In a functional MRI study, this SNP was associated with<br />

abnormal patterns <strong>of</strong> neural connectivity (Esslinger et al.<br />

2009; Science, 324:605). To further functionally characterize<br />

this SNP, we tested rs1344706 for association with phenotypes<br />

that include brain structure, neurocognitive performance, and<br />

clinical symptomatology. We first assessed the relationship <strong>of</strong><br />

rs1344706 to risk for illness in our sample <strong>of</strong> Caucasian<br />

patients with schizophrenia/schizoaffective disorder (n=277)<br />

and Caucasian controls (n=247). Allele frequencies matched<br />

those in the original sample <strong>of</strong> O’Donovan et al. (2008) and<br />

confirmed an association with illness; T allele frequency was<br />

66% in cases vs 59% in controls (?2=4.3, p


O9 EARLY CAREER INVESTIGATOR<br />

TRACK: CANDIDATE GENES,<br />

EPIGENETICS, AND PHENOMICS<br />

O9.1 THE VAL 158 MET POLYMORPHISM IN THE<br />

COMT GENE IS ASSOCIATED WITH DEPRESSION<br />

AND MOTIVATION<br />

E. Åberg* (1), L. Sjöholm (1), Y. Forsell (2), C. Lavebratt (1)<br />

1. Department <strong>of</strong> Molecular Medicine and Surgery, CMM,<br />

Karolinska Institutet at Karolinska University Hospital Solna,<br />

Stockholm, Sweden 2. Department <strong>of</strong> Public Health Science,<br />

Karolinska Institutet at Karolinska University Hospital<br />

Solna, Stockholm, Sweden<br />

* elin.aberg@ki.se<br />

Environmental risk factors together with genetic vulnerability<br />

create a complex background to develop depression. In this<br />

study we investigate the association between the Val158Met<br />

polymorphism in the COMT gene and depression by using a<br />

Swedish population-based longitudinal study (PART). We<br />

also investigate if the polymorphism could be involved in the<br />

low motivational level found in depressed individuals. The<br />

PART study includes data from childhood to adulthood and<br />

contains scales on psychological well being. We found that<br />

depressed people displayed a lower frequency <strong>of</strong> the val/val<br />

genotype than controls (OR=1.50, 95% CI=1.11-2.02,<br />

P=0.0096). In analysis <strong>of</strong> men and women separately, the<br />

association was found among men only (OR=2.26, 95% CI=<br />

1.26-4.05, p=0.0086). Regression analysis including potential<br />

risk factors for depression further indicated that the genotypes<br />

met/met or met/val was associated with depression (P=0.01).<br />

Since COMT is involved in dopamine metabolism and is<br />

expressed in brain areas important for the brain reward<br />

system, we thought that variations in this gene could be<br />

involved in motivational disturbances. Individuals with the<br />

met/met or met/val genotype had more negative motivation<br />

symptoms (MDI scale) than individuals homozygous for the<br />

val-allel (P=0.01). Further, regression analyses including risk<br />

factors for depression showed that individuals with the<br />

genotype met/met or met/val had more negative motivation<br />

symptoms than individuals with the val/val combination<br />

(P=0.007). In conclusion, the COMT-polymorphism<br />

Val158Met has so far primarily been associated with<br />

schizophrenia, working memory and anxiety related<br />

behaviour. We can in this study present that variations in the<br />

COMT-gene is associated with depression and motivation.<br />

O9.2 RE-SEQUENCING ANALYSIS OF THE<br />

SCHIZOPHRENIA-ASSOCIATED MICRODELETION<br />

REGION ON 1Q21.1<br />

F. Basmanav* (1), A. Forstner (1), J. Freudenberg (2), L.<br />

Priebe (1), M. Alexander (1), V. Nieratschker (3), R. Breuer<br />

(3), S. Moebus (4), M. Rietschel (3), M. Nöthen (1), T.<br />

Mühleisen (1), S. Cichon (1)<br />

1. Institute <strong>of</strong> Human Genetics, Department <strong>of</strong> Genomics, Life<br />

& Brain Center, University <strong>of</strong> Bonn, Bonn, Germany 2. Center<br />

<strong>of</strong> Genomics and Human Genetics, Feinstein Research<br />

Institute, New York, USA 3. Department <strong>of</strong> Genetic<br />

Epidemiology, Central Institute <strong>of</strong> Mental Health, Mannheim,<br />

Germany 4. Institute for <strong>Medical</strong> Informatics, Biometry and<br />

Epidemiology, University Hospital <strong>of</strong> Essen, University<br />

Duisburg-Essen, Essen, Germany<br />

* basmanav@uni-bonn.de<br />

We and others have recently identified rare microdeletions on<br />

chromosome 1q21.1 as the strongest genetic risk factor for<br />

schizophrenia known to date (odds ratio ~15). In the present<br />

study, we aimed to explore to what extent common and rare<br />

susceptibility variants in the region spanned by the<br />

microdeletion might contribute to the disease allele spectrum.<br />

Analysis <strong>of</strong> common SNPs was done using Illumina HH550<br />

data and did not provide evidence for association with<br />

schizophrenia. To identify potential rare coding variants, we<br />

applied an exon-based re-sequencing approach covering seven<br />

RefSeq genes and three predicted genes. Approximately 25 kb<br />

<strong>of</strong> sequence information per individual were generated from<br />

96 DSMIV-diagnosed schizophrenia patients (derived from<br />

parent-<strong>of</strong>fspring trios) and 96 sex-matched controls.<br />

Re-sequencing has been completed for 50% <strong>of</strong> the sample and<br />

provides some evidence for an over-representation <strong>of</strong><br />

previously unknown exonic single base exchanges in patients<br />

(n=5 in 48 patients and n=1 in 48 controls). Two <strong>of</strong> the rare<br />

variants detected in patients represent non-synonymous amino<br />

acid changes with a predicted effect on protein structure, one<br />

is a stop mutation. We hypothesize that such rare exonic single<br />

base changes may contribute to the schizophrenia disease<br />

allele spectrum at this particular locus. We are currently<br />

analysing the second half <strong>of</strong> the sample. All rare variants<br />

identified in the patients will be tested for parental inheritance<br />

and will be followed-up in large, independent samples <strong>of</strong><br />

schizophrenia patients and controls. F.B.Basmanav and<br />

A.J.Forstner contributed equally to this work.


O9.3 FAMILIAL FORMS OF PSYCHIATRIC<br />

DISORDERS IN A LARGE, NATIONALLY<br />

REPRESENTATIVE SAMPLE<br />

N. Low* (1)<br />

1. McGill University<br />

* nancy.low@mcgill.ca<br />

Introduction: Evidence from clinical populations <strong>of</strong> mood and<br />

anxiety disorders has demonstrated familial aggregation.<br />

Clinical characteristics <strong>of</strong> the familial forms <strong>of</strong> disorders have<br />

also been described (egs, earlier age <strong>of</strong> onset, worse<br />

impairment, treatment refractoriness). These findings require<br />

confirmation in population-based samples where the majority<br />

<strong>of</strong> the burden <strong>of</strong> illness rests. Examination <strong>of</strong> the specificity <strong>of</strong><br />

the familial clinical characteristics across the disorders is also<br />

understudied. Sample: The Canadian Community Health<br />

Survey was cross-sectional survey <strong>of</strong> 36,984 subjects<br />

randomly selected from the population <strong>of</strong> household residents.<br />

The response proportion nationally was 77%. Subjects were<br />

interviewed in-person using Composite International<br />

Diagnostic Interview which included family history. Statistical<br />

Analyses: Multivariable logistic regression models were<br />

constructed to identify clinical characteristic variables<br />

associated with a family history <strong>of</strong> depression, mania, panic<br />

and social phobia. Results: Earlier age <strong>of</strong> onset and age <strong>of</strong> 1st<br />

consultation were common to all the familial forms. Earlier<br />

age <strong>of</strong> 1st treatment was associated with all familial forms<br />

except social phobia. Comorbid disorders: social phobia and<br />

panic were associated with mood disorders; PTSD was<br />

associated with the anxiety disorders, but not mood disorders;<br />

psychosis was associated with depression only. Current<br />

medication use: anxiolytics and antidepressants was associated<br />

with all forms except mania; antipsychotics were associated<br />

the mood disorders; mood stabilizers were associated with<br />

depression, but not mania. Additionally, familial depression<br />

was associated with stressful precipitants, suicide attempts,<br />

impairment, and number <strong>of</strong> consultants seen. Conclusion: The<br />

familial forms <strong>of</strong> depression, mania, panic and social phobia<br />

have common and unique clinical characteristics.<br />

O9.4 RESPONSE TO LITHIUM IN BIPOLAR<br />

DISORDER: INTER-RATER RELIABILITY AND<br />

DISTRIBUTIONAL PROPERTIES<br />

M. Manchia* (1), C. Consortium on Lithium Genetics (2), I.<br />

International Group for The Study <strong>of</strong> Lithium Treated Patients<br />

(3), M. Alda (1)<br />

1. Department <strong>of</strong> Psychiatry, Dalhousie University, Halifax,<br />

Nova Scotia, Canada 2. www.ConLiGen.org 3.<br />

http://www.igsli.org<br />

* mirko.manchia@cdha.nshealth.ca<br />

The Consortium on Lithium Genetics (ConLiGen) has been<br />

created with the aim to investigate the genetic basis <strong>of</strong><br />

response to lithium prophylaxis in more than 1,200 bipolar<br />

patients followed in research centers across the world. In this<br />

context, it is important to assess the key phenotypic measures<br />

and the response to long-term lithium treatment reliability<br />

across the participating centres. To this end, we measured the<br />

inter-rater reliability <strong>of</strong> the response definition based on a 0-10<br />

scale published and validated previously (Gr<strong>of</strong> et al. 2002,<br />

Garnham et al. 2007). 23 investigators from 8 participating<br />

sites rated 12 standardized case vignettes. We analyzed the<br />

concordance <strong>of</strong> ratings by means <strong>of</strong> Kappa (?) and intra-class<br />

correlation coefficient (ICC). In addition, we performed<br />

mixture analysis <strong>of</strong> the empirical distribution <strong>of</strong> the total scale<br />

score from a sample <strong>of</strong> 623 patients studied previously.<br />

Kappa scores and ICC showed a good level <strong>of</strong> agreement<br />

across sites (?=0.67, z=36.87; ICC=0.73). Leaving out the<br />

clear cut non-responders (score=0), modelling <strong>of</strong> score<br />

distribution suggested the presence <strong>of</strong> a best fitting model <strong>of</strong><br />

two normal curves (1 component: ?2= 32.1, p0.05) with the following parameters:<br />

partial responders (mean=3.1, SD=1.5, proportion=0.36) and<br />

full responders (mean=7.7, SD=1.6, proportion=0.64) and<br />

suggested cut-<strong>of</strong>f for full response ≥6. These findings indicate<br />

a good inter-rater reliability <strong>of</strong> assessments <strong>of</strong> the response to<br />

lithium and provide analytical support for the definition <strong>of</strong> the<br />

full response phenotype. These results will be used in a<br />

subsequent genome wide association study <strong>of</strong> the ConLiGen<br />

samples.


O9.5 PROFILING EPIGENETIC CHANGES IN THE<br />

BRAIN ASSOCIATED WITH SCHIZOPHRENIA,<br />

BIPOLAR DISORDER, AND MAJOR DEPRESSIVE<br />

DISORDER<br />

E. Dempster (1), J. Mill (1), C. Wong (1), S. Docherty (1), R.<br />

Pidsley* (1)<br />

1. MRC Social, Genetic and Developmental Psychiatry<br />

Centre, Institute <strong>of</strong> Psychiatry, King’s College, London, UK<br />

* ruth.pidsley@kcl.ac.uk<br />

Traditional aetiological studies <strong>of</strong> psychiatric conditions have<br />

focused primarily on the interplay between genetic and<br />

environmental risk factors. However, a growing body <strong>of</strong><br />

evidence suggests that epigenetic factors also play an<br />

important role in mediating susceptibility to psychiatric<br />

conditions. Epigenetics refers to heritable changes in genomic<br />

function, which are not caused by changes in the nucleotide<br />

sequence <strong>of</strong> the DNA. DNA methylation is one <strong>of</strong> the most<br />

stable and well-characterised forms <strong>of</strong> epigenetic control.<br />

Whilst studies <strong>of</strong> epigenetic dysfunction in neuropsychiatric<br />

conditions are in their infancy, research to date indicates that<br />

there may be widespread DNA-methylation changes in the<br />

brains <strong>of</strong> affected patients. In this study we used a unique<br />

collection <strong>of</strong> post-mortem brain samples to identify<br />

DNA-methylation changes associated with schizophrenia,<br />

bipolar disorder and major depressive disorder. We used a<br />

candidate gene-approach to focus specifically on the<br />

DNA-methylation status across a set <strong>of</strong> genes that have been<br />

previously implicated in psychiatric conditions via<br />

transcriptomic and gene association studies. We used a<br />

highly-sensitive DNA methylation pr<strong>of</strong>iling strategy to assess<br />

epigenetic variation across the known promoter regulatory<br />

regions <strong>of</strong> these genes to detect epigenetic changes associated<br />

with disease. Our studies uncovered evidence <strong>of</strong> DNA<br />

methylation changes in several genes, either globally or at<br />

specific CpG sites, with a number <strong>of</strong> psychiatric patients being<br />

clear epigenetic outliers.<br />

O9.6 DIRAS2 IS ASSOCIATED WITH ADULT ADHD<br />

AND RELATED PERSONALITY TRAITS<br />

A. Reif* (1), L. Weissflog (1), T. Nguyen (2), C. Jacob (1), M.<br />

Romanos (1), S. Liedel (1), H. Schäfer (2), A. Warnke (1), B.<br />

Cormand (3), J. Haavik (4), B. Franke (5), K. Lesch (1)<br />

1. University <strong>of</strong> Würzburg 2. University <strong>of</strong> Marburg 3.<br />

University <strong>of</strong> Barcelona 4. University <strong>of</strong> Bergen 5. University<br />

<strong>of</strong> Nijmegen<br />

* Reif_A@klinik.uni-wuerzburg.de<br />

Attention-deficit/hyperactivity disorder (ADHD) is a clinically<br />

heterogeneous childhood behavioral neurodevelopmental<br />

disorder which is highly persistent into adulthood and <strong>of</strong>ten<br />

goes along with substantial comorbidity. Although it is<br />

assumed that adult ADHD (aADHD) has an even higher<br />

genetic background than childhood ADHD, studies on the<br />

genetics <strong>of</strong> aADHD are scarce. Linkage analyses and<br />

genome-wide association studies (GWAS) identified several<br />

susceptibility loci for this genetically complex disorder. The<br />

9q22 region has been detected by both linkage and GWAS as<br />

a candidate locus for ADHD (Lesch et al., 2008; Zhou et al.,<br />

2008). This region contains the GTP-binding RAS-like 2 gene<br />

(DIRAS2; MIM: 607863) which is expressed in the basal<br />

ganglia and the cerebellum. The function <strong>of</strong> the gene product<br />

is still unknown but might include the regulation <strong>of</strong> cell<br />

morphogenesis. As DIRAS2 thus can be considered a<br />

positional as well as functional candidate gene for ADHD, we<br />

conducted a case-control association study in 624 patients<br />

suffering from adult ADHD diagnosed according to<br />

DSM-IV criteria and 424 controls recruited in the Lower<br />

Franconia area in Germany, all <strong>of</strong> Caucasian origin. Six out <strong>of</strong><br />

the 14 tested SNPs covering all haplotype blocks <strong>of</strong> DIRAS2<br />

were associated with ADHD, with nominal p-values ranging<br />

from p= 0.009 to p= 0.01. Two <strong>of</strong> these SNPs were also<br />

associated with NEO Extraversion and TPQ Harm Avoidance<br />

in affected subjects. Within each <strong>of</strong> two haploblocks, there<br />

was one associated haplotype. A replication attempt is<br />

currently made in the IMpACT sample, consisting <strong>of</strong> an<br />

additional 1,000 patients from The Netherlands, Norway and<br />

Spain. Our findings in adult ADHD are confirmed by a<br />

family-based study on childhood ADHD (166 families, 576<br />

children), where SNPs in the gene were also associated with<br />

disease. These findings indicate consistently that DIRAS2<br />

may play a role in the pathomechanisms <strong>of</strong> ADHD.<br />

Re-sequencing <strong>of</strong> the gene as well as functional studies will be<br />

required to further explore the role <strong>of</strong> DIRAS2 in ADHD.


O9.7 A LONGITUDINAL STUDY OF EPIGENETIC<br />

VARIATION IN TWINS<br />

C. Wong* (1), A. Caspi (2), B. Williams (2), A. Ambler (1),<br />

T. M<strong>of</strong>fitt (2), J. Mill (1)<br />

1. 1-MRC Social, Genetic and Developmental Psychiatry<br />

Centre, Institute <strong>of</strong> Psychiatry, King’s College, London, UK<br />

2. 1-MRC Social, Genetic and Developmental Psychiatry<br />

Centre, Institute <strong>of</strong> Psychiatry, King’s College, London, UK;<br />

2- Departments <strong>of</strong> Psychology and Neuroscience, Psychiatry<br />

and Behavioral Sciences, and Institute for Genome Sciences<br />

and Policy Duke University<br />

* chloe.wong@kcl.ac.uk<br />

Twin studies have been widely used to provide an estimate <strong>of</strong><br />

the relative contribution <strong>of</strong> genetic and environmental factors<br />

to phenotypic traits. In many psychiatric conditions, it is<br />

commonly observed that monozygotic (MZ) twins are more<br />

concordant than dizygotic (DZ) twins, strongly implicating<br />

inherited genetic factors in pathogenesis. Given that MZ twins<br />

are assumed to be genetically identical, any discordance<br />

between the twins is attributed to ‘non-shared’ environmental<br />

factors in the classical twin-study approach. Emerging<br />

evidence suggests that epigenetic factors, such as DNA<br />

methylation, may also contribute to phenotypic<br />

discordance between genetically identical individuals.<br />

Few studies have investigated the extent <strong>of</strong> epigenetic<br />

variation between genetically-identical individuals, and no<br />

study has yet investigated longitudinal changes in DNA<br />

methylation <strong>of</strong> MZ and DZ twins. In this study we assessed<br />

the level <strong>of</strong> within-pair epigenetic discordance in a sample <strong>of</strong><br />

MZ and DZ twins pairs (MZ=46; DZ=45) obtained from the<br />

Environmental-Risk Study, in which samples from the same<br />

individuals were collected at two different time-points (age 5<br />

and 10). The levels <strong>of</strong> DNA methylation across seven<br />

candidate genes related to psychiatric disorders were<br />

quantified using high-resolution bisulfite-based mapping.<br />

Significant gene-specific DNA methylation differences were<br />

observed within both MZ and DZ twin-pairs at both age 5 and<br />

10. Moreover DNA methylation was shown to change<br />

dynamically over-time. Findings from this study will allow us<br />

to elucidate the contribution <strong>of</strong> genetic and environmental<br />

factors to longitudinal changes in DNA methylation and will<br />

have implications for understanding the causes <strong>of</strong> phenotypic<br />

lability.<br />

O9.8 INTERACTION BETWEEN CRHR1 GENE AND<br />

CHILDHOOD TRAUMA PREDICTS ADOLESCENT<br />

DEPRESSIVE AND ANXIETY SYMPTOMS<br />

K. Thode* (1), R. Olvera (1), C. Walss-Bass (1), D.<br />

Williamson (1)<br />

1. UTHSCSA<br />

* thode@uthscsa.edu<br />

The role <strong>of</strong> the hypothalamic-pituitary-adrenal (HPA) axis in<br />

stress-related psychiatric disorders has been well-established.<br />

Recent research identified an interaction between a SNP<br />

(rs1876831) in the CRH receptor 1 (CRHR1) gene and<br />

stressful life events for heavy alcohol use in adolescents. We<br />

identified a CRHR1 promoter SNP (rs12938031) that<br />

predicted increased HPA axis response to CRH stimulation.<br />

The aims <strong>of</strong> our current study were to examine rs12938031's<br />

relationship to rs1876831 as well as whether they moderate<br />

the effects <strong>of</strong> exposure to stress for symptoms <strong>of</strong> depression<br />

and anxiety in adolescents. Data were available from the Teen<br />

Alcohol Outcomes Study (TAOS), an ongoing cohort study<br />

examining alcohol use in adolescents. A total <strong>of</strong> 340<br />

adolescents aged 12-15 years (184 males, 156 females), were<br />

genotyped, and a subset <strong>of</strong> 321 completed the Childhood<br />

Trauma Questionnaire (CTQ) assessing lifetime stressful<br />

events and self-report questionnaires measuring depressive<br />

and anxiety symptoms. rs12938031 and rs1876831 were<br />

found to be in high linkage disequilibrium (D’0.951, LOD<br />

35.6, r2 0.388). Adolescents carrying the major A allele <strong>of</strong><br />

rs12938031 reported greater depressive symptoms in relation<br />

to higher CTQ scores than adolescents homozygous for the<br />

minor G allele (p=0.002). Females with high CTQ scores<br />

reported more depressive symptoms compared to males<br />

regardless <strong>of</strong> genotype (p=0.012). Adolescents carrying the<br />

minor T allele <strong>of</strong> rs1876831 reported greater anxiety<br />

symptoms in relation to higher CTQ scores than adolescents<br />

homozygous for the major C allele (p=0.015). These results<br />

provide further evidence for genetic moderation <strong>of</strong> the stress<br />

response directly involving SNPs in the CRHR1 gene.


O10 MOOD DISORDERS 3<br />

O10.1 ASSOCIATION BETWEEN DISTRACTIBILITY<br />

AND P2RX7 IN SWEDISH BIPOLAR DISORDER<br />

PATIENTS<br />

L. Backlund (1), P. Nikamo (2), L. Frisén (3), I. Römer Ek<br />

(1), L. Träskman-Bendz (4), H. Ågren (5), G. Edman (6), M.<br />

Landén (1), M. Schalling (2), U. Ösby (1)<br />

1. Dpt Clin Neurosc, Karolinska Institutet, Stockholm,<br />

Sweden 2. Div Neurosc, Dpt Mol Med and Surg, Karolinska<br />

Institutet, Stockholm, Sweden 3. Div <strong>of</strong> Psych, Dpt Clin Sc,<br />

Karolinska Institutet, Danderyd Univ Hosp, Stockholm,<br />

Sweden 4. Div Psych, Dpt Clin Sc, Univ Hosp, Lund, Sweden<br />

5. Sahlgrenska Ac,Univ Gothenburg, Inst Neurosc and<br />

Physiol, Sweden 6. Dpt Psych, R&D Sect, Danderyd Univ<br />

Hosp, Sweden<br />

Introduction: Bipolar disorder is a severe psychiatric disorder<br />

(heritability around 80%). At least a dozen genes have been<br />

shown to be involved in the etiology, although replication has<br />

been difficult. We investigated 36 SNPs and their association<br />

with different symptoms in bipolar disorder. Methods: A<br />

Swedish sample <strong>of</strong> 646 patients with bipolar disorder (BP1:<br />

79%, BP2: 21%), was phenotyped with lifetime assessment <strong>of</strong><br />

specific symptoms <strong>of</strong> mania and depression and analyzed with<br />

36 SNPs in the genes S100A10, BDNF, P2RX7, CANKK2,<br />

DAOA, SLC6A4, ADRBK2 and COMT. At first, 171 patients<br />

were analyzed. A second set <strong>of</strong> 475 patients was used for<br />

replication. Genotyping was carried out with Applied<br />

Biosystems TaqMan using ABI PRISM 7900HT Sequence<br />

Detecting System. Statistical analysis (Chi-square tests) was<br />

performed with the Unphased program 3.0.10. Results: The<br />

manic symptom distractibility (set 1: 44%, set 2: 59%, total:<br />

52 %) was significantly associated with two SNPs in the<br />

P2RX7 gene coding for a purinergic ATP-binding channel<br />

implicated in calcium-signaling and neurotransmitter release,<br />

rs1718119 (set 1: p=0.016, set 2: p=0.006, total p=0.0001,<br />

OR=1.7) and rs1621388 (set 1: p=0.038, set 2: =0.054, total:<br />

p=0.001, OR=1.4). No other consistent associations with<br />

distractibility were found. Conclusions: Genetic associations<br />

were identified between distractibility and two SNPs in the<br />

P2RX7 gene in Swedish bipolar patients. These two SNPs are<br />

7 kb and 0.4 kb away from the rs2230912, previously<br />

associated with bipolar disorder. These SNPs might be<br />

associated to dysfunctional P2RX7 receptors and therefore<br />

impairment <strong>of</strong> neurotransmitter systems implicated in bipolar<br />

disorder.<br />

O10.2 THE INTERACTION BETWEEN SEROTONIN<br />

TRANSPORTER GENE (SLC6A4) SINGLE<br />

NUCLEOTIDE POLYMORPHISMS (SNPS) AND<br />

DEPRESSION RISK FACTORS<br />

Z. Samaan* (1), C. Xie (1), M. Zhang (1), B. Keavney (2), J.<br />

Engert (3), G. Pare (1), S. Rangarajan (1), A. Rosengren (4),<br />

K. Sliwa (5), M. Zubaid (6), S. Yusuf (1), S. Anand (1),<br />

Interheart Investigators<br />

1. McMaster University 2. Newcastle University 3. McGill<br />

University 4. Sahlgrenska University 5. Chris Hani<br />

Baragwanath Hospital 6. Kuwait University<br />

* samaanz@mcmaster.ca<br />

Hypothesis: The relationship between genetic variants and<br />

depression is mediated by depression risk factors (DRF).<br />

Background: Genetic studies <strong>of</strong> depression have lacked<br />

consistency at least partly due to the heterogeneous nature <strong>of</strong><br />

depression. We investigated the association between SLC6A4<br />

variants and DRF. Methods: The INTERHEART study was<br />

conducted in 52 countries and identified myocardial infarction<br />

(MI) risk factors. A sub-sample (n = 8717, 80% male) from 5<br />

ethnic groups (South Asian, European, Arab, Iranian and<br />

Nepalese) was studied for depression (based on DSM IV<br />

criteria). Ten SNPs from SLC6A4 were genotyped and tested<br />

for association with depression, DRF, and MI using a<br />

dominant model adjusted for ethnicity, age and sex. Results:<br />

Depression occurred in 1002 (12%) individuals. DRF<br />

included: smoking, stressful life events, chronic disease (i.e.<br />

prior stroke, diabetes), unemployment, sleep, locus <strong>of</strong> control<br />

and BMI. Five SNPs were associated with sleep (rs1042173,<br />

rs4583306, rs7224199, rs3794808 and rs140701, p-values<br />

range from 0.008-0.02) and history <strong>of</strong> stroke (p-values range<br />

from 0.001-0.006). Two additional SNPs were associated<br />

with BMI (rs16965628 p = 0.0007, rs2020933 p = 0.003).<br />

rs2020942 was associated with MI (p = 0.0037) and showed a<br />

trend with depression (p = 0.09). Analysis <strong>of</strong> SNP*DRF on<br />

depression showed a significant interaction between<br />

rs2020942 and sleep (p = 0.003, β -0.14) and rs140700 and<br />

BMI (p = 0.0015, β 0.06). Conclusions: The association<br />

between SLC6A4 variants and depression may be mediated in<br />

part by its association with depression-related risk factors.


O10.3 GABAA RECEPTOR GENE VARIATION AND<br />

SCHIZOAFFECTIVE DISORDER, BIPOLAR TYPE:<br />

EVIDENCE IMPLICATING GABRR1<br />

E. Green (1), D. Grozeva (1), V. Moskvina (1), M. Hamshere<br />

(1), I. Jones (1), L. Jones (2), G. Kirov (1), I. Nikolov (1), D.<br />

Vukcevic (3), S. Caesar (2), K. Gordon-Smith (2), C. Fraser<br />

(1), E. Russell (1), D. St Clair (4), A. Young (5), N. Ferrier<br />

(5), A. Farmer (6), P. McGuffin (6), Wellcome Trust Case<br />

Consortium , P. Holmans (1), M. Owen (1), M. O'Donovan<br />

(1), N. Craddock (1)<br />

1. Cardiff University 2. University <strong>of</strong> Birmingham 3.<br />

University <strong>of</strong> Oxford 4. University <strong>of</strong> Aberdeen<br />

5. Newcastle University 6. Institute <strong>of</strong> Psychiatry<br />

Previously, we have shown that variation in the GABAA<br />

receptor genes, GABRB1, GABRA4, GABRB3, GABRA5<br />

and GABRR3, influence risk for research diagnostic<br />

Schizoaffective disorder, bipolar type (RDC SABP) in our<br />

Wellcome Trust Case Consortium (WTCCC) bipolar disorder<br />

genome-wide association study (GWAS) (279 RDC SABP<br />

cases and 2938 controls). To ascertain if within our data any<br />

additional GABAA receptor gene family members were<br />

potentially associated with susceptibility to risk for RDC<br />

SABP, we went on to examine the imputed GWAS data<br />

(frequentist additive model). Ten genes were studied; omitting<br />

the 5 previously identified associated genes, the 3 X<br />

chromosome genes and GABRD on chromosome 1 for which<br />

no SNPs were genotyped in our dataset. SNPs were selected<br />

with a minor allele frequency > 1% and that were located<br />

within the reference sequence or that lay within 20kb,<br />

upstream and downstream, <strong>of</strong> the genes. Imputation provided<br />

superior additional evidence for association with SNP<br />

rs9451173 (frequentist additive p value = 1.5x10-4), an<br />

intronic SNP within GABAA rho 1 gene (GABRR1). We<br />

genotyped the rs9451173 polymorphism in a subset <strong>of</strong> our<br />

WTCCC and new samples, 298 RDC SABP cases (279<br />

WTCCC & 19 new) and 2270 controls (1442 WTCCC & 828<br />

new). We observed a significant association for rs9451173, C<br />

allele: cases, 7.1%; controls: 3.8%; Allelic p value = 4x10-4;<br />

odds ratio OR, [95% CI] = 051, [0.36-0.74] Our findings<br />

suggest that variation in GABRR1 gene potentially influences<br />

risk for the RDC SABP phenotype.<br />

O10.4 IDENTIFYING PANELS OF GENES FOR<br />

GENETIC RISK PREDICTION IN BIPOLAR<br />

DISORDER USING CONVERGENT FUNCTIONAL<br />

GENOMICS<br />

H. Le-Niculescu (1), S. Patel (1), D. Koller (1), J. Nurnberger<br />

(1), N. Schork (2), A. Niculescu (3)<br />

1. Indiana University 2. Scripps Research Institute 3. Indiana<br />

University/ Indianapolis VA <strong>Medical</strong> Center<br />

Mounting convergent evidence implicates many more genes in<br />

complex disorders such as bipolar disorder than the small<br />

number identified unambiguously by Genome-Wide<br />

Association studies (GWAS) to date. We previously proposed<br />

and provided pro<strong>of</strong> <strong>of</strong> principle for the use <strong>of</strong> a<br />

complementary approach, Convergent Functional Genomics<br />

(CFG), as a way <strong>of</strong> mining the existing GWAS datasets for<br />

signals that are there already, but did not reach significance<br />

using a genetics-only approach. We have now extended our<br />

previous work to include more datasets <strong>of</strong> GWAS, and more<br />

recent evidence from other lines <strong>of</strong> work. In essence our<br />

analysis is the most comprehensive integration <strong>of</strong> genetics and<br />

functional genomics to date in the field <strong>of</strong> bipolar disorder,<br />

identifying aseries <strong>of</strong> best candidate genes for bipolar disorder.<br />

Moreover, we have put together, based on these top genes, a<br />

comprehensive list <strong>of</strong> the best Single Nucleotide<br />

Polymorphisms (SNPs). Panels <strong>of</strong> such SNPs can be used for<br />

genetic testing for bipolar disorder before the illness manifests<br />

itself. We have developed a Genetic Risk Prediction Score<br />

(GRPS) based on such panels, and demonstrate how in an<br />

independent test cohort for which we had both genotypic and<br />

clinical information, the GRPS differentiates between patients<br />

with bipolar disorder and normal controls. Such testing could<br />

be used for diagnostics and personalized medicine approaches,<br />

as well as for early detection and prevention efforts in<br />

high-risk individuals.


O10.5 MINOR ALLELE OF RS2230912 IS<br />

PROTECTIVE AGAINST RAPID CYCLING IN<br />

A SWEDISH SAMPLE OF BIPOLAR DISORDER<br />

PATIENTS<br />

P. Nikamo* (1), L. Backlund (2), L. Frisén (3), I. Römer Ek<br />

(2), L. Träskman-Bendz (4), H. Ågren (5), G. Edman (6), M.<br />

Landén (2), U. Ösby (2), M. Schalling (1)<br />

1. Neurogenetics Unit, Department <strong>of</strong> Molecular Medicine and<br />

Surgery, Karolinska Institutet, Stockholm, Sweden 2.<br />

Department <strong>of</strong> Clinical Neuroscience, Karolinska Institutet,<br />

Stockholm, Sweden 3. Division <strong>of</strong> Psychiatry, Department <strong>of</strong><br />

Clinical Sciences, Karolinska Institutet, Danderyd University<br />

Hospital, Stockholm, Sweden 4. Division <strong>of</strong> Psychiatry,<br />

Department <strong>of</strong> Clinical Sciences, University Hospital, Lund,<br />

Sweden 5. Academy at the University <strong>of</strong> Gothenburg,<br />

Institution <strong>of</strong> Neuroscience and Physiology, Sweden<br />

6. Department <strong>of</strong> Psychiatry, R&D Section, Danderyd<br />

University Hospital, Sweden<br />

* pernilla.nikamo@ki.se<br />

Background: Bipolar disorder is a common and severe<br />

psychiatric illness. Of bipolar patients, 15-20% suffers from<br />

the rapid cycling subdiagnosis, defined by at least four<br />

episodes <strong>of</strong> a mood disturbance during a twelve month period.<br />

In previous studies the gene P2RX7 which is a purinergic<br />

ATP-binding calcium channel expressed in neurons, has been<br />

associated with bipolar disorder. Methods: We selected<br />

three non-synonymous SNPs in the P2RX7 gene; RS7958311<br />

(His270Arg), RS1718119 (Thr348Ala) and RS2230912<br />

(Gln460Arg) as well as one synonymous SNP; RS1621388.<br />

The RS2230912 polymorphism is non-conservative such that<br />

the minor allele results in a glutamine to arginine change<br />

(Gln460Arg), which is likely to affect P2RX7 dimerization<br />

and protein–protein interactions. We performed an association<br />

analysis <strong>of</strong> 646 Swedish patients, diagnosed with bipolar<br />

disorder according to DSM-IV, comparing those with a rapid<br />

cycling subdiagnosis (n=115) with those with other<br />

subdiagnoses (n=375). Results: For the rapid cycling<br />

subdiagnosis the RS2230912_G allele in the P2RX7 gene<br />

showed a negative association (P=0.004; OR=0.46).<br />

RS2230912 and RS1621388 are 367 bases apart and were<br />

together as haplotype AT positively associated with rapid<br />

cycling (P=0.01; OR=1.24) whereas the GT haplotype was<br />

negatively associated (P=0.01; OR 0.49). Conclusion: In<br />

Swedish bipolar patients with a rapid cycling subdiagnosis, we<br />

identified in the P2RX7 gene two protective associations and<br />

one association with an increased rate <strong>of</strong> rapid cycling. Thus,<br />

our findings support previous findings <strong>of</strong> an association<br />

between bipolar disorder and RS2230912_G allele in the<br />

P2RX7 gene, although our findings were specifically related<br />

to the rapid cycling subdiagnosis. Corresponding author:<br />

O11 EARLY CAREER INVESTIGATOR<br />

TRACK: WHOLE-GENOME APPROACHES<br />

O11.1 ADDRESSING ALLELIC HETEROGENEITY IN<br />

THE WHOLE-GENOME STUDY ERA: A<br />

POPULATION-BASED LINKAGE STUDY<br />

INVESTIGATING UNIPOLAR DEPRESSION<br />

S. Cohen-Woods* (1), C. Lewis (1), M. NG (1), A. Butler (1),<br />

K. Pirlo (1), A. Schosser (1), R. Uher (1), M. Owen (2), M.<br />

Barnes (3), P. Muglia (4), M. Lathrop (3), S. Heath (3), K.<br />

Aitchison (5), A. Farmer (1), I. Craig (1), P. McGuffin (1), G.<br />

Breen (1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King’s College<br />

London 2. Department <strong>of</strong> Psychological Medicine, Cardiff<br />

University 3. Centre National de Génotypage, 91057 Evry,<br />

France 4. GSK Research & Development, Genetics, Drug<br />

Discovery, Via Fleming 4, Verona, Italy and Greenford Road,<br />

Greenford, Middlesex UB6 OHE, UK 5. Institute <strong>of</strong><br />

Psychiatry, King's College London<br />

* sarah.cohen@kcl.ac.uk<br />

Although the substantial genetic contribution to depression<br />

predisposition is well-established, specific genes have failed to<br />

be consistently identified through ‘traditional’ single-gene<br />

association studies. Genome-wide association studies<br />

(GWAS) have recently become viable through the<br />

advancements in genotyping platforms and reduction in<br />

chip-genotyping costs. A complementary strategy to the<br />

standard case-control association analysis <strong>of</strong> genome-wide<br />

association study (GWAS) data is population-based linkage<br />

(PBL) analysis. PBL identifies relatively rare variants, as<br />

opposed to common variants, and also accounts for the<br />

possibility <strong>of</strong> allelic heterogeneity. Case samples from three<br />

genetic studies in depression were combined; the Depression<br />

Case-Control (DeCC) study, the Depression Network (DeNT)<br />

study, and the Genome Based Therapeutic Drugs for<br />

Depression (GENDEP) study. Controls screened by<br />

questionnaire and/or telephone interview for absence <strong>of</strong><br />

psychiatric disorders, and lack <strong>of</strong> family history <strong>of</strong> psychiatric<br />

disorders, were also genotyped. In total the sample presented<br />

includes approximately 3,200 cases and 1,800 controls. Each<br />

case individual was diagnosed with DSM-IV/ICD-10<br />

depression, <strong>of</strong> at least moderate severity, as assessed by SCAN<br />

interview. Samples were genotyped using the Illumina Human<br />

610-quad DNA analysis bead chip. Loci showing evidence for<br />

linkage will be presented, alongside a clear description <strong>of</strong> the<br />

quality control applied. This represents the first clinical<br />

case-control GWAS study applying population-based linkage<br />

to date investigating recurrent unipolar depression. With<br />

important and far-reaching implications for pharmaco-genetic<br />

studies, more directed and economical research may be<br />

applied to further elucidate the underlying aetiology, and<br />

ultimately treatment, <strong>of</strong> unipolar depression.


O11.2 IMPROVEMENT OF PHENOTYPING IN<br />

GENOME WIDE ASSOCIATION STUDIES ON<br />

SCHIZOPHRENIA: AN APPLICATION OF FACTOR<br />

ANALYSIS<br />

E. Derks* (1), M. Boks (1), J. Allardyce (2), R. Oph<strong>of</strong>f (3), R.<br />

Kahn (1), W. Cahn (1)<br />

1. Department <strong>of</strong> Psychiatry, Rudolf Magnus Institute <strong>of</strong><br />

Neuroscience, University <strong>Medical</strong> Centre Utrecht, Utrecht,<br />

The Netherlands 2. Psychiatry and Neuropsychology, School<br />

for Mental Health and Neuroscience, EURON, SEARCH,<br />

Maastricht University <strong>Medical</strong> Centre, Maastricht, The<br />

Netherlands 3. Complex Genetics Section, Department <strong>of</strong><br />

Biomedical Genetics, University <strong>Medical</strong> Center Utrecht,<br />

Utrecht, the Netherlands<br />

* e.m.derks@umcutrecht.nl<br />

Genetic factors explain a large proportion (81%) <strong>of</strong> the<br />

variation in schizophrenia. Nevertheless, the identification <strong>of</strong><br />

specific genes is not very successful as the enormous<br />

investments that are made on the genotypic level (e.g.,<br />

genotyping ~1,000,000 SNPs) are not matched by similar<br />

investments on the phenotypic level. We aim to improve the<br />

assessment <strong>of</strong> individual differences in schizophrenia by<br />

performing factor analysis on 66 Comprehensive Assessment<br />

<strong>of</strong> Psychiatric History (CASH) symptoms in a sample <strong>of</strong> 2290<br />

patients with schizophrenia, schizophreniform, schizoaffective<br />

disorder, bipolar disorder or depression, and 1888 healthy<br />

controls from the Netherlands. Variation in schizophrenia<br />

symptoms is best represented by five latent dimensions<br />

(positive, negative, mania, disorganisation, and depression).<br />

Diagnostic subgroups show different patterns on the five latent<br />

dimensions. Compared to healthy controls, patients diagnosed<br />

with depression show elevated scores on each <strong>of</strong> the five<br />

dimensions. Despite the fact that schizophrenia is a highly<br />

heterogeneous disorder, gene finding studies collapse patients<br />

with different symptom patterns into one group. However, if<br />

in reality these patients have distinct genetic vulnerabilities,<br />

this would dramatically decrease the statistical power to detect<br />

functional genetic variants. Furthermore, case-control studies<br />

on schizophrenia should exclude patients diagnosed with<br />

depression as healthy controls. Even though these patients do<br />

not meet diagnostic criteria for schizophrenia, they obtain high<br />

scores on the five latent dimensions which explain variation in<br />

schizophrenia symptoms suggesting that they share part <strong>of</strong> the<br />

genetic vulnerability to schizophrenia. We recommend using<br />

continuously distributed factor scores as quantitative outcome<br />

measures in future gene finding studies.<br />

O11.3 HIGH COVERAGE WHOLE-GENOME<br />

SEQUENCING IN SCHIZOPHRENIA: PROOF OF<br />

FEASIBILITY AND VARIANT ANNOTATION<br />

PROTOCOLS<br />

A. Need* (1), D. Ge (1), K. Shianna (1), J. Thrall (2), J. Maia<br />

(1), J. McEvoy (2), D. Goldstein (1)<br />

1. Institute for Genome Sciences & Policy, Duke University,<br />

Durham, NC 27710, USA 2. Department <strong>of</strong> Psychiatry and<br />

Behavioral Sciences, Duke University <strong>Medical</strong> Center,<br />

Durham, NC 27509, USA<br />

* need0001@duke.edu<br />

Large-scale genome-wide association studies have suggested<br />

that common genetic variants make at most a modest<br />

contribution to the high heritability <strong>of</strong> schizophrenia. These<br />

findings, together with the association with schizophrenia <strong>of</strong> a<br />

number <strong>of</strong> rare copy number variants, have led to the<br />

hypothesis that the much <strong>of</strong> the heritability may be explained<br />

by rare variants with strong effects. In order to<br />

comprehensively screen for such variation it is necessary to<br />

sequence entire patient genomes (or exomes). Here we present<br />

the first whole genome sequence <strong>of</strong> a patient with<br />

schizophrenia, generated as part <strong>of</strong> a larger schizophrenia<br />

sequencing study at the Center for Human Genome Variation<br />

at Duke University. We use these data to illustrate the<br />

procedures involved in generating and analyzing<br />

whole-genome sequence data in an academic setting. We<br />

present a novel s<strong>of</strong>tware program, called Sequence Variant<br />

Analyzer, which is used to annotate and assign function to<br />

variants identified from the sequence data. We will report the<br />

total number <strong>of</strong> novel coding variants and their predicted<br />

function (nonsense, missense, synonymous, splice-affecting,<br />

etc), as well as detailed analyses <strong>of</strong> candidate genes, regions<br />

and pathways that have been associated with schizophrenia<br />

though family studies or animal models. Collectively, these<br />

analyses confirm the feasibility <strong>of</strong> whole-genome sequencing<br />

as a discovery tool in psychiatric genomics.


O11.4 GENETICS OF ALCOHOL DEPENDENCE<br />

COMORBIDITY IN BIPOLAR DISORDER:<br />

GENOME-WIDE ASSOCIATION STUDY<br />

E. Nwulia* (1), M. Hipolito (1), W. Lawson (1), P. Zandi (2),<br />

J. Nurnberger Jr (3), Bipolar Genome Study (BiGS)<br />

1. Howard University 2. Johns Hopkins University 3.<br />

University <strong>of</strong> Indiana<br />

* enwulia@howard.edu<br />

The identification <strong>of</strong> bipolar disorder (BD) susceptibility<br />

genes has been hindered by genetic heterogeneity , among<br />

other factors. Genetic heterogeneity may be a reflection <strong>of</strong><br />

phenotypic variability. Several lines <strong>of</strong> inquiry have suggested<br />

that genetic predisposition to BD might also be expressed as<br />

liability to alcohol dependence (AD) in subgroups <strong>of</strong> families.<br />

To further explore this relationship, we performed a<br />

Genomewide Association Study (GWAS) for comorbid AD in<br />

a sample <strong>of</strong> unrelated BD individuals (n=1001) <strong>of</strong> European<br />

ancestry (EA) recruited from 11 US institutions as part <strong>of</strong> the<br />

NIMH Collaborative Genetics <strong>of</strong> Bipolar Disease Study. BD<br />

cases were classified into those with AD (BDAD, N = 460)<br />

and those without AD (BDNAD, n = 528) for the purpose <strong>of</strong><br />

case-only GWA. Genotyping using Affymetrix 6.0 array was<br />

provided by the Genetic Analysis Information Network<br />

(GAIN). Analyses involved logistic and polytomous<br />

regressions; we also adjusted for sex, which was significantly<br />

associated with alcohol dependence (51.2% in males versus<br />

41.9% in females; P = 0.003). The strongest statistical signal<br />

(OR, 1.64; P = 1.19 x 10-6) from this case-only analysis was<br />

found in the WDR59 gene on chromosome 16q23.1. We also<br />

found strong association between a SNP in the alphaN-catenin<br />

(CTNNA2) gene on 2p12 with BDAD vs. BDNAD (OR, 1.51;<br />

P < 7.0 x 10-6). The risk allele for this locus was however<br />

inversely associated with BDNAD vs. normal controls (NC)<br />

(OR, 0.77;P < 10-3), but not with BDAD vs. NC (OR, 1.16; P<br />

< 0.08). Confirmation <strong>of</strong> these putative associations,<br />

coherence with other lines <strong>of</strong> study, and demonstration <strong>of</strong><br />

biological plausibility are needed for causal inferences to be<br />

applied to these findings.<br />

O11.5 GENE BASED AND PATHWAY ANALYSIS<br />

REVEAL GENOME-WIDE SIGNIFICANT GENES AND<br />

MOLECULAR PATHWAYS ASSOCIATED WITH<br />

BIPOLAR DISORDER<br />

I. Pedroso* (1), D. Collier (2), A. Farmer (1), P. McGuffin<br />

(1), G. Breen (1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King’s College<br />

London, De Crespigny Park, London SE5 8AF, UK 2.<br />

Division <strong>of</strong> Psychological Medicine, Institute <strong>of</strong> Psychiatry,<br />

King’s College London, De Crespigny Park, London SE5<br />

8AF, UK<br />

* intipedroso@gmail.com<br />

Despite the high heritability (~80%) and familial aggregation<br />

observed in Bipolar Disorder (BD), its molecular genetic<br />

underpinnings are still largely unknown. Candidate gene<br />

findings have yielded few replicated associations. Data is now<br />

available from several Genome Wide Association Scans<br />

(GWAS) <strong>of</strong> BD. We reanalyzed four <strong>of</strong> these GWAS using<br />

gene based and pathway analysis. We combined the<br />

association statistics <strong>of</strong> SNPs on a gene-wide basis while<br />

correcting by the number <strong>of</strong> tests and the correlation between<br />

them (i.e. LD between SNPs). Our method has advantages<br />

over alternatives: it is neither biased by the gene-size nor<br />

inflated by the correlation between the tests, it allows the use<br />

<strong>of</strong> summary statistics and LD information from HapMap and it<br />

is computationally inexpensive. We identified several<br />

genome-wide significant genes, which were below multiple<br />

testing correction on the single SNP analysis, and several<br />

pathways passed multiple testing correction. Our results<br />

suggest that combining the effect <strong>of</strong> genetic variants enhance<br />

the power to detect genetic associations and provide replicated<br />

evidence for common genetic variation on genes and<br />

biological processes on the aetiology <strong>of</strong> BD.


O11.6 A GENOME-WIDE ASSOCIATION STUDY OF<br />

OBSESSIVE-COMPULSIVE DISORDER USING DNA<br />

POOLING<br />

J. Wendland* (1), F. McMahon (1), D. Denys (2), S. Walitza<br />

(3), M. Arcos-Burgos (4), S. Cichon (5), T. Schulze (6), M.<br />

Rietschel (6), D. Murphy (1)<br />

1. NIMH 2. Univ. <strong>of</strong> Amsterdam 3. Univ. <strong>of</strong> Zurich 4. Univ.<br />

<strong>of</strong> Miami 5. University <strong>of</strong> Bonn 6. Central Institute <strong>of</strong> Mental<br />

Health, Mannheim<br />

* wendlandj@mail.nih.gov<br />

Obsessive-compulsive disorder (OCD) is a severe<br />

neuropsychiatric illness characterized by persistent, intrusive<br />

thoughts and repetitive behaviors. There is compelling<br />

evidence for major genetic contributions to the etiology <strong>of</strong><br />

OCD from twin and family studies. The exact molecular<br />

mechanisms, however, are not fully elucidated, and few genes<br />

or polymorphisms have been consistently replicated in genetic<br />

studies. Here, we used a genome-wide association study<br />

approach to identify common genetic variants involved in<br />

OCD. We pooled equimolar amounts <strong>of</strong> 40 – 60 unrelated<br />

individuals’ genomic DNA and used Illumina single<br />

nucleotide polymorphism (SNP) microarray chips<br />

interrogating over 320,000 markers distributed across the<br />

genome. We tested 4 OCD pools derived from US probands<br />

and 4 OCD pools from Dutch probands and an equal number<br />

<strong>of</strong> ethnically and geographically matched control pools.<br />

Inferred allele frequencies between case and control pools<br />

were compared with a two-sided t test. We identified 25 SNPs<br />

that were nominally significantly associated at P < .01 in both<br />

the US and European case-control comparison and had the<br />

same direction <strong>of</strong> effect. One <strong>of</strong> these markers, rs1409294,<br />

was replicated in an independent collection <strong>of</strong> 103 German<br />

early-onset OCD trios which were individually genotyped by<br />

5’ exonuclease methodology. The replicated variant is located<br />

on chromosome 6q22.31 in an intergenic region between the<br />

non-coding RNA gene STL (six-twelve leukemia) and<br />

NKAIN2, a protein-coding gene expressed in the brain. In<br />

addition to further replication attempts, functional studies are<br />

now warranted to determine the biological mechanisms<br />

through which the newly-identified locus may be relevant to<br />

OCD etiology.<br />

O12 SCHIZOPHRENIA 3<br />

O12.1 FOLLOW-UP FINE MAPPING AND DEEP<br />

RE-SEQUENCING OF ZNF804A IN SCHIZOPHRENIA.<br />

H. Williams (1), N. Norton (1), S. Dwyer (1), L. Carroll (1),<br />

L. Georgieva (1), T. Peirce (1), V. Moskvina (1), M.<br />

Hamshere (1), I. Nikolov (1), P. Holmans (1), S. Zammit (1),<br />

A. Hartmann (2), H. Moller (2), D. Morris (3), E. Quinn (3),<br />

N. Williams (1), I. Giegling (2), N. Craddock (1), M. Gill (3),<br />

A. Corvin (3), D. Rujescu (2), G. Kirov (1), M. Owen (1), M.<br />

O'Donovan (1)<br />

1. Cardiff University 2. Ludwig-Maximilians-University 3.<br />

Trinity College Dublin<br />

The use <strong>of</strong> genome wide association studies (GWAS) to<br />

uncover genetic variants that are robustly associated with<br />

complex diseases is well established. However, further work is<br />

required to identify which genetic variant or variants actually<br />

confer disease risk and the mechanism involved. We have<br />

previously published the results <strong>of</strong> a schizophrenia GWAS<br />

(O’Donovan, et al 2008) where an intronic SNP (rs1344706)<br />

in the gene ZNF804A was our most significant finding (meta<br />

P=1.61x10-7). Here we describe follow-up studies aimed at a)<br />

confirming the evidence for robust association obtained in our<br />

and c) determining how this might confer risk to the disorder.<br />

We have performed a meta analysis including additional data<br />

from recently published schizophrenia GWAS and show that<br />

marker rs1344706 now exceeds genome wide levels <strong>of</strong><br />

significance. We performed mutation screening <strong>of</strong> all coding<br />

sequence, association analysis <strong>of</strong> functional and tag SNPs as<br />

well as deep re-sequencing <strong>of</strong> the genomic region around<br />

ZNF804A. Our results show that although we have<br />

discovered and characterised many new novel polymorphisms,<br />

rs1344706 is still the most significant variant at this locus.<br />

Finally using the GeneVar database we shown that the<br />

rs1344706 risk allele (T) is associated with an over expression<br />

<strong>of</strong> the ZNF804A transcript.


O12.2 SCHIZOPHRENIA-RELATED BEHAVIORAL<br />

CHANGES IN G72/G30 TRANSGENIC MICE<br />

D. Otte* (1), A. Bilkei-Gorzo (1), Ö. Yilmaz (1), C. Turck<br />

(2), M. Holst (3), K. Schilling (4), R. Abou Jamra (5), P.<br />

Propping (5), A. Zimmer (1)<br />

1. Department <strong>of</strong> Molecular Psychiatry, University <strong>of</strong> Bonn,<br />

Germany 2. MPI Psychiatry, Munich, Germany 3. Institute <strong>of</strong><br />

Anatomy, University <strong>of</strong> Bonn, Germany 4. Institute <strong>of</strong><br />

Anatomy, University <strong>of</strong> Bonn, Germany 5. Institute <strong>of</strong> Human<br />

Genetics, University <strong>of</strong> Bonn, Germany<br />

* d.otte@uni-bonn.de<br />

Genetic studies have implicated the evolutionary novel,<br />

anthropoid primates-specific gene locus G72/G30 in<br />

psychiatric diseases like schizophrenia, bipolar- and<br />

panic-disorders. It encodes for a protein LG72 that has been<br />

controversially discussed as putative activator or inhibitor <strong>of</strong><br />

the peroxisomal enzyme D-amino-acid-oxidase (DAO), or as a<br />

mitochondrial protein. Here we have generated “humanized”<br />

BAC transgenic mice (G72Tg) that express G72/G30<br />

transcripts. In-situ hybridization analysis revealed that G72<br />

mRNAs are prominently expressed in granular cells<br />

throughout the brain. Transgenic mice show a number <strong>of</strong><br />

behavioral phenotypes that are relevant to psychiatric<br />

disorders. These phenotypes include deficits in sensorimotor<br />

gating, locomotor deficits, increase compulsive behaviors and<br />

deficits in smelling. These results identify LG72 as a possible<br />

key protein in pathomechanism <strong>of</strong> psychiatric diseases.<br />

O12.3 DEEP RESEQUENCING AND ASSOCIATION<br />

ANALYSIS OF SCHIZOPHRENIA CANDIDATE<br />

GENES<br />

P. Sullivan (1), J. Crowley* (1), M. Morgan (2), P. Sklar (3),<br />

S. Purcell (4), J. Lieberman (5), R. Gibbs (2)<br />

1. UNC Chapel Hill 2. Baylor College <strong>of</strong> Medicine 3.<br />

Massachusetts General Hospital 4. The Broad Institute 5.<br />

Columbia University<br />

* crowley@unc.edu<br />

In 2006, there were ~10 genes for which there was reasonable<br />

evidence for involvement in the etiology <strong>of</strong> schizophrenia. The<br />

pattern <strong>of</strong> results for these genes suggests that there may be<br />

sequence variants that have not yet been discovered, and<br />

existing re-sequencing has been limited. To address this<br />

limitation, we re-sequenced the coding regions, 5’ and 3’<br />

UTRs, splice sites, promoters and conserved intronic regions<br />

<strong>of</strong> these 10 genes in 727 cases with schizophrenia from the<br />

CATIE clinical trial and 729 matched controls. A total <strong>of</strong> 778<br />

single nucleotide polymorphisms (SNPs) were identified by<br />

resequencing, including 620 novel variants. A total <strong>of</strong> 30 high<br />

priority variants were deemed worthy <strong>of</strong> genotyping in a larger<br />

population, based on a combination <strong>of</strong> factors including:<br />

association with schizophrenia in CATIE, novelty, SNP type<br />

(non-sense and missense mutations) and minor allele<br />

frequency in cases <strong>of</strong> European ancestry. These 30 variants are<br />

currently being examined for association with schizophrenia in<br />

a distinct sample <strong>of</strong> 3,391 schizophrenia cases and 3,181<br />

controls, all <strong>of</strong> European ancestry and collected by the<br />

International Schizophrenia Consortium (ISC). In conclusion,<br />

schizophrenia candidate genes possess a great deal <strong>of</strong> common<br />

and rare variation not covered on current genome-wide<br />

association platforms. Further analysis <strong>of</strong> these variants in<br />

distinct schizophrenia samples will provide a clearer picture <strong>of</strong><br />

the role these genes play in schizophrenia etiology.


O12.4 FINE MAPPING OF AHI1 ON 6Q23 AS A<br />

SUSCEPTIBILITY GENE FOR SCHIZOPHRENIA<br />

FROM ASSOCIATION TO EVOLUTIONARY<br />

EVIDENCES<br />

F. Torri (1), A. Akelai (2), S. Lupoli (3), M. Sironi (4), M.<br />

Fumagalli (4), E. Osimo (1), C. Dal Fiume (1), D. ,<br />

Amann-Zalcenstein (5), E. Ben-Asher (5), K. Kanyas (2), R.<br />

Cagliani (4), D. Lancet (5), P. Cozzi (6), L. , SStrik Lievers<br />

(1), E. Salvi (1), A. Orro (7), J. Beckmann (8), B. Lerer (9), F.<br />

Macciardi (10)<br />

1. University <strong>of</strong> Milan, Filarete Foundation, Milan, Italy<br />

2. Hadassah–Hebrew University <strong>Medical</strong> Center, Jerusalem,<br />

Israel 3. INSPE, Scientific Institute San Raffaele, Milan, Italy<br />

4. Scientific Institute IRCCS E. Medea, Parini (LC), Italy 5.<br />

Weizmann Institute <strong>of</strong> Science, Rehovot, Israel 6. ITB, CNR,<br />

Milan, Italy 7. ITB, CNR, Milan, Italy;CILEA Consortium,<br />

Segrate, Milan, Italy 8. Centre Hospitalier Universitaire<br />

Vaudois and University <strong>of</strong> Lausanne, Switzerland 9.<br />

Hadassah–Hebrew University <strong>Medical</strong> Center, Jerusalem,<br />

Israel;Miller School <strong>of</strong> Medicine, University <strong>of</strong> Miami<br />

10. University <strong>of</strong> Milan, Filarete Foundation, Milan,<br />

Italy;Department <strong>of</strong> Psychiatry and Human Behavior,<br />

University <strong>of</strong> California, Irvine, CA, USA<br />

In previous linkage studies we identified a susceptibility<br />

region on 6q23.3 in a set <strong>of</strong> Arab-Israeli families. In a<br />

follow-up study we proposed the Abelson Helper Integration<br />

Site 1 (AHI1), which plays a key role in neurodevelopment, as<br />

a gene associated with SCZ. To definitively confirm AHI1 as<br />

the best finding in the region, we performed a dense mapping<br />

genotyping 2019 SNPs in 58 families <strong>of</strong> the previously studied<br />

sample. Our strongest associated SNPs and haplotypes lay<br />

within a 500 kb genomic region (135.6–136.1 Mb)<br />

encompassing AHI1 and BC040979. Within the AHI1 gene,<br />

rs11154801 (p=6.23E-06) and rs7759971 (p=1.84E-06)<br />

showed the strongest associations, with haplotypes having<br />

p-values in the 10E-8 to 10E-10 range. The second most<br />

highly significant region maps immediately distally from<br />

AHI1 and includes the intergenic region between BC040979<br />

and PDE7B, and both the PDE7B and MAP7 genes, which<br />

show brain expression. Given a possible involvement <strong>of</strong><br />

positive selection <strong>of</strong> SCZ genes we resequenced a 10kb region<br />

within AHI1 in ethnically defined populations and found<br />

evidence for a selective sweep in Europeans. Network analysis<br />

indicated the presence <strong>of</strong> two haplotype clades, with<br />

SCZ-susceptibility haplotypes clustering within the major<br />

clade. In line with new wave <strong>of</strong> genetic investigations<br />

focusing now on the implication <strong>of</strong> rare and common copy<br />

number variants (CNVs) in SCZ, we have also carried out a<br />

whole-genome CNV analysis in the same Arab-Israeli sample<br />

detect CNVs investigate the pathways in which the<br />

SCZ-associated CNVs are mainly involved to build a potential<br />

model <strong>of</strong> interaction with the SCZ-associated genes we found<br />

in our study. Our data confirm and definitely support the role<br />

<strong>of</strong> AHI1 as a susceptibility gene for SCZ.<br />

O12.5 COPY NUMBER VARIANTS AFFECTING<br />

MYELIN GENES IN SCHIZOPHRENIA<br />

L. Georgieva* (1), G. Kirov (1), P. Holmans (1), V.<br />

Moskvina (1), V. Haroutunian (2), T. Sakurai (1), J. Young<br />

(3), J. Buxbaum (3), N. Craddock (1), M. Owen (1), M.<br />

O'Donovan (1)<br />

1. MRC Centre for Neuropsychiatric Genetics and Genomics,<br />

Department <strong>of</strong> Psychological Medicine, Cardiff University,<br />

Heath Park, Cardiff, United Kingdom 2. Department <strong>of</strong><br />

Psychiatry, Mount Sinai School <strong>of</strong> Medicine, New York,<br />

USA;Mental Illness Research, <strong>Education</strong> and Clinical Centres,<br />

Bronx Veterans Affairs <strong>Medical</strong> Centre, Bronx, New York,<br />

USA 3. Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong><br />

Medicine, New York, USA<br />

* georgievaln@cardiff.ac.uk<br />

Background: Abnormal oligodendrocyte function and<br />

myelination have been implicated in schizophrenia (SZ) by a<br />

diverse range <strong>of</strong> experimental approaches including gene<br />

expression analysis, neuropathology, neuroimaging and<br />

genetic association. Some <strong>of</strong> the most convincing findings in<br />

SZ genetics in the last 18 months concern the involvement <strong>of</strong><br />

low frequency copy number variants (CNVs) in that disorder.<br />

Aims: To assess whether rare CNVs (


O13 AUTISM<br />

O13.1 ALTERED SYNAPTIC PLASTICITY IN MICE<br />

LACKING ONE COPY OF THE SHANK3 GENE, A<br />

GENE MUTATED IN AUTISM SPECTRUM<br />

DISORDERS<br />

J. Buxbaum* (1), T. Sakurai (1), X. Wang (1), Q. Zhou (1),<br />

D. Papapetrou (1), P. H<strong>of</strong> (1), O. Bozdagi (1), L. Ospina (1)<br />

1. Mount Sinai School <strong>of</strong> Medicine<br />

* joseph.buxbaum@mssm.edu<br />

Background: Studies have demonstrated that rare, highly<br />

penetrant genetic variants can lead to autism spectrum<br />

disorders (ASDs). With the identification <strong>of</strong> genes<br />

contributing to the development <strong>of</strong> ASD we are in a position<br />

to develop animal models to understand the developmental<br />

consequences <strong>of</strong> alterations in these genes. One recurrent<br />

finding is that loss <strong>of</strong> one copy <strong>of</strong> SHANK3, either by copy<br />

number variation (CNV) or by mutation, leads to global<br />

developmental delay, frequently with an ASD. Methods: We<br />

have used gene-targeting methods to disrupt Shank3 in mice<br />

and have characterized these mice from biochemical,<br />

electrophysiological, neuropathological and behavioral<br />

standpoints. Results: Mice lacking one copy <strong>of</strong> Shank3<br />

develop normally and are born with Mendelian ratios.<br />

Developmental milestones are normal in the heterozygotes as<br />

determined by detailed observation and behavioral testing.<br />

The frequency <strong>of</strong> miniature excitatory postsynaptic currents is<br />

reduced in these mice indicating a deficit in spontaneous<br />

neurotransmitter release. Moreover, hippocampal long-term<br />

potentiation (LTP) is deficient, while long-term depression<br />

(LTD) is unchanged. Microscopic examination <strong>of</strong> synaptic<br />

spines demonstrates that the normal, stable modulation <strong>of</strong><br />

spine volume in response to LTP is not present in the<br />

heterozygous animals, although transient changes are detected<br />

Discussion: Our analyses support a role for abnormal synaptic<br />

development and function in at least some forms <strong>of</strong> ASDs.<br />

These studies lead to direct predictions in human studies and<br />

to potential new avenues for ASD therapeutics.<br />

O13.2 NARROWING THE CRITICAL DELETION<br />

REGION FOR AUTISM SPECTRUM DISORDERS IN<br />

CHROMOSOME 16P11.2<br />

A. Crepel* (1), W. De La Marche (2), J. Fryns (1), I. Noens<br />

(3), J. Steyaert (2), K. Devriendt (1), H. Peeters (1)<br />

1. Center for Human Genetics, University Hospital Leuven,<br />

Leuven, Belgium 2. Department for Child and Youth<br />

Psychiatry, University <strong>of</strong> Leuven, Leuven, Belgium<br />

3. Center for Parenting, Child Welfare and Disabilities,<br />

Department <strong>of</strong> <strong>Education</strong>al Sciences, University <strong>of</strong> Leuven,<br />

Leuven, Belgium<br />

* an.crepel@med.kuleuven.be<br />

One <strong>of</strong> the most common genomic disorders associated to<br />

autism to date is a ~600kb deletion containing 25 genes at<br />

chromosome 16p11.2. It was reported to occur in up to 1% <strong>of</strong><br />

autistic patients in 3 large autism studies and was recently<br />

studied in MR/MCA patients, extending the phenotype to<br />

other developmental disorders and speech disorders. The<br />

inheritance pattern is similar to that <strong>of</strong> other major effect<br />

variants implicated in complex disorders: the deletion is not<br />

limited to sporadic patients with de novo mutations, but may<br />

be inherited from unaffected parents. This microdeletion is<br />

typically ~600kb in size and flanked by segmental<br />

duplications, which explains its recurrent nature. Of the 25<br />

genes in this region, many represent good autism candidate<br />

genes based on expression pr<strong>of</strong>iles, pathway analysis and<br />

other available functional data. Although no firm association<br />

has been established for any one <strong>of</strong> these genes, mutation<br />

studies yielded suggestive evidence for the SEZ6L2 gene.<br />

We report the discovery <strong>of</strong> a smaller, 115kb deletion within<br />

the known 16p11.2 CNV in a 3 generation family with<br />

4 patients with autism and normal intelligence. Segregation<br />

was studied by independent psychiatric evaluation and<br />

collecting scores on the Social Responsiveness Scale (SRS) in<br />

9 family members. From these data we have evidence for<br />

segregation <strong>of</strong> the small deletion with autism or autistic traits<br />

in this family. We conclude that this observation narrows the<br />

critical deletion region for autism on chromosome 16p11.2 to<br />

only 5 genes.


O13.3 GENETIC MAPPING OF TEMPORAL MEMORY<br />

COMPONENTS IN MICE REVEALS A<br />

HOMOLOGOUS AUTISM LOCUS<br />

H. Bruining (1), O. Stiedl (2), F. Meye (1), E. Pjetri (1), H.<br />

Oppelaar (1), L. Nonkens (1), G. Ramakers (1), C. Fernandez<br />

(3), H. Swaab (4), H. van Lith (5), H. van Engeland (1), M.<br />

Kas (5)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neurosciences, Department <strong>of</strong><br />

Psychiatry, University <strong>Medical</strong> Centre Utrecht, the<br />

Netherlands 2. Center for Neurogenomics and Cognitive<br />

Research, Neuroscience Campus Amsterdam, VU<br />

University Amsterdam, The Netherlands 3. Social, Genetic<br />

and Developmental Psychiatry Centre, Institute <strong>of</strong> Psychiatry,<br />

King's College London, London, United Kingdom<br />

4. University <strong>of</strong> Leiden, Department <strong>of</strong> Social Sciences, The<br />

Netherlands 5. Rudolf Magnus Institute <strong>of</strong> Neurosciences,<br />

Department <strong>of</strong> Neuroscience and Pharmacology University<br />

<strong>Medical</strong> Centre Utrecht, The Netherlands<br />

Chromosome substitution strains (CSSs) <strong>of</strong> mice are<br />

extremely useful to identify genetic factors underlying<br />

neurobiological mechanisms <strong>of</strong> complex behaviors. The<br />

current study mapped chromosomes affecting the regulation <strong>of</strong><br />

memory formation in a social discrimination (SD) task in 21<br />

strains <strong>of</strong> the CSS panel (C57BL/6J-Chr#A/NaJ). Selected<br />

CSSs with altered SD performance were tested in various<br />

hippocampus-dependent learning tasks, and long-term<br />

potentiation (LTP) was assessed as a measure <strong>of</strong> hippocampal<br />

plasticity. We identified particular CSSs with either<br />

impairment in short-term or long-term SD performance. For<br />

example, one CSS was unable to discriminate conspecifics at<br />

5-min training-test interval (TTI), but showed SD at 24 hr<br />

TTI. In contrast, another CSS showed SD after 5 min, but not<br />

after a TTI <strong>of</strong> 24 hr. These CSS-specific temporal<br />

performance differences were confirmed in object<br />

discrimination and fear conditioning. No differences were<br />

found in the Morris water maze when compared to C57BL/6J<br />

controls, indicating that spatial memory was not affected.<br />

Electrophysiological measurements in the hippocampus<br />

revealed an elevated threshold for LTP induction only in the<br />

CSS with long-term memory impairments. These findings<br />

indicate that temporal processes <strong>of</strong> memory formation can be<br />

dissected at the genetic, behavioral and electrophysiological<br />

level. Additional phenotypic characterization <strong>of</strong> specific<br />

performance differences (e.g., encoding and extinction <strong>of</strong><br />

memory) indicates a common genetic contribution <strong>of</strong><br />

short-term versus long-term memory components in both<br />

social and non-social discrimination capacity and fear<br />

conditioning. Interestingly, subsequent genetic mapping <strong>of</strong><br />

these social and non-social memory phenotypes revealed a<br />

mouse QTL-interval homologous to an autism CNV locus.<br />

O13.4 DISSECTING THE CLINICAL<br />

HETEROGENEITY OF AUTISM SPECTRUM<br />

DISORDERS THROUGH DEFINED GENOTYPES.<br />

H. Bruining (1), L. de Sonneville (2), H. Swaab (2), M. de<br />

Jonge (1), M. Kas (3), H. van Engeland (1), J. Vorstman (1)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Children and Adolescent Psychiatry,<br />

University <strong>Medical</strong> Centre Utrecht, the Netherlands 2.<br />

Institute <strong>of</strong> Social Sciences, Department <strong>of</strong> Developmental<br />

disorders, University <strong>of</strong> Leiden, the Netherlands 3. Rudolf<br />

Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Neuroscience and Pharmacology, University <strong>Medical</strong> Centre<br />

Utrecht, Utrecht, the Netherlands<br />

The increasing evidence for a causative role <strong>of</strong> genetic<br />

variants with large effect in autism spectrum disorders (ASD)<br />

raises the question whether specific genetic subtypes lead to<br />

ASD phenotypes that are distinguishable from the remainder<br />

<strong>of</strong> the ASD population. We examined autistic symptoms in<br />

two samples with a genetic variant previously shown to cause<br />

ASD: patients with the 22q11.2 deletion (22q11DS) or with<br />

Klinefelter syndrome (KS). These assessments aim to provide<br />

pro<strong>of</strong>-<strong>of</strong>-concept <strong>of</strong> the existence <strong>of</strong> genotype-specific ASD<br />

phenotypes. ASD symptom pr<strong>of</strong>iles in 22q11DS (n=90) and<br />

KS (n=51) were statistically compared at two different levels:<br />

1) with a large group <strong>of</strong> idiopathic ASD patients (n=371) and<br />

2) with the non-ASD subjects among the 22q11DS and KS<br />

groups. Discriminant analyses <strong>of</strong> the autistic symptom pr<strong>of</strong>iles<br />

clearly showed that autistic phenotypes in the two subgroups<br />

could be delineated from the idiopathic ASD. Furthermore, the<br />

two subgroups (22q11DS and KS) displayed enhanced<br />

symptom homogeneity compared to the idiopathic ASD<br />

patients.To our knowledge, this is the first reversed<br />

phenotyping approach that provides robust evidence for the<br />

existence <strong>of</strong> genetic subtype-specific ASD phenotypes within<br />

the ASD population. We propose that evidence for the<br />

existence <strong>of</strong> a phenotype related to a specific genetic variant is<br />

provided when 1) the ASD phenotypes <strong>of</strong> a genetic subtype<br />

can be delineated from the remainder <strong>of</strong> the ASD population<br />

based on ASD symptomatology, and 2) the variance <strong>of</strong> ASD<br />

symptoms within a genetic subtype is significantly smaller in<br />

comparison to the variance <strong>of</strong> the remainder <strong>of</strong> the ASD<br />

population.


O13.5 THE EFFECT OF CHR16P11.2<br />

MICRODELETIONS AND MICRODUPLICATIONS<br />

ON GENE EXPRESSION IN AUTISM SPECTRUM<br />

DISORDERS AND SCHIZOPHRENIA<br />

M. Kusenda, V. Vacic , S. Yoon, J. Sebat<br />

* Kusenda@cshl.edu<br />

The number <strong>of</strong> rare variants found to be associated with<br />

multiple psychiatric disorders is growing. One such locus is a<br />

recurrent ~600kb copy number variant (CNV) at 16p11.2,<br />

occurring in approximately 1% <strong>of</strong> autism and 0.3% <strong>of</strong><br />

schizophrenia cases, as compared to 0.01% <strong>of</strong> the general<br />

population. We hypothesize that one or more <strong>of</strong> the 25 genes<br />

at this locus contribute to the neurodevelopmental phenotype<br />

observed in patients with psychiatric disorders. To determine<br />

how gene function is altered by this CNV, we analyzed<br />

genome wide expression data from Epstein Barr Virus (EBV)<br />

transformed Lymphoblast cell lines (LCL), <strong>of</strong> patients with<br />

autism or schizophrenia who have a de novo or inherited<br />

16p11.2. Using RNA expression pr<strong>of</strong>iling by Affymetrix<br />

Human Genome U133 Plus 2.0 chip, we examined differential<br />

cis and trans gene expression in individuals with 1, 2, or 3<br />

copies <strong>of</strong> the genomic region (6, 19, 16 respectively). Our<br />

results highlighted 140 genes located both within and outside<br />

<strong>of</strong> the mutation which expression correlates with genotype.<br />

Some <strong>of</strong> these genes play a role in development while others<br />

have been associated with psychiatric disorders. We are<br />

currently analyzing our list <strong>of</strong> 140 dysregulated genes to<br />

identify pathways and functions relevant to<br />

neurodevelopment, and psychiatric disorders. Data generated<br />

by this study will give insight into dosage sensitive genes<br />

within the risk variant, and may help pinpoint genes which are<br />

relevant to pathology <strong>of</strong> the psychiatric disorders associated<br />

with this region.<br />

O14 EPIGENETICS<br />

O14.1 INVESTIGATING GENOME-WIDE<br />

EPIGENETIC DIFFERENCES IN TWINS<br />

DISCORDANT FOR ALCOHOL DEPENDENCE<br />

C. Nymberg* (1), A. Lourdusamy (1), J. Mill (1), S.<br />

Desrivieres (1), R. Rose (2), J. Kaprio (3), D. Dick (4), G.<br />

Schumann (5)<br />

1. Social, Genetic and Developmental Psychiatry Centre 2.<br />

University <strong>of</strong> Indiana 3. University <strong>of</strong> Helsinki 4. Virginia<br />

Institute for Psychiatric and Behavioral Genetics 5. Social,<br />

Genetic and Developmental Psychiatry Centre<br />

* charlotte.nymberg@kcl.ac.uk<br />

Alcohol dependence (AD) is the result <strong>of</strong> interacting<br />

environmental and genetic factors. However, to date<br />

researchers have been unable to identify specific causal<br />

genetic or environmental risk factors underlying the disorder.<br />

Recent studies suggest that DNA methylation, or the silencing<br />

<strong>of</strong> gene expression, may play an important role for<br />

understanding psychiatric disorders such as AD. At least two<br />

factors influence DNA methylation: 1) environmental factors<br />

may produce epigenetic marks which speed up the process <strong>of</strong><br />

adaptation to a changing and less predictable environment; 2)<br />

genes may encode enzymes and proteins that help construct<br />

and maintain methylation-patterns. To control for the genetic<br />

components underlying DNA methylation, the current study<br />

adopted a twin design. The study aims to investigate DNA<br />

methylation patterns in blood samples from 36 monozygotic<br />

twins, diagnostically discordant for AD. To date psychiatric<br />

research has mainly taken a candidate gene approach to the<br />

investigation <strong>of</strong> DNA methylation. However, this focus may<br />

be too constrained. This study takes a genome-wide approach<br />

to investigating DNA methylation using a methylation<br />

sensitive array with 385.000 features covering promoter<br />

regions and CpG islands. Although the results are preliminary,<br />

they suggest that methylation differences related to psychiatric<br />

disorders such as alcohol dependence are worthy <strong>of</strong> further<br />

study.


O14.3 IDENTIFICATION OF IMPRINTED GENES<br />

CONTRIBUTING TO SPECIFIC BRAIN REGIONS<br />

USING HIGH THROUGHPUT SEQUENCING<br />

C. Barr (1), K. Wigg (1), E. Dempster (1), L. Gomez (1), Y.<br />

Feng (1), P. Monnier (1), R. Logan (1), J. Eubank (1)<br />

1. The Toronto Western Research Institute<br />

Current estimates suggest that there are around 100 imprinted<br />

genes, but this number may be as high as 600, with some<br />

imprinted in a tissue/cell type specific pattern and little is<br />

known <strong>of</strong> these genes. We followed up evidence that suggests<br />

imprinted genes influence the development <strong>of</strong> specific brain<br />

regions. We predicted that key genes expressed from the<br />

paternal genome will contribute to the development <strong>of</strong> the<br />

hypothalamus while key maternally expressed genes will<br />

influence the development <strong>of</strong> the neocortex. We developed a<br />

strategy using crosses and reverse crosses <strong>of</strong> mouse strains to<br />

determine the parent-<strong>of</strong>-origin <strong>of</strong> expressed genes. We used<br />

next generation sequencing <strong>of</strong> mRNA tags from the cortex and<br />

the thalamus/hypothalamus dissected from the F1 to identity<br />

tags differing in expression between the crosses. We<br />

identified tags that were differentially expressed between the<br />

crosses showing the same pattern <strong>of</strong> parent-<strong>of</strong>-origin effect in<br />

both brain regions including known imprinted genes (Meg3,<br />

Snrpn, Rian) and genes with no prior evidence for<br />

parent-<strong>of</strong>-origin effects (Pctk3, Klf10). We also observed<br />

genes that displayed a parent <strong>of</strong> origin effect in only one brain<br />

region with biallelic expression in the other region (Pon2,<br />

Itgb1bp1). Our results show the feasibility <strong>of</strong> using next<br />

generation sequencing <strong>of</strong> RNA as a genome-wide approach to<br />

identify novel imprinted genes as well as to identify tissue<br />

specific effects. The understanding <strong>of</strong> parent-<strong>of</strong>-origin effects<br />

in specific brain regions will make a significant contribution to<br />

our understanding <strong>of</strong> the role <strong>of</strong> imprinting in brain<br />

development and function.<br />

O14.4 EPIGENETIC DYSREGULATION OF HTR2A IN<br />

POST-MORTEM BRAIN OF PATIENTS WITH<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER<br />

H. Mostafavi-Abdolmaleky (1), S. Yaqubi (2), P.<br />

Papageorgis (2), S. Thiagalingam (3)<br />

1. Boston University, Department <strong>of</strong> Medicine and Iran<br />

University <strong>of</strong> <strong>Medical</strong> Sciences, Department <strong>of</strong> Psychiatry &<br />

Mental Health Research Center 2. Boston University,<br />

Department <strong>of</strong> Medicine, Genetics Programs 3. Boston<br />

University, Department <strong>of</strong> Medicine<br />

Introduction: Pharmacologic and genetic studies provided<br />

evidence for the involvement <strong>of</strong> HTR2A in schizophrenia<br />

(SCZ) and bipolar disorder (BD) pathogenesis. However,<br />

HTR2A polymorphisms have minimal effect in the<br />

pathogenesis <strong>of</strong> these diseases. Hypothesizing that epigenetic<br />

dysregulation <strong>of</strong> 5HTR2A may have a role in SCZ and BD we<br />

analyzed promoter DNA methylation <strong>of</strong> HTR2A in<br />

post-mortem brains. Method:DNA derived from dorsolateral<br />

frontal cortex <strong>of</strong> patients with SCZ and BD and normal<br />

controls (each 35) obtained from the Stanley <strong>Medical</strong><br />

Research Institute. HTR2A polymorphisms was determined<br />

using enzymatic restriction <strong>of</strong> PCR product. qRT-PCR was<br />

used to quantify gene expression level and bisulfite<br />

sequencing to assess DNA methylation level <strong>of</strong> CpGs at<br />

HTR2A promoter. Results: We found strong evidence for: 1)<br />

epigenetic fine tuning <strong>of</strong> HTR2A; 2) inverse correlation<br />

between promoter DNA methylation and gene expression<br />

levels; 3) higher expression <strong>of</strong> HTR2A in individuals carrying<br />

the G allele <strong>of</strong> -1438A/G polymorphism; 4) hypo-expression<br />

<strong>of</strong> HTR2A in SCZ; 5) DNA hypermethylation <strong>of</strong> cytosine<br />

located before -1438A/G polymorphic site (determined in<br />

individuals with the GG genotype) in SCZ and BD versus the<br />

controls; 6) correlation between methylation level <strong>of</strong> this<br />

cytosine and neighboring cytosines followed by guanine and<br />

several other cytosines followed by adenine which their<br />

methylation levels are inversely correlated with gene<br />

expression level. Conclusion: Aberrant DNA methylation <strong>of</strong><br />

CpG and CpA sites <strong>of</strong> the HTR2A promoter region may<br />

contribute to SCZ and BD pathogenesis, particularly in<br />

individuals carrying the G allele <strong>of</strong> -1438A/G polymorphism.


O14.5 ALLELIC SKEWING OF DNA METHYLATION<br />

IS WIDESPREAD ACROSS THE GENOME<br />

L. Schalkwyk (1), E. Meaburn (1), R. Smith (1), E. Dempster<br />

(1), R. Plomin (1), J. Mill* (1)<br />

1. Institute <strong>of</strong> Psychiatry, King's College London<br />

* j.mill@iop.kcl.ac.uk<br />

DNA methylation is a key epigenetic mechanism involved in<br />

the developmental regulation <strong>of</strong> gene expression. Across the<br />

majority <strong>of</strong> the genome, DNA methylation is assumed to be<br />

complementary on both alleles, although there are several<br />

exceptions, most notably in regions subject to genomic<br />

imprinting. Although mounting evidence supports the notion<br />

that differential allelic expression is widespread, the full extent<br />

<strong>of</strong> allele-specific DNA methylation (ASM) in the human<br />

genome has not been assessed. We present the first large-scale<br />

survey <strong>of</strong> the degree <strong>of</strong> allelic skewing <strong>of</strong> DNA methylation<br />

with the aim <strong>of</strong> identifying novel differentially methylated<br />

regions (DMRs) associated with either genomic imprinting or<br />

DNA sequence variation. We used high-resolution<br />

microarrays to quantitatively assess changes in normalized<br />

relative allele signals (RAS) for single nucleotide<br />

polymorphisms (SNPs) in amplicons covering 7.6% <strong>of</strong> the<br />

human genome following cleavage with a cocktail <strong>of</strong><br />

methylation-sensitive restriction enzymes (MSREs). Our<br />

approach detected clear examples <strong>of</strong> ASM in the vicinity <strong>of</strong><br />

known imprinted loci, highlighting the validity <strong>of</strong> the method,<br />

and selected findings were independently verified using<br />

bisulfite-mapping and gene expression analyses. In total 2,705<br />

(1.5%) <strong>of</strong> our informative SNPs demonstrated an average RAS<br />

change >0.10 following MSRE digestion. These data suggest<br />

that >35,000 such sites exist across the genome, and that some<br />

degree <strong>of</strong> ASM is a widespread phenomenon.<br />

O15 GENE ENVIRONMENT<br />

INTERACTIONS<br />

O15.1 THE INTERACTION OF VARIATIONS IN THE<br />

FKBP5 GENE AND ADVERSE LIFE EVENTS IN<br />

PREDICTING THE FIRST ONSET OF DEPRESSION<br />

DURING A TEN-YEAR FOLLOW-UP<br />

P. Zimmermann* (1), T. Brueckl (1), H. Pfister (1), E.<br />

Binder (1), M. Uhr (1), R. Lieb (2), F. Holsboer (1), M. Ising<br />

(1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry, Germany 2. University<br />

<strong>of</strong> Basel, Switzerland<br />

* pzimmer@mpipsykl.mpg.de<br />

Background: The co-chaperone FKBP5 <strong>of</strong> hsp-90 is an<br />

important modulator <strong>of</strong> glucocorticoid receptor (GR) function,<br />

the main receptor <strong>of</strong> the stress hormone system. By<br />

modulating this system, FKBP5 is in a key position to<br />

modulate gene-environment interactions (GxE) critical for the<br />

pathogenesis <strong>of</strong> major depression. In fact, associations<br />

between single nucleotide polymorphisms (SNPs) in the<br />

FKBP5 gene and susceptibility to mood disorders as well as<br />

interaction with early trauma to predict post traumatic stress<br />

disorder (PTSD) have been reported (Binder et al., 2004;<br />

2008). Aim:To explore GxE between variations in the FKBP5<br />

gene and adverse (separation and traumatic) events in<br />

predicting the first onset <strong>of</strong> a major depressive episode (MDE)<br />

in a ten-years prospective-longitudinal community survey.<br />

Methods:Analyses are based on 884 Caucasians (14-24 years<br />

at baseline) without baseline MDE who completed the<br />

ten-year follow-up. Data were assessed with the M-CIDI<br />

according to its DSM-IV algorithms. Five SNPs within the<br />

FKBP5 gene were selected for genotyping according to the<br />

results <strong>of</strong> previous studies (Binder et al., 2004, 2008; Ising et<br />

al., 2008). Results:While we did not observe genetic main<br />

effects, we found interactions between the five SNPs and<br />

traumatic (but not separation) events with the strongest effect<br />

for severe trauma (pcorrected: 1.8x10-3 to


O15.2 INTERACTION BETWEEN CHILDHOOD<br />

ADVERSITY AND COMT VAL158MET<br />

POLYMORPHISM IN PSYCHOTIC DISORDER<br />

H. Fisher* (1), C. Morgan (1), S. Luzi (1), M. Di Forti (1), P.<br />

Dazzan (1), C. Pariante (1), D. Collier (1), K. Aitchison (1), A.<br />

David (1), R. Murray (1), P. McGuffin (1)<br />

1. Institute <strong>of</strong> Psychiatry, King's College London<br />

* helen.fisher@iop.kcl.ac.uk<br />

Childhood adversity has frequently been associated with<br />

psychosis but not all exposed individuals develop this disorder<br />

suggesting that some may have a genetic vulnerability. The<br />

valine (Val) allele <strong>of</strong> the catechol-O-methyltransferase<br />

(COMT) Val158Met polymorphism has been linked to<br />

psychosis in those exposed to stress in adulthood. Therefore,<br />

we predicted this allele would also interact with childhood<br />

adversity in the development <strong>of</strong> clinically-relevant psychotic<br />

disorders. In a cross-sectional study, 161 first-presentation<br />

psychosis patients and 100 unaffected controls completed the<br />

Childhood Experience <strong>of</strong> Care and Abuse Questionnaire<br />

(CECA.Q) to determine experience <strong>of</strong> at least one adverse<br />

event before 17 years <strong>of</strong> age (physical or sexual abuse,<br />

parental separation or death, taken into care, disrupted living<br />

arrangements). Blood samples were genotyped for the COMT<br />

Val158Met polymorphism using polymerase chain reaction.<br />

Reported exposure to any adversity in childhood was<br />

significantly associated with being a psychosis case in this<br />

sample (unadjusted OR=2.80, 95% CI 1.66-4.74, p


O15.4 SEARCHING FOR GENES WITH<br />

ENVIRONMENTAL INTERACTION IN COMPLEX<br />

DISORDERS<br />

K. Merikangas*<br />

*merikank@mail.nih.gov<br />

O16 ALCOHOL & SUBSTANCE ABUSE<br />

O16.1 ASSOCIATION OF SUBSTANCE USE<br />

DISORDERS WITH CHILDHOOD TRAUMA BUT NOT<br />

AFRICAN GENETIC HERITAGE IN AN AFRICAN<br />

AMERICAN COHORT<br />

F. Ducci* (1), A. Roy (2), P. Shen (3), Q. Yuan (3), N. Yuan<br />

(4), C. Hodgkinson (3), L. Goldman (5), D. Goldman (3)<br />

1. Institute <strong>of</strong> Psychiatry, Kings College, London, UK<br />

2. The Social, Genetic, and Developmental Psychiatry Centre,<br />

Division <strong>of</strong> Psychological Medicine, Institute <strong>of</strong> Psychiatry;<br />

the Psychiatry Service, Department <strong>of</strong> Veterans Affairs, New<br />

Jersey VA 3. Laboratory <strong>of</strong> Neurogenetics, National Institute<br />

on Alcohol Abuse and Alcoholism, Bethesda, Md 4. Mel and<br />

Enid Zuckerman College <strong>of</strong> Public Health, University <strong>of</strong><br />

Arizona, Tucson 5. Department <strong>of</strong> Environmental Health<br />

Sciences, Johns Hopkins Bloomberg School <strong>of</strong> Public Health,<br />

Baltimore<br />

*francesca.ducci@kcl.ac.uk<br />

Objective: Genetic variation influences differential<br />

vulnerability to addiction within populations. However, it<br />

remains unclear whether differences in frequencies <strong>of</strong><br />

vulnerability alleles contribute to disparities between<br />

populations and to what extent ancestry correlates with<br />

differential exposure to environmental risk factors, including<br />

poverty and trauma. Method: The authors used 186<br />

ancestry-informative markers to measure African ancestry in<br />

407 addicts and 457 comparison subjects self-identified as<br />

African Americans. The reference group was 1,051<br />

individuals from the Human Genome Diversity Cell Line<br />

Panel, which includes 51 diverse populations representing<br />

most worldwide genetic diversity. Results: African Americans<br />

varied in degrees <strong>of</strong> African, European, Middle Eastern, and<br />

Central Asian genetic heritage. The overall level <strong>of</strong> African<br />

ancestry was actually smaller among cocaine, opiate, and<br />

alcohol addicts (proportion=0.76–0.78) than nonaddicted<br />

African American comparison subjects (proportion=0.81).<br />

African ancestry was associated with living in impoverished<br />

neighborhoods, a factor previously associated with risk. There<br />

was no association between African ancestry and exposure to<br />

childhood abuse or neglect, a factor that strongly predicted all<br />

types <strong>of</strong> addictions. Conclusions: These results suggest that<br />

African genetic heritage does not increase the likelihood <strong>of</strong><br />

genetic risk for addictions. They highlight the complex<br />

interrelation between genetic ancestry and social, economic,<br />

and environmental conditions and the strong relation <strong>of</strong> those<br />

factors to addiction. Studies <strong>of</strong> epidemiological samples<br />

characterized for genetic ancestry and social, psychological,<br />

demographic, economic, cultural, and historical factors are<br />

needed to better disentangle the effects <strong>of</strong> genetic and<br />

environmental factors underlying interpopulation differences<br />

in vulnerability to addiction and other health disparities.


O16.2 ASSOCIATIONS OF DRD1-D5, SLC18A2,<br />

SLC6A3, DDC, AND TH WITH ALCOHOL<br />

DEPENDENCE (AD) AND RELATED DISORDERS IN<br />

THE IRISH AFFECTED SIB PAIR STUDY OF<br />

ALCOHOL DEPENDENCE (IASPSAD)<br />

L. Hack* (1), G. Kalsi (1), F. Aliev (1), P. Kuo (2), C.<br />

Prescott (3), D. Patterson (4), D. Walsh (5), D. Dick (1), B.<br />

Riley (1), K. Kendler (1)<br />

1. Virginia Institute for Psychiatric and Behavioral Genetics,<br />

Department <strong>of</strong> Psychiatry, Virginia Commonwealth<br />

University, Richmond, VA, USA 2. Institute <strong>of</strong> Clinical<br />

Medicine, College <strong>of</strong> Medicine, National Cheng Kung<br />

University, Tainan, Taiwan 3. Department <strong>of</strong> Psychology,<br />

University <strong>of</strong> Southern California, Los Angeles, CA, USA<br />

4. Shaftesbury Square Hospital, Belfast, Northern Ireland,<br />

United Kingdom 5. Health Research Board, Dublin, Ireland<br />

* hacklm@vcu.edu<br />

The mesocorticolimbic dopaminergic system is central to the<br />

reward pathway. As a result, variation in dopamine genes may<br />

contribute to liability for AD and related disorders. We<br />

examined 10 dopamine genes (DRD1-D5, SLC18A2,<br />

SLC6A3, DDC, TH and COMT) for association with AD and<br />

alcohol-related traits in PLINK 1.05. The sample included 575<br />

independent cases selected from the IASPSAD sample and<br />

530 ethnically matched and screened controls. After data<br />

cleaning, 101 SNPs were available for analysis. Only SNPs<br />

surviving 5000 gene-based permutations are reported here. We<br />

reasoned that gene-based correction was a sufficiently<br />

conservative correction method because all selected genes<br />

have a priori evidence <strong>of</strong> association with AD and/or related<br />

phenotypes. For the dichotomous trait AD, we observed the<br />

strongest signal within DRD4 (rs12280580: P=0.003;<br />

OR=1.33) and a nominal signal within the dopamine<br />

transporter, SLC6A3 (rs27048: P=0.04; OR=0.83). Analysis<br />

<strong>of</strong> the quantitative alcohol-related traits (age at onset, initial<br />

sensitivity, tolerance, max24, and withdrawal) yielded<br />

association with initial sensitivity and DRD3 (p=0.002),<br />

withdrawal and DRD5 (p=0.002), and weaker associations<br />

with other traits. Because additional evidence indicates that<br />

polymorphisms in dopamine genes contribute to other<br />

externalizing phenotypes, we conducted secondary association<br />

analyses on 6 <strong>of</strong> these traits, including symptoms for alcohol<br />

dependence, antisocial personality disorder, attention<br />

deficit/hyperactivity disorder, conduct disorder, drug<br />

dependence, and scores for novelty seeking. We found that 7<br />

DA genes were associated with one or more externalizing<br />

phenotypes. Our study provides evidence that variation in<br />

dopamine genes contributes to AD as well as a range <strong>of</strong><br />

externalizing traits.<br />

O16.3 SPECIFIC AND NON-SPECIFIC GENETIC RISK<br />

FACTORS FOR ALCOHOL USE: ANALYSES OF<br />

DEVELOPMENTAL TRENDS AND<br />

GENE-ENVIRONMENT INTERACTIONS<br />

K. Kendler* (1), C. Prescott (2), D. Dick (1)<br />

1. Virginia Institute for Psychiatric and Behavioral Genetics 2.<br />

University <strong>of</strong> Southern California<br />

* Kendler@vcu.edu<br />

Although we know that the risk for alcohol dependence (AD)<br />

is influenced by genetic factors both specific to alcohol and<br />

for more general externalizing traits, we know little about how<br />

these two classes <strong>of</strong> genetic risk act during development.<br />

Furthermore, while environmental exposures can moderate the<br />

impact <strong>of</strong> genetic risk factors on alcohol consumption (AC),<br />

such studies have typically explored one environmental<br />

exposure during a single developmental period. Use life-event<br />

history retrospective data from 1,796 male twins from the<br />

Virginia Twin Register, we found that non-specific genetic<br />

risk for externalizing disorders impacts maximally on AC at<br />

ages 15-17 and then declines in importance. By contrast,<br />

specific genetic risks for AD have maximal and more enduring<br />

effects on AC starting at ages 18-21. We also find interactions<br />

between these genetic risks and 5 key environmental variables<br />

(parental monitoring, peer deviance, alcohol availability,<br />

prosocial behaviors and parental bonding) early in<br />

adolescence. As individuals age, the main effects <strong>of</strong> these<br />

environments on AC become more prominent while their<br />

interactions with genetic effects diminish. Genetic effects on<br />

AC appear to be much more environmentally sensitive at early<br />

developmental stages. Finally, in 5,072 male and female adult<br />

drinking twins from the Virginia Registry, we examined the<br />

relationship between genetic risk for a common factor made<br />

up <strong>of</strong> four measures <strong>of</strong> alcohol consumption during the period<br />

<strong>of</strong> lifetime heaviest drinking (frequency, quantity, maximum<br />

single-day consumption and frequency <strong>of</strong> drinking to<br />

intoxication) and AD. A structural twin model produced very<br />

high genetic correlations between the common factor and AD<br />

(+0.88 in females and +0.91 in males). These quantitative<br />

indices <strong>of</strong> alcohol consumption accurately reflected the<br />

genetic risk for AD.


O16.4 RESEQUENCING OF NICOTINIC RECEPTOR<br />

GENES AND ANALYSIS OF RARE AND COMMON<br />

VARIATIONS WITH NICOTINE DEPENDENCE<br />

J. Wessel* (1), D. Hinds (2), M. Kennemer (2), S. Pitts (2), R.<br />

Stokowski (2), D. Ballinger (2), J. McClure (3), L. Jack (1), J.<br />

Hardin (1), G. Swan (1), A. Bergen (1)<br />

1. SRI International 2. Perlegen Sciences 3. Group Health<br />

*jennifer.wessel@sri.com<br />

Genome-wide association studies have identified a few<br />

common single nucleotide polymorphisms (SNPs) associated<br />

with nicotine dependence but only a small portion <strong>of</strong> the<br />

heritability can be explained. We set out to determine if rare<br />

and common SNPs identified from sequencing <strong>of</strong> nicotinic<br />

receptor genes in a large sample <strong>of</strong> individuals could further<br />

explain the genetic basis <strong>of</strong> nicotine dependence. Exons <strong>of</strong> ten<br />

nicotinic receptor genes (CHRNA2-7, CHRNB1-4) were<br />

resequenced with 454 technology using DNA from saliva in<br />

448 European-Americans <strong>of</strong> a smoking cessation trial. We<br />

tested for associations <strong>of</strong> common and rare variations with the<br />

Fagerström Test <strong>of</strong> Nicotine Dependence (FTND) using<br />

standard linear and logistic regression methods for common<br />

SNPs and recently developed methods for rare SNPs, the<br />

cohort allelic sum test (CAST), the weighted sum statistic<br />

(WSS), and the multivariate distance matrix regression<br />

(MDMR) method. 166 SNPs were identified, 16.3% had<br />

reduced completion rate and were excluded from analyses,<br />

74.1% had minor allele frequencies .05), but the<br />

MDMR showed significant associations with all CHRNB2<br />

SNPs by allele sharing (p=0.004) or weighted allele frequency<br />

(p=0.003). CHRNB2 common and rare variants exhibited<br />

significant association to FTND in this sample.<br />

O17 DEMENTIA AND MEMORY<br />

O17.1 GENOME-WIDE ASSOCIATION STUDY OF<br />

ALZHEIMER'S DISEASE<br />

D. Harold* (1), R. Abraham (1), P. Hollingworth (1), R. Sims<br />

(1), A. Gerrish (1), M. Hamshere (1), S. Lovestone (2), C.<br />

Brayne (3), D. Rubinsztein (3), M. Gill (4), B. Lawlor (4), K.<br />

Morgan (5), P. Passmore (6), M. Nöthen (7), W. Maier (7), G.<br />

Livingston (8), N. Bass (8), A. Goate (9), S. Younkin (10), A.<br />

Al-Chalabi (11), R. Gwilliam (12), P. Deloukas (12), P.<br />

Holmans (1), M. O'Donovan (1), M. Owen (1), J. Williams (1)<br />

1. Cardiff University 2. Institute <strong>of</strong> Psychiatry 3. University <strong>of</strong><br />

Cambridge 4. Trinity College Dublin 5. University <strong>of</strong><br />

Nottingham 6. Queen's University Belfast 7 University <strong>of</strong><br />

Bonn 8. University College London 9. Washington University<br />

10. Mayo Clinic 11. Kings College London 12. Sanger<br />

Institute<br />

* wpcdhh@cf.ac.uk<br />

Alzheimer’s disease (AD) is the most common form <strong>of</strong><br />

dementia and is highly heritable but genetically complex.<br />

Neuropathologically, the disease is characterised by<br />

extracellular senile plaques containing β-amyloid and<br />

intracellular neur<strong>of</strong>ibrillary tangles containing<br />

hyperphosphorylated τ protein. Mutations <strong>of</strong> the amyloid<br />

precursor protein (APP) gene and the presenilin 1 and 2 genes<br />

(PSEN1, PSEN2) cause rare, Mendelian forms <strong>of</strong> the disease<br />

usually with an early-onset. However, in the more common<br />

form <strong>of</strong> AD, only apolipoprotein E (APOE) has been<br />

established unequivocally as a susceptibility gene. Aiming to<br />

identify novel AD loci, we undertook a two-stage<br />

genome-wide association study <strong>of</strong> Alzheimer’s disease<br />

involving over 16,000 individuals genotyped on Illumina<br />

platforms. In stage 1 (3,941 cases and 7,848 controls), we<br />

replicated the established association with the APOE locus<br />

and observed genome-wide significant association with SNPs<br />

at two novel independent loci (p


O17.2 IMPACT OF ALCOHOL ABUSE ON<br />

ALZHEIMER DISEASE<br />

M. Slifer (1), M. Cuccaro (1), J. Haines (2), M. Pericak-Vance<br />

(1)<br />

1. MIHG 2. Vanderbilt University<br />

Background: Efforts to understand the Alzheimer Disease<br />

(AD) phenotype have identified cardiovascular,<br />

neuropsychiatric, and lifestyle factors that are associated with<br />

the onset and course <strong>of</strong> AD. The relationship between alcohol<br />

use and cognitive impairment is controversial. While generally<br />

believed to be deleterious given its independent role in<br />

promoting cognitive decline, several studies have found<br />

moderate alcohol consumption to be protective.<br />

Understanding the impact <strong>of</strong> history <strong>of</strong> excessive alcohol use<br />

on individuals at risk for AD is critical given that alcohol use<br />

is common in the aging population. Methods: This<br />

retrospective study examined the relationship between remote<br />

history <strong>of</strong> alcohol substance abuse (SA) and age <strong>of</strong> onset in<br />

LOAD. These results were further analyzed as a function <strong>of</strong><br />

gender. Based on chart review <strong>of</strong> 294 cases, we identified 65<br />

unrelated participants with LOAD and a remote history <strong>of</strong> SA.<br />

History <strong>of</strong> SA was based on medical history data. All<br />

participants were in sustained remission. All participants had<br />

history, clinical presentation, and imaging (if available)<br />

consistent with classic AD; all are classified as probable AD.<br />

Age at onset (AAO) was defined on the basis <strong>of</strong> chart review<br />

<strong>of</strong> age at functional impairment. Results: Individuals with a<br />

history <strong>of</strong> SA and LOAD demonstrate a statistically<br />

significant earlier age <strong>of</strong> onset (69.9y, SD=6.8y) vs. those<br />

with LOAD and no history <strong>of</strong> SA (73.3y, SD=6.6y).<br />

Sex-stratified analysis revealed that males with LOAD were<br />

significantly more likely to have a history <strong>of</strong> SA vs. females<br />

(odds ratio=4.5 (95% CI 2.4-8.3)). Depression was also more<br />

prevalent among those with a history SA (69% vs. 40%).<br />

There was no statistically significant difference in APOE<br />

genotypes, or allele frquencies between those with SA and<br />

LOAD versus those with LOAD alone. Conclusons: These<br />

findings suggest that history <strong>of</strong> alcohol abuse may result in an<br />

earlier age <strong>of</strong> onset for individuals at risk for developing<br />

LOAD. There is no evidence that alcohol use results in AD<br />

pathophysiology. However, alcohol abuse may exacerbate a<br />

developing pattern <strong>of</strong> cognitive impairment in AD or<br />

compromise/reduce cognitive reserves resulting in an earlier<br />

onset. Given the tremendous health care costs associated with<br />

AD, a widely cited objective is to delay onset <strong>of</strong> disease.<br />

Factors which accelerate the course <strong>of</strong> the illness have the<br />

potential to result in significant economic burden. Similarly,<br />

quality <strong>of</strong> life for individuals and families is equally<br />

compromised with an earlier onset.<br />

O17.3 CNV GWAS FINDS RARE DELETIONS IN<br />

THYROID HORMONE GENE ASSOCIATED WITH<br />

ALZHEIMER’S DISEASE<br />

C. Lambert* (1), G. Linse Peterson (1)<br />

1. Golden Helix, Inc.<br />

* lambert@goldenhelix.com<br />

In numerous studies <strong>of</strong> Alzheimer’s disease (AD), thyroid<br />

hormones and the genes that regulate them have been<br />

associated with an increased risk <strong>of</strong> the disease. However,<br />

few, if any, genome-wide Copy Number Variation (CNV)<br />

studies have found associations with either rare or common<br />

variants for AD. The presence <strong>of</strong> rare CNVs has been<br />

suggested in the increased risk <strong>of</strong> schizophrenia and other<br />

psychiatric disorders, and we hypothesize CNVs could be<br />

associated with increased risk <strong>of</strong> AD as well. To this end, we<br />

performed a genome-wide association test to identify CNVs<br />

using log ratio data from an Affymetrix 500k array. The<br />

cohort consisted <strong>of</strong> approximately 1600 Caucasians, with<br />

roughly equal numbers <strong>of</strong> cases and controls (the average age<br />

<strong>of</strong> onset was 72 years). While no common CNVs appeared to<br />

be associated with AD, there were three femalecases with<br />

large overlapping deletions (116 kilobases to 230 kilobases),<br />

where the consensus region was 65 kilobases and spanned<br />

approximately one-third <strong>of</strong> a thyroid hormone gene, with the<br />

two larger deletions spanning the entire gene. The deletion<br />

did not exist in any controls or in any males. Studies have<br />

reported a link between thyroid hormones and AD,<br />

particularly in women, further supporting this finding.<br />

Replication studies are underway to confirm the variant’s<br />

association with AD; results will be reported at this meeting.


O17.4 COMMON VARIANTS ASSOCIATED WITH<br />

BRAIN ATROPHY IN ALZHEIMER’S DISEASE<br />

PATIENTS<br />

S. Furney (1), A. Simmons (1), K. Lunnon (1), G. Breen (1),<br />

A. Hodges (1), S. Lovestone (1)<br />

1. NIHR Biomedical Research Centre for Mental Health,<br />

Institute <strong>of</strong> Psychiatry, King’s College London.<br />

Alzheimer’s disease is the most common form <strong>of</strong> dementia<br />

and affects more than 400,000 people in the U.K. In this study<br />

we have tested the hypothesis that common genetic variation<br />

has an affect on brain atrophy in Alzheimer’s disease patients<br />

from the AddNeuroMed project. Over 300 European<br />

individuals were categorised as Control, Mild Cognitive<br />

Impairment (MCI) or Alzheimer’s disease (AD) by psychiatric<br />

assessment. The individuals had T1 structural MRI scanning<br />

from which entorhinal cortical thicknesses and whole brain<br />

volumes were calculated. In addition, blood was taken from<br />

the individuals and extracted DNA was run on the Illumina<br />

Human 610-Quad Beadchip. We used 494419 SNPs from<br />

across the genome (484465 autosomal) that had passed<br />

stringent quality control thresholds <strong>of</strong> minor allele frequency<br />

>0.05, a sample calling rate <strong>of</strong> 0.985, and a SNP calling rate <strong>of</strong><br />

0.99. Relatedness <strong>of</strong> the individuals was assessed by identity<br />

by descent estimates. Population stratification was conducted<br />

to identify any potential outlying individuals. HapMap Phase 2<br />

SNPs from CEU founders were imputed in the samples.<br />

A total <strong>of</strong> 239 individuals categorised as Control, Mild<br />

Cognitive Impairment or Alzheimer’s disease passed the<br />

genotyping quality control, were genotyped for ApoE and had<br />

scanning data that passed quality control protocols. Entorhinal<br />

cortical thicknesses and whole brain volumes were corrected<br />

for age, sex, disease status and ApoE4 dosage. An additive<br />

model was applied to investigate the interaction between<br />

individual allele dosage and MCI or AD status. A number <strong>of</strong><br />

SNPS were assigned p-values


POSTER<br />

ABSTRACTS


ADHD<br />

1 THE ROLE OF THE Y CHROMOSOME IN<br />

NEURODEVELOPMENTAL DISORDERS<br />

E. Stergiakouli* (1), H. Williams (1), K. Langley (1), A.<br />

Thapar (1), M. Owen (1)<br />

1. Department <strong>of</strong> Psychological Medicine, Cardiff University,<br />

Henry Wellcome Building, Heath Park CF14 4XN, Cardiff,<br />

U.K.<br />

*stergiakouliE@cardiff.ac.uk<br />

ADHD and schizophrenia are neurodevelopmental disorders<br />

that are more prevalent in males. Both disorders also show sex<br />

differences in age <strong>of</strong> onset or severity. The Y chromosome is<br />

potentially an important influence on male susceptibility to<br />

neuropsychiatric disorders. The way the Y chromosome can<br />

increase risk to neuropsychiatric disorders is directly or<br />

indirectly by interacting with autosomal genes expressed in<br />

the brain or with environmental factors. In addition, it can<br />

modify the disease phenotype by influencing cognitive<br />

performance. However, due to difficulties arising from the<br />

lack <strong>of</strong> recombination and widely accepted nomenclature, the<br />

Y chromosome has been largely excluded from genetic and<br />

genomic studies <strong>of</strong> neuropsychiatric disorders. In order to<br />

overcome this lack <strong>of</strong> knowledge, we chose to study the Y<br />

chromosome in a sample <strong>of</strong> 210 cases with ADHD, 310 cases<br />

with schizophrenia and 700 U.K. controls. In total, 11 Y<br />

chromosome markers were selected to represent the main Y<br />

chromosome haplogroups that are present in the U.K.<br />

according to data from the Y chromosome Consortium and<br />

personal communication with Y chromosome researchers.<br />

Statistical analysis <strong>of</strong> Y chromosome haplogroups revealed<br />

that although there was no significantly increased<br />

representation <strong>of</strong> any haplogroup in cases with ADHD or<br />

schizophrenia compared to controls, specific Y chromosome<br />

haplogroups were associated with higher performance and full<br />

scale IQ within the ADHD sample. Our results provide<br />

evidence that Y chromosome variation may modify ADHD<br />

phenotype by influencing cognitive performance.<br />

2 INFLUENCE OF DAT1 POLYMORPHISM ON<br />

STRIATAL ACTIVATION IN ADULT ADHD<br />

M. Hoogman* (1), E. Aarts (2), M. Zwiers (2), D.<br />

Slaats-Willemse (3), R. Cools (2), C. Kan (1), J. Buitelaar (1),<br />

B. Franke (4)<br />

1. Department <strong>of</strong> Psychiatry, Donders Institute for Brain,<br />

Cognition and Behaviour, Radboud University Nijmegen<br />

(<strong>Medical</strong> Centre), Nijmegen 2. Donders Centre for Cognitive<br />

Neuroimaging, Donders Institute for Brain, Cognition and<br />

Behaviour, Radboud University Nijmegen (<strong>Medical</strong> Centre),<br />

Nijmegen 3. Karakter Child and Adolescent Psychiatric<br />

Centre, Nijmegen, The Netherlands 4. Department <strong>of</strong> Human<br />

Genetics, Donders Institute for Brain, Cognition and<br />

Behaviour, Radboud University Nijmegen (<strong>Medical</strong> Centre),<br />

Nijmegen<br />

*m.hoogman@psy.umcn.nl<br />

Background: Attention Deficit/Hyperactivity Disorder<br />

(ADHD) is characterised by altered reward processing and<br />

striatal hypoactivity during reward tasks. The striatal reward<br />

processing centre has a high dopamine transporter (DAT)<br />

density. A VNTR in the 3’ UTR <strong>of</strong> DAT1, a risk factor for<br />

ADHD, alters gene expression and influences dopamine<br />

transporter density in the striatum and levels <strong>of</strong> synaptic<br />

dopamine. Our objective is to explore the influence <strong>of</strong> this<br />

polymorphism on functionality <strong>of</strong> the striatum in adult ADHD.<br />

Methods: So far, twenty-eight adults meeting criteria for<br />

DSM-IV ADHD and nineteen healthy subjects have been<br />

recruited. Recruitment is still ongoing. Participants perform a<br />

reward anticipation task inside a 1.5 Tesla MR scanner<br />

(adapted from Knutson et al., 2001). Statistical analysis<br />

compares the ventral striatal BOLD response <strong>of</strong> groups based<br />

on genotype (10/10-repeat homozygotes versus 9-repeat<br />

carriers). Results: Interim analysis already reveals more<br />

ventral striatal activity in 10/10-repeat homozygotes in ADHD<br />

patients (p=0.03). The exact opposite effect is found in healthy<br />

individuals (p=0.02; interaction genotype x disorder:<br />

p=0.002). Genotype or disorder does not affect behavioral<br />

measures. Discussion: These results could be explained by the<br />

added effect <strong>of</strong> an excess <strong>of</strong> striatal phasic dopamine (DA) in<br />

ADHD patients and less DAT availability in 9-repeat carriers<br />

to transport DA. Therefore an overdose <strong>of</strong> DA might lead to<br />

less striatal activation. This study can help to explain why the<br />

9-repeat allele <strong>of</strong> the DAT1 VNTR is a risk factor for adult<br />

ADHD (Franke et al, 2008).


3 META-ANALYSIS OF THE SLC6A3/DAT1 VNTR<br />

HAPLOTYPE IN PERSISTENT ADHD SUGGESTS<br />

DIFFERENTIAL INVOLVEMENT OF THE GENE IN<br />

CHILDHOOD AND PERSISTENT ADHD<br />

B. Franke* (1, 2), S. Johansson (3, 4), K. Lesch (5), P.<br />

Asherson (6), S. Faraone (7), E. Mick (8), J. Buitelaar (2), J.<br />

Haavik (3, 9), B. Cormand (11), J. Ramos-Quiroga (10), A.<br />

Reif (5)<br />

1. Department <strong>of</strong> Human Genetics, Donders Institute for<br />

Brain, Cognition and Behavior, Centre for Neuroscience,<br />

Radboud University Nijmegen <strong>Medical</strong> Centre, Nijmegen,<br />

The Netherlands 2. Department <strong>of</strong> Psychiatry, Donders<br />

Institute for Brain, Cognition and Behavior, Centre for<br />

Neuroscience, Radboud University Nijmegen <strong>Medical</strong> Centre,<br />

Nijmegen, The Netherlands 3. Department <strong>of</strong> Biomedicine,<br />

University <strong>of</strong> Bergen 4. Center for <strong>Medical</strong> Genetics and<br />

Molecular Medicine, Haukeland University Hospital, Bergen,<br />

Norway 5. Department <strong>of</strong> Psychiatry and Psychotherapy,<br />

Clinical and Molecular Psychobiology, and Interdisciplinary<br />

Center for Clinical Research (IZKF), University <strong>of</strong> Würzburg,<br />

Würzburg, Germany 6. MRC Social Genetic and<br />

Developmental Psychiatry, Institute <strong>of</strong> Psychiatry, Kings<br />

College London, London, UK 7. Departments <strong>of</strong> Psychiatry<br />

and Neuroscience and Physiology, SUNY Upstate <strong>Medical</strong><br />

University, Syracuse, New York, USA 8. Department <strong>of</strong><br />

Psychiatry, Massachusetts General Hospital and Harvard<br />

<strong>Medical</strong> School, Boston, MA, USA 9. Department <strong>of</strong><br />

Psychiatry, Haukeland University Hospital, Bergen, Norway<br />

10. Department de Genètica, Facultat de Biologia, Universitat<br />

de Barcelona, CIBER Enfermedades Raras, Instituto de Salud<br />

Carlos III, Barcelona, and Institut de Biomedicina de la<br />

Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain<br />

11. Department <strong>of</strong> Psychiatry, Hospital Universitari Vall<br />

d’Hebron, Barcelona, and Department <strong>of</strong> Psychiatry and Legal<br />

Medicine, Universitat Autònoma de Barcelona, Catalonia,<br />

Spain<br />

*b.franke@antrg.umcn.nl<br />

Attention-deficit/hyperactivity disorder (ADHD) is one <strong>of</strong> the<br />

most common neuropsychiatric disorders with a worldwide<br />

prevalence around 4-5% in children and 1-4% in adults.<br />

Although ADHD is highly heritable and familial risk may<br />

contribute most strongly to the persistent form <strong>of</strong> the disorder,<br />

there are few studies <strong>of</strong> the genetics <strong>of</strong> ADHD in adults. In the<br />

current paper, we present results <strong>of</strong> the International<br />

Multicentre Persistent ADHD Genetics CollaboraTion<br />

(IMpACT) that has been set up with the goal <strong>of</strong> performing<br />

research into the genetics <strong>of</strong> persistent ADHD. A<br />

meta-analysis <strong>of</strong> the SLC6A3/DAT1 gene in persistent ADHD<br />

was carried out in 1440 patients and 1769 controls from<br />

IMpACT and one earlier report. DAT1, encoding the<br />

dopamine transporter, is one <strong>of</strong> the most frequently studied<br />

genes in ADHD, though results have been inconsistent. A<br />

variable number tandem repeat polymorphism (VNTR) in the<br />

3’ untranslated region (UTR) <strong>of</strong> the gene and, more recently, a<br />

haplotype <strong>of</strong> this VNTR with another VNTR in intron 8 have<br />

been the target <strong>of</strong> most studies. Whereas the 10/10 genotype <strong>of</strong><br />

the 3’ UTR VNTR and the 10-6 haplotype <strong>of</strong> the two VNTRs<br />

are thought to be risk factors for ADHD in children, we found<br />

the 9/9 genotype (p=0.03) and the 9-6 haplotype (p=0.03)<br />

associated with persistent ADHD. A differential association <strong>of</strong><br />

DAT1 with ADHD in children and in adults might help<br />

explain the inconsistencies observed in earlier association<br />

studies. However, the data might also imply that DAT1 plays<br />

a modulatory rather than causative role in ADHD.<br />

4 FURTHER INVESTIGATION OF THE STEROID<br />

SULFATASE (STS) GENE AND ADHD: ASSOCIATION<br />

AND MOLECULAR ANALYSES<br />

K. Brookes* (1), Z. Hawi (2), J. Park (1), S. Scott (1), A.<br />

Kirley (2), E. Barry (2), D. Coghill (3), M. Gill (2), L. Kent<br />

(1)<br />

1. Bute <strong>Medical</strong> School, University <strong>of</strong> St Andrews, St<br />

Andrews, UK 2. Department <strong>of</strong> Psychiatry, Trinity Centre for<br />

Health Sciences, St James’s Hospital, Dublin, Ireland 3.<br />

Division <strong>of</strong> <strong>Medical</strong> Sciences, Centre for Neuroscience,<br />

University <strong>of</strong> Dundee, Scotland<br />

*kjb11@st-andrews.ac.uk<br />

Attention Deficit Hyperactivity Disorder (ADHD) is one <strong>of</strong><br />

the most heritable and prevalent behavioural disorders<br />

affecting 3-5% <strong>of</strong> school-aged children. Evidence from animal<br />

and human studies suggests steroid sulphatase (STS) may<br />

influence aspects <strong>of</strong> attention. Using tagging SNP’s we<br />

previously demonstrated that genetic variation in the STS gene<br />

is associated with ADHD. This study extends those initial<br />

findings by fine mapping the 5’ region <strong>of</strong> the STS gene using<br />

twelve SNP markers in an extended sample <strong>of</strong> 450 probands,<br />

<strong>of</strong> which the majority were male (90.1%) between the ages <strong>of</strong><br />

4 – 15 years. The majority <strong>of</strong> the sample was diagnosed as<br />

ADHD combined subtype (82.3%), with comorbidity rates <strong>of</strong><br />

46.9% for Oppositional Defiant Disorder, and 15.3% for<br />

Conduct Disorder. Additionally, STS protein and RNA were<br />

isolated from twenty-eight normal post-mortem brains in order<br />

to investigate the functionality <strong>of</strong> associated polymorphisms,<br />

by western blotting and mRNA gene expression. Results<br />

indicated that six SNP’s were associated with ADHD (with a<br />

significance value <strong>of</strong> P less than 0.05 for each individual<br />

marker) spanning the first two introns <strong>of</strong> the STS gene and one<br />

within a known alternative first exon. Haplotype analysis<br />

using the Gabriel method indicated two haplotype blocks, both<br />

<strong>of</strong> which were significantly associated with ADHD (P=0.001<br />

and P=0.004). mRNA expression and protein levels were not<br />

related to any <strong>of</strong> the significantly associated SNP markers.


5 CADHERIN 13 IS IMPLICATED IN ADULT ADHD,<br />

ITS ENDOPHENOTYPES AND CO-MORBID<br />

CONDITIONS<br />

A. Reif* (1), T. Nguyen (2), L. Weissflog (1), C. Jacob (1), M.<br />

Romanos (1), S. Liedel (1), H. Schäfer (2), A. Warnke (1), B.<br />

Cormand (3), J. Haavik (4), B. Franke (5), K. Lesch (1)<br />

1. University <strong>of</strong> Würzburg 2. University <strong>of</strong> Marburg 3.<br />

University <strong>of</strong> Barcelona 4. University <strong>of</strong> Bergen 5. University<br />

<strong>of</strong> Nijmegen<br />

*reif_a@klinik.uni-wuerzburg.de<br />

The atypical cadherin 13 (CDH13) has hitherto mainly gained<br />

attention regarding its function in the heart, where it has an<br />

important role in cell-cell adhesion. However, it is also<br />

expressed in the brain with a remarkable expression pattern,<br />

and with transcripts found in the prefrontal cortex, thalamus,<br />

the raphe nuclei and the locus coeruleus, CDH13 is in the<br />

center <strong>of</strong> monoaminergic neurotransmission. In the last year,<br />

several genome-wide association studies (GWAS) provided<br />

evidence that CDH13 is associated with different substance<br />

abuse disorders and quite interestingly, CDH13 was also<br />

amongst the top hits <strong>of</strong> two GWAS in ADHD (Lesch et al.,<br />

2008; Lasky-Su et al., 2008). Further evidence for a role <strong>of</strong><br />

CDH13 in ADHD was provided by a meta-analysis <strong>of</strong> ADHD<br />

linkage studies, where the locus containing CDH13 was the<br />

only one to reach genome-wide significance (Zhou et al.,<br />

2008). Thus, CDH13 is a highly relevant candidate gene for<br />

ADHD and related conditions. We have tested an association<br />

<strong>of</strong> CDH13 with adult ADHD (aADHD) interrogating 86<br />

tagging SNPs in a sample <strong>of</strong> 624 aADHD cases and 422<br />

controls. Thereby, several SNPs scattered across the gene<br />

were found to be associated with disease, co-morbid<br />

conditions such as depression, as well as NEO Extraversion<br />

and TPQ Novelty Seeking. 14 SNPs had p-values < 0.01, and<br />

within two haploblocks a significant association <strong>of</strong> a<br />

haplotype with aADHD was observed. To replicate this<br />

finding, we additionally tested a family-based sample<br />

consisting <strong>of</strong> 170 families with at least one child affected with<br />

ADHD. Again, seven SNPs in this sample were associated<br />

with disease. Joint analysis revealed that five SNPs and two<br />

haplotypes, each including one <strong>of</strong> the risk alleles, were<br />

associated with disease in both samples. Further replication<br />

was aimed for in the IMpACT sample consisting <strong>of</strong> more than<br />

1,000 additional aADHD cases from three European countries.<br />

Taken all findings together, CDH13 appears to be a highly<br />

relevant gene for disorders with the shared feature <strong>of</strong> deficient<br />

impulse control such as ADHD and substance abuse. The<br />

corresponding pathomechanism might include defective<br />

monoaminergic neurotransmission. This latter hypothesis has<br />

yet to be established, and respective analyses are underway.<br />

6 RIGHT-SIDED SPATIAL DIFFICULTIES IN ADHD<br />

DEMONSTRATED IN CONTINUOUS MOVEMENT<br />

CONTROL<br />

K. Johnson* (1), A. Dáibhis (2), C. Tobin (2), R. Acheson<br />

(2), A. Watchorn (2), A. Mulligan (3), E. Barry (2), J.<br />

Bradshaw (4), M. Gill (2), I. Robertson (2)<br />

1. Queen's University <strong>of</strong> Belfast 2. Trinity College Dublin 3.<br />

Mater Hospital 4. Monash University<br />

* k.johnson@qub.ac.uk<br />

Attention Deficit Hyperactivity Disorder (ADHD) is a<br />

prevalent neurodevelopmental psychiatric disorder<br />

characterised by hyperactivity, inattention and impulsiveness.<br />

Children with ADHD <strong>of</strong>ten show spatial attentional deficits,<br />

exhibiting a subtle rightwards bias, possibly due to<br />

dysfunction within the right hemisphere fronto-parietal<br />

network. Approximately 50% <strong>of</strong> children with ADHD also<br />

show signs <strong>of</strong> movement dysfunction. This movement<br />

dysfunction, however, has not been clearly described. Some<br />

areas <strong>of</strong> the brain involved in movement control (the<br />

fronto-striatal circuits) show anatomical and physiological<br />

dysfunction in ADHD. This study compared 31 children with<br />

and 31 children without ADHD on a movement kinematic task<br />

that tested hand-drawing movement precision, using the same<br />

procedure as Mattingley et al., (1994) and Phillips et al.,<br />

(1996). Participants used an electronic pen on a WACOM<br />

Intuos3 PTZ930 A4-sized digitizing tablet. The pen tip<br />

position was sampled as X and Y coordinates at 200 Hz, with<br />

an accuracy <strong>of</strong> ±0.25 mm. Data were acquired using the<br />

MovAlyzer® s<strong>of</strong>tware package, on a laptop computer. The<br />

task was to join targets <strong>of</strong> either 10 or 20mm diameter that<br />

were separated by a distance <strong>of</strong> 62.5 or 125mm. Constant<br />

error in the X and Y planes, peak vertical velocity and<br />

acceleration, and movement time were analysed. Apart from a<br />

significantly increased rate <strong>of</strong> acceleration across all<br />

conditions, the children with ADHD demonstrated no<br />

temporal difficulties with the task; rather they showed subtle<br />

spatial difficulties. The children with ADHD showed<br />

difficulties in accuracy <strong>of</strong> movement towards the right. They<br />

were less accurate in the X plane when moving towards the<br />

right-sided targets over the long distance. Greater variability<br />

in target accuracy was shown when moving towards the small<br />

target on the right side. These results suggest that the subtle<br />

spatial bias towards the right that has been demonstrated in<br />

ADHD on visual spatial tasks also extends into the movement<br />

domain.


7 COMMON VARIANTS IN THE TPH1 AND TPH2<br />

REGIONS ARE NOT ASSOCIATED WITH<br />

PERSISTENT ADHD IN 3559 INDIVIDUALS FROM<br />

FOUR EUROPEAN POPULATIONS<br />

S. Johansson* (1, 2), A. Halmøy (1), T. Mavroconstanti (1),<br />

K. Jacobsen (1), E. Landaas (1), A. Reif (3), C. Jacob (3), A.<br />

Boreatti-Hummer (3), S. Kreiker (3), K. Lesch (3), C. Kan (4),<br />

J. Kooij (5), L. Kiemeney (6), J. Buitelaar (4), B. Franke (4),<br />

M. Ribases (7), R. Bosch (7), M. Bayes (8), M. Casas (7), J.<br />

Ramos-Quiroga (7), B. Cormand (9), P. Knappskog (2), J.<br />

Haavik (1, 10)<br />

1. Department <strong>of</strong> Biomedicine, University <strong>of</strong> Bergen, Norway<br />

2. Center for <strong>Medical</strong> Genetics and Molecular Medicine,<br />

Haukeland University Hospital. Bergen, Norway 3.<br />

Department <strong>of</strong> Psychiatry, Psychosomatics and<br />

Psychotherapy, University <strong>of</strong> Würzburg, Würzburg, Germany<br />

4. Department <strong>of</strong> Psychiatry, Donders Institute for Brain,<br />

Cognition and Behavior, Radboud University Nijmegen<br />

<strong>Medical</strong> Centre, Nijmegen, The Netherlands 5. Parnassia,<br />

Psycho-<strong>Medical</strong> Centre, The Hague, The Netherlands 6.<br />

Department <strong>of</strong> Epidemiology & Biostatistics, Radboud<br />

University Nijmegen <strong>Medical</strong> Centre, Nijmegen, The<br />

Netherlands 7. Department <strong>of</strong> Psychiatry, Hospital<br />

Universitari Vall d’Hebron, Barcelona, Catalonia, Spain 8.<br />

Genes and Disease Program, Center for Genomic Regulation<br />

(CRG-UPF), Barcelona, Catalonia, Spain 9. Departament de<br />

Genètica, Facultat de Biologia, Universitat de Barcelona,<br />

Catalonia, Spain 10. Division <strong>of</strong> Psychiatry, Haukeland<br />

University Hospital, Bergen, Norway.<br />

* stefan.johansson@biomed.uib.no<br />

The tryptophan hydroxylase 1 and 2 (TPH1 and TPH2) genes<br />

which encode the rate-limiting enzymes in the serotonin<br />

biosynthesis have been implicated in several psychiatric<br />

disorders. Results from children with<br />

attention-deficit/hyperactivity disorder (ADHD) are<br />

conflicting, and little is known about TPH1 and TPH2 in adult<br />

ADHD patients. We therefore first genotype-tagged all<br />

common variants within both genes in a Norwegian sample <strong>of</strong><br />

451 patients with a diagnosis <strong>of</strong> adult ADHD and 584<br />

controls. Six <strong>of</strong> the SNPs were subsequently genotyped in<br />

three additional independent European Caucasian samples <strong>of</strong><br />

adult ADHD cases and controls from the International<br />

Multicenter persistent ADHD CollaboraTion (IMpACT).<br />

None <strong>of</strong> the SNPs reached formal study-wide significance in<br />

the total meta-analysis sample <strong>of</strong> 1636 cases and 1923<br />

controls, despite having a power <strong>of</strong> >80% to detect a variant<br />

conferring an OR=1.25 at P=0.001-level. However,<br />

rs17794760 located in TPH1 showed nominal significance<br />

[OR=0.84 (0.71-1.00), P=0.05]. In conclusion, in the single<br />

largest ADHD genetic study <strong>of</strong> TPH1 and TPH2 variants<br />

presented to date (n=3559 individuals) we did not find<br />

consistent evidence for a major effect <strong>of</strong> common genetic<br />

variants on persistent ADHD.<br />

8 CASE-CONTROL STUDY OF SIX GENES<br />

ASYMMETRICALLY EXPRESSED IN THE TWO<br />

CEREBRAL HEMISPHERES: ASSOCIATION OF<br />

BAIAP2 WITH ADHD<br />

M. Ribasés* (1), R. Bosch (1), A. Hervás (2), J.<br />

Ramos-Quiroga (1), C. Sánchez-Mora (1), A. Bielsa (1), X.<br />

Gastaminza (1), S. Guijarro-Domingo (2), M. Nogueira (1), N.<br />

Gómez-Barros (1), S. Kreiker (3), S. Groß-Lesch (3), C. Jacob<br />

(3), K. Lesch (3), A. Reif (3), S. Johansson (4), K. von Plessen<br />

(5), P. Knappskog (6), J. Haavik (4), X. Estivill (7), M. Casas<br />

(1), M. Bayés (7), B. Cormand (8)<br />

1. Department <strong>of</strong> Psychiatry, Hospital Universitari Vall<br />

d’Hebron, Barcelona, Catalonia, Spain 2. Child and<br />

Adolescent Mental Health Unit, Hospital Mútua de Terrassa,<br />

Barcelona, Spain 3. Department <strong>of</strong> Psychiatry,<br />

Psychosomatics and Psychotherapy, University <strong>of</strong> Würzburg,<br />

Füchsleinstr. Würzburg, Germany 4. Department <strong>of</strong><br />

Biomedicine, University <strong>of</strong> Bergen, Norway 5. Division <strong>of</strong><br />

Psychiatry, Haukeland University Hospital, Bergen, Norway<br />

6. Center <strong>of</strong> <strong>Medical</strong> Genetics and Molecular Medicine,<br />

Haukeland University Hospital, Norway 7. Genes and Disease<br />

Program, Center for Genomic Regulation (CRG-UPF,)<br />

Barcelona, Spain 8. Departament de Genètica, Facultat de<br />

Biologia, Universitat de Barcelona, Spain<br />

* marta.ribases@gmail.com<br />

Attention-deficit hyperactivity disorder (ADHD) is a<br />

childhood-onset neuropsychiatric disease that persists into<br />

adulthood in at least 30% <strong>of</strong> ADHD children. Different lines<br />

<strong>of</strong> evidence suggest that abnormal left-right brain asymmetries<br />

in ADHD patients may be involved in a variety <strong>of</strong><br />

ADHD-related cognitive processes. Although mechanisms<br />

underlying cerebral lateralization are unknown, left-right<br />

cortical asymmetry in humans has been associated with<br />

transcriptional asymmetry at embryonic stages and a number<br />

<strong>of</strong> genes differentially expressed between hemispheres have<br />

been identified. We selected six functional candidate genes<br />

showing at least 1.9-fold differential expression between<br />

hemispheres (BAIAP2, DAPPER1, LMO4, NEUROD6,<br />

ATP2B3 and ID2) and performed a case-control analysis in an<br />

initial Spanish sample <strong>of</strong> 587 ADHD patients (270 adults and<br />

317 children) and 587 controls. The single- and<br />

multiple-marker analysis provided evidence for a contribution<br />

<strong>of</strong> BAIAP2 to adulthood ADHD (P=0.0026, OR=1.69<br />

(1.20-2.44) and P=0.0016, OR=1.64 (1.20-2.22), respectively).<br />

We then tested BAIAP2 for replication in two independent<br />

adult samples from Germany (639 ADHD patients and 612<br />

controls) and Norway (417 ADHD cases and 469 controls)<br />

contributed by the International Multicentre Persistent ADHD<br />

CollaboraTion (IMpACT). While no significant results were<br />

observed in the Norwegian sample, we replicated the initial<br />

association between BAIAP2 and adulthood ADHD in the<br />

German population (P=0.0062; OR=1.21 (1.03-1.42)). Our<br />

results support the participation <strong>of</strong> BAIAP2 in the continuity<br />

<strong>of</strong> ADHD across lifespan, at least in some <strong>of</strong> the populations<br />

analyzed, and suggest that genetic factors potentially<br />

influencing abnormal cerebral lateralization may be involved<br />

in this neurodevelopmental disorder.


9 META-ANALYSIS OF BRAIN-DERIVED<br />

NEUROTROPHIC FACTOR P.VAL66MET IN ADULT<br />

ADHD IN FOUR EUROPEAN POPULATIONS<br />

C. Sánchez-Mora* (1), M. Ribasés (1), J. Ramos-Quiroga<br />

(1), M. Casas (2), R. Bosch (2), A. Boreatti-Hümmer (3), M.<br />

Heine (3), C. Jacob (3), K. Lesch (3), O. Fasmer (4), P.<br />

Knappskog (5), J. Kooij (6), C. Kan (7), J. Buitelaar (7), E.<br />

Mick (8), P. Asherson (9), S. Faraone (10), B. Franke (7), S.<br />

Johansson (5), J. Haavik (4), A. Reif (4), M. Bayés (11), B.<br />

Cormand (12)<br />

1. Department <strong>of</strong> Psychiatry, Hospital Universitari Vall<br />

d’Hebron, Barcelona, Catalonia, Spain 2. Hospital Universitari<br />

Vall d’Hebron, Barcelona, Catalonia, Spain 3. Department <strong>of</strong><br />

Psychiatry, Psychosomatics and Psychotherapy, University <strong>of</strong><br />

Würzburg, Füchsleinstr. 15, 97080 Würzburg, Germany 4.<br />

Division <strong>of</strong> Psychiatry, Haukeland University Hospital,<br />

Bergen, Norway 5. Center <strong>of</strong> <strong>Medical</strong> Genetics and Molecular<br />

Medicine, Haukeland University Hospital, Norway 6. PsyQ,<br />

Psycho-<strong>Medical</strong> Programs, Program Adult ADHD, The<br />

Hague, The Netherlands 7. Department <strong>of</strong> Psychiatry,<br />

Radboud University Nijmegen <strong>Medical</strong> Centre, Nijmegen,<br />

The Netherlands 8. Department <strong>of</strong> Psychiatry, Massachusetts<br />

General Hospital and Harvard <strong>Medical</strong> School, Boston, MA,<br />

USA 9. Social, Genetic, and Developmental Psychiatry<br />

Centre, Institute <strong>of</strong> Psychiatry, King’s College London,<br />

London, UK 10. Department <strong>of</strong> Neuroscience, SUNY Upstate<br />

<strong>Medical</strong> University, Syracuse, New York 11. Genes and<br />

Disease Program, Center for Genomic Regulation<br />

(CRG-UPF,) Barcelona, Catalonia, Spain 12. Departament de<br />

Genètica, Facultat de Biologia, Universitat de Barcelona,<br />

Catalonia, Spain<br />

* crissanchez@ir.vhebron.net<br />

Attention-deficit hyperactivity disorder (ADHD) is a<br />

multifactorial, neurodevelopmental disorder that <strong>of</strong>ten persists<br />

into adolescence and adulthood and is characterized by<br />

inattention, hyperactivity and impulsiveness. Before the<br />

advent <strong>of</strong> the first genome-wide association studies in ADHD,<br />

genetic research had mainly focused on candidate genes<br />

related to the dopaminergic and serotoninergic systems,<br />

although several other genes had also been assessed.<br />

Pharmacological data, analysis <strong>of</strong> animal models and<br />

association studies suggest that Brain-Derived Neurotrophic<br />

Factor (BDNF) is also a strong candidate gene for ADHD.<br />

Several polymorphisms in BDNF have been reported and<br />

studied in psychiatric disorders but the most frequent is the<br />

p.Val66Met (rs6265G>A) single nucleotide polymorphism<br />

(SNP), with functional effects on the intracellular trafficking<br />

and secretion <strong>of</strong> the protein. To deal with the inconsistency<br />

raised among different case–control and family-based<br />

association studies regarding the p.Val66Met contribution to<br />

ADHD, we performed a meta-analysis <strong>of</strong> published as well as<br />

unpublished data from four different centres that are part <strong>of</strong><br />

the International Multicentre Persistent ADHD CollaboraTion<br />

(IMpACT). A total <strong>of</strong> 1,445 adulthood ADHD patients and<br />

2,247 sex-matched controls were available for the study. No<br />

association between the p.Val66Met polymorphism and<br />

ADHD was found in any <strong>of</strong> the four populations or in the<br />

pooled sample. The meta-analysis also showed that the overall<br />

gene effect for ADHD was not statistically significant when<br />

gender or comorbidity with mood disorders were considered.<br />

Despite the potential role <strong>of</strong> BDNF in ADHD, our data do not<br />

support the involvement <strong>of</strong> p.Val66Met in the pathogenesis <strong>of</strong><br />

this neuropsychiatric disorder.<br />

10 EVIDENCE THAT GENETIC VARIATION IN THE<br />

OXYTOCIN RECEPTOR GENE INFLUENCES SOCIAL<br />

COGNITION SCORES IN ADHD<br />

J. Park* (1), G. Vetuz (1), C. Toye (1), E. Barry (2), A.<br />

Kirley (2), Z. Hawi (2), K. Brookes (2), M. Gill (2), L. Kent<br />

(1)<br />

1. Bute <strong>Medical</strong> School, University <strong>of</strong> St Andrews, St<br />

Andrews, Scotland 2. Department <strong>of</strong> Psychiatry, Trinity<br />

Centre for Health Sciences, St James’s Hospital, Dublin,<br />

Ireland<br />

* jp81@st-andrews.ac.uk<br />

Attention Deficit Hyperactivity Disorder (ADHD) is a highly<br />

heritable, common disorder <strong>of</strong> childhood. Recent evidence<br />

demonstrates that ADHD and autistic traits share some genetic<br />

liability, and both children with ADHD and autism have social<br />

cognition deficits. Evidence from animal and human studies<br />

implicates the oxytocin system in the development <strong>of</strong> social<br />

cognition and previous association studies have reported<br />

significant findings for genetic variation in the oxytocin<br />

receptor (OXTR) gene with autism. This study therefore<br />

investigated whether polymorphisms in the OXTR gene were<br />

associated with either the ADHD phenotype or social<br />

cognition traits in ADHD probands.This study employed a<br />

family association sample consisting <strong>of</strong> 450 probands, aged 4<br />

– 15 years, <strong>of</strong> which 90.1% were male. The majority <strong>of</strong> the<br />

sample was diagnosed as ADHD combined subtype (82.3%),<br />

with 46.9% comorbid for Oppositional Defiant Disorder, and<br />

15.3% for Conduct Disorder. Additionally, 119 <strong>of</strong> the<br />

probands completed the Social and Communication Disorders<br />

Checklist (SCDC), with higher scores indicating poorer social<br />

cognition. No statistical significant difference in SCDC score<br />

between males and females was found. 5 single nucleotide<br />

polymorphisms (SNP’s) in the OXTR gene previously<br />

investigated in autism susceptibility were genotyped. No<br />

significant associations with the ADHD phenotype were found<br />

but a significant association between rs53576 genotype groups<br />

and SCDC scores was discovered (F= 4.566, p= 0.013).<br />

Post-hoc tests demonstrated the G allele <strong>of</strong> rs53576 to be<br />

associated with poorer social cognition. This SNP lies in an<br />

intronic 3 haplotype previously associated with autism.<br />

Further investigation <strong>of</strong> these findings is warranted.


11 GENOMEWIDE ASSOCIATION SCAN OF<br />

REACTION TIME DATA IN COMBINED TYPE ADHD<br />

A. Vasquez (1), P. Asherson* (2), A. Wood (2), N. Rommelse<br />

(1), F. Rijsdijk (2), B. Franke (1), K. Zhou (2), T.<br />

Banaschewski (3), A. Rothenberger (4), R. Oades (5), M. Gill<br />

(6), R. Ebstein (7), H. Roeyers (8), I. Manor (9), A. Miranda<br />

(10), F. Mulas (10), H. Steinhausen (11), E. Sonuga-Barke<br />

(12), J. Buitelaar (1), J. Sergeant (13), S. Faraone (14), J.<br />

Kuntsi (2)<br />

1. Department <strong>of</strong> Psychiatry, Radboud University, Nimjmegan<br />

<strong>Medical</strong> Centre, Nijmegan, Netherlands 2. MRC Social<br />

Genetic and Developmental Psychiatry, Institute <strong>of</strong><br />

Psychiatry, Kings College London, London, UK 3.<br />

Department <strong>of</strong> child and adolescent psychiatry and<br />

psychotherapy, University <strong>of</strong> Mannheim, Mannheim,<br />

Germany 4. Child and adolescent psychiatry, University <strong>of</strong><br />

Goettingen, Goettingen, Germany 5. University Clinic for<br />

child and adolescent psychiatry, Essen, Germany 6.<br />

Department <strong>of</strong> Psychiatry, Trinity Centre for Health Sciences,<br />

Dublin, Ireland 7. Herzog Memorial Hospital, Jerusalem,<br />

Israel 8. Department <strong>of</strong> Experimental Clinical Health<br />

Psychology, Ghent University, Ghent, Belgium 9. ADHD<br />

clinic, Geha Mental Health Center, Petak-Tikvah, Israel 10.<br />

Department <strong>of</strong> developmental and educational psychology,<br />

University <strong>of</strong> Valencia, Valencia, Spain 11. Department <strong>of</strong><br />

child and adolescent psychiatry, University <strong>of</strong> Zurich, Zurich,<br />

Switzerland 12. Institute for disorders <strong>of</strong> impulsivity and<br />

attention, University <strong>of</strong> Southampton 13. Vrije Universiteit, de<br />

Boelelaan, Amsterdam, Netherlands 14. Departments <strong>of</strong><br />

Psychiatry, Neuroscience and Physiology, SUNY Upstate<br />

<strong>Medical</strong> University, Syracuse, United States<br />

* p.asherson@iop.kcl.ac.uk<br />

Results from multivariate familial model fitting on a large<br />

sample <strong>of</strong> ADHD and control sibling-pairs indicate the<br />

presence <strong>of</strong> two familial cognitive impairment factors (Kuntsi<br />

et al., in preparation). The larger factor, reflecting 70% <strong>of</strong> the<br />

familial variance with ADHD, captured all familial influences<br />

on mean reaction time (RT) and RT variability. The second<br />

factor, reflecting 20% <strong>of</strong> the familial variance with ADHD,<br />

captured all familial influences on omission errors on the<br />

go/no-go task and 60% <strong>of</strong> those on commission errors. These<br />

findings suggest partially separable genetic influences that<br />

contribute to the heritability <strong>of</strong> ADHD; RT variables represent<br />

important indices <strong>of</strong> underlying genetic liability. We therefore<br />

adopted a genome-wide scan approach to search for genes<br />

associated with RT measures. The sample consisted <strong>of</strong> two<br />

datasets from the International ADHD Multicentre Genetics<br />

(IMAGE) project: the 8-team collaborative dataset derived RT<br />

data from go/no-go and 4-choice reaction time tasks; the<br />

Dutch dataset derived RT data from the Stop task. To evaluate<br />

whether the two datasets could be combined we used Full<br />

Information Maximum Likelihood Estimation to estimate<br />

phenotypic (0.73–0.75) and familial (0.91–0.81) correlations<br />

for mean RT and RT variability, respectively, between the<br />

two-datasets. The final sample consists <strong>of</strong> around 500 ADHD<br />

combined type probands plus their siblings for whom we had<br />

both genomewide scan and RT data. Missing sibling GWAS<br />

data is imputed using known IBD information from linkage<br />

scan data. Further studies will aim to replicate these findings<br />

through international collaborations <strong>of</strong> RT data in ADHD.<br />

12 INVESTIGATION OF CANDIDATE GENE<br />

INTERACTIONS INFLUENCING RISK FOR ADHD<br />

R. Segurado* (1), M. Bellgrove, M. Gill (2), Z. Hawi (2)<br />

1. Trinity College Dublin 2. T.C.D.<br />

* rsegurdo@tcd.ie<br />

A number <strong>of</strong> genes with function related to synaptic<br />

neurochemistry have been associated with an<br />

Attention-Deficit/Hyperactivity Disorder. However,<br />

susceptibility to the development <strong>of</strong> common psychiatric<br />

disorders by single variants acting alone, have so far only<br />

explained a small proportion <strong>of</strong> the heritability <strong>of</strong> the<br />

phenotype. It has been postulated that the unexplained “dark<br />

heritability” may at least in part be due to epistatic effects,<br />

which would explain the small observed main effects and the<br />

difficulties in replication <strong>of</strong> positive findings. We undertook a<br />

comprehensive exploration <strong>of</strong> pair-wise interactions between<br />

150 genetic variants in 25 candidate genes involved in<br />

monoaminergic catabolism, anabolism, release, re-uptake and<br />

signal transmission, in a sample <strong>of</strong> 177 parent-affected child<br />

trios using a case-only design. Marker-pairs showing large<br />

effects and statistical significance were further explored with a<br />

case-pseudocontrol design using conditional logistic<br />

regression. We detected a number <strong>of</strong> moderate interaction<br />

odds ratios (1.7 – 2.8), including between markers in the<br />

ADRA1A and HTR2A genes, and markers in the HTR1B and<br />

SLC18A2 genes. These effects are no larger than you would<br />

expect by chance under the assumption <strong>of</strong> independence <strong>of</strong> all<br />

pair-wise relations, however independence is unlikely.<br />

Furthermore, the size <strong>of</strong> these effects is <strong>of</strong> interest and<br />

attempts to replicate these results in other samples are<br />

anticipated.


13 PHARMACOGENETICS ANALYSIS BETWEEN<br />

THE DOPAMINE TRANSPORTER GENE AND<br />

METHYLPHENIDATE RESPONSE IN CHILDREN<br />

WITH ATTENTION-DEFICIT/HYPERACTIVITY<br />

DISORDER<br />

J. Genro* (1), C. Zeni (1), G. Polanczyk (1), T. Roman (1), R.<br />

Luis (1), M. Hutz (1)<br />

1. UFRGS<br />

* juliagenro@hotmail.com<br />

The gene DAT1 (SLC6A3) is the main candidate in<br />

pharmacogenetics studies with ADHD because this transporter<br />

is the major site <strong>of</strong> methylphenidate (MPH) action. All<br />

association studies between DAT1 and MPH response have<br />

focused in the 3’ VNTR <strong>of</strong> the gene presenting conflicting<br />

results. The objective in this study is to verify if the variant<br />

-839 C/T (rs2652512) located in the promoter region <strong>of</strong> the<br />

gene is associated with response to MPH. A sample <strong>of</strong> 200<br />

children from an ADHD outpatient clinic was genotyped for<br />

the promoter polymorphism. The outcome measure was the<br />

parent-rated oppositional subscale <strong>of</strong> the Swanson, Nolan and<br />

Pelham Scale - version IV. The scale was applied by child<br />

psychiatrists blinded to genotype at baseline and in the first<br />

and third months <strong>of</strong> treatment. Data were analyzed using an<br />

ANOVA model for repeated measures. Patients homozygous<br />

for the T allele had a greater reduction in symptoms <strong>of</strong><br />

opposition (p = 0.003) and inattention (p = 0.004) compared<br />

with patients with C allele over the 3 months <strong>of</strong> treatment.<br />

These results suggest that response to MPH in children with<br />

ADHD is influenced by genotype <strong>of</strong> the polymorphism -839<br />

C/T <strong>of</strong> the DAT1 gene. The DAT1 promoter region may<br />

influence response to treatment with MPH as well as<br />

susceptibility to ADHD.<br />

14 LACK OF ASSOCIATION BETWEEN THREE DAT1<br />

POLYMORPHISMS AND RESPONSE TO<br />

METHYLPHENIDATE IN ADULTS WITH ADHD<br />

V. Contini* (1), M. Victor (2), G. Bertuzzi (1), E. Grevet (3),<br />

C. Salgado (3), P. Belmonte-de-Abreu (2)<br />

1. Department <strong>of</strong> Genetics, Instituto de Biociências,<br />

Universidade Federal do Rio Grande do Sul 2. Department <strong>of</strong><br />

Psychiatry, Faculdade de Medicina, Universidade Federal do<br />

Rio Grande do Sul 3. Adult ADHD Outpatient Clinic, Hospital<br />

de Clínicas de Porto Alegre<br />

* veronica.contini@gmail.com<br />

Attention-deficit hyperactivity disorder (ADHD) is a highly<br />

heritable disorder manifesting itself in symptoms <strong>of</strong><br />

inattention and/or hyperactivity/impulsivity that result in<br />

impairment in multiple domains <strong>of</strong> adaptive functioning. The<br />

pharmacotherapy is central in ADHD treatment and the main<br />

pharmacological drug prescribed is methylphenidate<br />

hydrochloride (MPH). Pharmacogenetics studies <strong>of</strong> MPH<br />

response in ADHD have mainly focused on the 40-bp variable<br />

number <strong>of</strong> tandem repeat (VNTR) in the 3’ untranslated region<br />

(3’-UTR) <strong>of</strong> the DAT1. Most studies were performed in<br />

children samples, and obtained conflicting findings. Only two<br />

studies assessed the 3´VNTR in adult samples, one with<br />

positive and the other with negative findings. In the present<br />

study, we investigate three polymorphisms in the DAT1 gene<br />

(3´VNTR; VNTR in intron 8 and -839 C>T), and their<br />

possible role in the therapeutic response to MPH treatment in<br />

a sample <strong>of</strong> 171 adults with ADHD. The diagnostic<br />

procedures followed the DSM-IV criteria and the outcome<br />

measures were the SNAP-IV subscales, applied at the<br />

beginning and after the 30th day <strong>of</strong> treatment. The<br />

polymorphisms were analyzed separately and considering<br />

haplotypes comprising the 3´VNTR and the VNTR in intron 8.<br />

No significant effects <strong>of</strong> any DAT1 polymorphisms or<br />

haplotypes on the response to MPH were found. The current<br />

report is the second to demonstrate no difference in MPH<br />

response based upon DAT1 genotypes in adults with ADHD.<br />

If there is a significant effect <strong>of</strong> DAT1 polymorphisms in adult<br />

ADHD, this effect is likely to be very small.


15 FAMILY-BASED GENOME-WIDE ASSOCIATION<br />

STUDY OF ATTENTION-DEFICIT/HYPERACTIVITY<br />

DISORDER<br />

E. Mick* (1), A. Todorov (2), S. Loo (3), X. Hu (4), B.<br />

Dechairo (4), S. Hall (4), S. Nelson (3), C. Shtir (3), S.<br />

Smalley (3), R. Todd (2), B. Neale (1), S. Faraone (5)<br />

1. Massachusetts General Hospital 2. Washington University<br />

School <strong>of</strong> Medicine 3. UCLA 4. Molecular Medicine, Pfizer<br />

Inc. 5. SUNY Upstate <strong>Medical</strong> University<br />

* emick1@partners.org<br />

Genes likely play a substantial role in the etiology <strong>of</strong><br />

attention-deficit hyperactivity disorder (ADHD) but the<br />

genetic architecture <strong>of</strong> the disorders is unknown and prior<br />

genome-wide association studies have not identified a<br />

genome-wide significant association. We have conducted a<br />

third, independent multi-site GWAS <strong>of</strong> DSM-IV-TR ADHD<br />

using the Illumina Human1Mand Human1M-Duo BeadChip<br />

platforms. After applying quality control filters association<br />

with ADHD was tested with 835,136 SNPs in 735 DSM-IV<br />

ADHD trios from in 733 families. Our smallest p-value<br />

(6.7E-07) did not reach the threshold for genome-wide<br />

statistical significance (5.0E-08) but one <strong>of</strong> the 20 most<br />

significant associations was located in a candidate gene <strong>of</strong><br />

interest for ADHD, (SLC9A9, rs9810857, p=6.4E-6). We also<br />

conducted gene-based tests <strong>of</strong> candidate genes identified in the<br />

literature and found additional evidence <strong>of</strong> association with<br />

SLC9A9. We and our colleagues in the Psychiatric GWAS<br />

Consortium are working to pool together GWAS samples to<br />

establish the large data sets needed to follow-up on these<br />

results and to identify genes for ADHD and other disorders.<br />

16 ATTACHMENT DIFFICULTIES IN CHILDREN<br />

WITH ATTENTION DEFICIT HYPERACTIVITY<br />

DISORDER – A PHENOTYPE ANALYSIS STUDY<br />

FROM THE INTERNATIONAL MULTICENTRE<br />

ADHD GENETICS STUDY<br />

A. Mulligan* (1), F. Motala (2), R. Anney (3), M. Gill (3)<br />

1. University College Dublin 2. Mater Misericordiae<br />

University Hospital 3. Trinity College Dublin<br />

* aisling.mulligan@ucd.ie<br />

Introduction: Children with attachment difficulties such as<br />

reactive attachment disorder may present with hyperactivity.<br />

This study tested the hypothesis that attachment difficulties<br />

may occur in ADHD and may exacerbate symptoms <strong>of</strong><br />

ADHD. Aims: (1) To measure attachment difficulties in<br />

children with ADHD and ascertain if children with ADHD and<br />

attachment difficulties have more severe symptoms <strong>of</strong> ADHD<br />

than those without attachment difficulties. (2) To ascertain if<br />

there is an association between disorders comorbid to ADHD<br />

and attachment difficulties in ADHD. Methods:Attachment<br />

difficulties were measured for 966 children with ADHD,<br />

combined type participating in the International Multicentre<br />

ADHD Genetics study (IMAGE), using questions from the<br />

Parental Account <strong>of</strong> Children’s Symptoms (PACS)<br />

semi-structured interview. Diagnoses <strong>of</strong> oppositional defiant<br />

disorder (ODD), conduct disorder (CD) and <strong>of</strong> anxiety<br />

disorder were generated using the PACS. Current symptoms<br />

<strong>of</strong> ADHD and <strong>of</strong> oppositional behaviour were measured using<br />

the Conners’ parent and teacher rating scales. The group was<br />

divided into those with no attachment difficulties and those<br />

with some attachment difficulties, and ADHD symptom<br />

severity was compared in each group. Attachment symptoms<br />

were compared in those with and without comobid CD, ODD,<br />

and anxiety disorder. Results: Parents <strong>of</strong> children with ADHD<br />

and attachment difficulties (N=432) noted slightly more severe<br />

symptoms <strong>of</strong> ADHD (mean Conners’ score = 79.07 vs. mean<br />

Conners’ score = 75.77, p


17 FAMILY-BASED COPY NUMBER VARIATION<br />

ASSOCIATION WITH ATTENTION-DEFICIT<br />

HYPERACTIVITY DISORDER<br />

C. Shtir* (1), S. Smalley (1), E. Mick (2), A. Todorov (3), B.<br />

Dechairo (4), S. Faraone (3), S. Hall (4), S. Loo (1), J.<br />

McCracken (1), J. MvGough (1), B. Neale (3), R. Todd (3), S.<br />

Nelson (1)<br />

1. University <strong>of</strong> California Los Angeles 2. Harvard <strong>Medical</strong><br />

School 3. Washington University 4. Pfizer Inc<br />

* cshtir@mednet.ucla.edu<br />

Several family and twin-based studies explored the genetic<br />

landscape attributed with risk <strong>of</strong> developing attention-deficit<br />

hyperactivity disorder (ADHD). However, major risk factors<br />

are yet to be defined. Despite strong heritability, the complex<br />

etiology <strong>of</strong> ADHD indicates that rare structural variants may<br />

play a determinant role. We present a family-based<br />

genome-wide copy number variation (CNV) screen followed<br />

by CNV association tests, with the purpose <strong>of</strong> identifying<br />

inherited and de novo ADHD-associated structural variants.<br />

To our knowledge this is the second, yet largest ADHD study<br />

characterizing effects <strong>of</strong> CNVs. A total <strong>of</strong> 733 trios<br />

ascertained through the University <strong>of</strong> California at Los<br />

Angeles, Washington University at St.Louis, and<br />

Massachusetts General Hospital were analyzed for excess<br />

transmission <strong>of</strong> CNVs and de novo mutations. Children<br />

between 6-17 years old were diagnosed according to the<br />

DSM-IV-TR criteria. UCLA trios were genotyped on the<br />

Illumina Human 1M-Duo bead array, while WASH-U and<br />

MGH samples were genotyped on the Illumina Human1M<br />

bead array. Normalized logR Ratios <strong>of</strong> total SNP signal<br />

intensities, quantile normalization and eigenstrat-based<br />

principal component analyses were used to eliminate sample<br />

and batch effects. Hidden-Markov-Model derived algorithms<br />

were employed for detection <strong>of</strong> CNVs. Associations <strong>of</strong><br />

structural variants with ADHD were assessed through linear<br />

regression models and Mann Whitney tests. We observe<br />

evidence for CNV association with ADHD for genes involved<br />

in neurological disorders and neurodevelopment pathways, for<br />

transcription factors previously reported to be susceptible for<br />

ADHD, and for a set <strong>of</strong> putative genes that may play an<br />

additional role in the biogenesis <strong>of</strong> ADHD.<br />

18 POSTNATAL EXPOSURE TO PCB153 ALTERS<br />

CATECHOLAMINE AND SECOND MESSENGER<br />

SIGNALING IN RATS: IMPLICATIONS FOR ADHD<br />

T. Das Banerjee* (1), F. Middelton (1), E. Borgå Johansen<br />

(2), T. Sagvolden (2), S. Faraone (1)<br />

1. SUNY Upstate <strong>Medical</strong> university 2. University <strong>of</strong> Oslo<br />

* dast@upstate.edu<br />

ADHD is a neurobehavioral disorder with both genetic and<br />

environmental components. PCB153 is one <strong>of</strong> the most<br />

commonly detected polychlorinated biphenyl congeners in<br />

human milk. Studies have shown that PCB153-exposed male<br />

rats exhibit hyperactivity, a characteristic behavioral symptom<br />

<strong>of</strong> ADHD. In this study we wanted to investigate the<br />

molecular mechanism by which PCB153 induces ADHD-like<br />

symptoms. The study was conducted in Wistar Kyoto (WKY)<br />

male and female rats dosed via gavage with corn oil vehicle, 3<br />

mg/kg b.w. <strong>of</strong> PCB153 from PND11-13. Brains were<br />

harvested from the experimental and control animals at<br />

PND65. For microarray screening, RNA from four dorsal<br />

striatum samples was obtained per treatment group per gender,<br />

with a total <strong>of</strong> 16 samples. RNA was processed using the<br />

recommended Affymetrix protocol and hybridized to the<br />

GeneChip Rat Gene 1.0 ST array. Gene-level as well as<br />

exon-level analyses were performed for both males and<br />

females and with them combined (in a 2 way ANOVA). Our<br />

results show that there are major disruptions <strong>of</strong> catecholamine<br />

and second messenger signaling in the brains <strong>of</strong> male and<br />

female rats exposed to PCB153. These changes appear to be<br />

gender specific. Altogether, the data suggests that postnatal<br />

exposure to PCB153 significantly alters brain circuits involved<br />

in ADHD.


19 NO EXCESS RATES OF CNV IN MALES<br />

COMPARED TO FEMALES IN<br />

NEURODEVELOPMENTAL DISORDERS AND<br />

CONTROL INDIVIDUALS<br />

I. Zaharieva* (1), A. Martin (1), K. Langley (1), C. Davies<br />

(1), E. Evans (1), S. Syed (1), R. Roberts (1), K.<br />

Mantripragada (1), N. Buttigieg (1), D. Grozeva (1), G. Kirov<br />

(1), M. O'Donovan (1), M. Owen (1), N. Williams (1), A.<br />

Thapar (1)<br />

1. Cardiff University, MRC Centre for Neuropsychiatric<br />

Genetics and Genomics<br />

* zaharievait@cardiff.ac.uk<br />

Recent studies implicate rare, large copy number variants in<br />

neurodevelopmental disorders such as schizophrenia, autism<br />

and attention deficit hyperactivity disorder (ADHD). Given<br />

the fact that neurodevelopmental disorders more commonly<br />

affect males, we wanted to investigate whether rare,<br />

large-scale copy number variants (CNV) (greater than100 kb)<br />

have higher rates in males compared to females. We used<br />

three datasets for the analysis: a dataset <strong>of</strong> 316 ADHD male<br />

patients and 50 ADHD female patients, a set <strong>of</strong> 314<br />

schizophrenia male patients and 156 schizophrenia female<br />

patients and a control set <strong>of</strong> 535 males and 512 females from<br />

1958 birth cohort. No statistically significant difference was<br />

observed when the total number <strong>of</strong> CNVs greater than 100 kb<br />

was compared between males and females in each dataset. The<br />

male to female ratio and p-values for each dataset are: ADHD<br />

male/female ratio=1.1, p=0.66; Schizophrenia male/female<br />

ratio=1.17, p=0.28; Controls male/female ratio=1.19, p=0.1.<br />

Furthermore, we saw no significant difference after stratifying<br />

the CNVs according to size. The minimum p-value observed<br />

is 0.07 in controls for the size range 100kb to 500kb<br />

(male/female ratio=1.23). In conclusion, our results show no<br />

evidence that males have an increased burden <strong>of</strong> CNVs<br />

compared to females in either ADHD or schizophrenia, and<br />

the rates are very similar to those in normal controls.<br />

20 ASSOCIATION OF DRD4 WITH CHILDHOOD<br />

ADHD COMORBIDITY IN WOMEN WITH BULIMIA<br />

NERVOSA<br />

Z. Yilmaz* (1), A. Kaplan (1), R. Levitan (1), J. Kennedy (1)<br />

1. University <strong>of</strong> Toronto<br />

* zeynep.yilmaz@utoronto.ca<br />

Purpose: Bulimia nervosa (BN) is a psychiatric disorder that<br />

predominantly affects women, and is characterized by regular<br />

binge eating, purging and characteristic psychopathology. Past<br />

research has shown that a proportion <strong>of</strong> patients with BN<br />

report a history <strong>of</strong> attention deficit/hyperactivity disorder<br />

(ADHD), and the dopaminergic genes, especially the<br />

dopamine receptor D4 (DRD4), have been associated<br />

separately with both BN and ADHD. The purpose <strong>of</strong> this<br />

study is to explore the possible role <strong>of</strong> DRD4 in predicting<br />

ADHD comorbidity in women with BN. Methods: 81 women<br />

with current or past BN purging subtype were genotyped for<br />

the presence or absence <strong>of</strong> the hyp<strong>of</strong>unctional 2-repeat<br />

and 7-repeat alleles <strong>of</strong> DRD4. Patients also completed the<br />

Wender Utah Rating Scale (WURS) for the assessment <strong>of</strong><br />

childhood ADHD history and the Brown Adult ADD Scale for<br />

current ADHD symptoms. Results: The presence either the<br />

2-repeat or 7-repeat alleles <strong>of</strong> DRD4 was associated with<br />

childhood ADHD in BN patients, as assessed by the WURS.<br />

There was no relationship between the Brown scores and the<br />

DRD4 genotype. Conclusion: The pathophysiology <strong>of</strong> both<br />

BN and ADHD may in part be related to an underlying<br />

hypodopaminergic state, and these finding may have<br />

implications for treatment, especially with dopaminergic<br />

agents, for BN patients with a childhood history <strong>of</strong> ADHD.


21 A CIS-ACTING POLYMORPHISM IN SLC9A9 IS<br />

NOT ASSOCIATED WITH ADHD IN AN IRISH<br />

SAMPLE.<br />

M. Hill* (1), Z. Hawi (2), M. Gill (2), R. Anney (2)<br />

1. Centre for the Cellular Basis <strong>of</strong> Behaviour, Department <strong>of</strong><br />

Neuroscience, The James Black Centre, Institute <strong>of</strong><br />

Psychiatry, Kings College London, 125 Coldharbour Lane,<br />

London, SE5 9NU, UK 2. Neuropsychiatric Genetics<br />

Research Group, Department <strong>of</strong> Psychiatry, Trinity College<br />

Dublin, Ireland<br />

* matthew.hill@kcl.ac.uk<br />

Introduction: Attention-deficit hyperactivity disorder (ADHD)<br />

is a common neuropsychiatric disorder with a strong genetic<br />

component. A recent genome-wide association study<br />

implicates SLC9A9 as a potential susceptibility locus. The<br />

nominally associated polymorphisms reside within non-coding<br />

regions <strong>of</strong> the gene and are thus hypothesised to confer<br />

vulnerability via altering gene expression. This study aimed to<br />

identify cis-acting variants in this promising candidate gene<br />

and test for their association with ADHD in an Irish sample.<br />

Methods: Using RNA from the HapMap CEU lymphoblasts<br />

we measured allelic expressing imbalance (AEI) <strong>of</strong> the<br />

SLC9A9 transcript using a transcribed single nucleotide<br />

polymorphism (SNP). Transmission dysequilibrium testing in<br />

an ADHD parent-child sample was performed to test for<br />

association with ADHD. Results: Moderate imbalance <strong>of</strong> the<br />

SLC9A9 transcript was observed in 7 <strong>of</strong> the 45 individuals<br />

suitable for investigation. Correlation between AEI and ~1000<br />

SNPs across the gene and flanking regions revealed a single<br />

SNP that was highly correlated with imbalance and remained<br />

significant after correction for multiple testing. Subsequent<br />

association testing revealed that this SNP was not associated<br />

with ADHD in an Irish sample. Conclusion: This study has<br />

identified moderate and infrequent allelic expression<br />

imbalance <strong>of</strong> the SLC9A9 transcript in lymphoblasts indicting<br />

the presence <strong>of</strong> cis-acting variants. A likely cis-acting variant<br />

identified by this screen was not associated with ADHD. The<br />

role <strong>of</strong> this SNP in other diseases, e.g. autism and<br />

Alzheimer’s, in which SLC9A9 is implicated is <strong>of</strong> future<br />

interest.<br />

290 BIPOLAR DISORDER CANDIDATE<br />

REGULATORY MUTATIONS IN NR2E1<br />

FUNCTIONALLY EVALUATED IN VIVO IN MICE<br />

J. Schmouth* (1), R. Bonaguro (1), C. de Leeuw (1), L.<br />

Dreolini (2)<br />

1. Centre for Molecular Medicine and Therapeutics, Child &<br />

Family Research Institute, University <strong>of</strong> British Columbia,<br />

Department <strong>of</strong> Genetics 2. Canada's Michael Smith Genome<br />

Sciences Centre, British Columbia Cancer Agency,<br />

Vancouver, Canada<br />

Background: NR2E1 is a gene coding an orphan nuclear<br />

receptor expressed in vertebrate forebrain and in the adult<br />

brain. Previous case-control studies in the Simpson laboratory<br />

associated NR2E1 with bipolar disorder and resequencing <strong>of</strong><br />

NR2E1 revealed 14 novel candidate regulatory mutations in<br />

patients suffering from either bipolar disorder or cortical<br />

malformation (microcephaly). Adult mice lacking Nr2e1<br />

(‘Fierce’ mice) display hyperactivity, increased aggression,<br />

and altered neurogenesis. A transgenic “rescue” study using<br />

random-insertion mice harboring multiple copies <strong>of</strong> the human<br />

NR2E1 gene on the Nr2e1-null background has shown that<br />

NR2E1 is sufficient to correct the fierce phenotype.<br />

Hypothesis: Candidate mutations in human NR2E1 regulatory<br />

regions will cause brain and behaviour disorders in Nr2e1-null<br />

mice. Material and methods: Five mouse models harboring<br />

human NR2E1 BAC DNA, introduced in the genome as a<br />

single copy, using the Hprt1 locus are being generated. These<br />

will be bred onto the fierce background and assessed for<br />

rescue <strong>of</strong> behavioural and histological phenotypes. Results:<br />

We have generated the positive control (wild-type) mouse<br />

strain, which is currently undergoing breeding to the fierce<br />

background. One variant sequence g.2078G>C has generated<br />

chimeric animals and the negative control (deletion allele) has<br />

been generated and been electroporated in embryonic stem<br />

cells (ESCs). Conclusion: This work may demonstrate that the<br />

candidate mutations identified in regulatory regions <strong>of</strong> human<br />

NR2E1 are capable <strong>of</strong> causing brain and behaviour disorders<br />

in mice. This will support NR2E1 as a new drug target, and<br />

the role <strong>of</strong> genetic testing as part <strong>of</strong> diagnosis and treatment <strong>of</strong><br />

bipolar disorder.


292 DOPAMINE D4 RECEPTOR GENE: POSSIBLE<br />

INFLUENCE ON ATTENTION DEFICIT<br />

HYPERACTIVITY DISORDER IN BRAZILIAN<br />

POPULATION<br />

G. Akutagava-Martins (1), G. Ferraz (1), J. Genro (1), G.<br />

Polanczyk (2), C. Zeni (3), M. Schmitz (3), L. Rohde (3), M.<br />

Hutz (1), T. Roman* (1)<br />

1. Department <strong>of</strong> Genetics, Federal University <strong>of</strong> Rio Grande<br />

do Sul, Brazil 2. Department <strong>of</strong> Psychology and Neuroscience,<br />

Duke University, USA 3. Department <strong>of</strong> Psychiatry and Legal<br />

Medicine, Federal University <strong>of</strong> Rio Grande do Sul, Brazil<br />

The dopamine D4 receptor gene (DRD4) is one <strong>of</strong> the most<br />

investigated genes in Attention deficit/hyperactivity disorder<br />

(ADHD) and has already been accepted as a susceptibility<br />

gene. The aim <strong>of</strong> present study was to search for a possible<br />

association between ADHD and the 120 bp tandem<br />

duplication <strong>of</strong> promoter region, the SNPs -616C>G (rs747302)<br />

and -521C>T (rs1800955) and the 48 bp VNTR <strong>of</strong> DRD4 gene<br />

in a Brazilian sample composed by 488 ADHD children and<br />

adolescents, diagnosed according to DSM-IV criteria, and<br />

their biological parents. The 120 bp tandem duplication and<br />

the 48 bp VNTR were genotyped by PCR, while the SNPs<br />

-616C>G and -521C>T were genotyped by PCR-RFLP. The<br />

possibility <strong>of</strong> linkage disequilibrium among the studied<br />

polymorphisms was tested using MLocus s<strong>of</strong>tware. The<br />

hypothesis <strong>of</strong> association was verified using FBAT s<strong>of</strong>tware.<br />

There was no evidence <strong>of</strong> association between each<br />

polymorphism and ADHD for the whole sample (P values<br />

ranging from 0.200 to 0.974). However, when the analysis was<br />

restricted to families where the proband presented the<br />

combined type <strong>of</strong> the disorder, the VNTR 2R allele was<br />

preferentially not transmited (P=0.033). Haplotype analyses<br />

did not show any association (P values ranging from 0.063 to<br />

0.977). The supposed protective effect <strong>of</strong> 2R allele observed<br />

herein is intriguing. This result must be interpreted cautiously<br />

and investigated through other approaches, as dimensional<br />

analyses. It is also possible that the findings, divergent from<br />

the literature, are due to DRD4 structural complexity, which<br />

should be understood to better characterize its association with<br />

ADHD.


EARLY CAREER INVESTIGATOR TRACK:<br />

ALTERNATIVE PHENOTYPES, GENE-<br />

GENE, AND GENE-ENVIRONMENT<br />

INTERACTION<br />

ECI 1 ASSOCIATION OF CAROTENOID<br />

ANTIOXIDANT AND SCHIZOPHRENIA USING<br />

NON-INVASIVE MEASUREMENT<br />

T. Chow (1), S. Tee (1), H. Loh (1), P. Tang (1), S. Soon (1)<br />

1. Universiti Tunku Abdul Rahman<br />

During stress, abnormally high neuronal activities in human<br />

brain would trigger excessive free radicals as part <strong>of</strong> the body<br />

immune response. This impairs antioxidant defense system<br />

and causes detrimental effects on signal transduction.<br />

Antioxidants are hypothesized to counteract oxidative stress<br />

and thus are suspected to be involved in stress-related<br />

disorders including schizophrenia. Carotenoids serve as a<br />

powerful free radical-neutralizing antioxidant which is<br />

absorbed in human plasma and tissue, therefore providing<br />

reliable indicator <strong>of</strong> a person’s overall antioxidant level. This<br />

study aims to investigate the possible relationship between<br />

carotenoid antioxidant level and schizophrenia. A total <strong>of</strong> 524<br />

schizophrenic subjects and 289 healthy control subjects were<br />

recruited. Subject skin carotenoid score were determined using<br />

non-invasive Raman spectroscopy technique that measures<br />

stable carotenoids in the human skin. Statistical analyses<br />

compared carotenoid score: (i) between patients and controls<br />

in different age and gender, and (ii) among patients with<br />

different subtypes, medications, and duration <strong>of</strong> illness. The<br />

results showed significant (p


at TTC12 and CHRNA3 appears to be additive. TTC12 seems<br />

to influence mainly initiation <strong>of</strong> use in adolescence. In<br />

contrast, genetic variation at CHRNA3 appears to be mainly<br />

involved in the transition towards regular/heavy use in<br />

adulthood.<br />

ECI 3 ASSOCIATION OF CHROMOSOME 20 LOCI<br />

WITH CATEGORICAL DIAGNOSES AND CLINICAL<br />

DIMENSIONS OF SCHIZOPHRENIA IN THE IRISH<br />

STUDY OF HIGH DENSITY SCHIZOPHRENIA<br />

FAMILIES<br />

T. Bigdeli* (1), B. Maher (1, 2), Z. Zhao (1, 2), E. van den<br />

Oord (3), B. Webb (3), R. Amdur (4), S. Bergen (1), F. O'Neill<br />

(5), D. Walsh (6), B. Riley (1, 2), K. Kendler (1, 2)<br />

1. Department <strong>of</strong> Human and Molecular Genetics, Virginia<br />

Institute for Psychiatric and Behavioral Genetics, Virginia<br />

Commonwealth University, Richmond, VA, USA 2.<br />

Department <strong>of</strong> Psychiatry, Virginia Institute for Psychiatric<br />

and Behavioral Genetics, Virginia Commonwealth University,<br />

Richmond, VA, USA 3. Virginia Institute for Psychiatric and<br />

Behavioral Genetics, Center for Biomarker Research and<br />

Personalized Medicine, Virginia Commonwealth University,<br />

Richmond, VA, USA 4. Mental Health Service Line,<br />

Washington VA <strong>Medical</strong> Center, Washington, DC 5.<br />

Department <strong>of</strong> Psychiatry, Queens University, Belfast, UK 6.<br />

The Health Research Board, Dublin, Ireland<br />

* bigdelitb@vcu.edu<br />

Background: Prior genomewide scans <strong>of</strong> schizophrenia<br />

support evidence <strong>of</strong> linkage to regions <strong>of</strong> chromosome 20.<br />

However, association analyses have yet to provide support for<br />

any etiologically relevant variants. Methods: We analyzed<br />

2988 LD-tagging single nucleotide polymorphisms (SNPs) in<br />

327 genes on chromosome 20, to test for association with<br />

schizophrenia in 270 Irish high-density families (ISHDSF,<br />

N=270 families, 1408 subjects). These SNPs were genotyped<br />

using an Illumina iSelect genotyping array which employs the<br />

Infinium assay. Given a previous report <strong>of</strong> novel linkage with<br />

chromosome 20p using latent classes <strong>of</strong> psychotic illness,<br />

association analysis was also conducted for each <strong>of</strong> five<br />

factor-derived scores based on the Operational Criteria<br />

Checklist for Psychotic Illness (delusions, hallucinations,<br />

mania, depression, and negative symptoms). Tests <strong>of</strong><br />

association were conducted using the PDTPHASE and<br />

QPDTPHASE packages <strong>of</strong> UNPHASED. Results: While no<br />

single variant was significant after Bonferroni-correction,<br />

initial gene-dropping simulations identified loci which<br />

exceeded empirical significance criteria for minimum<br />

genewise P value or the truncated product <strong>of</strong> P values. In<br />

particular, TOX2 shows association with both intermediate<br />

and broad diagnoses <strong>of</strong> schizophrenia. Additionally,<br />

R3HDML and C20orf39 appear to be associated with<br />

depressive symptoms <strong>of</strong> schizophrenia. Conclusions:<br />

Preliminary results overall provide evidence that chromosome<br />

20 may harbor schizophrenia susceptibility or modifier loci.<br />

Ongoing gene-dropping analyses aim to assess the empiric<br />

significance <strong>of</strong> these associations.<br />

ECI 4 EFFECTS OF MEDICATION ON<br />

MONOAMINE-RELATED GENE EXPRESSION IN<br />

BLOOD OF CHILDREN WITH TOURETTE<br />

SYNDROME<br />

I. Liao* (1), L. Lit (1), B. Corbett (1), D. Gilbert (2), S. Bunge<br />

(3), F. Sharp (1)<br />

1. University <strong>of</strong> California, Davis 2. University <strong>of</strong> Cincinnati<br />

3. University <strong>of</strong> California, Berkeley<br />

* isaacliao@gmail.com<br />

Tourette syndrome (TS), a highly heritable<br />

neurodevelopmental disorder <strong>of</strong> unknown etiology resulting in<br />

motor and verbal tics, is frequently treated with<br />

psychopharmacological agents affecting monoamine<br />

neurotransmission, such as dopamine receptor antagonists.<br />

Previous studies have demonstrated differences in gene<br />

expression in blood between TS and controls; however, some<br />

<strong>of</strong> these differences might reflect medication effects. To<br />

address this, we examined gene expression in the peripheral<br />

blood <strong>of</strong> 28 medicated TS subjects (MED) and 26<br />

unmedicated TS subjects (UNMED) using whole-genome<br />

Affymetrix microarrays. Expression <strong>of</strong> the D2 dopamine<br />

receptor gene (DRD2) was correlated with tic severity in MED<br />

but not UNMED, suggesting an effect <strong>of</strong> medication on<br />

DRD2. Expression <strong>of</strong> the epsilon subunit <strong>of</strong> the GABA<br />

receptor (GABRE), which results in insensitivity to<br />

anesthetics and increases the opening probability <strong>of</strong> the<br />

GABA receptor, was negatively correlated with tic severity in<br />

UNMED but not MED. In addition, expression <strong>of</strong><br />

phenylethanolamine N-methyltransferase (PNMT), an enzyme<br />

responsible for the synthesis <strong>of</strong> epinephrine from<br />

norepinephrine (NE), was positively correlated with tic<br />

severity in UNMED but not MED. Because GABRE is found<br />

on NE-ergic neurons in the locus coeruleus, and the locus<br />

coeruleus receives PNMT-rich projections, these findings<br />

support current pharmacological clinical data that suggest an<br />

abnormality in locus coeruleus/NE circuits in TS. In addition,<br />

these findings demonstrate the effects <strong>of</strong> medication on blood<br />

gene expression in TS.


ECI 5 ANALYSIS OF DAT1 AND 5HTT GENES ADN<br />

PSYCHOSOCIAL ADVERSITIES IN ADOLESCENT<br />

INHABITANTS OF MEXICO CITY<br />

G. Martinez Levy* (1), M. Cardenas-Godinez (1), M.<br />

Briones Velasco (1), A. Gomez-Sanchez (1), E. Mendez (1),<br />

B. Corina (1)<br />

1. Instituto Nacional De Psiquiatria Ramon De La Fuente<br />

Muñiz<br />

* gaaml82@yahoo.com.mx<br />

Attention Deficit Hyperactivity Disorder (ADHD) is a<br />

clinically multifactorial mental disorder. Its etiology is<br />

unknown, however a complex interplay between genetic and<br />

environmental factors has been proposed. Three thousand and<br />

five subjects (12-18 years old) inhabitants <strong>of</strong> the metropolitan<br />

area <strong>of</strong> Mexico City, were interviewed face to face using the<br />

Adolescent-CIDI-CAPI, which generates a psychiatric<br />

diagnosis based on the DSM IV-R and ICD-10 criteria.<br />

Psychosocial adversity information was also gathered from<br />

this survey. Three hundred and two individual fulfill diagnosis<br />

criteria for ADHD, with 252 cases that agreed to give a saliva<br />

sample. They were typed for the LPR localized in the<br />

promoter region <strong>of</strong> 5HTT and the VNTR <strong>of</strong> the 5 region <strong>of</strong> the<br />

DAT1 gene, and compared with 284 DNA samples randomly<br />

chosen <strong>of</strong> the 833 participants who did not fulfill diagnostic<br />

criteria for any psychiatric disorder and gave saliva sample.<br />

Physical abuse, negligence, substance use and criminal<br />

conduct in parents as well as family violence were adversities<br />

clearly associated with the diagnosis <strong>of</strong> ADHD. No<br />

differences in genotypic or allelic frequencies for these<br />

particular variants were observed between cases and controls.<br />

Evaluations <strong>of</strong> the interaction among these molecular and<br />

psychosocial variables, as well as the analysis <strong>of</strong> other<br />

candidate genes are in progress.<br />

ECI 6 MATERNAL SMOKING DURING PREGNANCY,<br />

OFFSPRING BEHAVIORAL DISORDERS, AND THE<br />

SEROTONIN 1B RECEPTOR<br />

A. Talati* (1), S. Hamilton (2), P. Wickramaratne (1), S.<br />

Hodge (1), M. Weissman (1)<br />

1. Columbia University 2. University <strong>of</strong> California San Diego<br />

* at2071@columbia.edu<br />

Background: Smoking by mothers during pregnancy is<br />

associated with a number <strong>of</strong> adverse physiological and<br />

behavioral outcomes among their <strong>of</strong>fspring, including<br />

premature birth, lower birth-weight, cognitive impairment, and<br />

behavioral and substance use disorders. We present here<br />

findings on the long-term effects <strong>of</strong> maternal smoking<br />

exposure on <strong>of</strong>fspring, and further investigate genetic<br />

underpinnings <strong>of</strong> the association. Method: Mothers and<br />

<strong>of</strong>fspring were interviewed at multiple waves spanning more<br />

than twenty years. Psychiatric diagnoses were determined at<br />

each wave using the age-appropriate version <strong>of</strong> the Schedule<br />

for Affective Disorders and Schizophrenia (SADS). Smoking<br />

during pregnancy was ascertained by maternal self-report;<br />

<strong>of</strong>fspring were classified based on whether or not their mother<br />

smoked ≥10 cigarettes per day, nearly every day. Final<br />

diagnoses <strong>of</strong> mothers and <strong>of</strong>fspring were made independently,<br />

using the best estimate procedure, and blind to maternal<br />

smoking and previous child and maternal assessments. Single<br />

nucleotide polymorphisms (SNPs) were tested within the<br />

serotonin transporter (SLC6A4) and 1B (HTR1B) receptor,<br />

and the monoamine oxidase A variable nucleotide repeat<br />

region (MAOAVNTR). Results: After adjusting for multiple<br />

potential confounders, maternal smoking during pregnancy<br />

was associated with a 3-fold increase in drug and alcohol use<br />

disorders among female, and a two-fold increase in conduct<br />

disorder among male, <strong>of</strong>fspring. One SNP rs6298, located<br />

within the coding region <strong>of</strong> HTR1B was associated with a<br />

five-fold increase in AUD and a nine-fold increase in CD.<br />

When the association between smoke exposure and <strong>of</strong>fspring<br />

psychopathology was further stratified by the genotype at the<br />

above polymorphism, exposure to smoking was associated<br />

with CD among <strong>of</strong>fspring with TT but not CC/CT genotypes.<br />

Conclusion: Our data show that HTR1B is associated with<br />

conduct and alcohol use disorders, and suggest that HTR1B<br />

may further moderate the impact <strong>of</strong> exposure to maternal<br />

prenatal smoking on <strong>of</strong>fspring psychopathology.


ANIMAL MODELS<br />

22 THE DOG AS A GENETIC MODEL FOR SOCIAL<br />

BEHAVIORS: PRELIMINARY INVESTIGATION OF<br />

SOCIAL INTERACTIVE BEHAVIORS IN DOGS<br />

USING GENOME WIDE ANALYSIS<br />

L. Lit* (1), S. Boyd (2), D. Hessl (1), D. Bannasch (3)<br />

1. Dept <strong>of</strong> Psychiatry & M.I.N.D. Institute, University <strong>of</strong><br />

California Davis 2. Chief, Section <strong>of</strong> Genetics; Children's<br />

Miracle Network Endowed Chair; M.I.N.D. Institute,<br />

University California Davis 3. Dept <strong>of</strong> Population Health &<br />

Reproduction, School <strong>of</strong> Veterinary Medicine, University <strong>of</strong><br />

California Davis<br />

* llit@ucdavis.edu<br />

Currently, there are no animal models that can provide insight<br />

into the genetic underpinnings <strong>of</strong> normal social behaviors that<br />

are impaired in neurodevelopmental disorders. Social<br />

interactive behaviors between domestic dogs (Canis<br />

familiaris) and humans have been well documented<br />

experimentally. It has been hypothesized that a limited subset<br />

<strong>of</strong> wild canids originally displayed the social characteristics<br />

necessary to interact with humans, ultimately resulting in<br />

domestication. This suggests that some genes contributing to<br />

social interactive behaviors might predate breed formation,<br />

and that these genes would be associated with social behaviors<br />

irrespective <strong>of</strong> breed. We asked owners <strong>of</strong> 14 dogs (1 Basset<br />

Hound, 1 Black Russian Terrier, 2 Dalmatians, 1 Petit Basset<br />

Griffon Vendeen, 7 Nova Scotia Duck Tolling Retrievers, and<br />

2 mixed breed dogs) to complete surveys comprising owner<br />

report <strong>of</strong> social interactive behaviors observed in their dogs.<br />

Subscales representing initiation <strong>of</strong> reciprocal social behaviors<br />

(INIT), response to social interactions with humans (RSPNS),<br />

communication with humans (COMM), and attention (ATT)<br />

were derived from responses using factor analysis.<br />

Genome-wide SNP data for these dogs was generated using<br />

127K Affymetrix canine SNP arrays. Using subscales as<br />

input, we performed genome-wide quantitative trait<br />

association analysis with 10,000 permutations to determine<br />

SNPs associated with each subscale. SNPs were identified<br />

that corresponded to genes or regions homologous to those in<br />

humans associated with autism, ADHD, and schizophrenia.<br />

This contributes to the validity <strong>of</strong> the domestic dog as a model<br />

to investigate naturally occurring variation in social behaviors<br />

impaired in neurodevelopmental disorders in humans.<br />

23 SUSCEPTIBILITY TO ANTIPSYCHOTIC-INDUCED<br />

VACUOUS CHEWING MOVEMENTS AND OTHER<br />

EXTRAPYRAMIDAL SYMPTOMS ARE HIGHLY<br />

HERITABLE TRAITS IN MICE<br />

J. Crowley* (1), C. Quackenbush (1), A. Pratt (1), D. Adkins<br />

(2), E. van den Oord, M. Bogue (3), H. McLeod (1), P.<br />

Sullivan (1)<br />

1. UNC Chapel Hill 2. VCU 3. The Jackson Laboratory<br />

* crowley@unc.edu<br />

Around 50% <strong>of</strong> patients with schizophrenia discontinue<br />

assigned treatments due to intolerable adverse drug reactions<br />

(ADRs), but there are no clinically useful tests to predict<br />

individual risk. The primary ADRs limiting the utility <strong>of</strong><br />

typical antipsychotic drugs are extrapyramidal side effects<br />

(EPS), most notably tardive dyskinesia (TD), characterized by<br />

involuntary or<strong>of</strong>acial movements. The confident identification<br />

<strong>of</strong> TD susceptibility genes in human patients has proven to be<br />

very difficult and it remains unclear if TD susceptibility is<br />

sufficiently heritable to expect the identification <strong>of</strong> useful<br />

genetic predictors. To address these limitations, we have used<br />

a genetically diverse panel <strong>of</strong> mice to calculate the heritability<br />

<strong>of</strong> antipsychotic-induced EPS. A total <strong>of</strong> 18 inbred mouse<br />

strains (N = 5 mice per strain) were chronically treated with<br />

haloperidol (3 mg/kg/day) for 120 days and monitored for the<br />

development <strong>of</strong> vacuous chewing movements (VCMs; the<br />

mouse analog <strong>of</strong> TD) and other movement phenotypes derived<br />

from open field activity and an inclined screen test. These<br />

longitudinal data allowed us to calculate the heritability <strong>of</strong> the<br />

over-time response trajectories induced by haloperidol<br />

treatment (ratio <strong>of</strong> strain-level variance to strain-level +<br />

mouse-level variance). Every movement phenotype examined<br />

showed an across-time drug response heritability >50% and<br />

VCMs demonstrated an 81% heritability. Furthermore,<br />

confirmatory factor analysis revealed two significant factors<br />

underlying the movement response trajectories, with one<br />

factor explaining most <strong>of</strong> the variance in the locomotor<br />

activity response trajectories and the other primarily<br />

explaining the or<strong>of</strong>acial movement response trajectories.<br />

These data suggest that mapping <strong>of</strong> susceptibility alleles for<br />

EPS is a worthwhile endeavor. To our knowledge, this is the<br />

first demonstration that VCMs, the murine analog <strong>of</strong> TD, is a<br />

highly heritable mammalian trait.


24 EFFECTS OF PRENATAL NICOTINE EXSPOSURE<br />

ON DOPAMINERGIC GENE EXPRESSION AND<br />

METHYLATION STATUS<br />

N. Ilott* (1), J. Mill (1), P. Asherson (1), I. Stolerman (2), L.<br />

Bizarro (3)<br />

1. MRC SGDP Research Centre, Institute <strong>of</strong> Psychiatry, Kings<br />

College, London 2. Section <strong>of</strong> Behavioural Pharmacology,<br />

Institute <strong>of</strong> Psychiatry, King's College London 3.<br />

Universidade Federal Do Rio Grande do Sul, Departamento de<br />

Psicologia do Desenvolvimento e da Personalidade, Instituto<br />

de Psicologia, Rua Ramiro Barcellos 2600, Porto Alegre - -<br />

RS, Brasil<br />

* nicholas.ilott@iop.kcl.ac.uk<br />

Smoking during pregnancy (SDP) is an important risk factor<br />

for a number <strong>of</strong> developmental conditions including sudden<br />

infant death syndrome (SIDS) and behavioural abnormalities.<br />

Human studies have identified SDP as contributing to the<br />

development <strong>of</strong> externalizing behaviours such as Attention<br />

Deficit Hyperactivity Disorder (ADHD) and Antisocial<br />

Behaviour (ASB) although the causal role <strong>of</strong> tobacco exposure<br />

remains unclear. Animal studies have provided an insight into<br />

the behavioural consequences <strong>of</strong> gestational nicotine with<br />

suggested associations with increased locomotor activity,<br />

heightened anxiety as well as learning deficits. Molecular<br />

mechanisms underlying these behavioural consequences have<br />

been considered, with evidence suggesting long-lasting effects<br />

on nicotinic acetylcholine receptor (nAchR) subunit<br />

expression and neurotransmitter signaling. Here we investigate<br />

altered regulation <strong>of</strong> dopamine system genes because <strong>of</strong> the<br />

central role <strong>of</strong> dopaminergic transmission in ADHD. We<br />

studied expression and methylation patterns <strong>of</strong> five<br />

dopaminergic genes in the striatum and frontal cortex <strong>of</strong> adult<br />

rats prenatally exposed to nicotine. We found no major<br />

differences in either mRNA expression or promoter<br />

methylation for any <strong>of</strong> the genes investigated, although there<br />

was a small but significant increase in dopamine receptor D5<br />

(DRD5) mRNA expression in striatum <strong>of</strong> exposed animals<br />

compared to controls. Therefore we report no major effects <strong>of</strong><br />

gestational nicotine on the expression or methylation <strong>of</strong> the<br />

genes we have investigated. This work is being extended to an<br />

investigation at the level <strong>of</strong> the transcriptome and methylome<br />

to provide an unbiased approach for looking at gene regulation<br />

by gestational nicotine.<br />

25 BRAIN GENE EXPRESSION IN MICE<br />

HYPOMORPHIC FOR NEUREGULIN 1 (NRG1) AND<br />

THE EFFECTS OF THE ATYPICAL ANTIPSYCOTIC<br />

CLOZAPINE<br />

A. Byrnes* (1), J. Crowley (2), T. Konneker (2), J. Dackor<br />

(2), F. Wright (1), E. Anton (3), P. Sullivan (2)<br />

1. Department <strong>of</strong> Biostatistics, University <strong>of</strong> North Carolina at<br />

Chapel Hill, NC 2. Department <strong>of</strong> Genetics, University <strong>of</strong><br />

North Carolina at Chapel Hill, NC 3. Department <strong>of</strong> Cell and<br />

Molecular Physiology, University <strong>of</strong> North Carolina at Chapel<br />

Hill, NC<br />

* abyrnes@bios.unc.edu<br />

Schizophrenia (SCZ) is a severe and highly heritable<br />

psychiatric disorder for which neuregulin 1 (NRG1) may play<br />

an etiological role. Mice hypomorphic for Nrg1 have been<br />

shown to exhibit SCZ-like characteristics that are ameliorated<br />

with clozapine and represent a potential animal model for<br />

SCZ. The study aims to explore the effects <strong>of</strong> a Nrg1 mutation<br />

on forebrain gene expression and the effects <strong>of</strong> clozapine. We<br />

compared gene expression in forebrain from 8 Nrg1<br />

hypomorphic and 8 wild type mice using the Illumina and<br />

Affymetrix expression assays treated with vehicle along with<br />

8 Nrg1 hypomorphic and 8 wild type mice treated with the<br />

atypical antipsychotic clozapine. Statistical analyses<br />

contrasted the effects <strong>of</strong> strain (Nrg1 hypomorph vs wild type)<br />

and drug (vehicle vs clozapine). As expected, we found that<br />

Nrg1 expression was significantly higher in wild type mice as<br />

compared to hypomorphic mice. Other differentially expressed<br />

transcripts include ADP-ribosylation factor-like 3 (Arl3) and<br />

Hermansky-Pudlak syndrome 1 homolog (Hps1).<br />

Transcription <strong>of</strong> mitochondrial tryptophanyl tRNA synthetase<br />

2 (Wars2) and interleukin-1 receptor-associated kinase (Irak)<br />

appear to be altered by treatment with clozapine. Irak also<br />

showed significant interaction effects, as did member RAS<br />

oncogene family (Rab). Numerous pathways were also<br />

significant in these analyses. These include immune function<br />

and response to stimulus and olfactory receptors which<br />

appears to have convergent validity with respect to recent SCZ<br />

GWAS results.


26 THE KNOCKOUT MOUSE REPOSITORY<br />

HTTP://WWW.KOMP.ORG/<br />

R. O'Neill* (1), P. de Jong (2), K. Lloyd (3)<br />

1. DCM, NCRR, NIH 2. Children's Oakland Research Institute<br />

3. University <strong>of</strong> California, Davis<br />

* oneillr@mail.nih.gov<br />

Genome wide association studies (GWAS) and other<br />

investigations <strong>of</strong> human psychiatric conditions are discovering<br />

the association <strong>of</strong> specific phenotypes with various gene<br />

alleles and single nucleotide polymorphisms (SNPs). In<br />

followup experiments, knocking out one specific gene in an<br />

animal model can determine the function <strong>of</strong> a gene suspected<br />

<strong>of</strong> causing the human condition. More than 2,000 different<br />

mouse Embryonic stem (ES) cell lines carrying specific gene<br />

knockouts (KOs) can be obtained from the Knockout Mouse<br />

Program (KOMP) Repository. In addition, KO vectors<br />

useful for targeting recombination in ES cells, and KO mice,<br />

are currently available. In 2011, ~8,500 such individual gene<br />

KOs will be available from the KOMP repository.<br />

http://www.komp.org/can be queried to see if an individual<br />

investigator's gene <strong>of</strong> interest is already available, or to<br />

register interest in a specific KO that will or could be made.<br />

More than 100 <strong>of</strong> the 2,000 KOs currently available have<br />

annotations in Online Mendelian Inheritance in Man. Several<br />

vector constructs are used to make KOs, and conditional<br />

vectors allow tissue-specific or cell-type-specific perturbations<br />

within the nervous system. This is done by temporally or<br />

spatially restricting expression <strong>of</strong> the KO vector phenotype via<br />

combination with various patterns <strong>of</strong> Cre-recombinase<br />

expression (>100 Cre-drivers from<br />

http://www.credrivermice.org/). Nineteen Institutes and<br />

Centers at NIH fund these efforts, with the repository located<br />

at the University <strong>of</strong> California, Davis and the Children's<br />

Oakland Research Institute; with major partners at the Sanger<br />

Institute in the U.K.; Velocigene at Regeneron, Inc.; and the<br />

Jackson Laboratory.<br />

294 A RECENT SINE INSERTION CREATED A<br />

COMT-OVEREXPRESSING, BEHAVIOUR<br />

MODIFYING ALLELE IN MANY INBRED MOUSE<br />

STRAINS.<br />

R. Kember* (1), C. Fernandes (1), E. Tunbridge (2), L. Liu<br />

(1), J. Payo-Cano (1), H. Lad (1), M. Parsons (1), L.<br />

Schalkwyk (1)<br />

1. Institute <strong>of</strong> Psychiatry, Kings College London 2.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Oxford<br />

Comt is a key enzyme <strong>of</strong> neurotransmitter catabolism and a<br />

perennial candidate gene for human psychiatric disorders. In<br />

mouse it is located in a large genomic region <strong>of</strong> extremely low<br />

variation among the classical inbred strains, with only four<br />

known putative SNPS in 600 Kb. The strain C57BL/6J and<br />

others including 129 (multiple substrains) have a B2 SINE<br />

insertion in the 3' UTR which is not present in others<br />

including DBA/2J and C3HeB/FeJ. A stringent association<br />

test, using over 700 highly outbred mice with data from an<br />

extensive behavioural battery, indicates that this insertion<br />

allele is responsible for differences in behaviour related to<br />

exploration and anxiety. Differences in the gene expression<br />

level and in the activity <strong>of</strong> the enzyme itself (74% increase in<br />

activity with the insertion) indicate a mechanism for these<br />

behavioural effects. This finding robustly shows that a<br />

relatively modest difference in Comt expression (comparable<br />

to the human Val/Met polymorphism) can have demonstrable<br />

phenotypic effects across behaviours in a human-like variety<br />

<strong>of</strong> outbred genetic backgrounds.


ANXIETY DISORDERS<br />

27 A PH SENSITIVE SODIUM CHANNEL IS A<br />

POTENTIAL CANDIDATE GENE FOR PANIC<br />

DISORDER<br />

N. Gregersen* (1), H. Nørmølle Buttenschøn (2), M.<br />

Nyegaard (1), H. Dahl (3), A. Hedemand (1), A. Suhl<br />

Kristensen (2), D. Woldbye (4), S. Joensen (5), T. A. Kruse<br />

(3), A. Børglum (1), O. Mors (2)<br />

1. Inst. <strong>of</strong> Human Genetics, Aarhus University, Denmark 2.<br />

Center for Psychiatric Research, Aarhus University Hospital,<br />

Risskov, Denmark 3. Dept. <strong>of</strong> Clinical Biochemistry and<br />

Genetics, University <strong>of</strong> Southern Denmark 4.<br />

Neuropsychiatric Recearch Laboratory, University <strong>of</strong><br />

Copenhagen, Denmark 5. Dept. <strong>of</strong> Psychiatry, National<br />

Hospital, Faroe Islands<br />

* noomigregersen@mac.com<br />

INTRODUCTION: A genome wide scan conducted on 13<br />

patients with panic disorder, PD, from the isolated population<br />

<strong>of</strong> the Faroe Islands, observed significant association with<br />

chromosome 17p11.2. This chromosomal region contains the<br />

pH sensitive sodium channel ACCN1. Markers within this<br />

gene have shown to be significantly associated with PD. The<br />

aim <strong>of</strong> current study was to replicate the findings on<br />

chromosome 17 in a new Faroese PD sample and in an<br />

outbred Danish sample <strong>of</strong> patients with PD and control<br />

individuals. METHODS: We have genotyped 50tagSNPs in<br />

the ACCN1 gene region using the Sequenom platform<br />

(Sequenom, Inc.San Diego, USA) and the Lightcycler 480<br />

Genotyping master (RocheApplied Science, Mannheim,<br />

Germany). DNA from 14 patients with PD and 119 controls<br />

was obtained from the Genetic Biobank <strong>of</strong> the Faroe Islands.<br />

The Danish population consisted <strong>of</strong> 365 cases and 649 control<br />

individuals. For statistical analysis we used PLINK<br />

(https://gene.gram.au.dk/bcos/index.html). RESULTS: One <strong>of</strong><br />

the markers was significantly associated with PD in the new<br />

Faroese sample (rs9906088: p=0.0268). In the combined<br />

Faroese case-control sample consisting <strong>of</strong> 27 cases and 162<br />

controls four SNP´s were significantly associated with PD<br />

(p-values ranging from 0.008p to 0.0144). Three SNP´s were<br />

significantly associated with PD in the Danish<br />

case-controlsample (p-values ranging from 0.0002 to 0.0139).<br />

DISCUSSION: These results indicate ACCN1 to be a<br />

potential candidate gene for PD. However, further studies are<br />

needed. Furthermore, we were not able to replicate the most<br />

significantly associated marker from the previous study.<br />

ACKNOWLEDGEMENTS: Genetic Biobank <strong>of</strong> the Faroes<br />

for samples and the Lundbeck foundation for financing the<br />

project.<br />

28 G ALLELE CARRIERS AT 5HT1A HAVE AN<br />

INCREASED RISK FOR MIXED ANXIETY AND<br />

DEPRESSION<br />

E. Molina (1), J. Cervilla (2), M. Rivera (3), B. Gutiérrez (1)<br />

1. CIBERSAM UGR, Spain 2. CIBERSAM UGR, HU San<br />

Cecilio, Spain 3. CIBERSAM UGR, Institute <strong>of</strong> Psychiatry<br />

London<br />

Serotonin 1A receptors are key regulators <strong>of</strong> serotonin activity<br />

and their dys-regulation might be involved in the pathology <strong>of</strong><br />

major depression (MD) and generalized anxiety disorder<br />

(GAD). Previous studies have yielded inconclusive results as<br />

to whether the 5-HT1A receptor gene has a role in the<br />

aetiology <strong>of</strong> MD and no study up to date had studied this<br />

polymorphism on either pure MD or MD comorbid with<br />

GAD. We aim to clarify the putative role <strong>of</strong> the C (-1019) G<br />

polymorphism at the serotonin 1A receptor gene in the<br />

aetiology <strong>of</strong> both MD and GAD and comorbid MD/GAD<br />

states. DSM-IV MD and GAD diagnoses were ascertained<br />

using the Composite International Diagnostic Interview and<br />

the Primary Care Evaluation <strong>of</strong> Mental Disorders Patient<br />

Health Questionary, respectively. 1059 sujects who took part<br />

in the PREDICT-GENE study (1) were included for molecular<br />

analyses using Sequenom platform. Carrying the G allele <strong>of</strong><br />

the C (-1019) G polymorphism was associated with MD (OR<br />

= 1.67, 95%CI = 1.14 - 2.44, p = 0.008) but this association<br />

became non-significant after adjusting by presence <strong>of</strong> GAD<br />

(OR = 1.43, 95%CI = 0.958-2.141, p = 0.080). Similarly, the<br />

C (-1019) G polymorphism was univariately associated with<br />

GAD (Crude: OR = 2.54, 95%CI = 1.28-4.861, p = 0.003),<br />

although adjusting by MD made results no longer significant<br />

(OR =1.97, 95%CI = 0.991-3.910, p=0.05). However, we<br />

found a solid significant association between carrying the G<br />

allele and comorbid MD and GAD (OR =3.41, 95% CI =<br />

1.444-8.048, p = 0.005) which remained robust and<br />

statistically significant after adjusting by sex, age and family<br />

history <strong>of</strong> psychological problems (OR =2.82, 95%CI =<br />

1.177-6.772, p = 0.020). Our results suggest that the C (-1019)<br />

G serotonin 1A polymorphism confers a risk for the frequent<br />

clinical presentation <strong>of</strong> comorbid MD and GAD but not for<br />

either <strong>of</strong> these disorders alone after adjusting by presence <strong>of</strong><br />

one another.


29 REPLICATION OF A GENOME-WIDE<br />

ASSOCIATION STUDY OF PANIC DISORDER IN A<br />

JAPANESE POPULATION<br />

T. Otowa* (1), T. Minato (1), N. Sugaya (2), K. Inoue (3), T.<br />

Shimada (1), Y. Kawamura (1), M. Tochigi (1), T. Umekage<br />

(1), H. Tanii (4), T. Miyagawa (5), N. Nishida (5), K.<br />

Tokunaga (5), Y. Okazaki (6), H. Kaiya (7), T. Sasaki (8)<br />

1. Department <strong>of</strong> Neuropsychiatry, Graduate School <strong>of</strong><br />

Medicine, the University <strong>of</strong> Tokyo 2. Outpatient Clinic for<br />

Anxiety Disorders, Akasaka Mental Clinic 3. Department <strong>of</strong><br />

Public Health, Fujita Health University School <strong>of</strong> Medicine 4.<br />

Department <strong>of</strong> Neuropsychiatry, Graduate School <strong>of</strong><br />

Medicine, Mie University 5. Department <strong>of</strong> Human Genetics,<br />

Graduate School <strong>of</strong> Medicine, the University <strong>of</strong> Tokyo 6.<br />

Department <strong>of</strong> Neurology, Tokyo Metropolitan Matsuzawa<br />

Hospital 7. Research Center for Panic Disorder, Nagoya<br />

Mental Clinic 8. Office for Mental Health Support and<br />

Graduate School <strong>of</strong> <strong>Education</strong>, the University <strong>of</strong> Tokyo<br />

* totowa-psy@umin.ac.jp<br />

Panic disorder (PD) is an anxiety disorder characterized by<br />

recurrent and unexpected panic attacks, subsequent worry, and<br />

phobic avoidance. Although a number <strong>of</strong> association and<br />

linkage studies have been conducted, no gene has been<br />

identified as a susceptibility locus. We previously conducted a<br />

genome-wide association analysis <strong>of</strong> PD in 200 Japanese<br />

patients and the same number <strong>of</strong> controls, using a 500K single<br />

nucleotide polymorphisms (SNPs) chip. Here we report a<br />

replication analysis <strong>of</strong> PD using the DigTag2 assay. DigTag2<br />

assay is suitable for genotyping an intermediate number <strong>of</strong><br />

SNPs with high accuracy. The second stage sample consisted<br />

<strong>of</strong> 558 Japanese patients and 566 controls. Thirty-two markers<br />

were tested in a replication sample. As a result, no significant<br />

association could be found after correction for multiple<br />

testing. However, the difference was observed at the nominal<br />

allele P-value < 0.05 for two SNPs (rs6733840 and rs132617).<br />

Further studies on these variants with a larger sample size may<br />

be worth testing to confirm the results.<br />

30 GENETICS OF PANIC DISORDER: A<br />

GENOME-WIDE ASSOCIATION STUDY<br />

J. Schumacher* (1), C. Allgulander (2), E. Binder (3), J.<br />

Deckert (4), K. Domschke (5), A. Erhardt (3), E. Eriksson (6),<br />

J. Ioannidis (7), J. Kennedy (8), A. Kristensen (9), E. Maron<br />

(10), A. Metspalu (10), O. Mors (9), J. Nurnberger (11), T.<br />

Otowa (12), R. Philibert (13), A. Reif (4), M. Rietschel (14),<br />

T. Sasaki (12), T. Schulze (1), D. Woldbye (15), F. McMahon<br />

(1)<br />

1. NIMH Bethesda 2. Karolinska Institute 3. MPI Munich 4.<br />

Univ. Wuerzburg 5. Univ. Muenster 6. Univ. Goteborg 7.<br />

Univ. Ioannina 8. Univ. Toronto 9. Univ. Aarhus 10. Univ.<br />

Tartu 11. Univ. Indiana 12. Univ. Tokyo 13. Univ. Iowa 14.<br />

Univ. Heidelberg 15. Univ. Copenhagen<br />

* schumacherj@mail.nih.gov<br />

Panic disorder (PD) is a severe, heritable condition affecting<br />

about two percent <strong>of</strong> the population. The mode <strong>of</strong> inheritance<br />

is complex and involves multiple contributing genes, each <strong>of</strong><br />

small to moderate effect. Although genetic linkage has been<br />

reported to a number <strong>of</strong> chromosomal regions, no linkage<br />

finding has been replicated consistently throughout all studies.<br />

In addition, all candidate gene association studies applied to<br />

PD so far have failed to deliver definitive results. There is<br />

increasing evidence that genome-wide association (GWA)<br />

studies represent a promising approach to the identification <strong>of</strong><br />

genes involved in complex disorders, such as PD. The Panic<br />

International Consortium (PANIC) was established as a<br />

collaborative approach involving most <strong>of</strong> the leading<br />

international PD research groups (from the Universities <strong>of</strong><br />

Aarhus, Copenhagen, Goteborg, Heidelberg, Indiana, Iowa,<br />

Münster, Tartu, Tokyo, Toronto, Würzburg, the Karolinska<br />

Institute in Stockholm, the MPI in Munich, and the NIMH),<br />

with statistical advice from John Ioannidis (University <strong>of</strong><br />

Ionnina). The initial goal was to complete a GWA on one <strong>of</strong><br />

the largest PD samples. In total, the PANIC sample consists <strong>of</strong><br />

>2,000 patients and > 2,000 controls from two different<br />

populations, which were collected at 12 different research<br />

sites. The GWA results on the full PANIC sample will be<br />

presented. The identified genes may represent promising<br />

targets for functional studies aimed at improved diagnosis and<br />

treatment.


AUTISM<br />

31 SOCIAL RESPONSIVENESS SCALE:<br />

STANDARDIZATION AND VALIDATION OF THE<br />

DUTCH ADULT VERSION<br />

W. De la Marche* (1), E. Scholte (2), J. Steyaert (1, 3), M.<br />

Dorst (2), I. van Berckelaer-Onnes (2), I. Noens (4)<br />

1. Child and Adolescent Psychiatry Dep., UPC-K.U.Leuven,<br />

Belgium . Faculty <strong>of</strong> Social and Behavioral Sciences, Leiden<br />

University, Netherlands 3. Clinical Genetics Dep., University<br />

Maastricht, Netherlands 4. Faculty <strong>of</strong> Psychology and<br />

<strong>Education</strong>al Studies, K.U.Leuven, Belgium<br />

* wouter.delamarche@uz.kuleuven.be<br />

Background: Autistic traits tend to be continuously distributed<br />

in the general population. The Social Responsiveness Scale<br />

(SRS) (Constantino et al., 2003) is a 65-item reporter based<br />

questionnaire that has proven to quantify autistic traits in a<br />

reliable way. We translated the adult research SRS into Dutch<br />

and generated a self-report version <strong>of</strong> this questionnaire,<br />

approved by Dr. Constantino and the original publisher.<br />

Objectives: 1. To confirm the validity <strong>of</strong> the Dutch informant-<br />

and self-report adult SRS in the general Dutch-speaking<br />

population (the Netherlands and Flanders, Belgium). 2. To<br />

describe the distribution <strong>of</strong> autistic traits as measured by the<br />

SRS in the general population. Methods: Randomly selected<br />

adults in the Netherlands and Flanders (Belgium) were asked<br />

to fill out SRS questionnaires about themselves and their<br />

partner and vice versa. Only questionnaires without missing<br />

items were taken into account for the analyses. Results:<br />

Preliminary results from the Netherlands (N=866 for<br />

self-report, N=694 for partner-report) show a good test-retest<br />

reliability (self-report: 0.90; partner-report: 0.91). Total<br />

internal consistency is high for both versions <strong>of</strong> the Dutch<br />

SRS (0.93 and 0.95), with two items not (or even negatively)<br />

correlating with the total score. More results will be available<br />

at the ISPG conference. Conclusions: Our results suggest that<br />

the Dutch Social Responsiveness Scale for Adults and the<br />

newly developed self-report version <strong>of</strong> this questionnaire have<br />

psychometric properties comparable to the original SRS for<br />

children, with a few small exceptions.<br />

32 ASSOCIATION BETWEEN A HIGH-RISK AUTISM<br />

LOCUS AND AUTISM-SPECTRUM<br />

DISORDER-RELATED PHENOTYPES IN THE AVON<br />

LONGITUDINAL STUDY OF PARENTS AND<br />

CHILDREN (ALSPAC)<br />

B. Glaser* (1), K. Wang (2), J. Glessner (2), J. Golding (3),<br />

C. Steer (3), S. Grant (2), H. Hakonarson (2), G. Davey Smith<br />

(1)<br />

1. <strong>Medical</strong> Research Council Centre for Causal Analysis in<br />

Translational Epidemiology and Department <strong>of</strong> Social<br />

Medicine, University <strong>of</strong> Bristol, Bristol, UK 2. Children’s<br />

Hospital <strong>of</strong> Philadelphia, Philadelphia, US 3. Centre for Child<br />

and Adolescent Health, Department <strong>of</strong> Community Based<br />

Medicine, University <strong>of</strong> Bristol, Bristol, UK<br />

* B.Glaser@bris.ac.uk<br />

Autism spectrum disorders (ASDs) are highly heritable<br />

childhood neuro-developmental conditions, which are<br />

characterised by impairment <strong>of</strong> social interaction and<br />

communication, and repetitive interests, behaviours, and<br />

activities. Recent genome-wide analysis revealed evidence for<br />

a genetic variant on 5p14.1 (rs4307059), which is associated<br />

with risk for ASD. It is unclear however if this risk variant<br />

also relates to a quantitative trait locus underlying a broader<br />

autism phenotype. We therefore studied association between<br />

ASD-related phenotypes and variation in rs4307059 in up to<br />

7313 ALSPAC children, representative <strong>of</strong> the general<br />

population. For this we explored a battery <strong>of</strong> cognitive tests<br />

and questionnaires, administered between the age <strong>of</strong><br />

15-months and 12-years, measuring early vocabulary and<br />

communication patterns<br />

(MacArthur-Communicative-Development-Inventory/MacArt<br />

hur-Toddler-Communication-Questionnaire), intelligibility<br />

(ALSPAC measure), social behaviour and behavioural<br />

difficulties<br />

(Emotionality-Activity-Sociability-Temperament-Survey;<br />

Revised-Rutter-Parent-Scale for Preschool Children;<br />

Standard-Assessment-Tests <strong>of</strong> social skills;<br />

Strengths-and-Difficulties-Questionnaire, Cambridge<br />

Hormones-and-Moods-Project-Friendship-Questionnaire;<br />

Special-<strong>Education</strong>al Needs assessment), social communication<br />

(Social-and-Communications-Disorders-Checklist;<br />

Children’s-Communication-Checklist), verbal intelligence,real<br />

world pragmatics and working memory<br />

(Wechsler-Intelligence-Scale-for-Children-III). To overcome<br />

the non-independent nature <strong>of</strong> our data and to account for<br />

multiple testing and missing genotypes, we adopted a<br />

permutation approach. Our study finds empirical evidence for<br />

a joint association effect between ASD-related phenotypes and<br />

rs4307059 (pEmp = 0.0015; SE = 0.0004), which is further<br />

strengthened when the direction <strong>of</strong> each single effect is taken<br />

into account (pEmp = 0.0004; SE = 0.0002). Our results show<br />

that the observed pr<strong>of</strong>ile <strong>of</strong> associations is unlikely to be the<br />

result <strong>of</strong> chance suggesting that common variation in<br />

rs4307059 is also associated with a broader ASD-phenotype.


33 SOCIAL RESPONSIVENESS: A QUANTITATIVE<br />

INTERMEDIATE PHENOTYPE IN PARENTS OF<br />

CHILDREN WITH AUTISM SPECTRUM DISORDERS.<br />

W. De la Marche* (1)<br />

1. Child and Adolescent Psychiatry Dep., UPC-K.U.Leuven,<br />

Belgium<br />

* wouter.delamarche@uz.kuleuven.be<br />

Background: Although heritability <strong>of</strong> Autism Spectrum<br />

Disorders (ASD) is up to 90%, finding specific genetic origins<br />

has proven to be very difficult. A combination <strong>of</strong> several<br />

genetic variants (Copy Number Variants, Single Nucleotide<br />

Polymorphisms) with each a small effect on the ASD<br />

susceptibility, inherited from both parents, might be the cause<br />

<strong>of</strong> most <strong>of</strong> the ASD cases (Steyaert & De la Marche, 2008).<br />

As a consequence, most parents <strong>of</strong> a child with ASD should be<br />

carriers <strong>of</strong> some <strong>of</strong> those risk factors without having ASD<br />

themselves. Carrying such a risk factor might result in<br />

subclinical autistic traits. The Social Responsiveness Scale<br />

(Constantino et al., 2003) has proven to reliably measure<br />

autistic traits in a quantitative way in the general as well as in<br />

the clinical population. Objectives: To assess if parents <strong>of</strong> a<br />

child with ASD show autistic traits, intermediate between<br />

control adults and adults with ASD. Methods: We asked<br />

control adults, parents <strong>of</strong> children with ASD and adults with<br />

ASD to fillout the Dutch self-report version <strong>of</strong> the Social<br />

Responsiveness Scale and we asked their partners or parents to<br />

fill out the other-report form. (Preliminary) Results: Total SRS<br />

scores differ significantly between the three groups, both in<br />

males and females (except for other-report in females,<br />

possibly due to smaller sample), with parents <strong>of</strong> children with<br />

ASD showing intermediate quantitative autistic traits.<br />

Conclusions: These results support the hypothesis <strong>of</strong> multiple<br />

genetic variants with a small effect each (additive polygenic<br />

effect) inherited by the parents as a cause <strong>of</strong> ASD.<br />

34 HLA AND AUTISM: CASE REPORT<br />

M. Cojocaru* (1), C. Albu (1), D. Albu (1), E. Severin (1)<br />

1. "Carol Davila" Univ Med Pharm<br />

* genetics.dentistry@gmail.com<br />

Background: Certain HLA alleles are known to be associated<br />

with disease susceptibility. Few previous studies have shown<br />

the association and linkage <strong>of</strong> HLA-A2 class 1 allele with<br />

autism and other studies suggested that DR4 and DR13 alleles<br />

are linked to autism spectrum disorder. Objective: our aim was<br />

to evaluate HLA haplotype in a Caucasian family with a twin<br />

pair discordant for autism. Subjects and Methods: A pair <strong>of</strong><br />

4-year-old twin male was evaluated and diagnosed by a<br />

multidisciplinary team. Diagnostic criteria for autism were as<br />

follows: impairment in social interaction, impairments in<br />

communication and restricted repetitive and stereotyped<br />

patterns <strong>of</strong> behavior, interests, and activities. The twin<br />

zygosity was determined according to blood test: ABO blood<br />

type system, Rh blood group system and HLA haplotype. The<br />

parents had been typed for HLA haplotypes too. Results:<br />

There was no family history <strong>of</strong> autism or autoimmune<br />

disorders. The diagnosis <strong>of</strong> autism was early made because <strong>of</strong><br />

the language delay was present. Both twins showed language<br />

impairment. One twin met criteria for autism and his co-twin<br />

did not. Both twins had HLA A2 B18 DR13. The haplotype<br />

was inherited from their mother. Conclusions: our results<br />

suggest the role <strong>of</strong> HLA A2 and DR13 alleles as a genetic<br />

susceptibility factor <strong>of</strong> autism.


35 THE OXYTOCIN RECEPTOR GENE (OXTR) IS<br />

ASSOCIATED WITH AUTISM IN JAPANESE<br />

POPULATION<br />

X. Liu* (1), Y. Kawamura (2), T. Shimada (2), T. Otowa (2),<br />

S. Koishi (2), Y. Kano (3), N. Kato (4), K. Tokunaga (5), T.<br />

Sasaki (6)<br />

1. Department <strong>of</strong> Human Genetics, Graduate School <strong>of</strong><br />

Medicine, University <strong>of</strong> Tokyo 2. Department <strong>of</strong><br />

Neuropsychiatry, Graduate School <strong>of</strong> Medicine, University <strong>of</strong><br />

Tokyo 3. Department <strong>of</strong> Child Psychiatry, The University <strong>of</strong><br />

Tokyo Hospital 4. Department <strong>of</strong> Psychiatry, School <strong>of</strong><br />

Medicine, Showa University 5. Department <strong>of</strong> Human<br />

Genetics, Graduate School <strong>of</strong> Medicine, University <strong>of</strong> Tokyo<br />

6. Health Service Center, University <strong>of</strong> Tokyo<br />

* liuxiaoxi@m.u-tokyo.ac.jp<br />

Purpose: Autism spectrum disorder (ASD) (OMIM: 209850)<br />

is a neurodevelopment disorder characterized by impairment<br />

in social interaction and communication as well as restricted<br />

and stereotyped behaviors and interests. Previous research has<br />

suggested the involvement <strong>of</strong> OXTR in the susceptibility to<br />

ASD, and positive associations between OXTR and ASD were<br />

observed in 4 independent studies from different ethnics<br />

groups, In addition genome-wide linkage studies also have<br />

indentified the chromosomal region Chr3 p25.3 as a candidate<br />

region which harbors the OXTR gene, Despite these emerging<br />

evidence, partial inconsistency <strong>of</strong> the previous findings<br />

together with statistical issue regarding sample size and<br />

statistical method still deserve further study In this study,we<br />

attempted to replicate these recently published findings.<br />

Method: we conducted 1) family based association test and 2)<br />

population-based case-control test by using a total <strong>of</strong> 1210<br />

Japanese samples, and 14 SNPs <strong>of</strong> OXTR gene were<br />

genotyped. Result: Significant associations with ASD for both<br />

single SNPs and haplotypes were observed in the present<br />

study. We confirmed the previous associations <strong>of</strong> 1)<br />

rs2254298 and rs53576 observed in Chinese population and <strong>of</strong><br />

the 2) 5’-flanking region <strong>of</strong> the exon 4 and a 3.5 kb region<br />

within the third intron which were found in Caucasian<br />

population. Conclusion: Our study demonstrates that SNPs<br />

and haplotypes in the OXTR gene confer risk for ASD.<br />

36 THE AUTISM GENOME PROJECT:<br />

GENOME-WIDE ASSOCIATION STUDIES IN AUTISM<br />

R. Anney* (1), o. the Autism Genome Project<br />

1. Trinity College Dublin<br />

* anneyr@tcd.ie<br />

Autism spectrum disorders (ASD) are early onset<br />

neurodevelopmental disorders affecting approximately 1/150<br />

individuals and characterized by deficits in reciprocal social<br />

interaction, communication and restricted and repetitive<br />

patterns <strong>of</strong> behaviours and interests. Evidence to date supports<br />

high heritability and a complex genetic aetiology. The Autism<br />

Genome Project (AGP) is a large international collaboration<br />

facilitating gene identification through access to a large<br />

phenotypic and genetic dataset. We will present results from<br />

genome-wide association (GWA) analyses from<br />

approximately 3000 families genotyped using the Illumina 1M<br />

BeadChip. To date results are available from over 1500 ASD<br />

families. This resource is sufficiently large to implement<br />

multiple analytical strategies for localizing CNVs and SNP<br />

loci affecting risk for ASD. We are presenting data from<br />

association analysis from the additive model across ancestral<br />

groups (all, European) and partitions <strong>of</strong> the data split along<br />

two diagnostic groups; any ASD (spectrum) and a narrow<br />

diagnostic group (autism).Results to date implicate one gene<br />

from the main analyses, namely MACROD2. While<br />

significant for a single GWA, the p-value falls just below the<br />

standard for GWA <strong>of</strong> approximately p=7.2x10-8. Other genes<br />

previously described as associated to autism have also been<br />

implicated by these analyses. The AGP is currently expanding<br />

the dataset to complete a replication analysis. The analyses we<br />

present will highlight both new loci and loci that continue to<br />

show compelling evidence for association.


37 AUTISM SPECTRUM DISORDER AND PARENTAL<br />

AGE IN JAPAN<br />

T. Shimada* (1), Y. Kawamura (1), X. Liu (2), T. Otowa (1),<br />

C. Kakiuchi (1), T. Umekage (3), H. Nishida (4), T. Sugiyama<br />

(5), Y. Kano (6), K. Kasai (1), T. Sasaki (7)<br />

1. Department <strong>of</strong> Neuropsychiatry, Graduate School <strong>of</strong><br />

Medicine, University <strong>of</strong> Tokyo 2. Department <strong>of</strong> Human<br />

Genetics, Graduate School <strong>of</strong> Medicine, University <strong>of</strong> Tokyo<br />

3. Division for Environment, Health and Safety, University <strong>of</strong><br />

Tokyo 4. Asunaro Hospital for Child and Adolescent<br />

Psychiatry 5. Aichi Children’s Health and <strong>Medical</strong> Center 6.<br />

Department <strong>of</strong> Child Psychiatry, University <strong>of</strong> Tokyo 7.<br />

Office for Mental Health Support, University <strong>of</strong> Tokyo<br />

* shimada-t@umin.net<br />

Background: Genetic factor are strongly associated with<br />

autism spectrum disorder (ASD), but other factors may also<br />

play a significant role. Recent studies suggest that increased<br />

maternal age and paternal age at birth are both associated with<br />

an elevated risk <strong>of</strong> ASD. The biological mechanisms<br />

underlying these relationships are not known. The association<br />

maternal age with ASD may be because <strong>of</strong> the increased risk<br />

<strong>of</strong> chromosomal abnormalities in ova <strong>of</strong> increased age or as a<br />

result <strong>of</strong> unstable trinucleotide repeats. Paternal age may be<br />

associated with ASD due to de novo spontaneous mutations<br />

that accumulate with advancing age in spermatagonia, the<br />

association may be confounded by sociocultural<br />

environmental factors. We investigated parental age at birth <strong>of</strong><br />

children with ASD participating genetic study. Methods:<br />

Maternal and paternal age at birth <strong>of</strong> 96 ASD children born in<br />

1965-2002 were compared with those <strong>of</strong> children in the<br />

Japanese general population using national statistics. Results:<br />

The mean maternal age and paternal age at birth <strong>of</strong> ASD<br />

children were 31.3 year and 34.4 year, respectively, which<br />

were significantly higher than the expected parental age (29.0<br />

year and 31.5 year, respectively) according to the national<br />

statistics. Conclusions: Advanced parental age at birth might<br />

be associated with the risk <strong>of</strong> ASD in the Japanese population.


EARLY CAREER INVESTIGATOR TRACK:<br />

CANDIDATE GENES<br />

ECI 7 USING DIMENSIONAL MODELS OF NICOTINE<br />

DEPENDENCE TO AID IN GENE IDENTIFICATION ---<br />

AN EXAMINATION OF THE NICOTINIC RECEPTOR<br />

GENES<br />

L. Chen* (1), R. Grucza (1), E. Johnson (2), N. Breslau (3),<br />

D. Hatsukami (4), N. Saccone (5), T. Baker (6), S. Smith (6),<br />

A. Goate (1), J. Rice (1), L. Bierut (1)<br />

1. Department <strong>of</strong> Psychiatry, Washington University School <strong>of</strong><br />

Medicine, St. Louis, MO, USA 2. Research Triangle Institute<br />

International3, Research Triangle Park, North Carolina 3.<br />

Department <strong>of</strong> Epidemiology, Michigan State University, East<br />

Lansing, Michigan 4. Department <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Minnesota, Minneapolis, Minnesota, USA 5. Department <strong>of</strong><br />

Genetics, Washington University School <strong>of</strong> Medicine, St.<br />

Louis, MO, USA 6. Department <strong>of</strong> Medicine, Center for<br />

Tobacco Research and Intervention, University <strong>of</strong> Wisconsin<br />

School <strong>of</strong> Medicine. Madison, WI<br />

* chenli@psychiatry.wustl.edu<br />

Introduction: Existing evidence suggests that nicotine<br />

dependence is associated with genetic variants in nicotinic<br />

receptor genes with phenotypic measures that strongly reflect<br />

smoking heaviness such as cigarettes per day and FTND.<br />

Nicotine dependence is multifactorial and it is unknown how<br />

the nicotinic receptor genes relate to the range <strong>of</strong> dependence<br />

factors. Methods: A community sample <strong>of</strong> 2073 European<br />

American subjects participated in the COGEND study.<br />

Phenotypic definitions including FTND, DSM, NDSS,<br />

WISDM scores and subphenotypes including PDM, SDM,<br />

daily cigarette use, and urgency to smoke in the morning were<br />

assessed. Two hundred and twenty four SNPs in the α5-α3-β4<br />

nicotinic receptor genes were analyzed. Results: The<br />

multifactorial WISDM, and NDSS dependence measures<br />

shared numerous significant associations with genetic variants,<br />

while the DSM4 diagnostic phenotype shared few relations.<br />

We found cigarette per day, NDSS drive subscale, WISDM<br />

PDM subscales were highly associated with SNP rs16969968<br />

and WISDM PDM subscale was highly associated with SNP<br />

rs578776. We found the subphenotype craving showing the<br />

strongest associations with both rs16969968 (involved in<br />

amino acid changes in nicotinic receptor) and rs588765<br />

(involved in mRNA expression in nicotinic receptor). We<br />

demonstrated the differential associations between other<br />

nicotinic receptors genes and dimensional phenotypes.<br />

Conclusion: The phenotypic associations with rs16969968 are<br />

strongest with a heavy smoking phenotype. Both SNPs<br />

involved in biological mechanisms <strong>of</strong> nicotinic receptor<br />

expression is validated by their convergent phenotypic<br />

presentation in the craving subphenotype <strong>of</strong> WISDM. These<br />

dimensional phenotypes and subphenotypes can be helpful in<br />

refining the phenotypic characteristics associated with specific<br />

gene clusters.<br />

ECI 8 FUNCTIONAL POLYMORPHISMS IN THE<br />

INTERLEUKIN 6 AND TUMOR NECROSIS FACTOR<br />

GENES IN OBSESSIVE-COMPULSIVE DISORDER<br />

C. Cappi (1), R. Muniz (1), A. Sampaio (1), S. Palácios (1), Q.<br />

Cordeiro* (1), H. Brentani (1), A. Marques (2), H. Vallada<br />

(1), E. Miguel (1), L. Guilherme (1), A. Hounie (1)<br />

1. Sao Paulo University 2. National Institute <strong>of</strong> Health<br />

* qcordeiro@yahoo.com<br />

Obsessive-compulsive disorder (OCD) is characterized by<br />

recurrent unwanted thoughts (obsessions), usually<br />

accompanied by repetitive behaviors (compulsions) intended<br />

to alleviate anxiety caused by obsessions. The role <strong>of</strong> the<br />

immune system in central nervous system processes has long<br />

been under investigation in the pathogenesis <strong>of</strong> a variety <strong>of</strong><br />

neuropsychiatric disorders. In this study, we investigated<br />

polymorphisms <strong>of</strong> the genes that codify the cytokines TNF<br />

and IL6 using a trio analysis approach, which minimizes<br />

population stratification bias. Blood samples were obtained<br />

from 71 OCD patients recruited at the University <strong>of</strong> São Paulo<br />

Clinical Hospital and their 166 biological first-degree<br />

relatives. All participants gave written informed consent. All<br />

patients who had DSM-IV criteria for OCD and one or more<br />

first-degree biological relatives (142 parents and 36 siblings)<br />

willing to participate in the study were included. The analyses<br />

were completed with PLINK s<strong>of</strong>tware. The transmission<br />

disequilibrium test was performed to measures the<br />

over-transmission <strong>of</strong> an allele from heterozygous parents to<br />

affected <strong>of</strong>fspring. We found a significant association between<br />

TNF rs1800795 polymorphism and OCD. The TNF rs1800629<br />

was significantly associated only on the parental discordance<br />

test. We haven’t found a significant association between IL6<br />

rs13447446 polymorphism and OCD. This finding supports a<br />

role for TNF in OCD. Plasma cytokine evaluation in OCD<br />

patients as well as their polymorphisms may contribute to the<br />

understanding <strong>of</strong> the role <strong>of</strong> immune factors in OCD etiology.


ECI 9 DRD1 PROMOTER REGION POLYMORPHISM<br />

(RS4532) IS RECESSIVELY ASSOCIATED WITH<br />

SCHIZOPHRENIA MARKED BY MILD SYMPTOMS<br />

AND SPARED WORKING MEMORY<br />

P. DeRosse* (1), T. Lencz (2), K. Burdick (2), A. Malhotra<br />

(1)<br />

1. Division <strong>of</strong> Psychiatry Research, The Zucker Hillside<br />

Hospital 2. Center for Translational Psychiatry, The Feinstein<br />

Institute for <strong>Medical</strong> Research<br />

* pderosse@lij.edu<br />

Background: A recent meta-analysis (Allen et al., Nat Gen, 40,<br />

827-834) indicated that a promoter region variant (rs4532,<br />

-48A/G) in the DRD1gene was one <strong>of</strong> only four SNPs with<br />

strong evidence for association to schizophrenia (SZ). Even<br />

so, the effect size was modest (allelic OR=1.18,<br />

95%CI=1.01-1.38), with relatively modest sample size (725<br />

cases). Additionally, mode <strong>of</strong> transmission was not clarified,<br />

and effects on clinical phenotypes such as symptom scores and<br />

cognitive performance has not been reported. Methods:<br />

Caucasian SZ cases (n=280) and controls (n=250) were<br />

genotyped using the Affymetrix 500K array, and rs4532<br />

genotype was imputed with high confidence (>0.87) using<br />

HapMap/Mach. Symptom scores were assessed using the<br />

SCID-IV, based on lifetime presence indicated by self-report,<br />

and, in most cases, verified by informant report and/or<br />

extensive chart records. A brief neurocognitive battery was<br />

administered containing 6 primary measures. It was<br />

hypothesized that digit span performance would be<br />

specifically affected by DRD1 genetic variation based on<br />

long-standing evidence <strong>of</strong> a relationship between prefrontal<br />

D1 receptor function and working memory performance.<br />

Results: Patients with schizophrenia were significantly<br />

(p=0.005; OR=1.44; 95%CI=1.11-1.86) more likely to carry<br />

the minor (G) allele, strengthening the prior meta-analytic<br />

results and increasing the patient sample by nearly 40%.<br />

Additionally, results were most consistent with a recessive<br />

model <strong>of</strong> transmission (OR=2.2; 95%CI=1.25-3.90). While<br />

GG homozygotes were thus more than twice as likely to be<br />

classified as cases, these patients were marked by reduced<br />

severity across multiple symptom domains (auditory<br />

hallucinations, alogia, and disorganized behavior). Moreover,<br />

patients carrying the GG genotype demonstrated relatively<br />

spared working memory (approximately 1 point difference on<br />

digits backward, p=.003). Conclusions: A SNP in the<br />

promoter region <strong>of</strong> DRD1 is reliably associated with<br />

schizophrenia, possibly in a recessive mode <strong>of</strong> transmission.<br />

Interestingly, this SNP appears to be associated with a<br />

relatively mild clinical and cognitive presentation.<br />

ECI 10 CHILDHOOD MALTREATMENT AND MAOA<br />

GENOTYPE: AN INVESTIGATION OF MAIN AND<br />

INTERACTIVE EFFECTS ON DIVERSE CLINICAL<br />

EXTERNALIZING OUTCOMES<br />

J. Derringer* (1), R. Krueger (1), D. Irons (2), W. Iacono (2)<br />

1. Department <strong>of</strong> Psychology, Washington University in St.<br />

Louis 2. Department <strong>of</strong> Psychology, University <strong>of</strong> Minnesota<br />

* derringer@wustl.edu<br />

We studied the impact <strong>of</strong> MAOA genotype and retrospectively<br />

reported childhood maltreatment (physical and/or sexual) on<br />

young adult lifetime clinical externalizing symptoms<br />

(substance problems, adult antisocial behavior, and conduct<br />

disorder). Participants were 841 individual twins from the<br />

community-based Minnesota Twin Family Study assessed<br />

through age 25. Childhood maltreatment was associated with<br />

increasing symptoms for substance problems, antisocial<br />

behavior, and conduct disorder. No significant main effects <strong>of</strong><br />

MAOA were observed on any phenotype, nor were there<br />

significant GxE interactions between MAOA and harsh<br />

discipline on any phenotype or a significant interaction<br />

between MAOA and childhood sexual assault on substance<br />

problems. We did, however, find evidence that childhood<br />

sexual assault interacted with MAOA genotype to predict<br />

antisocial behavior (p=0.014, R2=0.4%) and conduct disorder<br />

symptoms (p=0.007, R2=0.7%). Individuals with the low<br />

MAOA activity genotype who reported childhood sexual<br />

assault had more symptoms than individuals with either the<br />

high MAOA activity genotype and/or no history <strong>of</strong> childhood<br />

sexual assault. These findings suggest that the previously<br />

reported interaction between MAOA and childhood<br />

maltreatment is specific to the aggressive subset <strong>of</strong><br />

externalizing disorders.


ECI 11 ASSOCIATION BETWEEN SIGMA1<br />

RECEPTOR GENE A61C (GLN2PRO) AND<br />

SEROTONIN RECEPTOR 1A PROMOTER -1019C>G<br />

POLYMORPHISM AND PANIC DISORDER IN<br />

JAPANESE POPULATION.<br />

Y. Konishi* (1), H. Tanii (1), T. Otowa (2), T. Sasaki (3), H.<br />

Kaiya (4), Y. Okazaki (5)<br />

1. Department <strong>of</strong> Psychiatry, Mie University 2. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> Tokyo 3. Health Service Center,<br />

University <strong>of</strong> Tokyo 4. Warakukai Panic Disorder Clinical<br />

Research Center 5. Metropolitan Matsuzawa Hospital<br />

* konikonikoni1029@hotmail.co.jp<br />

Panic disorder (PD) is a disease characterized by an uneasy<br />

feeling accompanied by two or more physical symptoms<br />

caused by abnormality <strong>of</strong> the autonomic nerve system, such as<br />

palpitation, perspiration, chest unpleasantness, a feeling <strong>of</strong><br />

dizziness, and the like. These symptoms are not necessarily<br />

repeated with each attack <strong>of</strong> PD. PD is considered to have<br />

significant genetic factors in the etiology. Sigma1 receptor<br />

(Sig-1R) is considered to modulate the function<br />

N-methyl-D-aspartate (NMDA) receptors and have strong<br />

affinity with fluvoxamine, a typical therapeutic drungs <strong>of</strong> PD.<br />

Serotonin receptor 1A (5-HT1A) gene is proposed to be one <strong>of</strong><br />

pathogenic factors for panic disorder related with serotonin<br />

system. Regarding 5-HT1A receptor promoter -1019C>G<br />

polymorphism, it was shown that the G allele significantly<br />

correlated with panic disorder with agoraphobia. A study <strong>of</strong><br />

PD patients using fMRI showed that for patients homozygous<br />

for G allele, fearful stimuli were associated with a decreased<br />

activation <strong>of</strong> the right prefrontal cortex. In these context, we<br />

studied in this study single-nucleotide polymorphisms (SNPs)<br />

<strong>of</strong> Sig-1R A61C (Gln2Pro) and 5-HT1A receptor promoter<br />

-1019C>G polymorphism in Japanese patients with PD and<br />

controls. No significant association was observed between the<br />

two SNPs and PD. We therefore conclude that the Sig-1R<br />

A61C (2Gln2Pro) and 5-HT1A receptor -1019C>G<br />

polymorphism may not play a major role in PD in Japanese<br />

populations.<br />

ECI 12 AMYLOID PRECURSOR PROTEIN-BINDING<br />

PROTEIN A2 (APBA2) IS AN AUTISM CANDIDATE<br />

GENE<br />

T. Babatz (1), R. Kumar* (1), J. Sudi (1), W. Dobyns (1), S.<br />

Christian (1)<br />

1. The University <strong>of</strong> Chicago<br />

* rkumar1@bsd.uchicago.edu<br />

Background: We have previously reported an autistic proband<br />

and affected brother harboring a maternally inherited 289 kb<br />

microduplication <strong>of</strong> 15q13.1, a region previously associated<br />

with autism, schizophrenia and mental retardation. APBA2 is<br />

located within 15q13.1 and encodes a synaptic protein that<br />

interacts directly with the autism-associated protein NRXN1.<br />

Mice deleted for APBA2 show abnormal social behaviors. We<br />

hypothesize that rare genetic variation <strong>of</strong> APBA2 underlies<br />

risk for autism. Objectives: To screen APBA2 for rare<br />

nucleotide variation in autistic individuals and controls.<br />

Methods: Sanger sequencing was used to screen the coding<br />

regions <strong>of</strong> APBA2 in 512 autism samples and 463 control<br />

individuals. Bioinformatics approaches were used to evaluate<br />

the functional effects <strong>of</strong> APBA2 variants. Results: We<br />

identified seven novel autism-specific non-synonymous<br />

coding variants, all <strong>of</strong> which are predicted to affect protein<br />

function and/or alter residues that are conserved across all 28<br />

species examined. We identified four non-synonymous<br />

variants specific to controls, indicating no significant<br />

difference in mutation burden between cases and controls. Of<br />

particular interest were two non-synonymous coding variants<br />

that were identified in two sibs with autism: (1) a<br />

paternally-inherited heterozygous 6-bp deletion and (2) a<br />

maternally-inherited heterozygous missense mutation. These<br />

results suggest compound heterozygous mutations <strong>of</strong> APBA2<br />

in this autism sibship. Conclusions: This work represents the<br />

first sequence-level evaluation <strong>of</strong> APBA2 as an autism<br />

candidate. The co-occurrence <strong>of</strong> two non-synonymous<br />

mutations in both affected siblings in a single family, each<br />

transmitted from a different unaffected parent, may indicate a<br />

causative role for APBA2 mutations in this isolated case.


CANDIDATE GENES<br />

38 GWAS PROGRAM OF CHINA AND ITS<br />

APPLICATION IN THE STUDY OF PSYCHIATRIC<br />

DISEASES<br />

L. He* (1), Y. Shi (1), G. He (1), G. Feng (1)<br />

1. Bio-X Center, Shanghai Jiao Tong University<br />

* helinhelin@gmail.com<br />

GWAS, a powerful measure although it can be not perfect<br />

from some recent overviews, has geared up association studies<br />

<strong>of</strong> causative genes to common complex diseases, including<br />

psychiatric diseases that have caused severe problems for<br />

societies, in the West particularly. As a slow coach, however,<br />

China finally decided to initiate its own nationwide GWAS<br />

program. It can be pretty late but fortunately as the etiologies<br />

<strong>of</strong> those diseases are versatile and the genetic backgrounds<br />

vary in different populations, it is very necessary to study the<br />

genetic factors in each population individually. In recent<br />

years, Chinese researchers have started to generate some data<br />

based on GWAS including genome-wide association, cnv,<br />

epigenomics, and microRNA-based work to dig out genetic<br />

etiology <strong>of</strong> some psychiatric diseases with the Chinese<br />

samples. In my presentation, I will show recent advances with<br />

several GWAS-based studies <strong>of</strong> psychiatric diseases.<br />

39 MAJOR DEPRESSION, ANXIETY AND THE<br />

NOREPINEPHRINE TRANSPORTER<br />

H. Buttenschøn* (1), H. Buch (1), A. Kristensen (1), J.<br />

Thomsen (2), S. Mikkelsen (2), J. Bonde (3), H. Kolstad (4),<br />

L. Kærlev (4), A. Kærgaard (4), J. Andersen (4), A. Hansen<br />

(5), R. Rugulies (5), A. Børglum (6), O. Mors (1)<br />

1. Centre for Psychiatric Research, Aarhus niversitetshospital,<br />

Risskov, Denmark 2. Department <strong>of</strong> Occupational Medicine,<br />

Glostrup University Hospital, Denmark 3. Bispebjerg<br />

University Hospital, Denmark 4. Danish Ramazzini Centre,<br />

Department <strong>of</strong> Occupational Medicine, Aarhus University<br />

Hospital, Denmark 5. National Research Centre for the Work<br />

Environment, Copenhagen, Denmark 6. Inst. <strong>of</strong> Human<br />

Genetics, Aarhus University, Denmark<br />

* henrbutt@rm.dk<br />

Background: Genetic factors play important roles in the<br />

etiology <strong>of</strong> anxiety and depressive disorders. Symptoms <strong>of</strong><br />

anxiety and depression commonly co-occur, and high rates <strong>of</strong><br />

comorbidity among anxiety and depressive disorders are<br />

well-established. The aim <strong>of</strong> the present study was to explore<br />

321 tagSNPs in 20 candidate genes for association with<br />

anxiety and/or depression, respectively. The genes were<br />

selected based on their function in norepinephrine signalling,<br />

serotonin signalling, the hypothalamic-pituitary-adrenal<br />

(HPA) axis or their involvement in neuroplasticity. Method:<br />

The case-control sample consisted <strong>of</strong> 331 patients with major<br />

depression, 219 patients with anxiety disorders and 297<br />

ethnically matched controls. Cases and controls were<br />

interviewed with the semi-structured diagnostic interview<br />

Schedules for Clinical Assessment in Neuropsychiatry<br />

(version 2.1). The genotyping were performed using the<br />

Sequenom platform (Sequenom, Inc, San Diego, USA). To<br />

test for allelic, genotypic, and haplotypic association the<br />

PLINK s<strong>of</strong>tware was used<br />

(http://pngu.mgh.harvard.edu/purcell/plink/). Results: A<br />

number <strong>of</strong> genes showed significant association with<br />

depression and anxiety. However, the norepinephrine<br />

transporter (NET) appeared to be the most significantly<br />

associated gene in the anxiety sample. Five SNPs within the<br />

gene appeared to be allelic, genotypic and haplotypic<br />

associated with anxiety. The NET gene also appeared to be<br />

significantly associated with depression. In total six SNPs,<br />

five <strong>of</strong> which also were associated with anxiety, were allelic,<br />

genotypic and haplotypic associated with depression.<br />

Discussion: The present study suggested the NET gene as a<br />

common susceptibility gene for depression and anxiety.


40 CNV OVERLAPPING GSK3beta (GLYCOGEN<br />

SYNTHASE KINASE-3beta) GENE AND ITS<br />

ASSOCIATION WITH MOOD DISORDERS<br />

PHENTOYPES IN SPANISH POPULATION<br />

E. Saus* (1), V. Soria (2), G. Escaramís (3, 5), J. Crespo (2,<br />

5), J. Valero (6), A. Guitiérrez-Zotes (6), L. Martorell (6), E.<br />

Vilella (6), J. Menchón (2, 5), X. Estivill (1, 3), M.<br />

Urretavizcaya (2, 5), M. Gratacòs (1, 3)<br />

1. Genetic Causes <strong>of</strong> Disease Group, Genes and Disease<br />

Program, Center for Genomic Regulation (CRG-UPF),<br />

Barcelona, Catalonia, Spain. 2. CIBERSAM (CIBER en Salud<br />

Mental), Mood Disorders Clinical and Research Unit,<br />

Psychiatry Department, Bellvitge University Hospital,<br />

Barcelona, Spain. 3. CIBER en Epidemiología y Salud Pública<br />

(CIBERESP), Instituto de Salud Carlos III, Madrid, Spain 4.<br />

Genetic Causes <strong>of</strong> Disease Group, Genes and Disease<br />

Program, Center for Genomic Regulation (CRG-UPF),<br />

Barcelona, Catalonia, Spain. 5. Department <strong>of</strong> Clinical<br />

Sciences, Bellvitge Campus, Barcelona University, Barcelona,<br />

Spain. 6. Grup d’Investigació en Psiquiatria. Hospital<br />

Universitari Institut Pere Mata, Rovira i Virgili University,<br />

Reus, Spain.<br />

* ester.saus@crg.es<br />

Glycogen synthase kinase-3β (GSK3β) is known to regulate<br />

critical cellular functions and recent findings support that may<br />

play a role in the pathophysiology and treatment <strong>of</strong> mood<br />

disorders (MD). Lachman et al. (2006) found an increased<br />

number <strong>of</strong> gains in a CNV (copy number variant) partially<br />

overlapping with GSK3β in bipolar patients compared with<br />

control individuals. Here, we replicated this experiment in a<br />

Spanish sample <strong>of</strong> MD under the hypothesis that this CNV<br />

could partially underlie the susceptibility to MD and also<br />

could be associated with specific clinical subphenotypes. The<br />

sample consisted <strong>of</strong> 444 MD patients (256 Unipolar<br />

Depressive Disorder, 188 Bipolar Disorder) and 428<br />

psychiatrically screened controls. All samples were genotyped<br />

in triplicate by qPCR. Triplicates undergone a quality control<br />

measure, and a linear mixed model was generated to<br />

normalize all the data allowing a posterior relative<br />

quantification <strong>of</strong> gains and losses. We found a total <strong>of</strong> 42<br />

samples with gains and 13 samples with losses (18 gains and 4<br />

losses in MD patients, and 24 gains and 9 losses in controls).<br />

An association study considering MD vs. controls adjusting by<br />

age and sex was performed, but no significant result was<br />

obtained. Then, we tested 5 clinical subphenotypes in MD:<br />

polarity, seasonality, chronotype, antidepressant treatment<br />

response and age at onset. Only seasonality scores were<br />

nominally associated. While we have not been able to confirm<br />

a direct involvement <strong>of</strong> GSK3β CNV in the susceptibility to<br />

MD, changes in copy number in GSK3β could partially<br />

contribute to seasonal pattern in MD.<br />

41 ASSOCIATION BETWEEN POLYMORPHISMS IN<br />

THE METALLOPHOSPHOESTERASE (MPPE1) GENE<br />

AND BIPOLAR DISORDER<br />

F. Loh<strong>of</strong>f* (1), T. Ferraro (1), E. Brodkin (1), A. Weller (1),<br />

P. Bloch (1)<br />

1. University <strong>of</strong> Pennsylvania<br />

* loh<strong>of</strong>f@mail.med.upenn.edu<br />

Genetic linkage studies in bipolar disorder (BPD) suggest that<br />

a susceptibility locus exists on chromosome 18p11. The<br />

metallophosphoesterase (MPPE1) gene maps to this region.<br />

Dysregulation <strong>of</strong> protein phosphorylation and subsequent<br />

abnormal cellular signaling has been postulated to be involved<br />

in neuropsychiatric disorders thus making MPPE1 a plausible<br />

biological candidate gene for BPD. In this study, we<br />

hypothesized that genetic variation in the MPPE1 gene<br />

contributes to BPD. We tested this hypothesis by genotyping 4<br />

SNPs (rs871044; rs3974590; rs593713; rs602201) in BPD<br />

patients (n=570) and healthy controls (n=725). Genotypes and<br />

allele frequencies were compared between groups using Chi<br />

square contingency analysis. Linkage disequilibrium (LD)<br />

between markers was calculated and estimated haplotype<br />

frequencies were compared between groups. Single marker<br />

analysis revealed an association <strong>of</strong> rs3974590 with BPD<br />

(p=0.009; permutation corrected p=0.046). Haplotype analysis<br />

did not show any significant association with disease after<br />

permutation correction. Our results provide evidence <strong>of</strong> an<br />

association between a polymorphism in the MPPE1 gene and<br />

BPD. Additional studies are necessary to confirm and<br />

elucidate the role <strong>of</strong> MPPE1 as a susceptibility gene for BPD<br />

on chromosome 18p.


42 A MULTI-DEMENSIONAL EVIDENCE-BASED<br />

CANDIDATE GENE PRIORITIZATION APPROACH<br />

FOR SCHIZOPHRENIA AND OTHER COMPLEX<br />

DISEASES<br />

J. Sun (1), P. Jia (1), A. Fanous (2), B. Webb (3), E. van den<br />

Oord (3), B. Riley (3), X. Chen (3), J. Bukszar (3), K. Kendler<br />

(3), Z. Zhao* (1)<br />

1. Vanderbilt University 2. Washington VA <strong>Medical</strong> Center 3.<br />

Virginia Commonwealth University<br />

* zhongming.zhao@vanderbilt.edu<br />

Introduction: During the past decade we have seen an<br />

exponential growth <strong>of</strong> vast amounts <strong>of</strong> genetic data generated<br />

for complex disease studies. Across a variety <strong>of</strong> complex<br />

biological problems currently, there is a strong trend towards<br />

the integration <strong>of</strong> data from multiple sources. So far, candidate<br />

gene prioritization approaches have been designed for specific<br />

purposes, by utilizing only some <strong>of</strong> the available sources <strong>of</strong><br />

genetic studies, or by using a simple weight scheme.<br />

Specifically to psychiatric disorders, there has been no<br />

prioritization approach that fully utilizes all major sources <strong>of</strong><br />

experimental data. Methods: Here we present a<br />

multi-dimensional evidence-based candidate gene<br />

prioritization approach for schizophrenia. In this approach, we<br />

first collect and curate genetic studies for schizophrenia from<br />

four major categories: association studies, linkage analyses,<br />

gene expression, and literature search. Genes in these data sets<br />

are initially scored by category-specific scoring methods.<br />

Then, an optimal weight matrix is searched by a two-step<br />

procedure (core genes and unbiased P values in independent<br />

genome-wide association studies). Finally, genes are<br />

prioritized by their combined scores using the optimal weight<br />

matrix. Results: The prioritized genes were evaluated by the<br />

enriched P values in two independent GWA studies and also<br />

by gene expression pattern in human tissues. The evaluation<br />

suggests this approach generates prioritized candidate genes<br />

that are promising for further analysis or replication. For<br />

example, our follow up gene network/pathway analysis using<br />

these prioritized candidate genes suggested a few novel<br />

candidate genes, one <strong>of</strong> which was successfully verified in our<br />

Irish Case-Control Study <strong>of</strong> Schizophrenia (ICCSS)<br />

sample. Conclusions: The approach can be applied to other<br />

complex diseases, especially other psychiatric disorders.<br />

43 MATERNAL MTHFR C677T GENOTYPE &<br />

DEPRESSED MOOD DURING PREGNANCY AFFECTS<br />

SLC6A4 METHYLATION IN INFANTS AT BIRTH<br />

R. Wade (1), T. Oberlander (1), U. Brain (1), J. Austin* (1),<br />

A. Devlin (1)<br />

1. UBC<br />

* jehannine.austin@ubc.ca<br />

In utero and early postnatal exposure to depressed maternal<br />

mood may program childhood behaviour via epigenetic<br />

processes. Methylenetetrahydr<strong>of</strong>olate reductase (MTHFR) is<br />

an enzyme important for methyl metabolism. A common<br />

variant in the MTHFR gene, C677T, is associated with<br />

depression. We investigated the effect <strong>of</strong> maternal MTHFR<br />

C677T genotype on maternal mood (Edinburgh Postnatal<br />

Depression Scale, EPDS), during pregnancy (n=82 women, all<br />

receiving folate supplements) and on promoter methylation <strong>of</strong><br />

two genes implicated in depression, brain derived<br />

neurotrophic factor, BDNF, and sodium-dependent serotonin<br />

transporter, SLC6A4, in the women and their infants at birth.<br />

Women with the MTHFR 677TT genotype had greater<br />

(P


44 HTR1B AS A RISK PROFILE MAKER IN<br />

PSYCHIATRIC DISORDERS: A REVIEW THROUGH<br />

MOTIVATION AND MEMORY<br />

A. Drago* (1), D. De Ronchi (1), A. Serretti (1)<br />

1. University <strong>of</strong> Bologna<br />

* antonio.drago@unibo.it<br />

HTR1B receptor is involved in the regulation <strong>of</strong> the serotonin<br />

system, playing different roles in specific areas <strong>of</strong> the brain.<br />

Consistently, HTR1B manipulation in animals results into<br />

altered behavior possibly related to psychiatric disorders. We<br />

here review HTR1B, its product and its functional role in the<br />

neuronal nets involved in motivation and memory. Then, we<br />

go the other way round and analyze how and how much<br />

HTR1B variations impact the psychiatric phenotypes mainly<br />

associated with these functions. The neuronal nets involved in<br />

motivational and mnemonic functions are described, and the<br />

role played by HTR1B in these systems is depicted: as a<br />

result, a picture <strong>of</strong> the relevance <strong>of</strong> HTR1B in the modulation<br />

<strong>of</strong> motivation and memory is attained. By contrast, the review<br />

<strong>of</strong> the genetic association studies resulted into partial evidence<br />

sustaining HTR1B relevance toward substance related and<br />

obsessive compulsive disorders and no solid evidence toward<br />

other psychiatric disorders.<br />

45 POLYMORPHIC VARIANTS AND DIFFERENTIAL<br />

EXPRESSION OF DPYSL2 IN SCHIZOPHRENIA<br />

C. Winchester* (1), M. Bailey (2), P. Johnson (2), L.<br />

O'Donovan (2), B. Morris (2), J. Pratt (1)<br />

1. University <strong>of</strong> Strathclyde 2. University <strong>of</strong> Glasgow<br />

* c.winchester@bio.gla.ac.uk<br />

We identified DPYSL2 as a candidate gene for schizophrenia<br />

from a microarray analysis <strong>of</strong> the prefrontal cortex <strong>of</strong> a rodent<br />

phencyclidine (PCP) model <strong>of</strong> schizophrenia, developed in our<br />

laboratory 1,2. This PCP model produces a pattern <strong>of</strong><br />

metabolic hyp<strong>of</strong>unction, neurochemical changes and<br />

behavioural deficits in the prefrontal cortex that closely mirror<br />

the cognitive deficits <strong>of</strong> schizophrenia 1,2. Location <strong>of</strong> the<br />

gene within a linkage and association peak in chr. 8p21, its<br />

function in neuronal development and its interactions with<br />

proteins otherwise implicated in the pathophysiology <strong>of</strong><br />

schizophrenia (e.g. SEMA3A, DISC1, GSK3, CTNNB1) also<br />

support DPYSL2 as a plausible candidate gene for<br />

schizophrenia. We performed a case-control association study<br />

<strong>of</strong> DPYSL2 in DNA samples from 500 UK patients with<br />

DSM-IV schizophrenia and 500 ethnically similar normal<br />

controls. ABI TaqMan assays were used to genotype 11 tag<br />

SNPs across several haplotype blocks. Association <strong>of</strong><br />

schizophrenia with individual SNPs was tested using a variety<br />

<strong>of</strong> Chi squared tests and the Armitage Trend test. Haplotypes<br />

have been predicted and analysis is underway. Five SNPs<br />

encompassing a region in the 3’ half <strong>of</strong> DPYSL2, previously<br />

implicated in schizophrenia, showed modest associations with<br />

schizophrenia in our sample (best P = 0.021 for rs13277175<br />

under a G allele-dominant model; O.R. = 1.35 [1.045-1.752,<br />

95% C.I.]). DPYSL2 transcripts have been analysed by<br />

quantitative RT-PCR in schizophrenia patient and control<br />

post-mortem dorsolateral prefrontal cortex and showed that<br />

transcript levels are associated with genotype at several <strong>of</strong> the<br />

associated SNPs. We present converging evidence for<br />

DPYSL2 as a risk factor for schizophrenia. 1. Cochran et<br />

al.2003. Neuropsychopharmacology. 28 (2):265-275 2. Pratt et<br />

al. 2008. British Journal <strong>of</strong> Pharmacology 153, S465-470 This<br />

work was funded by NHS Scotland R&D for Mental Health<br />

Research.


46 LACK OF SUPPORT FOR THE ASSOCIATION OF<br />

PER3, CSNK1E, AND CLOCK GENE WITH<br />

MORNINGNESS-EVENINGNESS IN YOUNG ADULT<br />

FEMALE<br />

E. Joo* (1), K. Lee (1), H. Kim (1), K. Choi (1)<br />

1. Eulji University<br />

* jej1303@gmail.com<br />

Circadian rhythm is known to be genetic. Multiple SNPs in<br />

clock genes have been searched for a possible association with<br />

circadian rhythm. The association was tested in normal<br />

population as well as psychiatric patients group such as<br />

bipolar disorder. Recent genetic studies reported that in<br />

unrelated bipolar patients some clock genes were associated<br />

with morningness-eveningness measurement scale. We<br />

explored possible association <strong>of</strong> several promising SNP<br />

showed positive association in previous studies with<br />

chronotype in a young adult female group. We collected<br />

phenotype data and blood from 804 young Korean adult<br />

females. Most <strong>of</strong> them were nurses working on shift schedule.<br />

Morningness-eveningness was measured with 13 items<br />

Composite Scale. DNA was extracted from the blood and<br />

genotyping was done in TaqMan assay method. We selected 4<br />

polymorphic sites from three different clock genes: 1 SNP and<br />

1 VNTR from PER3 gene, 1 SNP from CSNK1E gene, and 1<br />

SNP from CLOCK gene. Associations were analyzed based on<br />

individual and epistatic interaction <strong>of</strong> polymorphic sites. We<br />

could not find any association <strong>of</strong> PER3, CSNK1E, and<br />

CLOCK gene with Composite Scale scores in individual<br />

genotype as well as analysis for epistatic interaction between<br />

polymorphic sites. Morningness-eveningness seems complex<br />

as a trait. Clock genes could not support clock genes as a<br />

significant contributor for chronotypes, however the size <strong>of</strong><br />

SNPs to be tested were small in this study. More studies with<br />

bigger sample size including males would be required.<br />

47 ASSOCIATION STUDY OF CHRNA7 WITH<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER IN<br />

KOREAN POPULATION<br />

E. Joo* (1), K. Lee (1), S. Kim (2), Y. Ahn (2), Y. Kim (2)<br />

1. Eulji University 2. Seoul University<br />

* jej1303@gmail.com<br />

CHRNA7 has been known a strong candidate gene for<br />

schizophrenia. It is located on chromosome 15q13-q14, one <strong>of</strong><br />

the replicated linkage spot <strong>of</strong> schizophrenia. We conducted an<br />

association study to replicate previous positive findings in<br />

Korean population. We included 254 patients with<br />

schizophrenia, 193 patients with bipolar disorder type I, 38<br />

patients with bipolar disorder type II, 64 schizoaffective<br />

disorder patients, and 349 controls. Total 898 subjects were<br />

included and genotyping were done for three SNPs <strong>of</strong><br />

CHRNA7. Those 3 SNPs are rs2337506 (A/G, intron),<br />

rs6494223 (C/T, intron), and rs12916879 (A/G, intron).We<br />

found a marginally significant association with rs12916879<br />

with bipolar disorder type I. Both allele and genotype-wise,<br />

we found a weak signal (chisquare=3.568, df=1, p=0.059 for<br />

allele, chisquare=7.504, df=2, p=0.023 for genotype). Sliding<br />

window haplotype analysis using UNPHASED could not find<br />

any significant association. Other polymorphism was not<br />

associated with schizophrenia or bipolar spectrum disorders in<br />

this population. Further analysis with more SNPs is required<br />

to reveal the role <strong>of</strong> CHRNA7 in schizophrenia and bipolar<br />

spectrum disorders.


48 ALLELIC EXPRESSION IMBALANCE ANALYSIS<br />

OF ANK3 AND CACNA1C PROVIDES STRONG<br />

EVIDENCE OF CIS-ACTING VARIATION<br />

AFFECTING EXPRESSION OF BOTH GENES.<br />

E. Quinn* (1), M. Hill (1), R. Anney (1), M. Gill (1), A.<br />

Corvin (1), D. Morris (1)<br />

1. Neuropsychiatric Genetics Research Group, Department <strong>of</strong><br />

Psychiatry and Institute <strong>of</strong> Molecular Medicine, Trinity<br />

College Dublin, Ireland.<br />

* quinne5@tcd.ie<br />

Genome-wide association studies (GWAS) have identified<br />

ANK3 and CACNA1C as susceptibility genes for bipolar<br />

disorder. Available biological information on these genes<br />

suggests a potential molecular mechanism involving ion<br />

channel dysfunction. The associated SNPs at ANK3<br />

(rs10994336) and CACNA1C (rs1006737) are both intronic<br />

with no obvious impact on gene function. Instead <strong>of</strong> affecting<br />

the protein function, these risk variants may impact on gene<br />

regulation affecting expression. Testing for allelic expression<br />

imbalance (AEI) is a method <strong>of</strong> identifying such cis-acting<br />

regulatory polymorphisms by using a coding DNA marker<br />

SNP to assay relative allelic abundance in cDNA compared to<br />

genomic DNA. The primary aim <strong>of</strong> this study was to examine<br />

ANK3 and CACNA1C for regulatory polymorphisms<br />

contributing to differential gene expression in HapMap CEU<br />

lymphoblastoid cell lines. If detected, the secondary aim was<br />

to determine if the strong GWAS signals reported for these<br />

genes could be attributed to these functional SNPs. AEI was<br />

successfully detected at ANK3 and CACNA1C using coding<br />

marker SNPs rs3750800 and rs1544514 respectively,<br />

indicating the presence <strong>of</strong> cis-acting variants at both loci.<br />

There was considerable evidence <strong>of</strong> AEI at ANK3 with more<br />

than half <strong>of</strong> all heterozygous samples (21 out <strong>of</strong> 34) showing<br />

AEI and a small number <strong>of</strong> samples showing near<br />

mono-allelic expression. However, the AEI at either gene<br />

could not be attributed to the GWAS-associated SNPs. These<br />

data indicate that there is genetic variation local to both genes<br />

that is affecting their expression, but this variation is not<br />

responsible for increasing risk <strong>of</strong> bipolar disorder.<br />

49 RELEVANT FINDINGS ON THE GENES OF<br />

SEROTONERGIC SYSTEM AND SUICIDE IN<br />

SLOVENIA<br />

A. Videtic* (1), T. Zupanc (2), J. Balazic (2), P. Pregelj (3),<br />

R. Komel (1)<br />

1. Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, University<br />

<strong>of</strong> Ljubljana, Vrazov trg 2, Ljubljana, SI-1000, Slovenia 2.<br />

Institute <strong>of</strong> Forensic Medicine, Faculty <strong>of</strong> Medicine,<br />

University <strong>of</strong> Ljubljana, Korytkova ulica 2, Ljubljana,<br />

SI-1000, Slovenia 3. University Psychiatric Clinic Ljubljana,<br />

Studenec 48, Ljubljana Polje, SI-1260, Slovenia<br />

* alja.videtic@mf.uni-lj.si<br />

In Europe, countries with the highest suicide rates form a<br />

so-called J-curve, which starts in Finland and extends south to<br />

Slovenia – a country with one <strong>of</strong> the world’s highest suicide<br />

rates. In 2006 there were 529 suicides, with the rate <strong>of</strong> 26.3<br />

suicide victims per 100.000 citizens, most <strong>of</strong> the victims being<br />

between 35 and 64 years years <strong>of</strong> age. Suicide is a complex<br />

phenomenon, influenced by environmental and genetic<br />

factors, where for the latter the most interesting results were<br />

obtained for serotonergic system. In our study we performed<br />

molecular-genetic analysis on polymorphisms 68G>C and<br />

-995G>A in serotonin 2C receptor gene, and -1019 C>G in<br />

serotonin 1A receptor gene, on 334 suicide victims and 312<br />

controls. For 62 suicide victims psychological autopsy was<br />

available. A significantly different distributions for<br />

polymorphism 68G>C were observed in two cases: between<br />

genotypes <strong>of</strong> female suicide victims and controls<br />

(P(FET)=0.008) and in alleles <strong>of</strong> female and male populations<br />

(P=0.005). An excess <strong>of</strong> GG genotype and allele G was<br />

observed. Haplotype analysis showed marginal association <strong>of</strong><br />

haplotype G-C (-995G, 68C) with suicide only in female<br />

population. For polymorphism -1019C>G data on stressful life<br />

events <strong>of</strong> suicide victims was available. More stressful life<br />

events in the month prior to the suicide were reported for the<br />

subgroup with CC genotype (mean number <strong>of</strong> events=2.53;<br />

SD=1.50) in comparison to subgroup with CG/GG genotypes<br />

(mean number <strong>of</strong> events=1.58; SD=1.32; P


50 ASSOCIATION OF THE GABRD GENE AND<br />

CHILDHOOD-ONSET MOOD DISORDERS<br />

Y. Feng* (1), K. Kapornai (2), E. Kiss (2), Z. Tamás (3), L.<br />

Mayer (2), I. Baji (3), G. Daróczi (2), I. Benák (2), V.<br />

Kothencné (2), E. Dombovári (2), E. Kaczvinszk (2), M.<br />

Besnyo (3), J. Gádoros (3), J. Székely (4), M. Kovacs (5), Á.<br />

Vetró (2), J. Kennedy (6), C. Barr (1, 7)<br />

1. Toronto Western Hospital 2. Szeged University 3.<br />

Vadaskert Hospital 4. Semmelweis University 5. University <strong>of</strong><br />

Pittsburgh School <strong>of</strong> Medicine 6. Centre for Addiction and<br />

Mental Health 7. The Hospital for Sick Children<br />

* yfeng@uhnres.utoronto.ca<br />

The chromosome 1p36 region was previous indicated as a<br />

locus for susceptibility to recurrent major depressive disorder<br />

based on a linkage study in a sample <strong>of</strong> 497 sib pairs. We<br />

investigated the GABAA d receptor subunit gene, GABRD, as<br />

a susceptibility gene to childhood-onset mood disorders<br />

(COMD) because <strong>of</strong> substantial evidence implicating<br />

GABAergic dysfunction in mood disorders and the position <strong>of</strong><br />

this gene in the 1p36 linkage region. Using a sample<br />

consisting <strong>of</strong> 603 Hungarian families with a child/adolescent<br />

proband diagnosed with a mood disorder with the onset <strong>of</strong> the<br />

first episode before age 15, we found evidence for association<br />

<strong>of</strong> two polymorphisms located within the gene, rs2376805 and<br />

rs2376803, as well as haplotypes <strong>of</strong> these two markers.<br />

Further, significant evidence <strong>of</strong> association was only observed<br />

in male subjects when the results were analyzed by sex. This<br />

was in contrast with the previous linkage findings, as LOD<br />

scores exceeding 3 were only in female-female pairs in that<br />

study. These findings point to the GABRD gene as a<br />

susceptibility gene for COMD, however, this gene may not<br />

explain the previous linkage finding.<br />

51 GPR50, A CANDIDATE FOR BIPOLAR DISORDER,<br />

AFFECTS NEURITE OUTGROWTH<br />

E. Gruenewald (1), H. Kinnell (2), D. Porteous (1), P.<br />

Thomson* (1)<br />

1. <strong>Medical</strong> Genetics Section, MMC. The University <strong>of</strong><br />

Edinburgh.UK 2. MRC Human Reproductive Science Unit,<br />

Edinburgh, UK<br />

* pippa.thomson@ed.ac.uk<br />

In the search for candidate genes for major mental disorders,<br />

X-linked orphan G protein-coupled receptor 50 (GPR50), also<br />

termed melatonin-related receptor, was identified as a risk<br />

gene for bipolar disorder. G protein-coupled receptors are<br />

important susceptibility candidates for disease as they are<br />

involved in molecular signaling pathways and are major<br />

targets <strong>of</strong> drug treatment. The function <strong>of</strong> GPR50 is unclear<br />

and no ligand has yet been identified. In order to understand<br />

its function, a yeast two-hybrid study was performed,<br />

identifying putative interactors with the C-terminal region <strong>of</strong><br />

GPR50. Key interactors are known to be involved in neuronal<br />

development and lipid metabolism. In this study we report a<br />

follow up <strong>of</strong> one biologically interesting interactor: a potent<br />

neurite outgrowth inhibitor, which has also been implicated in<br />

apoptosis, schizophrenia, bipolar disorder and Alzheimer’s<br />

disease. We confirmed the interaction, by<br />

immunocytochemistry and co-immunoprecipitation.<br />

Furthermore, we performed expression analysis <strong>of</strong> both genes<br />

in the developing mouse brain. Interestingly, we have<br />

identified a possible functional interaction between GPR50<br />

and this protein. Transient GPR50 over-expression results in<br />

extended neurites (p


52 ASSOCIATION STUDY OF GENES INVOLVED IN<br />

A-TO-I RNA EDITING IN BIPOLAR DISORDER AND<br />

SCHIZOPHRENIA PATIENTS FROM AN ISOLATED<br />

NORTHERN SWEDISH POPULATION.<br />

S. Ceulemans* (1), M. Alaerts (1), D. Forero (1), S. De Zutter<br />

(1), K. Norrback (2), L. Moens (1), R. Adolfsson (2), J.<br />

Del-Favero (1)<br />

1. Applied Molecular Genomics Group, VIB Department <strong>of</strong><br />

Molecular Genetics, Belgium University <strong>of</strong> Antwerp (UA),<br />

Antwerpen, Belgium 2. Department <strong>of</strong> Clinical Sciences,<br />

Division <strong>of</strong> Psychiatry, Umeå University, Sweden<br />

* shana.ceulemans@molgen.vib-ua.be<br />

Bipolar (BP) disorder and schizophrenia (SZ) are among the<br />

most commonbrain diseases worldwide with a large impact on<br />

both the patients and theirrelatives’ lives. The observation that<br />

receptor subunits <strong>of</strong> twoimportant neurotransmitters <strong>of</strong> the<br />

central nervous system, glutamate andserotonin, undergo<br />

A-to-I RNA editing by “Adenosine Deaminases Acting on<br />

RNA” (ADARs) supports the hypothesis that this modification<br />

is potentially involvedin the etiology <strong>of</strong> psychiatric disorders.<br />

In our search for BP disorder and SZ susceptibility genes, we<br />

are exploring the malfunctioning and/or deficiency <strong>of</strong> the<br />

A-to-I RNA editingprocess. Hereto, a HapMapbased<br />

association study <strong>of</strong> the genes ADAR, ADARB1 and GRIA2<br />

was performed by genotyping a total <strong>of</strong> 90 (h)tSNPs inBP<br />

patients (N=316); SZ patients (N=486) and matched control<br />

individual (n=512) originating from a northern Swedish<br />

isolated population. Single marker and haplotype specific<br />

statistical analyses showed significant association <strong>of</strong> several<br />

ADARB1 SNPs withBP as well as with LD-blocks <strong>of</strong><br />

ADARB1with BP disorder. In addition, ADARB1gave also<br />

rise to an associated haplotype with SZ. Together, these<br />

resultsindicate that ADARB1 should befurther explored to<br />

unravel its genetic contribution to the etiology <strong>of</strong> BP disorder<br />

and SZ. Furthermore,single marker association was found for<br />

ADARwith SZ (P=0,027) and for GRIA2 withBP disorder<br />

(P=0,006).Taken together, the results <strong>of</strong> this association study<br />

show that A-to-I RNA editing ispotentially involved in the<br />

susceptibility to psychiatric disorders.<br />

53 ASSOCIATION OF NEUROTROPHIN RECEPTOR<br />

(NTRK-3) GENE POLYMORPHISMS WITH BIPOLAR<br />

DISORDERS<br />

S. Djurovic* (1, 2, 3), M. Mattingsdal (1), L. Athanasiu (2, 3),<br />

I. Melles (1, 3), I. Agartz (1, 4), S. Lorentzen (1, 5), G.<br />

Morken (6), O. Andreassen (1, 3)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Oslo, Oslo, Norway 2.<br />

Department <strong>of</strong> <strong>Medical</strong> Genetics; Oslo University Hospital -<br />

Ulleval, Oslo, Norway 3. Department <strong>of</strong> Psychiatry; Oslo<br />

University Hospital - Ulleval, Oslo, Norway 4. Department <strong>of</strong><br />

Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway<br />

5. Department <strong>of</strong> Psychiatry, Oslo University Hospital –Aker,<br />

Oslo, Norway 6. Østmarka Psychiatric Department, St Olavs<br />

Hospital and Institute <strong>of</strong> Neuroscience, Norwegian University<br />

<strong>of</strong> Technology and Science, Trondheim, Norway<br />

* srdjan.djurovic@medisin.uio.no<br />

Recent Genome Wide Association Studies (GWAS) have<br />

revealed novel candidate genes for bipolar disorder, but most<br />

<strong>of</strong> the genetic mechanisms are still unknown. In the present<br />

study we investigated possible genetic variants <strong>of</strong> NTRK-3<br />

associated with bipolar disorder in a homogenous Norwegian<br />

sample (the TOP study), based on its important role in brain<br />

development and plasticity and our recently reported findings<br />

for possible association with schizophrenia. We carried<br />

out GWAS <strong>of</strong> bipolar disorder using the Affymetrix 6.0 array<br />

in case-control samples <strong>of</strong> Norwegian origin (bipolar disorder<br />

(n=225) , healthy controls (n=385)). Twelve NTRK-3 single<br />

nucleotide polymorphisms (SNPs) were nominally associated<br />

with bipolar disorder. This has been followed by replication in<br />

the available schizophrenia sample and confirmed significant.<br />

The SNPs investigated in the current study are all located in<br />

intronic regions <strong>of</strong> NTRK-3. Thus, it is difficult to achieve a<br />

full understanding <strong>of</strong> the effect <strong>of</strong> the different SNPs on the<br />

level <strong>of</strong> diagnosis and brain biology. In order to evaluate the<br />

importance <strong>of</strong> NTRK-3 variants in psychosis, further efforts in<br />

identification <strong>of</strong> quantifiableclinical (endo)phenotypic<br />

variables are needed.


54 ASSOCIATION ANALYSIS WITH DYSBINDIN<br />

GENE: SHARED GENETIC SUSCEPTIBILITY<br />

BETWEEN BIPOLAR DISORDERS AND<br />

SCHIZOPHRENIA?<br />

A. Ruiz* (1), R. Murray (2), J. Powell (2), E. Miranda (1), P.<br />

Sham (3)<br />

1. Departamento de Psiquiatría, Facultad de Medicina,<br />

Hospital Clínico Universidad de Chile, Santiago, Chile. 2.<br />

Institute <strong>of</strong> Psychiatry, King’s College London, University <strong>of</strong><br />

London. UK 3. Genome Research Center, University <strong>of</strong> Hong<br />

Kong, Hong Kong.<br />

* airuiz@uchile.cl<br />

Introduction: Recent studies have suggested an overlap in<br />

genetic susceptibility for bipolar disorders and schizophrenia,<br />

including dysbindin gene. Susceptibility alleles for<br />

neuropsychiatric disorders, and patterns <strong>of</strong> linkage<br />

disequilibrium, are likely to differ in ethnically different<br />

populations. Admixed populations <strong>of</strong>fer an opportunity to<br />

evaluate the role <strong>of</strong> genetics in the aetiology <strong>of</strong> complex<br />

disorders. The objective <strong>of</strong> this study is to carry out an<br />

ssociation analysis <strong>of</strong> dysbindin gene with schizophrenia and<br />

bipolar disorders in a Chilean admixed population. This paper<br />

will present the results found in the association study between<br />

the dysbindin gene and schizophrenia. Method A case-control<br />

and schizophrenic family samples, according to DSM-IV<br />

criteria, were studied. Five hundred and sixty individuals were<br />

included. Ten dysbindin SNPs, previously associated with<br />

schizophrenia, were genotyped. Markers informative for<br />

ancestry were selected to study the Chilean population<br />

structure. Unphased program was used for the statistical<br />

analysis. Population stratification was studied with L-POP<br />

s<strong>of</strong>tware. WHAP program was used in the structured<br />

association analysis. Results No single marker and haplotype<br />

associations were detected (p>0.05). The Chilean samples<br />

showed the same pattern <strong>of</strong> allele frequencies and LD<br />

described for ethnically diverse samples. The analysis <strong>of</strong><br />

population structure found two ancestral populations. The<br />

structured association analysis <strong>of</strong> dysbindin gene, did not<br />

detect possible spurious findings in the Chilean case-control<br />

sample. Conclusion No association between dysbindin gene<br />

and schizophrenia was found. An appropriate interpretation <strong>of</strong><br />

the results must consider all possible limitations, including<br />

false results, due to insufficient statistical power, allelic<br />

heterogeneity, and clinical heterogeneity <strong>of</strong> schizophrenia<br />

55 RGS4 GENE AND SCHIZOPHRENIA: A<br />

CANDIDATE GENE STUDY IN A CHILEAN ADMIXED<br />

POPULATION<br />

A. Ruiz* (1), R. Murray (2), J. Powell (2), E. Miranda (1), P.<br />

Sham (3)<br />

1. Departamento de Psiquiatría, Facultad de Medicina,<br />

Hospital Clínico Universidad de Chile, Santiago, Chile. 2.<br />

Institute <strong>of</strong> Psychiatry, King’s College London, University <strong>of</strong><br />

London. UK 3. Genome Research Center, University <strong>of</strong> Hong<br />

Kong, Hong Kong.<br />

* airuiz@uchile.cl<br />

Introduction: It has been reported that polymorphisms in the<br />

RGS4 gene, on 1q21-22, are associated with schizophrenia.<br />

This study analysed this possible association in a Chilean<br />

admixed sample, ethnically different from populations<br />

examined in previous reports. Methods: The study was carried<br />

out in a case-control sample, composed <strong>of</strong> cases with DSM-IV<br />

schizophrenia and unaffected control subjects; and a sample<br />

<strong>of</strong> DSM-IV schizophrenics families. Five hundred and sixty<br />

individuals were recruited in Santiago, Chile. Four RGS4<br />

markers, previously associated with schizophrenia, and a<br />

number <strong>of</strong> ancestry informative markers were genotyped. The<br />

data were analysed by means <strong>of</strong> the UNPHASED program.<br />

Analysis <strong>of</strong> population stratification, in the case-control<br />

sample, was carried out using L-POP s<strong>of</strong>tware. A structured<br />

association analysis was performed with WHAP program.<br />

Results: The analysis <strong>of</strong> population structure detected<br />

stratification in the case-control sample; however the cases<br />

and controls were well matched. Significant LD was observed<br />

for all pairwise calculations. In both samples, none <strong>of</strong> the<br />

SNPs included in this analysis were found to be associated<br />

with illness (P >0.05), and no significant haplotypic<br />

association was observed (P > 0.05). The structured<br />

association analysis did not show confounding effects <strong>of</strong><br />

population stratification. Conclusions: No evidence was found<br />

to support an association between genetic variants in the<br />

RGS4 gene and schizophrenia in this sample; even though the<br />

genetic analysis suggested that the Chilean sample showed<br />

similarities with ethnically different samples, previously<br />

described as having association with this gene. Among other<br />

explanations, these results might be the consequence <strong>of</strong><br />

inadequate statistical power.


56 CIRCADIAN POLYMORPHISMS IN NIGHTOWLS<br />

D. Kripke* (1), C. Nievergelt (1), K. Rex (1), W. Klimecki<br />

(2), S. Ancoli-Israel (1), T. Shekhtman (1), J. Kelsoe (1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California, San<br />

Diego 2. Department <strong>of</strong> Pharmacology, University <strong>of</strong> Arizona<br />

* dkripke@ucsd.edu<br />

Patients with DSM-IV delayed sleep phase disorder, or<br />

nightowls, have trouble falling asleep until very late at night<br />

and trouble awakening in the morning. This <strong>of</strong>ten-disabling<br />

condition is believed to be partially genetic in origin. It has<br />

been hypothesized that polymorphisms in the genes<br />

comprising the intracellular circadian oscillator system may be<br />

involved, as has been demonstrated with the converse<br />

advanced sleep phase disorder. In our sample, delayed sleep<br />

phase disorder is comorbid with both unipolar depression and<br />

bipolar disorder. Cases were 362 mixed-ancestry volunteers<br />

recruited nationally by advertising and interview and 355<br />

ancestrally-balanced controls without any reported disorder <strong>of</strong><br />

sleep timing. DNA from blood or saliva samples was<br />

genotyped for 768 SNPs using a custom Illumina Golden Gate<br />

assay. At least some SNPs providing partial tag-SNP<br />

coverage <strong>of</strong> 36 genes involving the circadian system, its visual<br />

input, and melatonin synthesis were examined. A number <strong>of</strong><br />

polymorphisms were nominally associated with DSPS<br />

case-control status, but only rs2482705 in NFIL3 was<br />

significantly associated after control for multiple testing<br />

(nominal P=0.0002, EMP2=0.032, OR=0.46), and that only<br />

among self-identified Europeans, when preliminary controls<br />

for 10 ancestry factors and age were included in the model.<br />

NFIL3 is a negative transcription factor regulating<br />

transcription <strong>of</strong> several circadian genes and participating in<br />

circadian molecular feedback loops. Since the rare A allele<br />

had an odds ratio <strong>of</strong> 0.46, it may be protective against delayed<br />

sleep phase or may tend to produce advanced sleep phase.<br />

57 A FAMILY-BASED STUDY OF DNA SEQUENCE<br />

VARIANTS IN GRM7 WITH SCHIZOPHRENIA IN AN<br />

INDONESIAN POPULATION<br />

S. Schwab* (1), C. Ganda (1), N. Amir (2), H. Heriani (2), I.<br />

Irmansyah (2), A. Kusumawardhani (2), M. Nasrun (2), I.<br />

Widyawati (2), D. Wildenauer (1)<br />

1. University <strong>of</strong> Western Australia 2. University <strong>of</strong> Indonesia<br />

* sschwab@cyllene.uwa.edu.au<br />

Differences in clinical presentation and outcome <strong>of</strong><br />

schizophrenia in patients from developing countries as<br />

opposed to developed countries may suggest the presence <strong>of</strong><br />

specific environmental and/or genetic factors. In order to<br />

address this question, we have collected a sample <strong>of</strong> 124<br />

affected sib-pair families with schizophrenia in Indonesia. We<br />

have used this sample previously for genome wide linkage<br />

analysis and have reported a genome wide significant locus on<br />

chromosome 3p. In our current study we have performed an<br />

association study using single nucleotide polymorphisms<br />

(SNPs) located in the genomic region <strong>of</strong> the metabotropic<br />

glutamate receptor 7 (GRM7) which is located in the centre <strong>of</strong><br />

the linkage peak on chromosome 3p. Overall, 18 SNPs have<br />

been genotyped using fluorescence based genotyping in the<br />

sample <strong>of</strong> 124 families showing evidence for linkage.<br />

Transmission disequilibrium test revealed one SNP<br />

(rs17031835) with association at a nominal significance level<br />

(P=0.004, not corrected for multiple testing). In addition,<br />

multiple haplotypes containing this SNP showed nominal<br />

significant association as well. Our study supports the idea<br />

that glutamatergic transmission and specifically the GRM7<br />

gene might be important in the development <strong>of</strong> schizophrenia<br />

in the Indonesian population.


58 INTERACTION OF 5HTTLPR AND DRD4 VNTR ON<br />

SUSTAINED ATTENTION<br />

M. Yamamoto* (1), J. McGeary (1, 2), T. Wells (3), C.<br />

Beevers (3)<br />

1. Brown University Center for Alcohol and Addiction Studies<br />

2. Research Service, Providence Veterans Affairs <strong>Medical</strong><br />

Center 3. University <strong>of</strong> Texas at Austin<br />

* maki_yamamoto@brown.edu<br />

Previous research suggests an association <strong>of</strong> 5HTTLPR<br />

variation with regards to depression and biased attention to<br />

emotional stimuli. Separate lines <strong>of</strong> research indicate a similar<br />

association between DRD4 VNTR and attention related<br />

phenotypes. This study aims to examine how these two<br />

variations interact in a population <strong>of</strong> non-depressed,<br />

un-medicated, healthy adults. Participants completed a<br />

standardized spatial cueing task that assessed maintenance <strong>of</strong><br />

attention for happy, sad, fearful, and neutral facial<br />

expressions. Individuals were also genotyped for 5HTTLPR<br />

and DRD4 VNTR polymorphisms. Results indicate a clear<br />

gene x gene interaction for the prediction <strong>of</strong> maintained<br />

attention. In DRD4 VNTR short homozygotes, an allele dose<br />

pattern is observed such that each additional short 5HTTLPR<br />

allele is associated with increased sustained attention for all<br />

stimuli. However, the pattern is completely reversed for<br />

DRD4 VNTR individuals who carry a seven or greater repeat<br />

allele. Specifically, there is an allele dose response<br />

relationship <strong>of</strong> decreased attention for each additional short<br />

5HTTLPR allele. These results extend and expand on what<br />

has previously been examined in attentional bias studies. If<br />

replicated, these results suggest the need to jointly model<br />

5HTTLPR and DRD4 VNTR variation in such studies.<br />

59 POLYMORPHISMS IN THE TACR1 GENE ARE<br />

ASSOCIATED WITH<br />

ATTENTION-DEFICIT/HYPERACTIVITY DISORDER<br />

(ADHD), ALCOHOLISM, BIPOLAR DISORDER AND<br />

COMBINED ALCOHOLISM WITH BIPOLAR<br />

DISORDER<br />

S. Sharp* (1), A. McQuillin (1), G. Lydall (1), N. Bass (1), A.<br />

Anjorin (1), M. Marks (1), J. Lawrence (1), M. Morgan (1), P.<br />

Asherson (2), A. Thapar (3), S. Hunt (1), C. Stanford (1), H.<br />

Gurling (1)<br />

1. University College London 2. King's College London 3.<br />

Cardiff University<br />

* s.sharp@ucl.ac.uk<br />

Attention-deficit/hyperactivity disorder (ADHD) is a clinically<br />

and genetically heterogeneous syndrome which is comorbid<br />

with childhood conduct disorder, alcoholism, substance abuse,<br />

dis-social personality disorder, and affective disorders. It is<br />

becoming increasingly clear that genes causing bipolar mania<br />

overlap with genes for a subtype <strong>of</strong> ADHD. Mice with<br />

functional ablation <strong>of</strong> the tachykinin receptor 1, TACR1, gene<br />

have recently been proposed as a model <strong>of</strong> ADHD, presenting<br />

locomotor hyperactivity that is prevented by<br />

psychostimulants. These mice also have reduced voluntary<br />

alcohol consumption, an effect that is mimicked by systemic<br />

administration <strong>of</strong> the TACR1 antagonist L-703,606 in wildtype<br />

mice. We and others have found evidence <strong>of</strong> association<br />

between TACR1 and bipolar disorder in three independent<br />

samples. We have also detected association with TACR1<br />

SNPs with ADHD, alcoholism and in combined alcoholism<br />

with bipolar disorder. Increased susceptibility to subtypes <strong>of</strong><br />

ADHD, alcoholism and bipolar disorder may be caused by the<br />

same mutations in TACR1. An alternative hypothesis is that<br />

one set <strong>of</strong> mutations increases the risk for ADHD and that<br />

different sets increase the risk for bipolar disorder and<br />

alcoholism. In an attempt to address this issue, TACR1 in<br />

cases <strong>of</strong> ADHD and combined alcoholism with bipolar<br />

disorder have been sequenced in order to identify potentially<br />

aetiological changes.


60 HUMAN MINERALOCORTICOID RECEPTOR<br />

GENE VARIANTS MODULATE COGNITIVE<br />

VULNERABILITY FOR DEPRESSION<br />

M. Klok* (1), A. Van der Does (2), E. Giltay (3), F. Zitman<br />

(3), E. De Kloet (1), R. DeRijk (1)<br />

1. Division <strong>of</strong> <strong>Medical</strong> Pharmacology, Leiden University,<br />

Leiden/Amsterdam Center for Drug Research (LACDR), The<br />

Netherlands 2. Department <strong>of</strong> Psychology, Leiden University,<br />

The Netherlands 3. Department <strong>of</strong> Psychiatry, Leiden<br />

University <strong>Medical</strong> Center (LUMC), The Netherlands<br />

* m.d.klok@lacdr.leidenuniv.nl<br />

The mineralocorticoid receptor (MR) plays a central role in<br />

the regulation <strong>of</strong> hypothalamic-pituitary-adrenal (HPA) axis<br />

activity. Animal studies indicate that the MR mediates effects<br />

<strong>of</strong> cortisol on emotions and coping behaviour. We hypothesise<br />

that human MR-gene variants influence cognition and<br />

emotions. Previously, we showed that a single nucleotide<br />

polymorphism (SNP; MR I180V) related to more feelings <strong>of</strong><br />

depression among elderly and identified a MR haplotype<br />

(frequency .36) to relate to higher dispositional optimism in<br />

elderly women, not in men. In the present study 154 students<br />

(46 M/108 F; 23.9±5 yrs) completed a questionnaire that<br />

measures cognitive vulnerability to depression and that<br />

includes subscales for hopelessness, rumination and<br />

aggression (LEIDS-R; Leiden Index <strong>of</strong> Depression<br />

Sensitivity-Revised). Neuroticism was also measured<br />

(NEO-PI) as well as symptoms <strong>of</strong> depression (HADS-D). MR<br />

SNPs and haplotypes were assessed, resulting in three<br />

haplotypes with frequencies <strong>of</strong> .50; .35; .13. Significant<br />

associations were found only in females between the<br />

haplotype with a frequency .35 and lower scores for<br />

hopelessness, aggression, risk aversion, neuroticism (p< .05),<br />

and in particular for rumination (p= .001), persisting after<br />

adjustment for age and emotional abuse during childhood.<br />

Excluding currently depressed participants (n= 14)<br />

strengthened the results. Moreover, this haplotype<br />

significantly associated with less symptoms <strong>of</strong> depression (p<<br />

.05). The results fit with our previous study showing an<br />

association between this haplotype and higher dispositional<br />

optimism, also only in women. Together the data indicate that<br />

MR-gene variants modulate cognitive vulnerability for<br />

depression. Financial support: Hersenstichting Nederland<br />

(project No. 10F02(2).37), Rivierduinen, KNAW<br />

61 A COLLABORATIVE FAMILY-BASED<br />

ASSOCIATION STUDY OF A GLUTAMATE<br />

RECEPTOR GENE (GRIN2B) IN OBSESSIVE<br />

COMPULSIVE DISORDER.<br />

S. Shaheen* (1), P. Arnold (1), S. Stewart (2), S. Taillefer (3),<br />

E. Yamashita (3), J. Fagerness (3), A. Hounie (4), E. Cook (5),<br />

D. Pauls (2), M. Richter (6), J. Kennedy (7), C. Mathews (8),<br />

G. Hanna (9)<br />

1. Hospital for Sick Children, University <strong>of</strong> Toronto 2.<br />

Massachusetts General Hospital 3. Hospital for Sick Children<br />

4. University <strong>of</strong> Sao Paulo 5. University <strong>of</strong> Illinois at Chicago<br />

6. Sunnybrook Health Sciences Centre 7. Centre for Addiction<br />

and Mental Health 8. University <strong>of</strong> California at San Francisco<br />

9. University <strong>of</strong> Michigan<br />

* shaheen70@gmail.com<br />

Obsessive-compulsive disorder (OCD) is a common<br />

debilitating condition affecting 1-3% <strong>of</strong> the population. Recent<br />

investigation suggests that glutamate, an excitatory<br />

neurotransmitter, may play a critical role in the pathogenesis<br />

<strong>of</strong> OCD. Specifically, the candidate gene GRIN2B (ionotropic<br />

glutamate receptor NMDA 2B subunit) has been implicated in<br />

OCD based on evidence from neuroimaging and previous<br />

family based association studies. The purpose <strong>of</strong> this study<br />

was to examine a possible association between SNPs and<br />

haplotype blocks <strong>of</strong> GRIN2B and OCD in a larger sample that<br />

has been examined previously for this gene. The sample<br />

consisted <strong>of</strong> OCD-affected probands and their immediate<br />

family members from a combined sample <strong>of</strong> 1575 individuals<br />

(308 families), recruited from specialized OCD clinics by<br />

investigators at six sites. Tag SNPs were selected (n = 10)<br />

using publicly available databases (dbSNP, HapMap) and the<br />

Haploview program.Genotyping was performed on the<br />

sequenom iPlex platform. Data analysis was performed using<br />

a Family Based Association Test (FBAT) under two models <strong>of</strong><br />

inheritance. SNPs rs1019385 (p = 0.04), and rs1806191 (p =<br />

0.04) were significantly associated with OCD transmission in<br />

male probands. A haplotype block consisting <strong>of</strong> SNPs<br />

rs1806191 and rs1806201 (p < .05) was associated with OCD<br />

transmission in male and female probands. These findings<br />

suggest a possible role <strong>of</strong> the glutamatergic system,<br />

particularly GRIN2B, in the pathogenesis <strong>of</strong> OCD.


62 ASSOCIATION OF TRYPTOPHAN<br />

HYDROXYLASE-2 (TPH2) POLYMORPHISM WITH<br />

AGE AT FIRST SUICIDE ATTEMPT IN<br />

PSYCHIATRICALLY ADMITTED ADOLESCENTS<br />

A. Cohen* (1), J. McGeary (2), C. Esposito-Smythers (3), A.<br />

Spirito (4)<br />

1. Department <strong>of</strong> Biology, Brown University 2. Providence<br />

Veterans Affairs <strong>Medical</strong> Center, Providence, Rhode Island 3.<br />

Department <strong>of</strong> Psychology, George Mason University 4.<br />

Center for Alcohol and Addiction Studies, Brown University<br />

* andrew_cohen@brown.edu<br />

Background Tryptophan hydroxylase-2 (TPH2) is the<br />

rate-limiting enzyme in the production <strong>of</strong> serotonin (5-HT) in<br />

the brain. Genetic variation in the TPH2 gene has been<br />

associated with major depressive disorder, bipolar disorder,<br />

and suicide attempt and completion. However, results so far<br />

are inconclusive. Adolescents represent an at-risk population<br />

for suicide and thus examination <strong>of</strong> TPH2 variation within an<br />

adolescent sample could provide important information on the<br />

neurobiology underlying suicidal behavior. Methods 90<br />

psychiatrically admitted adolescents (ages 13-17) were<br />

genotyped for TPH2 SNP rs1386494 and associations with<br />

history <strong>of</strong> a suicide attempt, method <strong>of</strong> attempt (violent or<br />

non-violent), and age at first attempt were examined for<br />

statistical significance. Results No differences were found<br />

across groups on age, gender, race, or ethnicity. Among<br />

suicide attempters, A carriers were more likely to have a<br />

history <strong>of</strong> violent suicide attempts than GG homozygotes<br />

(50% vs. 32%, small effect). Conversely, among suicide<br />

attempters, GG homozygotes were more likely to have a<br />

history <strong>of</strong> non-violent suicide attempts (71% vs. 60%, small<br />

effect). Further, GG homozygotes reported an earlier age at<br />

first suicide (M = 13.2, SD= 2.8) attempt than A carriers (M =<br />

15.2, SD= 1.2) (large effect). No differences were found<br />

across groups on presence <strong>of</strong> any suicide attempt or total<br />

number <strong>of</strong> attempts. Conclusion Preliminary results suggest<br />

that variation within the TPH2 gene influences suicide-related<br />

phenotypes. It is possible that TPH2 variation impacts early<br />

brain development that influences suicidal behavior. Further<br />

study <strong>of</strong> this gene in larger samples is needed to continue this<br />

investigation.<br />

63 HIPPOCAMPAL ATROPHY AS A QUANTITATIVE<br />

TRAIT IN A GENOME-WIDE ASSOCIATION STUDY<br />

(GWAS) IDENTIFYING NOVEL SUSCEPTIBILITY<br />

GENES FOR ALZHEIMER’S DISEASE<br />

S. Potkin* (1), G. Guffanti (1), A. Lakatos (1), J. Turner (1),<br />

F. Kruggel (1), J. Fallon (1), A. Saykin (2), A. Orro (3), S.<br />

Lupoli (3), E. Salvi (3), M. Weiner (4), F. Macciardi (1), A.<br />

Neuroimaging Initiative<br />

1. University <strong>of</strong> California, Irvine 2. Indiana University 3.<br />

University <strong>of</strong> Milan 4. University <strong>of</strong> California, San Francisco<br />

* sgpotkin@uci.edu<br />

INTRODUCTION With the exception <strong>of</strong> APOE e4 allele, the<br />

common genetic risk factors for sporadic Alzheimer’s Disease<br />

(AD) are unknown. GWAS studies <strong>of</strong>fer the possibility <strong>of</strong><br />

discovering unanticipated risk genes. METHOD We<br />

completed a genome-wide association study on 381<br />

participants in the ADNI (Alzheimer’s Disease Neuroimaging<br />

Initiative) study. Samples were genotyped using the Illumina<br />

Human610-Quad BeadChip. 516,645 unique Single<br />

Nucleotide Polymorphisms (SNPs) were included in the<br />

analysis following quality control measures. Two analyses<br />

were completed: a standard case-control analysis, and a novel<br />

approach using hippocampal atrophy measured on MRI as an<br />

objectively defined, quantitative phenotype. A General Linear<br />

Model was applied to identify SNPs for which there was an<br />

interaction between the genotype and diagnosis on the<br />

quantitative trait. RESULTS The case-control analysis<br />

identified APOE and TOMM40 at a genome-wide significance<br />

level <strong>of</strong> £10-6. The quantitative trait analysis identified 21<br />

genes or chromosomal areas with a p-value £10-6, which can<br />

be considered potential “new” candidate loci to explore in the<br />

etiology <strong>of</strong> sporadic AD. These candidates included EFNA5,<br />

CAND1, MAGI2, ARSB, and PRUNE2, genes involved in the<br />

regulation <strong>of</strong> protein degradation, apoptosis, metabolism and<br />

neuronal loss. Supportive evidence based on case-control<br />

studies and biological plausibility by gene annotation is<br />

provided. CONCLUSION Using hippocampal atrophy as a<br />

quantitative phenotype in a genome-wide scan, we have<br />

identified new candidate risk genes for sporadic Alzheimer’s<br />

disease that merit further investigation. A QT analysis<br />

provides a powerful strategy for gene discovery in the context<br />

<strong>of</strong> a genome-wide survey.


64 A COMMON MECP2 HAPLOTYPE ASSOCIATES<br />

WITH REDUCED CORTICAL SURFACE AREA IN<br />

HUMANS IN TWO INDEPENDENT POPULATIONS<br />

A. Joyner* (1, 2), C. Roddey (1), C. Bloss (1), T. Bakken (1),<br />

L. Rimol (3), I. Melles (3), I. Agartz (3), S. Djurovic (3), E.<br />

Topol (2), N. Schork (2), O. Andreassen (3), A. Dale (1)<br />

1. <strong>UCSD</strong> 2. STSI 3. TOP<br />

* ajoyner@ucsd.edu<br />

The gene MECP2 is a well-known determinant <strong>of</strong> brain<br />

structure. Mutations in this gene cause microencephalopathy<br />

and are associated with several neurodevelopmental disorders<br />

that affect both brain morphology and cognition. Though<br />

mutations in MECP2 result in severe neurological phenotypes,<br />

the effect <strong>of</strong> common variation in this genetic region is<br />

unknown. We find that common sequence variations in a<br />

region in and around MECP2 show association with structural<br />

brain size measures in two independent cohorts, a discovery<br />

sample from the Thematic Organized Psychosis (TOP)<br />

research group, and a replication sample from the Alzheimer’s<br />

Disease Neuroimaging Initiative (ADNI). The most<br />

statistically significant replicated association (p


66 GENETIC VARIATIONS OF FYN-TYROSINE<br />

KINASE IN PSYCHIATRIC DISORDERS<br />

K. Hattori* (1), Y. IIjima (1), H. Uchiyama (1), N.<br />

Yamamoto (1), T. Fujii (1), H. Hori (1), T. Teraishi (1), R.<br />

Hashimoto (2), M. Tatsumi (3), M. Omori (4), N. Okamoto<br />

(4), K. Arima (4), T. Higuchi (5), H. Kunugi (1)<br />

1. Department <strong>of</strong> Mental Disorder Research, National Institute<br />

<strong>of</strong> Neuroscience, National Center <strong>of</strong> Neurology and Psychiatry<br />

2. Molecular Research Center for Children's Mental<br />

Development, Osaka University Graduate School <strong>of</strong> Medicine<br />

3. Yokohama psychosomatic clinic 4. Department <strong>of</strong><br />

Psychiatry, Musashi Hospital, National Center <strong>of</strong> Neurology<br />

and Psychiatry 5. National Center <strong>of</strong> Neurology and<br />

Psychiatry<br />

* hattori@ncnp.go.jp<br />

Introduction: Fyn kinase regulates NMDA-R function, and<br />

participates in learning, emotion and sensitivity to<br />

antipsychotics. Recently, we found that Fyn protein levels are<br />

decreased in the platelets <strong>of</strong> schizophrenic patients (Hattori et<br />

al., 2009). We also found splicing patterns <strong>of</strong> fyn mRNA are<br />

altered in schizophrenia; specifically, the ratio <strong>of</strong> a defective<br />

is<strong>of</strong>orm fyn delta-7, in which exon7 was absent, was elevated.<br />

Methods: To determine whether genetic variations <strong>of</strong> fyn gene<br />

are associated with psychiatric disorders such as schizophrenia<br />

and bipolar disorder, weanalyzed Japanese cohort <strong>of</strong> 497<br />

patients with schizophrenia, 528 major depression, 138 bipolar<br />

disorder, and 932 control subjects. Eight tagging SNPs were<br />

genotyped by TaqMan assays. A possible association between<br />

the fyn genotype with performance in the Wisconsin Card<br />

Sorting Test (WCST) and intelligence quotient (IQ) was also<br />

examined in 166 control subjects. Results: Although no<br />

association <strong>of</strong> those SNPs with schizophrenia or depression<br />

was found, a weak but significant association was found<br />

between bipolar disorder and a SNP. Haplotype analyses also<br />

revealed that the frequencies <strong>of</strong> the haplotype that contain<br />

above SNP were significantly different between bipolar<br />

disorder and the control. Next, among control subjects,<br />

significant differences were observed between two SNPs, both<br />

are on the same LD block, and WCST scores. Conclusion:<br />

Genetic variation <strong>of</strong> the fyn gene may contribute to molecular<br />

mechanisms <strong>of</strong> bipolar disorder and some aspects <strong>of</strong><br />

intelligence.<br />

67 DIFFERENTIAL GENE EXPRESSION ANALYSIS<br />

BY ACUTE AND CHRONIC LITHIUM TREATMENT<br />

IN HUMAN PRIMARY FIBROBLASTS<br />

C. Fukuhara* (1), S. Lee (1), K. Singhapakdi (2), S. Chen<br />

(3), C. Maddox (4), M. Iuvone (5)<br />

1. Neuroscience Institute, Morehouse School <strong>of</strong> Medicine 2.<br />

Emory University 3. Department <strong>of</strong> Dermatology, Emory<br />

University School <strong>of</strong> Medicine 4. CRC, Morehouse School <strong>of</strong><br />

Medicine 5. Department <strong>of</strong> Ophthalmology, Emory University<br />

School <strong>of</strong> Medicine<br />

* cfukuhara@msm.edu<br />

One <strong>of</strong> the difficulties treating psychiatric patients is that some<br />

patients are resistant to certain medications. To help<br />

developing personalized therapies, it would be beneficial to<br />

establish an assay by which clinical sensitivities to medicines<br />

can be evaluated in vitro. For many psychiatric drugs, it takes<br />

several weeks to find out whether or not they will be effective,<br />

a predictive diagnostic test would be useful. Dermal fibroblast<br />

may serve as a convenient model to study the long-term<br />

effects <strong>of</strong> psychiatric medications because they survive long<br />

enough in vitro for chronic drug experiments without<br />

immortalization, and they express certain transporters and<br />

receptors that are targets <strong>of</strong> psychiatric medicines. A goal <strong>of</strong><br />

the present study was to test the hypothesis that dermal<br />

fibroblasts obtained from human subjects can be used to study<br />

the effects <strong>of</strong> psychiatric medicines on differential gene<br />

expression patterns among subjects. The mood stabilizer<br />

lithium was used to test for differential gene expression<br />

patterns. Skin biopsies were obtained from healthy volunteers<br />

and fibroblast cell cultures were initiated. Fibroblasts were<br />

cultured in the presence <strong>of</strong> lithium at 0, 1, or 1.5 mM<br />

concentration for 1 or 4 weeks before harvest. No significant<br />

difference was observed in cell numbers among the three<br />

groups for up to 1 week <strong>of</strong> the treatments. Total RNA was<br />

isolated from those samples and subjected to Affymetrix<br />

GeneChip Human Genome Array analysis and further real<br />

time quantitative PCR for validation. Our preliminary data<br />

showed that 1738 and 1029 mRNA species were up- and<br />

down-regulated more than two fold by 1-week lithium<br />

treatment. After 4 weeks lithium treatment, 1227 and 962<br />

species were up- and down-regulated. Expressions <strong>of</strong> about<br />

5000 genes were differentially affected between 1 and 4 weeks<br />

lithium treatments. Consistently over expressed and<br />

suppressed genes between 1 and 4 weeks lithium treatments<br />

were 257 and 180, respectively. Success in establishing the<br />

dermal fibroblast model will enable us to identify biomarkers<br />

that can be used to develop personalized medicines.


68 TRYPTOPHAN HRYDROXYLASE-2 5'- AND<br />

3'-REGULATORY POLYMORPHISMS ARE<br />

DIFFERENTIALLY ASSOCIATED WITH HPA AXIS<br />

FUNCTION AND SELF-INJURIOUS BEHAVIOR IN<br />

RHESUS MONKEY<br />

G. Chen* (1), M. Novak (2), J. Meyer (2), B. Kelly (1), E.<br />

Vallender (1), G. Miller (1)<br />

1. Harvard <strong>Medical</strong> School 2. University <strong>of</strong> Massachusetts at<br />

Amherst<br />

* guo-lin_chen@hms.harvard.edu<br />

INTRODUCTION: Tryptophan hydroxylase-2 (TPH2)<br />

synthesizes neuronal serotonin and is linked to numerous<br />

behavioral traits. We have previously identified a constellation<br />

<strong>of</strong> polymorphisms in rhesus monkey TPH2 (rhTPH2) and<br />

characterized the functionality <strong>of</strong>3'-UTR polymorphisms. This<br />

study aims to functionally characterize the 5'-FR<br />

polymorphisms and comprehensively evaluate the effect <strong>of</strong><br />

rhTPH2 genotype on physiological and behavioral traits.<br />

METHODS: The functionality <strong>of</strong> rhTPH2 5'-FR<br />

polymorphisms was assessed by luciferase reporter assay,<br />

while the effects <strong>of</strong> rhTPH2 genotype on<br />

hypothalamic-pituitary-adrenal (HPA) axis function and<br />

self-injurious behavior (SIB) were evaluated by<br />

genotype-phenotype correlation. RESULTS: rhTPH2 5'-FR<br />

haplotype exhibits a significant, cell-dependent effect on gene<br />

expression. The 5'-FR -1485(AT)n polymorphism exerts<br />

significant effects on the afternoon cortisol level and nocturnal<br />

HPA negative feedback, while the 3'-UTR 2051A>C exhibits<br />

significant effects on the morning cortisol level and cortisol<br />

response to ACTH challenge, as well as marginally significant<br />

effects on the daytime HPA negative feedback and self-biting<br />

rate. Meanwhile, -1485(AT)n and 2051A>C interact to<br />

influence the plasma ACTH level. In addition, the<br />

genotype/allele frequency <strong>of</strong> 5'-FR -1325Ins>Del differed<br />

significantly between self-wounders and non-self-wounders,<br />

while 3'-UTR 2128S>L differed significantly in<br />

genotype/allele frequency between the high- and<br />

low-frequency biters. DISCUSSION: This study demonstrates<br />

the functionality <strong>of</strong> rhTPH2 5'-FR polymorphisms, and<br />

provides evidence for the differential association <strong>of</strong> rhTPH2<br />

5'-FR and 3'-UTR polymorphisms with HPA axis function and<br />

SIB. Our findings shed light on the role <strong>of</strong> TPH2 gene<br />

variance in physiology and behavioral traits, and also<br />

contribute to the understanding <strong>of</strong> the pathophysiology and<br />

genetics <strong>of</strong> SIB.<br />

69 CHRNB2 PROMOTER REGION: ASSOCIATION<br />

WITH SUBJECTIVE EFFECTS AND GENE<br />

EXPRESSION DIFFERENCES<br />

N. H<strong>of</strong>t* (1), J. Stitzel (1), K. Hutchison (2), J. Miyamoto (1),<br />

M. Ehringer (1)<br />

1. Institute for Behavioral Genetics, University <strong>of</strong> Colorado,<br />

Boulder, CO, 80309 2. The MIND, Albuquerque, NM 87131<br />

* h<strong>of</strong>t@colorado.edu<br />

The family <strong>of</strong> neuronal nicotinic acetylcholine receptors<br />

shows regulation <strong>of</strong> activity by both endogenous acetylcholine<br />

and exogenous nicotine, making sequence variations in these<br />

receptors likely candidates for association with tobacco<br />

phenotypes. Our group has previously identified a significant<br />

association between a rare SNP, rs2072658,and subjective<br />

effects reported by young adults after smoking their first few<br />

cigarettes (Ehringer et al, 2007). We have replicated this<br />

association in a sample <strong>of</strong> adult smokers reporting physical<br />

subjective effects after smoking the first <strong>of</strong> three cigarettes<br />

(1.1mg nicotine) in an experimental session (p=0.01). The<br />

association seems to be driven by sweating, heart pounding<br />

and nausea (p= 0.0075, 0.019, 0.032 respectively). This<br />

suggests variation in CHRNB2 may continue to affect<br />

subjective response long after the initiation period. To assess<br />

whether this SNP, immediately upstream <strong>of</strong> exon1 <strong>of</strong><br />

CHRNB2, and other variation in the promoter region <strong>of</strong><br />

CHRNB2 has an effect on gene expression, we cloned 3 kb<br />

upstream <strong>of</strong> the initiation codon <strong>of</strong> CHRNB2 into a luciferase<br />

reporter vector. Promoter activity was tested in three cell<br />

types. Preliminary results suggest that in SH-SY5Y and P19<br />

cells, those constructs containing the minor allele <strong>of</strong><br />

rs2072658 show reduced expression compared to those<br />

containing the common allele (21% decrease p=0.017 and<br />

19.4% decrease p=0.031 respectively). This difference<br />

indicates rs2072658may lead to functional differences in gene<br />

expression which may contribute to the subjective effects to<br />

nicotine.


70 GENETIC VARIATION IN PROMOTER REGION<br />

OF NPPA GENE IS ASSOCIATED WITH<br />

CARDIOEMBOLIC STROKE<br />

B. Chang (1), J. Kim (1), S. Ko (1), Y. Bu (2), S. Park* (1), J.<br />

Park (1)<br />

1. Kyunghee University 2. Kyunghee University Oriental<br />

<strong>Medical</strong> Science Center<br />

* sjpark422@khu.ac.kr<br />

Stroke, a complex and multigenic disease, is developed from<br />

the impaired supply <strong>of</strong> blood to the brain. The obvious genetic<br />

factors or variations which are associated with stroke have<br />

been not found. Atrial natriuretic peptide (ANP; also known as<br />

NPPA) was determined as a genetic factor responsible for<br />

several diseases including stroke. This study genotyped the<br />

single nucleotide polymorphisms (SNP) in promoter region <strong>of</strong><br />

NPPA gene using total 941 Koreans <strong>of</strong> stroke patients<br />

diagnosed from the oriental medicine and controls, and<br />

investigated its association associations. There was no<br />

significant association between four common SNPs and stroke<br />

including hemorrhagic and ischemic and controls. However, in<br />

the Trial <strong>of</strong> Org 10172 in Acute Stroke Treatment (TOAST)<br />

classification <strong>of</strong> ischemic stroke, cardioembolic stroke showed<br />

a significant increase in frequency <strong>of</strong> rs5067(A/G) compared<br />

to contols and small-vessel occlusion (SVO) (p < 0.05).<br />

Considering that natriuretic peptides, as polypeptide hormones<br />

primarily secreted from the heart, play an important role in the<br />

regulation <strong>of</strong> cardiac hemodynamics, our findings suggest that<br />

a possible association <strong>of</strong> rs5065C allele in promoter <strong>of</strong> NPPA<br />

with cardioembolic stroke could regulate the expression <strong>of</strong><br />

NPPA gene to develop into stroke. Further study addressed to<br />

the function <strong>of</strong> rs5067(A/G) is needed. This work was<br />

supported(researched)by the Second Stage <strong>of</strong> Brain Korea 21<br />

project in 2009.<br />

71 ANALYSIS OF POSITIONAL CANDIDATE GENES<br />

FOR SCHIZOPHRENIA ON CHROMOSOME 5P13<br />

I. Bespalova* (1), B. Ritter (1), G. Angelo (1), J. Hunter (1),<br />

J. Silverman (1)<br />

1. Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong> Medicine,<br />

New York, NY 10029, USA<br />

* irina.bespalova@mssm.edu<br />

We have previously described a large, high-density pedigree<br />

with schizophrenia-affected family members from a genetic<br />

isolate <strong>of</strong> Spanish origin in the central mountainous region <strong>of</strong><br />

Puerto Rico. A genome scan in the extended family revealed a<br />

linkage signal on chromosome 5p13 with a multipoint<br />

lodscore <strong>of</strong> 4.8. We narrowed the region <strong>of</strong> linkage to the<br />

minimal critical interval (MCI) <strong>of</strong> 2.8 Mb, containing 13<br />

annotated genes. We have previously reported on sequencing<br />

<strong>of</strong> one <strong>of</strong> these genes, the relaxin/insulin-like family peptide<br />

receptor 3, RXFP3. In the present study we examined all <strong>of</strong><br />

the remaining 12 positional candidate genes in MCI by<br />

sequencing their coding, 5’- and 3’-untranslated, and first 2 kb<br />

<strong>of</strong> putative promoter regions in affected members <strong>of</strong> the<br />

Puerto Rican family: PDZD2, GOLPH3, MTMR12, ZFR,<br />

SUB1, NPR3, C5orf23, TARS, ADAMTS12, SLC45A2,<br />

AMACR, and C1QTNF3. We detected 100 sequence variants,<br />

61 <strong>of</strong> which were shared by all affected members <strong>of</strong> the<br />

family. The shared variants are likely to contribute to the<br />

disorder in the Puerto Rican family. To the time, we have<br />

analyzed the variants detected in the C1q and tumor necrosis<br />

factor related protein 3 (C1QTNF3) gene by genotyping in<br />

unrelated 118 schizophrenia-affected and 136 healthy Puerto<br />

Rican individuals <strong>of</strong> Spanish origin from the same central<br />

mountainous region <strong>of</strong> Puerto Rico. None <strong>of</strong> the<br />

polymorphisms demonstrated an association with the disorder,<br />

suggesting that the genetic variants in C1QTNF3 are most<br />

likely do not play role in schizophrenia.


72 THE EXPRESSION OF THE SCHIZOPHRENIA<br />

CANDIDATE GENE RGS4 IS SUBJECT TO VARIABLE<br />

CIS-ACTING INFLUENCES IN BRAIN<br />

F. Buonocore (1), C. Troakes (1), F. Hill (1), J. Cooper (1), N.<br />

Bray (1)<br />

1. Institute <strong>of</strong> Psychiatry, King's College London<br />

Regulator <strong>of</strong> G-protein signaling 4 (RGS4) modulates the<br />

intracellular signalling <strong>of</strong> several neurotransmitter receptors<br />

implicated in schizophrenia pathophysiology or treatment,<br />

including those binding dopamine, serotonin and glutamate.<br />

The expression <strong>of</strong> RGS4 in schizophrenic brain has been<br />

reported to be altered at both the RNA and protein level.<br />

Reports <strong>of</strong> genetic association between non-coding SNPs in<br />

the RGS4 gene and schizophrenia suggest that altered<br />

expression may partly reflect genetic risk factors, and<br />

therefore constitute a primary etiological mechanism in the<br />

disorder. We sought to determine whether RGS4 contains<br />

common variation affecting its expression in human brain,<br />

and, if so, where these effects are manifest. In an initial screen,<br />

we assessed relative allelic expression <strong>of</strong> RGS4 across<br />

multiple discrete brain regions in 12 individuals. We observed<br />

significant departure from the expected 1:1 ratio <strong>of</strong> allelic<br />

expression in several individuals, indicating that RGS4 is<br />

indeed subject to common cis-acting regulatory influences in<br />

brain. Furthermore, we found that, within individuals, these<br />

effects varied between brain regions, with more pronounced<br />

allelic expression imbalance in sub-cortical areas. These data<br />

demonstrate the existence <strong>of</strong> cis-regulatory variation in RGS4,<br />

which could underlie genetic association with schizophrenia,<br />

and point to areas <strong>of</strong> the brain that are affected.<br />

73 GAB2, GSTP1, SORL1, BDNF, GAPDH AND APOE,<br />

POLYMORPHISMS ASSOCIATED WITH LATE<br />

ONSET ALZHEIMER DISEASE.<br />

G. Izzo (1), O. Forlenza (1), B. Diniz (1), C. Kukita (1), L.<br />

Talib (1), P. Bertolucci (1), W. Gattaz (1), E. Ojopi* (1)<br />

1. Laboratório de Neurociências (LIM 27), Institute <strong>of</strong><br />

Psychiatry, University <strong>of</strong> São Paulo (USP), São Paulo, Brazil<br />

* elida@usp.br<br />

We investigated genes with relevant neurological function<br />

such as oxidative stress, neuronal apoptosis, and neuronal<br />

differentiation in patients with late onset Alzheimer’s Disease<br />

(AD). At the present study, we searched for associations <strong>of</strong><br />

late onset AD and 8 single nucleotide polymorphisms (SNPs)<br />

on 5 different genes (GAB2, GSTP1, BDNF, GAPDH, and<br />

APOE). DNA was extracted from AD patients and elderly<br />

controls. We performed genotypic analysis by using allelic<br />

discrimination on real-time PCR. At this time were analyzed<br />

more than 400 subjects. It was found a strong association at<br />

the studied population between the APOE*E2 allele in<br />

controls and also between the APOE*E4 allele in AD patients<br />

(P = 0,00127), confirming data already published. For GAB2<br />

rs2373115 polymorphism, our results suggest that the TT<br />

genotype <strong>of</strong>fers a higher susceptibility for developing AD,<br />

with a strong association (P = 0,005). The other genes studied<br />

did not show any significant association between its genotypes<br />

and AD. Combining APOE*4 allele with all studied<br />

polymorphisms we detected a strong association for some<br />

genotypes. We have analyzed genes involved with relevant<br />

roles in neuronal homeostasis, such as oxidative stress,<br />

neuronal apoptosis and neuronal differentiation. We have<br />

confirmed the APOE*E4 allele and GAB2 rs2373115 ‘TT’<br />

genotype as important targets for development <strong>of</strong> Alzheimer’s<br />

disease. Our findings confirmed previous data that GAB2<br />

modifies late onset AD risk in APOE*E4 carriers and<br />

influences Alzheimer‘s neuropathology. Although any <strong>of</strong> the<br />

other genes analyzed has shown a direct association with AD<br />

when compared between patients and controls, it was possible<br />

to observe an association to AD in APOE*E4 carriers,<br />

suggesting a modifying effect <strong>of</strong> this allele. If those<br />

associations prove to be consistent in larger samples, the<br />

biological roles <strong>of</strong> these genes on AD will require further<br />

clarification.


COGNITION<br />

74 A GENOME-WIDE ASSOCIATION STUDY OF<br />

NON-PATHOLOGICAL COGNITIVE AGEING<br />

G. Davies* (1), K. McGhee (1) M. Luciano (1), A. Tenesa<br />

(2), A. Payton (3), X. Ke (3), S. Harris (1, 8), A. Gow (1), D.<br />

Liewald (1), L. Whalley (4), H. Fox (5), G. McNeill (6), W.<br />

Ollier (3), A. Pickles (7), D. Porteous (8), P. Visscher (9), J.<br />

Starr (10), M. Horan (11), N. Pendleton (11), I. Deary (1)<br />

1. University <strong>of</strong> Edinburgh Centre for Cognitive Ageing and<br />

Cognitive Epidemiology, Department <strong>of</strong> Psychology,<br />

University <strong>of</strong> Edinburgh, 7 George Square, Edinburgh, UK<br />

2. Colon Cancer Genetics Group, Institute <strong>of</strong> Genetics and<br />

Molecular Medicine, University <strong>of</strong> Edinburgh and MRC<br />

Human Genetics Unit, Edinburgh, UK 3. Centre for Integrated<br />

Genomic <strong>Medical</strong> Research, School <strong>of</strong> Translational<br />

Medicine, The University <strong>of</strong> Manchester, Manchester, UK<br />

4. University <strong>of</strong> Aberdeen Institute <strong>of</strong> Applied Health<br />

Sciences, Foresterhill, Aberdeen, UK 5. Scottish Dementia<br />

Research Network, Royal Victoria Hospital, Edinburgh, UK<br />

6. Environmental & Occupational Medicine, Liberty Safe<br />

Work Research Centre, Foresterhill Road, Aberdeen, UK<br />

7. Biostatistics Group, Community Based Medicine, SCAN<br />

Building Complex, The University <strong>of</strong> Manchester,<br />

Manchester, UK 8. <strong>Medical</strong> Genetics Section, University <strong>of</strong><br />

Edinburgh Molecular Medicine Centre and Institute <strong>of</strong><br />

Genetics and Molecular Medicine, Edinburgh, UK<br />

9. Genetic Epidemiology, Queensland Institute <strong>of</strong> <strong>Medical</strong><br />

Research, Brisbane, Australia 10. University <strong>of</strong> Edinburgh<br />

Centre for Cognitive Ageing and Cognitive Epidemiology,<br />

Geriatric Medicine, University <strong>of</strong> Edinburgh, Royal Victoria<br />

Hospital, Edinburgh, UK 11. Clinical Neurosciences, School<br />

<strong>of</strong> Translational Medicine, The University <strong>of</strong> Manchester,<br />

Hope Hospital, Salford, Greater Manchester, UK<br />

* Gail.Davies@ed.ac.uk<br />

Cognitive decline is the single most feared aspect <strong>of</strong> growing<br />

old. It is a major contributor to lower quality <strong>of</strong> life and loss <strong>of</strong><br />

independence. In the UK, it accounts for 40% <strong>of</strong> admissions to<br />

institutional care and is currently a major personal and social<br />

burden. Retaining cognitive functions must be a priority for<br />

older people. Cognition and age-related cognitive decline are<br />

determined by genetic and environmental factors. The<br />

heritability <strong>of</strong> intelligence changes across the lifespan: ~30%<br />

in early childhood to 70% in adulthood and may decline<br />

slightly over the age <strong>of</strong> 65. This indicates that there are genetic<br />

contributions to individual differences in normal cognitive<br />

ageing. We have taken a hypothesis-free approach and<br />

genotyped two <strong>of</strong> the largest longitudinal elderly cohorts in the<br />

world that have repeated cognitive test data: ~2,000 samples<br />

from the University <strong>of</strong> Edinburgh’s and University <strong>of</strong><br />

Aberdeen’s follow-up studies <strong>of</strong> the Scottish Mental Surveys<br />

(SMS) <strong>of</strong> 1932 and 1947, and ~2,000 DNA samples from the<br />

Dyne Steele DNA bank <strong>of</strong> University <strong>of</strong> Manchester Age and<br />

Cognitive Performance Research Centre volunteers (DSDb).<br />

The genetic contribution to cognitive ability traits, and to<br />

age-related change in general cognitive ability, processing<br />

speed, and memory will be studied in the SMS and DSDb<br />

cohorts using simple least squares and assuming an additive<br />

genetic model. The best supported 30 SNPs will be genotyped<br />

in an independent replication set comprising over 6,000<br />

samples. Preliminary analyses have shown possible regions <strong>of</strong><br />

interest. Further analyses are in progress and results will be<br />

presented.


75 DECREASE IN IMMEDIATE EARLY GENE<br />

EXPRESSION ONE MONTH AFTER<br />

ELECTROCONVULSIVE SEIZURES.<br />

J. Correia-Pinto (1, 2), J. Quevedo (2), S. Valvassori (3), G.<br />

Feier (3), T. Barichello (3), M. Athie (1), S. Ribeiro (4), W.<br />

Gattaz (1), E. Ojopi* (1)<br />

1. Laboratório de Neurociências (LIM-27), Hospital das<br />

Clínicas, Instituto de Psiquiatria, Faculdade de Medicina da<br />

Universidade de São Paulo, USP, São Paulo, SP, Brazil<br />

2. Laboratório Cesar Timo-Iaria, Instituto de Ensino e<br />

Pesquisa, Hospital Sírio Libanês, São Paulo, SP, Brazil.<br />

3. Laboratório de Neurociências e Instituto Nacional de<br />

Ciência e Tecnologia Translacional em Medicina, Programa<br />

de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica<br />

de Ciências da Saúde, Universidade do Extremo Sul<br />

Catarinense, 88806-000 Criciúma, SC, Brazil. 4. Edmond and<br />

Lily Safra International Institute <strong>of</strong> Neuroscience <strong>of</strong> Natal<br />

(ELS-IINN), Natal, RN, Brazil. Federal University <strong>of</strong> Rio<br />

Grande do Norte (UFRN), Natal, RN, Brazil. Instituto<br />

Nacional de Ciência e Tecnologia em Interfaces<br />

Cérebro-Máquina (INCEMAQ).<br />

* elida@usp.br<br />

Electroconvulsive therapy (ECT) is a commonly used<br />

psychiatric treatment for severe depression management.<br />

Despite its clinical utility, post-ECT memory deficits are a<br />

common side effect. Neuronal plasticity and memory<br />

consolidation are intimately related to the expression <strong>of</strong><br />

immediate early genes (IEG) such as Egr1, Fos and Arc. IEG<br />

are rapidly induced in response to extracellular stimuli, and<br />

may thereby initiate an intracellular cascade that underlies<br />

long-term adaptations in neuronal gene expression following<br />

chronic and acute electroconvulsive seizures (ECS), an animal<br />

model <strong>of</strong> ECT. Here, we examined how acute or chronic ECS,<br />

alter expression <strong>of</strong> IEGs Egr1, Fos and Arc in hippocampus, a<br />

brain region implicated in the pathophysiology <strong>of</strong> depression<br />

in humans and in animal models <strong>of</strong> depression. Forty-two<br />

male Wistar rats, weighting 250–300 mg were submitted to<br />

bilateral ECS. Rats were submitted to a single stimulation<br />

(acute) or to a series <strong>of</strong> eight stimulations, applied one every<br />

48 hours (chronic). Tissue from hippocampus was collected<br />

immediately after and 2, 7, 30 and 90 days after the last<br />

seizure. Sham-ECS groups were handled as ECS groups but<br />

without shock delivery. Hippocampus from seven animals was<br />

pooled for each time point. Real-time PCR experiments were<br />

performed for Arc, Fos and Egr1, as target genes, and B2m<br />

and Rpl19, as endogenous controls. A fixed-effect ANOVA<br />

was used to account for treatment (acute and chronic) and<br />

temporal effects in gene expression. Then, a one way ANOVA<br />

followed by Tukey’s test as a post-hoc was used for group<br />

comparisons. A significant decrease in expression was<br />

observed for all tested genes comparing chronic versus acute<br />

ECS administration. This decrease was 1.3 fold for Arc<br />

(F1,2=15.895, p=0.001), 1.4 fold for Egr1 (F1,2=13.113,<br />

p=0.001) and 1.5 fold for Fos (F1,2=58.169, p


77 HOW SPECIFIC ARE MATHS QTLS?:<br />

BEHAVIOURAL GENOMIC ANALYSIS.<br />

Y. Kovas* (1), S. Doherty (2), R. Plomin (2)<br />

1. Goldsmiths College<br />

2. SGDP, Institute <strong>of</strong> Psychiatry, King's College London<br />

*y.kovas@gold.ac.uk<br />

Introduction: Quantitative genetic research has revealed a<br />

strong genetic basis to mathematical ability, and indicates that<br />

largely the same genes contribute to all facets <strong>of</strong> the<br />

mathematical domain and to other cognitive domains, which<br />

supports a ‘generalist genes’ hypothesis. Our recently<br />

conducted genome wide association scan identified first<br />

quantitative trait loci (QTLs) together explaining 3% in<br />

general mathematical ability (p = 7.277e-14). The association<br />

is linear across the distribution consistent with a quantitative<br />

trait locus (QTL) hypothesis; the third <strong>of</strong> children in our<br />

sample who harbor 10 or more <strong>of</strong> the 20 risk alleles are nearly<br />

twice as likely to be in the lowest performing 15% <strong>of</strong> the<br />

distribution. Our latest analyses, reported here, explore the<br />

associations between these QTLs and other phenotypes<br />

collected as part <strong>of</strong> Twins Early Development Study. Method:<br />

Using longitudinal multivariate data we examined whether the<br />

SNP-set which explains 3% <strong>of</strong> the variance in mathematical<br />

ability in our sample at 10 years <strong>of</strong> age is also associated with<br />

mathematical ability at other ages; and with spatial ability,<br />

reading, and g. The N ranged from 1500 to over 4000.<br />

Results: Although weaker than with mathematical ability at<br />

10, the correlations between the Mathematics SNP Set and<br />

reading, and spatial ability were highly significant.<br />

Conclusions: Our results are consistent with the Generalist<br />

Genes Hypothesis, in that the SNPs found for mathematical<br />

ability are also involved in mathematical disability, in all<br />

components <strong>of</strong> mathematics tested, and in other cognitive<br />

abilities. Some genetic specificity is also implied by these<br />

results.<br />

DEMENTIA<br />

78 ACE, MTHFR AND APO E GENE POLYMORPHISM<br />

IN NORTH INDIAN PATIENTS WITH ALZHEIMER’S<br />

DISEASE<br />

D. Tewari *(1), U. Mishra (1), J. Kalita (1)<br />

1. Sanjay Gandhi Post Graduate Institute <strong>of</strong> <strong>Medical</strong> Sciences,<br />

Lucknow-226014 (INDIA)<br />

*deepshikha_pgi@yahoo.co.in<br />

Background & purpose: Alzheimer disease (AD) is the most<br />

common form <strong>of</strong> dementia. In the current study we evaluated<br />

the distribution <strong>of</strong> ACE, MTHFR & APO E genotype in<br />

patients with dementia because <strong>of</strong> growing interest <strong>of</strong><br />

multifactorial etiology <strong>of</strong> the disease. Methods: The study<br />

group included 80 patients (63 men and 17 women, mean age<br />

78 ± 2.6 years) and 90 normal subjects who visited Neurology<br />

OPD, SGPGIMS, matched according to age and sex, and have<br />

been taken. The dementia status <strong>of</strong> these patients was assessed<br />

by clinical psychologist using the Clinical Dementia Rating<br />

(CDR) score. These patients were genotyped for vascular<br />

disease–associated polymorphisms in the genes coding for<br />

methylenetetrahydr<strong>of</strong>olate reductase (MTHFR),<br />

angiotensin-converting enzyme (ACE) and a common genetic<br />

risk factor for AD, apolipoprotein E e4 (APOE e4).<br />

Results: There was no significant association between ACE &<br />

MTHFR genotypes with AD and with control subjects.<br />

However T allele frequency <strong>of</strong> MTHFR gene is found double<br />

in dementia patients 12.5% (p>0.897) when compared with<br />

controls as 6.2%.There was a higher frequency <strong>of</strong> APOE e4<br />

alleles in patients with AD (18.2%, P>1.6) compared with<br />

control subjects (1.7%). Conclusions— AD is not associated<br />

with the genetic risk factors for vascular disease examined in<br />

this study.


ENDOPHENOTYPES<br />

79 GENOTYPE-CONTROLLED ANALYSIS OF<br />

PLASMA DOPAMINE beta-HYDROXYLASE IN<br />

CIVILIAN POST-TRAUMATIC STRESS DISORDER<br />

Y. Tang* (1), W. Li (1), K. Mercer (1), B. Bradley (2), C.<br />

Gillespie (1), R. Bonsall (1), K. Ressler (3), J. Cubells (1)<br />

1. Emory University School <strong>of</strong> Medicine 2. Dept. <strong>of</strong><br />

Psychiatry and Behavioral Sciences, Atlanta VA <strong>Medical</strong><br />

Center 3. Dept <strong>of</strong> Psychiatry, Emory University School <strong>of</strong><br />

Medicine; Yerkes National Primate Research Center; Howard<br />

Hugh <strong>Medical</strong> Institute<br />

*ytang5@emory.edu<br />

Background: Norepinephrine (NE) plays multiple roles in<br />

regulating arousal and autonomic stress responses and has<br />

been a central candidate in studying the pathophysiology <strong>of</strong><br />

post-traumatic stress disorder (PTSD). Dopamine<br />

β-hydroxylase (DβH) catalyzes the conversion <strong>of</strong> dopamine<br />

(DA) to norepinephrine (NE). Serum (or plasma) DβH activity<br />

is a highly heritable trait largely under the control <strong>of</strong> the DBH<br />

locus. Mustapic and colleagues (2007, Am. J. Med. Genet.<br />

Part B 144B: 1087) reported a PTSD-associated deficit in<br />

plasma DβH activity in a genotype-controlled analysis <strong>of</strong><br />

combat veterans. We tested whether such a deficit would<br />

occur in a predominantly African American sample <strong>of</strong><br />

civilians exposed to a high level <strong>of</strong> poverty-related stress and<br />

trauma. Methods: The primary outcome measure was severity<br />

<strong>of</strong> current adult PTSD symptoms, and current DSM-IV<br />

diagnosis <strong>of</strong> PTSD, as determined by the modified PTSD<br />

Symptom Scale (MPSS). Adulthood trauma exposure was<br />

assessed using the Traumatic Experience Inventory (TEI).<br />

Plasma DβH activity was measured by measuring conversion<br />

<strong>of</strong> tyramine to octopamine, isolating octopamine by HPLC,<br />

and detecting it by electrochemical detection. Genotypes were<br />

determined using the Taqman® platform. Results: Two<br />

hundred and thirty eight African American subjects were<br />

enrolled in this study. The mean age (± SD) was <strong>of</strong> 44±13<br />

years, with 57.8% being female. We found a strong<br />

association between rs1611115 genotype and plasma DβH<br />

activity in a co-dominant manner (p0.05) in any genotype group. There<br />

were no significant correlations found between DβH and<br />

PTSD severity and no differences <strong>of</strong> genotype frequency<br />

between PTSD cases and controls. The current results do not<br />

replicate the findings reported by Mustapic et al (2007) in a<br />

group <strong>of</strong> combat veterans with and without PTSD.<br />

Conclusions: We have replicated in an African American<br />

sample the prior finding that DBH rs1611115 genotype<br />

strongly associates with DβH activity in a co-dominant<br />

manner. There are no associations between DβH activity and<br />

PTSD diagnosis or symptom severity in this civilian sample.<br />

80 SENSORY GATING ENDOPHENOTYPE AND<br />

NEURONAL OSCILLATORY PATTERNS IN<br />

SCHIZOPHRENIA PATIENTS AND THEIR<br />

UNAFFECTED RELATIVES<br />

M. Hall* (1), A. Gibbs (1), G. Taylor (3), M. Coleman (2), V.<br />

Krause (2), O. Krastoshevsky (2), D. Salisbury (3), D. Levy<br />

(2)<br />

1. Psychology Research Laboratory & Cognitive Neuroscience<br />

Laboratory, McLean Hospital, Harvard <strong>Medical</strong> School<br />

2. Psychology Research Laboratory, McLean Hospital,<br />

Harvard <strong>Medical</strong> School 3. Cognitive Neuroscience<br />

Laboratory, McLean Hospital, Harvard <strong>Medical</strong> School<br />

*mhall@mclean.harvard.edu<br />

P50 sensory gating deficit is a leading endophenotype for<br />

schizophrenia (SZ). The auditory stimulus used to elicit P50<br />

event potentials induces a wide spectrum <strong>of</strong> oscillatory<br />

responses. Among them, beta-band oscillation (13-30 Hz)<br />

appears to contribute most to auditory P50 gating (Hong et al.,<br />

2008a) 1. However, gating <strong>of</strong> the beta oscillation was not<br />

impaired in patients with SZ or in their unaffected relatives<br />

(Hong et al., 2008b)2. The relationship between beta-band<br />

gating and P50 gating and the patterns <strong>of</strong> beta oscillation<br />

responses across brain regions remain to be determined.<br />

51 SZ patients, 25 unaffected first-degree relatives, and 34<br />

healthy comparison subjects were tested using a paired click<br />

paradigm. Evoked power <strong>of</strong> beta-band responses using time<br />

frequency analyses to S1 and S2 stimuli, gating <strong>of</strong> beta<br />

oscillatory responses (betaS2/betaS1), and gating <strong>of</strong> the P50<br />

wave were the main outcome measures. A P50 gating deficit<br />

was found in patients (p=.02). Relatives also showed<br />

moderately reduced suppression compared to controls<br />

(p=.087). Widespread reductions in beta-band responses to<br />

both S1 and S2 stimuli and impairment in gating <strong>of</strong> beta-band<br />

oscillation were found in patients compared with control<br />

subjects. Relatives did not differ significantly from control<br />

subjects. Correlation between the two gating measures was<br />

low (r =0-0.18).These results suggest that deficits in beta<br />

oscillatory responses are not endophenotypes for SZ. Rather,<br />

these deficits are associated with illness-related factors<br />

including age-<strong>of</strong>-onset, medication and BPRS scores. The<br />

underlying information processing assessed by the two gating<br />

measures appears to be independent.


81 A GENOME-WIDE LINKAGE STUDY OF<br />

COGNITIVE ENDOPHENOTYPES FOR<br />

SCHIZOPHRENIA HIGHLIGHTS THE<br />

NEUROTROPHIN RECEPTOR GENE (NTRK3) AS A<br />

SUSCEPTIBILITY LOCUS<br />

M. Aukes* (1), B. Alizadeh (2), C. Kemner (1), R. van 't Slot<br />

(1), E. Strengman (1), M. Sitskoorn (3), R. Sinke (2), R. Kahn<br />

(1), R. Oph<strong>of</strong>f (1)<br />

1. University <strong>Medical</strong> Center Utrecht 2. University <strong>Medical</strong><br />

Center Groningen 3. Tilburg University<br />

*m.aukes@umcutrecht.nl<br />

Integration <strong>of</strong> multiple lines <strong>of</strong> evidence is warranted in<br />

complex genetics. Our aim was to find new candidate loci that<br />

contain genes affecting candidate endophenotypes and<br />

potentially schizophrenia. We performed a genome-wide<br />

linkage scan <strong>of</strong> six schizophrenia endophenotypes with<br />

moderate heritability: sensorimotor gating, openness, verbal<br />

fluency, early visual perception, spatial working memory, and<br />

intelligence. Seven extended multiply affected Dutch<br />

pedigrees (n=118, 649 relative pairs) were genotyped for<br />

6,090 single nucleotide polymorphism markers. Two-point<br />

and multipoint variance-component based linkage analyses<br />

were performed using MERLIN. No genome-wide significant<br />

findings were observed. However, linkage peaks with<br />

suggestive evidence were located at 8q24 for early visual<br />

perception (LOD= 1.8), 17p13 and 16q21-22 for openness<br />

(LOD= 1.7 and 1.5). Suggestive two-point hits (empirical<br />

p-value


83 THE EFFECT OF COMT, BDNF AND NRG1 ON<br />

THE P50 ENDOPHENOTYPE IN PSYCHOSIS<br />

M. Shaikh*(1), M. Hua Hall (2), K. Schulze (1), A. Dutt (1),<br />

I. Williams (1), M. Constante (1), M. Walshe (1), M. Picchioni<br />

(1), T. Toulopoulou (1), D. Collier (1), F. Rijsdijk (1), J.<br />

Powell (1), M. Arranz (1), R. Murray (1), E. Bramon (1)<br />

1. The Biomedical Research Centre for Mental Health at the<br />

Institute <strong>of</strong> Psychiatry, Kings College London and The South<br />

London and Maudsley NHS Foundation Trust 2. Harvard<br />

<strong>Medical</strong> School, Psychology Research Laboratory, McLean<br />

Hospital, Belmont<br />

*madiha.shaikh@kcl.ac.uk<br />

Introduction: P50 gating deficits correlate with genetic risk for<br />

schizophrenia and constitute a plausible endophenotype for the<br />

disease. The well-supported role <strong>of</strong> COMT, BDNF and NRG1<br />

in neurodevelopment and cognition makes a strong theoretical<br />

case for the influence <strong>of</strong> these genes on P50 endophenotypes.<br />

Methods: The possible role <strong>of</strong> NRG1, COMT Val158Met and<br />

BDNF Val66Met gene polymorphisms on the P50<br />

endophenotype was examined in a large sample consisting <strong>of</strong><br />

psychotic patients, their unaffected relatives and unrelated<br />

healthy controls using linear regression analyses.<br />

Results: No evidence for association between NRG1, COMT<br />

Val158Met or BDNF Val66Met genotype and P50<br />

endophenotype was found. Discussion: The available<br />

neurophysiological evidence suggests that any such<br />

association between P50 indices and NRG1, COMT<br />

Val158Met or BDNF Val66Met genotypes, if present, must be<br />

very subtle.<br />

84 GENETIC AND NEUROCHEMICAL MODULATION<br />

OF BEHAVIOURAL AND NEURAL MEASURES OF<br />

RESPONSE INHIBITION<br />

M. Bellgrove* (1), T. Cummins (1), S. Nandam (1), P. Nathan<br />

(2), J. Wagner (1), J. Mattingley (1), R. Hester (3), C.<br />

Chambers (4)<br />

1. University <strong>of</strong> Queensland 2. University <strong>of</strong> Cambridge &<br />

GlaxoSmithKline. 3. University <strong>of</strong> Melbourne<br />

4. Cardiff University<br />

*m.bellgrove@uq.edu.au<br />

The ability to inhibit behavior is a key aspect <strong>of</strong> executive<br />

function that allows humans to behave flexibly. Twin studies<br />

have shown that behavioral measures <strong>of</strong> inhibitory control,<br />

such as stop-signal reaction time (SSRT), are highly heritable,<br />

and inhibitory dysfunction has been touted as a candidate<br />

endophenotype for ADHD and OCD. The neurochemical<br />

substrates <strong>of</strong> inhibitory control in the human brain remain<br />

unresolved with evidence supporting modulation by<br />

dopamine, noradrenaline and sertonin. We surveyed the<br />

association between a theoretically motivated set <strong>of</strong><br />

catecholamine genes, including DAT1, DRD4, DBH, COMT,<br />

DRD2 and NET, and SSRT (SSRT) in a sample <strong>of</strong> 250<br />

non-clinical adults. Associations were found between markers<br />

on DRD4, DBH and DRD2 that accounted for significant<br />

variance in SSRT, after controlling for age and gender. The<br />

influence <strong>of</strong> these genetic markers was further investigated in<br />

a targeted fMRI study <strong>of</strong> inhibitory control in a subset <strong>of</strong> the<br />

larger cohort (n=50). A further double-blind, placebo<br />

controlled, cross-over study <strong>of</strong> the influence <strong>of</strong> an acute<br />

challenge <strong>of</strong> methylphenidate (30mg), atomoxetine (60mg)<br />

and citalopram (30mg) on SSRT in non-clinical subjects<br />

(n=24) was also conducted. A significant effect <strong>of</strong> drug<br />

condition was found with inhibitory ability significantly<br />

improved under methylphenidate, compared with all other<br />

conditions. Our genetic, brain imaging and neurochemical data<br />

suggest a prominent role for catecholamine, particularly<br />

dopamine, modulation <strong>of</strong> inhibitory control but little evidence<br />

for modulation by sertonin.


85 CORTICAL THICKNESS IN SCHIZOPHRENIA<br />

AND BIPOLAR DISEASE: A TWIN MRI STUDY OF<br />

DISEASE-SPECIFIC AND COMORBID<br />

ENDOPHENOTYPES<br />

G. Van Baal* (1), H. Hulsh<strong>of</strong>f Pol (1), H. Schnack (1), R.<br />

Brouwer (1), R. Kahn (1)<br />

1. UMC Utrecht<br />

*G.C.M.vanBaal@umcutrecht.nl<br />

In the last few years, the nosological dichotomy between<br />

schizophrenia and bipolar disease has repeatedly been<br />

challenged by genetic findings. Lichtenstein et al. (The<br />

Lancet, 2009) found substantive heritabilities for both diseases<br />

(64% and 59% respectively), which were mostly due to<br />

comorbid genetic factors (52% <strong>of</strong> total genetic variance in<br />

schizophrenia, 69% in bipolar disease) and only<br />

partly disease-specific. This strongly encourages research <strong>of</strong><br />

endophenotypic traits that shed light on the disrupted<br />

mechanisms responsible for both diseases. Data on cortical<br />

thickness measured on a 1.5 Tesla MRI scanner are<br />

presented, which were collected in a large twin sample <strong>of</strong><br />

discordant schizophrenic (MZ/DZ: 12/12), concordant and<br />

discordant bipolar (22/25) and healthy control twin pairs<br />

(45/39), adding up to a total <strong>of</strong> 330 participants. Large parts <strong>of</strong><br />

the cortex were affected differently in schizophrenic<br />

vs. bipolar patients: schizophrenia patients showed thicker<br />

right inferior temporal and sensory-motor cortices than<br />

controls which was accounted for by genetic factors, bipolar<br />

patients showed a thinner right inferior frontal cortex than<br />

controls, which seemed to be due to unique environmental<br />

influences. Other areas were affected in a similar fasion in<br />

both diseases: both types <strong>of</strong> patients displayed decreased<br />

thickness <strong>of</strong> parahippocampal gyri, and increased thickness <strong>of</strong><br />

left temporoparietal cortex (wernicke's area). In general, these<br />

latter associations were caused by genetic factors. Combined<br />

with a (moderate) heritability, cortical thickness in these<br />

areas may be a promising endophenotype for the genetic<br />

mechanisms <strong>of</strong> comorbidity <strong>of</strong> these psychosis-related<br />

diseases.<br />

86 EFFECT OF CACNA1C RS1006737 ON NEURAL<br />

CORRELATES OF VERBAL FLUENCY IN HEALTHY<br />

INDIVIDUALS<br />

V. Nieratschker* (1), A. Krug (2), V. Markov (3), S. Kracht<br />

(2), A. Jansen (2), K. Zerres (4), T. Eggermann (4), T. Stöcker<br />

(5), N. Shah (5), J. Treutlein (6), T. Mühleisen (7), T. Kircher<br />

(2), M. Rietschel (1)<br />

1. Division <strong>of</strong> Genetic Epidemiology in Psychiatry, Central<br />

Institute <strong>of</strong> Mental Health, J 5, 68159 Mannheim, Germany<br />

2. Department <strong>of</strong> Psychiatry and Psychotherapy,<br />

Philipps-University Marburg, Rudolf-Bultmann-Str.<br />

8, 35039 Marburg, Germany 3. Department <strong>of</strong> Psychiatry and<br />

Psychotherapy, RWTH Aachen University, Pauwelsstr. 30,<br />

52074 Aachen, Germany 4. Institute <strong>of</strong> Human Genetics,<br />

RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen,<br />

Germany 5. Institute <strong>of</strong> Neuroscience and Biophysics 3 –<br />

Medicine, Research Center Jülich, Germany 6. Division <strong>of</strong><br />

Genetic Epidemiology in Psychiatry, Central Institute <strong>of</strong><br />

Mental Health, J 5, 68159 Mannheim, Germany 7. Department<br />

<strong>of</strong> Genomics, Life & Brain Center, University <strong>of</strong> Bonn,<br />

Sigmund-Freud-Strasse 25, 53127 Bonn, Germany<br />

*vanessa.nieratschker@zi-mannheim.de<br />

Background: Recent genome-wide association studies<br />

implicated the alpha 1C subunit <strong>of</strong> the L-type voltage-gated<br />

calcium channel (CACNA1C) gene as a susceptibility locus<br />

for bipolar disorder, schizophrenia and major depression<br />

(Sklar et al., 2008; Ferreira et al., 2009; Green et al., 2009).<br />

While the functions underlying the pathophysiology <strong>of</strong> these<br />

psychiatric disorders are yet unknown, impaired performance<br />

in verbal fluency tasks is an <strong>of</strong>ten replicated finding. We<br />

investigated the influence <strong>of</strong> the CACNA1C single nucleotide<br />

polymorphism (SNP) rs1006737 on verbal fluency and its<br />

neural correlates. Methods: Brain activation was measured<br />

with functional magnetic resonance imaging (fMRI) during a<br />

semantic verbal fluency task in 63 healthy male individuals.<br />

They additionally performed more demanding verbal fluency<br />

tasks outside the scanner. All subjects were genotyped for<br />

CACNA1C rs1006737. Results: For the behavioral measures<br />

outside the scanner, rs1006737genotype had an effect on<br />

semantic but not on lexical verbal fluency with decreased<br />

performance in risk-allele carriers. In the fMRI experiment,<br />

while there were no differences in behavioural performance,<br />

increased activation in the left inferior frontal gyrus as well as<br />

the left precuneus was found in risk-allele carriers in the<br />

semantic verbal fluency task. Conclusions: The rs1006737<br />

variant does influence language production on a semantic<br />

level in conjunction with the underlying neural systems. These<br />

findings are in line with results <strong>of</strong> studies in bipolar disorder,<br />

schizophrenia and major depression and may explain some <strong>of</strong><br />

the cognitive and brain activation variation found in these<br />

disorders.


87 SYSTEMATIC MUTATION SCREENING STUDY<br />

FOR PPP3CC IN SCHIZOPHRENIA<br />

H. Hwu* (1), C. Liu (1), U. Yang (2), P. Hsu (2), C. Fann (3),<br />

Y. Liu (4), C. Wen (1), C. Chang (3)<br />

1. Department <strong>of</strong> Psychiatry, National Taiwan University<br />

Hospital, Taipei, Taiwan 2. Institute <strong>of</strong> Bioinformatics,<br />

National YangMing University, Taipei, Taiwan 3. Institute <strong>of</strong><br />

Biomedical Science, Academia Sinica, Taipei, Taiwan 4.<br />

Division <strong>of</strong> Mental Health and Substance Abuse Research,<br />

National Health Research Institutes, Taipei, Taiwan<br />

* haigohwu@ntu.edu.tw<br />

Protein phosphatase 3 (formerly 2B), catalytic subunit, gamma<br />

is<strong>of</strong>orm (PPP3CC) has been reported associated with<br />

schizophrenia in literature and our previous association tudies.<br />

The aim <strong>of</strong> the study is to search the risk polymorphisms <strong>of</strong><br />

the gene for schizophrenia using systematic mutation<br />

screening method. We have sequenced the exons and promoter<br />

regions <strong>of</strong> PPP3CC. The samples for direct sequencing are 78<br />

schizophrenic patients and 50 normal controls. We have<br />

identified 22 genetic variations, <strong>of</strong> which 11 SNPs were<br />

genotyped for further analysis. In a preliminary study <strong>of</strong> small<br />

sample association study, which included 170 schizophrenic<br />

patients and 144 controls, we found one SNP in the 5’-UTR<br />

and two SNPs in the 3’-UTR region were significantly<br />

associated with schizophrenia either through genotype-wise or<br />

allele-wise analysis. The genotyping work in a large<br />

case-control sample, including 600 normal controls and 912<br />

schizophrenic patients, is ongoing.<br />

88 APPLICATION OF MULTIVARIATE GENETIC<br />

ANALYSIS TO FUNCTIONAL BRAIN IMAGING<br />

A. Scott *(1), K. McNealy (2), M. Dapretto (2), N. Schork (1)<br />

1. Scripps Translational Science Institute 2. University <strong>of</strong><br />

California, Los Angeles<br />

*ashleys@scripps.edu<br />

The technologic advances in neuroscience and genetics over<br />

the past decade have dramatically changed the methodological<br />

issues facing neurogenetic researchers. The advent <strong>of</strong><br />

high-through put genotyping assays, fine resolution brain<br />

imaging, and in-depth neuropsychological scales have allowed<br />

researchers to acquire massive amounts <strong>of</strong> data per subject.<br />

The aim <strong>of</strong> this study was to translate existing multivariate<br />

genetic analysis methods to brain imaging data as a first step<br />

towards an integrated multivariate imaging genetics approach.<br />

Using methods borrowed from analysis <strong>of</strong> high-dimensional<br />

gene expression data, we analyzed functional magnetic<br />

resonance imaging (fMRI) scans from 18 boys with autism<br />

spectrum disorders (ASD) and 18 age- and IQ-matched<br />

typically developing (TD) boys. Children were scanned while<br />

listening to a well-validated implicit language learning<br />

paradigm (McNealy et al., 2006; McNealy et al. 2009). For<br />

each subject, the mean timeseries from 106 regions-<strong>of</strong>-interest<br />

(ROI) over the cerebral volume were extracted. Multivariate<br />

distance matrix regression (MDMR; Wessel and Schork,<br />

2006; Schork et al. 2008) identified a number <strong>of</strong> ROIs<br />

significantly associated with autism diagnosis. Further<br />

examination <strong>of</strong> these ROIs revealed a subset <strong>of</strong> regions in<br />

which TD children showed more similar activation pr<strong>of</strong>iles<br />

than children with ASD, particularly in language-relevant<br />

areas. These findings are consistent with aberrant language<br />

processing in children with ASD, and demonstrate the<br />

capability <strong>of</strong> MDMR to identify shared brain activation<br />

patterns across individuals. This method can be extended to<br />

detect shared brain activation patterns among individuals with<br />

similar genetic pr<strong>of</strong>iles, providing a foundation for integration<br />

<strong>of</strong> functional brain imaging data and high-dimensional genetic<br />

data.


89 STRATEGIES FOR PURSUING INTERMEDIATE<br />

PHENOTYPES FOR BIPOLAR DISORDER: THE<br />

HEINZ C. PRECHTER LONGITUDINAL PROJECT<br />

S. Langenecker *(1), M. Kamail (1), L. Franti (1), A. Prossin<br />

(1), E. Saunders (1), A. Vederman (1), M. McInnis (1)<br />

1. University <strong>of</strong> Michigan <strong>Medical</strong> School<br />

*slange@med.umich.edu<br />

Introduction: Intermediate cognitive and personality<br />

phenotypes in Bipolar Disorder have been pursued with<br />

somewhat limited success to date. In part, this is due to the<br />

fact that state changes during the course <strong>of</strong> bipolar illness as<br />

well as high comorbidity with other psychiatric disorders and<br />

subtypes (alcohol and substance abuse, anxiety, psychosis,<br />

attention deficit disorder). Extensive clinical, environmental,<br />

personality and neuropsychological data are gathered from all<br />

participants, including regular follow-up evaluations to<br />

address the possibility <strong>of</strong> identifying reliable intermediate<br />

phenotypes. Methods: Diagnosis is conducted via structured<br />

Diagnostc interview for Genetic Studies, followed by<br />

Best-Estimate diagnosis in a preliminary cross-sectional<br />

analysis <strong>of</strong> healthy control subjects (HC, n = 75) and euthymic<br />

(E), depressed (D), or hypomanic/mixed (HM) patients with<br />

bipolar disorder (BD, n = 270). We identified intermediate<br />

cognitive phenotypes (ICPs) and intermediate personality<br />

phenotypes (IPP) from assessments in executive functioning,<br />

attention, memory, fine motor function, emotion processing<br />

and personality. Results: Of the eight cognitive factor scores,<br />

the HC group outperformed the E group in three (Processing<br />

Speed with Interference Resolution, Visual Memory, Fine<br />

Motor Dexterity). Conscientiousness from the NEO-PI was<br />

lower in the D and E groups compared to the HC group. The<br />

E group was higher than the HC group in Neuroticism from<br />

the NEO-PI. Conclusions: Use <strong>of</strong> the factor scores from<br />

cognitive and personality measures can assist in determining<br />

ICPs and IPPs for BD and related disorders, and may provide<br />

more specific targets for development <strong>of</strong> new analytic<br />

approaches and treatments.<br />

90 ASSOCIATION OF EVENT-RELATED<br />

OSCILLATIONS AND GLUTAMATE SYSTEM<br />

CANDIDATE GENES<br />

M. Rangaswamy* (1), N. Manz (1), T. Foroud (2), H.<br />

Edenberg (2), B. Porjesz (1), COGA<br />

1. SUNY Downstate <strong>Medical</strong> Center, Brooklyn, NY 2. Indiana<br />

University, Indianapolis, IN<br />

* madhavi.rangaswamy@downstate.edu<br />

Brain oscillations provide sensitive measures <strong>of</strong> brain function<br />

with exquisite temporal resolution during cognitive tasks.<br />

These quantitative neurophysiological endophenotypes <strong>of</strong>fer a<br />

powerful tool to study genetics <strong>of</strong> complex psychiatric<br />

disorders. Event related brain oscillations (EROs) - delta (1-3<br />

Hz) and theta (4-7 Hz) - were recorded during a visual oddball<br />

task from 1312 individuals from 251 multiplex alcoholic<br />

families in the Collaborative Study on the Genetics <strong>of</strong><br />

Alcoholism (COGA) sample. Significant linkage (LOD =<br />

3.97) and linkage disequilibrium on chromosome 4 at 139 cM<br />

was detected. Association analysis with a glutamatergic gene<br />

(GRID2) under this linkage peak revealed strong association<br />

with a large number <strong>of</strong> single nucleotide polymorphisms<br />

(SNPs). The human glutamate receptor gene GRID2 on<br />

chromosome 4, is a member <strong>of</strong> the glutamate receptor family<br />

which are the predominant excitatory neurotransmitter<br />

receptors in the central nervous system. In a genome-wide<br />

association study (GWAS) based on the Illumina 1 million<br />

SNP array, we analyzed EROs in response to the target<br />

stimulus with the same paradigm in 1064 genetically unrelated<br />

case-control subjects from the COGA study. SNP in several<br />

glutamatergic genomic regions were found to be significantly<br />

associated using a linear regression model for a case-control<br />

study. These findings from both the family based and<br />

case-control studies highlight the involvement <strong>of</strong> the<br />

glutamatergic pathways associated with cognitive dysfunction<br />

underlying alcoholism.


91 AN ASSOCIATION BETWEEN PREPULSE<br />

INHIBITION AND DRD2 GENE IN A SAMPLE OF<br />

HEALTHY ISRAELIS<br />

M. Monakhov* (1), R. Levin (2), S. Israel (2), U.<br />

Heresco-Levy (2), R. Bachner-Melman (3), R. Ebstein (2)<br />

1. Engelhardt Institute <strong>of</strong> Molecular Biology, Russian<br />

Academy <strong>of</strong> Sciences, Moscow, Russia 2. Herzog Memorial<br />

Hospital, Jerusalem, Israel 3. Psychiatry Department,<br />

Hadassah <strong>Medical</strong> School, Hebrew University , Jerusalem,<br />

Israel<br />

* misha.monahov@gmail.com<br />

Introduction: Prepulse inhibition (PPI) is well-established<br />

measure <strong>of</strong> sensorimotor gating, that has been associated<br />

with a number <strong>of</strong> phenotypes including social behavior and<br />

schizophrenia. It reflects the brain’s ability to filter incoming<br />

information <strong>of</strong> different sensory modalities and so might<br />

indirectly affect social decision making. Some studies report<br />

that D2 dopamine receptor agonists may decrease PPI in<br />

rodents and humans. We performed an association study <strong>of</strong><br />

PPI and four SNPs from human DRD2gene. Methods<br />

Prepulse inhibition <strong>of</strong> the startle response to auditory stimuli<br />

was measured in 38 healthy volunteers (20 males, 18 females;<br />

mean age 26±3.7 years) recruited in Jerusalem, Israel.<br />

Prepulse-pulse intervals <strong>of</strong> 30, 60 or 120 ms (PPI30, PPI60<br />

and PPI120 values, respectively) were used. DNA was<br />

isolated from blood samples <strong>of</strong> all participants and their<br />

parents, and genotyping <strong>of</strong> four DRD2 SNPs (rs1800498,<br />

rs1801028, rs6277, rs1800497) was performed with<br />

5'-nuclease assay or high resolution melt assay. Data<br />

Were analyzed with ANOVA and transmission disequilibrium<br />

test. Results and discussion:ANOVA <strong>of</strong> PPI among 38<br />

volunteers revealed an association between rs6277 SNP and<br />

60 ms interval PPI (F=5.4 p=0.009), where CC-homozygotes<br />

demonstrated less inhibition (mean±S.D. is 24.8±10.3) than<br />

T-allele carriers (54.4±3.9; for comparison <strong>of</strong> T+ vs. T-<br />

groups F=10.51, p=0.003). CC homozygotes also had visibly<br />

lower PPI with 30 and 120 ms interval, but differences did not<br />

reach statistical significance. Haplotype T-C-T-C was<br />

associated with higher 30 ms interval PPI<br />

(family-based UNPHASED analysis chisq=4.92, p=0.027). A<br />

trends in same direction, but not significant associations were<br />

observed for 60 and 120 ms interval tests. Results indicate a<br />

possible role <strong>of</strong> DRD2 in sensorimotor gating.


EPIGENETICS<br />

93 COMBINING EVIDENCE FOR MATERNAL AND<br />

IMPRINTING EFFECTS FROM MULTIPLE STUDIES<br />

B. Glaser* (1), G. Davey-Smith (1), A. Ades (2)<br />

1. <strong>Medical</strong> Research Council Centre for Causal Analysis in<br />

Translational Epidemiology and Department <strong>of</strong> Social<br />

Medicine, University <strong>of</strong> Bristol 2. Department <strong>of</strong> Community<br />

Based Medicine, University <strong>of</strong> Bristol<br />

* B.Glaser@bris.ac.uk<br />

The detection <strong>of</strong> small-scale genetic effects such as those<br />

exerted by maternal genotype or imprinting is <strong>of</strong>ten hampered<br />

by lack <strong>of</strong> sufficient study power. Joint analysis and<br />

meta-analysis can enhance power through increase in sample<br />

size and has been successfully applied to combine evidence<br />

for allelic and genotypic association. However methods to<br />

combine studies <strong>of</strong> parental influences such as those<br />

detectable in parent-<strong>of</strong>fspring trios are sparse. We developed a<br />

simple Bayesian approach to derive combined estimates for<br />

maternal and/or imprinting effects from multiple parent-child<br />

trio samples and mother-child pairs. For this, we extended the<br />

single-sample Poisson model for categorical outcomes<br />

(Weinberg et al., 1999) into a multinomial framework. Our<br />

approach accounts for sample heterogeneity through random<br />

effects, allows the combination <strong>of</strong> different sample designs<br />

and enhances the precision <strong>of</strong> effects estimates by including<br />

prior information. Using simulations, we studied the proposed<br />

method under different scenarios including varying minor<br />

allele frequencies, and genetic effect sizes for both maternal<br />

and imprinting effects. We will report results comparing fixed<br />

effect and random effects models using simulated and real<br />

data.<br />

94 COMPREHENSIVE DNA METHYLATION<br />

ANALYSIS IN THE HUMAN BRAIN AND ITS<br />

APPLICATION TO PSYCHIATRIC DISEASES<br />

K. Iwamoto* (1), M. Bundo (1), J. Ueda (1), Y. Nakano (1),<br />

M. Oldham (2), W. Ukai (3), E. Hashimoto (3), T. Saito (3),<br />

D. Geschwind (2), T. Kato (1)<br />

1. RIKEN Brain Science Institute 2. University <strong>of</strong> California<br />

Los Angeles 3. Sapporo <strong>Medical</strong> University<br />

* kaziwamoto@brain.riken.jp<br />

Altered DNA methylation status in the brain is proposed to be<br />

involved in the pathogenesis <strong>of</strong> psychiatric diseases. Although<br />

several studies reported altered DNA methylation in the brains<br />

<strong>of</strong> patients, few considered the effect <strong>of</strong> cellular heterogeneity<br />

in the brain. We employed the cell-sorter based method for<br />

separation <strong>of</strong> neuronal and non-neuronal nuclei from<br />

postmortem brain, and have performed comprehensive DNA<br />

methylation analyses using various qualitative and quantitative<br />

techniques including global methylation assay targeting whole<br />

genome and LINE-1 sequences, bead-based array, promoter<br />

tiling arrays as well as extensive bisulfite sequencing analyses.<br />

We found that while non-neuronal nuclei showed similar<br />

DNA methylation pattern to bulk cortex, neuronal nuclei<br />

manifested qualitatively and quantitatively distinctive DNA<br />

methylation patterns. Through the bioinformatic analysis, we<br />

found that genes related to neuronal activity were methylated<br />

in non-neuronal nuclei, while genes expressed in astrocytes<br />

were methylated in neuronal nuclei. Such DNA methylation<br />

differences may contribute to the phenotypic differences<br />

between neurons and non-neurons. We are performing a<br />

large-scale case-control study using these techniques.


95 MAPPING GENE REGULATORY ELEMENTS FOR<br />

BRAIN EXPRESSED GENES USING<br />

HIP-SEQUENCING<br />

C. Barr* (1), Y. Feng (1), R. Bremner (1)<br />

1. The Toronto Western Research Institute<br />

* cbarr@uhnres.utoronto.ca<br />

Current evidence indicates that psychiatric disorders are more<br />

likely to result from genetic variation that changes gene<br />

expression rather than variations in the coding region.<br />

However, the role <strong>of</strong> variation in gene regulation as a<br />

contributor to psychiatric disorders has been largely ignored<br />

because <strong>of</strong> the difficulty in identifying the location <strong>of</strong> gene<br />

regulatory elements outside <strong>of</strong> the proximal promoter. We<br />

used a genome-wide approach to identify brain-relevant gene<br />

regulatory elements, in two cell lines, a neuroblastoma and a<br />

glioblastoma. We used chromatin immunoprecipitation<br />

(ChIP) to modified histones and transcription factors<br />

combined with high throughput sequencing (ChIP-sequencing)<br />

to identify the position <strong>of</strong> brain-relevant regulatory elements<br />

across the genome. To maximize the identification <strong>of</strong><br />

regulatory elements, we performed ChIP using antibodies to<br />

acetylated histone H3, (a marker <strong>of</strong> active chromatin), the<br />

enhancer-associated protein p300 (a marker <strong>of</strong> enhancers), and<br />

RNA polymerase II (a marker <strong>of</strong> regions <strong>of</strong> active<br />

transcription). This approach was very successful, and we<br />

now have genomic maps <strong>of</strong> putative regulatory elements for<br />

genes expressed in these cell lines including genes associated<br />

with psychiatric disorders (e.g. DTNBP1, SNAP25). We also<br />

mapped putative gene regulatory elements in gene “deserts”<br />

in the region <strong>of</strong> positive markers from the GWAS studies. We<br />

are currently screening the putative regulatory regions in<br />

associated genes for genetic variation and testing the<br />

relationship to the respective psychiatric disorder in DNA<br />

from families. We are following on the success <strong>of</strong> this<br />

approach by identifying regulatory regions in brain tissue,<br />

particularly the DLPFC and the hippocampus.<br />

97 HYPOMETHYLATION OF HTR2A PROMOTER AT<br />

T102C POLYMORPHIC SITE IN DNA DERIVED<br />

FROM THE SALIVA OF PATIENTS WITH<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER<br />

M. Ghadiri* (1), S. Nohesara (1), H. Ahmankhaniha (1), E.<br />

Shirazi (1)<br />

1. Iran University <strong>of</strong> <strong>Medical</strong> Sciences<br />

* ghadiri_mohamad@yahoo.com<br />

Introduction: Contribution <strong>of</strong> the 5HT2A gene in the<br />

pathogenesis <strong>of</strong> schizophrenia (SCZ) and bipolar disorder<br />

(BD) has been the focus <strong>of</strong> many studies, because LSD which<br />

resembles serotonin causes psychosis and atypical<br />

antipsychotic drugs elicit their effects by blocking the<br />

5HT2A receptors. There has been evidence for the role <strong>of</strong><br />

5HT2A polymorphism(s) in SCZ and BD pathogenesis.<br />

Furthermore, DNA methylation <strong>of</strong> HTR2A promoter DNA at<br />

T102C polymorphic site was correlated with the HTR2A<br />

expression level in the human brain. Also, DNA<br />

hypomethylation <strong>of</strong> this site was reported in the post-mortem<br />

brain <strong>of</strong> patients with SCZ and BD. Hypothesizing that<br />

epigenetic dysregulation <strong>of</strong> 5HT2A in peripheral tissues may<br />

be used as a diagnostic/therapeutic marker we analyzed<br />

HTR2A promoter DNA methylation status <strong>of</strong> DNA derived<br />

from the saliva <strong>of</strong> patients with SCZ and BD vs. normal<br />

controls (each 16). Method:Bisulfite sequencing was used to<br />

screen the CpG methylation status <strong>of</strong> the 5HT2A entire<br />

promoter region. qMSP was used to quantify the degree <strong>of</strong><br />

cytosine methylation at differentially methylated sites in<br />

individuals carrying the C allele <strong>of</strong> the HTR2a t102c<br />

polymorphism. Results: Based on bisulfite sequencing,<br />

HTR2A promoter DNA was generally unmethylated in the<br />

DNA derived from the saliva however cytosine at T102C<br />

polymorphic site and neighboring cytosines were partially<br />

methylated both in the patient and controls. qMSP analysis<br />

revealed that this cytosine was significantly hypomethylated<br />

in SCZ and BD versus the control subjects. Conclusion:<br />

Cytosine methylation <strong>of</strong> HTR2A at T102C polymorphic site in<br />

DNA derived from the saliva could be used as a diagnostic,<br />

prognostic and/or therapeutic marker in SCZ and BD.


98 GENOME-WIDE METHYLATION ANALYSIS OF<br />

POSTMORTEM BRAINS SUGGESTS<br />

HYPOMETHYLATION IS ASSOCIATED WITH<br />

MAJOR DEPRESSION<br />

L. Cheng* (1), D. Zhang (1), C. Chen (1), J. Badner (1), W.<br />

Luo (1)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Neuroscience,<br />

The University <strong>of</strong> Chicago<br />

* lcheng2@bsd.uchicago.edu<br />

DNA methylation plays an important role in neuronal<br />

development and differentiation, with potential implications<br />

for neuropsychiatric disease susceptibilities. We performed a<br />

genome-wide methylation study using Illumina Infinium<br />

HumanMethylation27 BeadChip with 27,578 DNA CpG<br />

methylation loci on 153 cerebellum brain DNA samples from<br />

individuals <strong>of</strong> European ancestry. These samples are from two<br />

collections <strong>of</strong> the Stanley <strong>Medical</strong> Research institute and<br />

include 45 Schizophrenia (SZ), 46 Bipolar Disorder (BD), 15<br />

Major Depression (MD) and 47 unaffected control samples.<br />

After removing covariate and batch effects and after<br />

quantile normalization, an ANOVA test was used to detect<br />

differential methylation comparing cases and controls in the<br />

top half CpG loci with high methylation variation. No CpG<br />

locus in SZ, MD and BD show significant difference<br />

compared to controls with FDR q < 0.05. There were<br />

796, 1000 and 706 loci that showed nominally significant<br />

methylation differences with P ≤ 0.05 in SZ, MD and BD<br />

compared to controls, respectively. 33 loci showing<br />

nominally significant methylation changes were shared across<br />

all three diseases. Significantly more hypomethylated<br />

loci in differentially methylated sites were detected in MD (P<br />


GENE X ENVIRONMENT INTERACTIONS<br />

100 GENE-ENVIRONMENT INTERACTION OF THE<br />

LEPTIN RECEPTOR (LEPR) GENE AND TRAUMATIC<br />

LIFE EVENTS MODERATES DEPRESSIVE<br />

SYMPTOMS AND HPA-AXIS FUNCTION<br />

P. Zimmermann (1), T. Brueckl (1), H. Pfister (1), H.<br />

Wittchen (2), F. Holsboer (1), M. Ising (1), S. Lucae (1), R.<br />

Lieb (3), S. Kloiber* (1)<br />

1. Max-Planck-Institute <strong>of</strong> Psychiatry 2. Technische<br />

Universitaet Dresden<br />

* stkloiber@mpipsykl.mpg.de<br />

The comorbidity <strong>of</strong> depressive and metabolic disorders led to<br />

the suggestion, that metabolic networks could be involved in<br />

the pathophysiology <strong>of</strong> depression. Leptin, a major regulator<br />

<strong>of</strong> metabolic networks and has been shown to influence sleep<br />

and cognition and to exert antidepressant like effects, possibly<br />

via exerting a negative feedback inhibition on the HPA-axis.<br />

In addition, stressful life events, such as trauma, are assumed<br />

to act as environmental triggers for the onset <strong>of</strong> depression<br />

and, therefore, are capable to moderate the genetic<br />

vulnerability for depression. Analyzing the leptin system<br />

genes (LEP, LEPR, LEPROT, and LEPROTL1; 50 SNPs,<br />

Illumina 550k Bead Chip) in our 10 year prospective<br />

epidemiological sample (EDSP) consisting <strong>of</strong> 160 nuclear<br />

families including <strong>of</strong>fspring with a lifetime diagnosis <strong>of</strong> a<br />

major depression (baseline: 3021 individuals aged<br />

14-24 years), we detected SNPs in the LEPR gene displaying a<br />

clear indirect effect on depressive symptoms, which is<br />

moderated by severe traumatic events as environmental risk<br />

factor. Furthermore, the cortisol and ACTH response in the<br />

Dex/CRH test was significantly elevated in depressed patients<br />

carrying the Q223R mutation in a sample <strong>of</strong> 250 patients with<br />

major depression (MARS). Traumatic events in the patients<br />

history amplified this difference. Results remained significant<br />

after correction for age, gender and multiple testing.<br />

It is conceivable that a malfunctional leptin regulation<br />

influences cortisol levels and glucocorticoid receptor function,<br />

which in turn may contribute to the development <strong>of</strong> insulin<br />

resistance as well as improper regulation <strong>of</strong> the HPA axis in<br />

response to stress. While the former increases the risk for<br />

diabetes or other metabolic diseases, the latter poses a risk<br />

factor for the development <strong>of</strong> depression, especially, in<br />

combination with traumatic events. Our findings, therefore,<br />

provide further evidence that vulnerability for mood disorders<br />

as well as metabolic disturbances might emanate from a<br />

common pathway for which the leptin system might play a<br />

crucial role.<br />

101 INFLUENCE OF CRHR1 POLYMORPHISMS AND<br />

ADVERSE EVENTS ON DEPRESSION AND ANXIETY<br />

IN A COMMUNITY SAMPLE<br />

T. Brückl* (1), M. Ising (1), H. Pfister (1), A. Nocon (1), R.<br />

Lieb (2), F. Holsboer (1), P. Zimmermann (1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry 2. University <strong>of</strong> Basel<br />

* brueckl@mpipsykl.mpg.de<br />

Background: Depression and anxiety are highly comorbid<br />

disorders that can be conceived as stress-related disorders<br />

(SRD). Both disorders are likely to develop after exposure to<br />

stressful life events and are characterized by abnormalities <strong>of</strong><br />

the stress hormone regulation. The physical stress<br />

response is mediated by the corticotropin-releasing hormone<br />

(CRH) and polymorphisms in the corticotropin-releasing<br />

hormone type 1 (CRHR1) receptor gene have been implicated<br />

in affective and anxiety disorders (Bradley et al., 2008; Keck<br />

et al., 2008; Liu et al., 2006). The aim <strong>of</strong> this study is to<br />

examine main and interaction effects <strong>of</strong> CRHR1<br />

polymorphisms and adverse events on the subsequent onset <strong>of</strong><br />

affective and anxiety disorders. Methods: Data come from the<br />

longitudinal-epidemiological Early Developmental Stages <strong>of</strong><br />

Psychopathology (EDSP) Study comprising four waves <strong>of</strong><br />

data collection (T0-T3) in a randomly selected community<br />

sample <strong>of</strong> adolescents and young adults aged 14 to 24 years at<br />

baseline and 21 to 34 at third follow-up. Analyses are based<br />

on Caucasian study members who completed the third<br />

follow-up investigation and who consented to participate in<br />

the molecular-genetic part <strong>of</strong> the study (N=981). DSM-IV<br />

diagnoses and adverse events (e.g., trauma, separation events)<br />

were assessed using the Munich-Composite International<br />

Diagnostic Interview (M-CIDI). The association between prior<br />

adverse event and subsequent affective/anxiety disorder was<br />

assessed through Cox regressions with hazard ratios (HRs)<br />

and time-dependent covariates stratified by sex and age.<br />

Genotyping <strong>of</strong> the 6 selected SNPs was performed on a<br />

Sequenom platform employing the iPlex technology. Results:<br />

Results showed both an environmental and a genetic main<br />

effect. Prior exposure to an adverse event predicted the<br />

subsequent onset <strong>of</strong> any affective or anxiety disorder (HR=1.9,<br />

95%CI: 1.5-2.2, p=.000) and heterozygotes <strong>of</strong> the SNP<br />

“rs242940” reported significantly higher rates <strong>of</strong> any affective<br />

or anxiety disorder than minor allele homozygotes (45.3% vs.<br />

34.6%, Odds Ratio=1.6. 95% CI: 1.1-2.3, corr. p=.024). We<br />

also found a significant gene-environment interaction for the<br />

same SNP (rs242940) that withstood multiple testing (HR for<br />

interaction = 1.5, 95%CI: 1.1-2.0, corr. p-value=.048): Among<br />

individuals who had experienced an adverse event, carriers <strong>of</strong><br />

the minor allele had significantly lower rates <strong>of</strong><br />

affective/anxiety disorders than heterozygotes or major allele<br />

homozygotes, while no genotype-dependent differences in the<br />

rate <strong>of</strong> affective/anxiety disorder could be observed for<br />

individuals without exposure to an adverse event. Conclusion:<br />

CRHR1 polymorphisms seem to moderate the effect <strong>of</strong><br />

adverse events on the onset <strong>of</strong> affective or anxiety disorders<br />

whereby minor allele carriers appear to be protected against<br />

the pathogenic effect <strong>of</strong> adverse events. Liu Z et al. (2006)<br />

Association <strong>of</strong> corticotropin-releasing hormone receptor1 gene<br />

SNP and haplotype with major depression. Neuroscience


Letters 404:358-362 Keck M et al. (2008) Combined effects <strong>of</strong><br />

exonic polymorphisms in CRHR1 and AVPR1B Genes in a<br />

case/control study for panic disorder. Am J Med Genet Part B<br />

147B:1196-1204. Bradley RG et al. (2008) Influence <strong>of</strong> child<br />

abuse on adult depression. Arch Gen Psychiatry 65:190-200<br />

102 STRESSFUL LIFE EVENTS AND BDNF IN<br />

BIPOLAR DISORDER<br />

G. Hosang* (1), R. Uher (1), R. Keers (1), N. Craddock (2),<br />

M. Owen (2), A. Korszun (3), L. Jones (4), I. Jones (5), P.<br />

McGuffin (6), A. Farmer (7)<br />

1. Social, Genetic and Developmental Psychiatry Centre,<br />

Institute <strong>of</strong> Psychiatry, King's College London 2. Cardiff<br />

University 3. Queen Mary, University <strong>of</strong> London<br />

4.Birmingham University<br />

5. Cardiff University 6. Social, Genetic and Developmental<br />

Psychiatry Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London 7. Social, Genetic and Developmental Psychiatry<br />

Centre, Institute <strong>of</strong> Psychiatry, King's College London<br />

*g.hosang@iop.kcl.ac.uk<br />

Background: Bipolar disorder is a highly heritable illness,<br />

although identification <strong>of</strong> risk genes remains elusive.<br />

Gene-environment interactions have been the focus <strong>of</strong> much<br />

research since they are included in the heritability parameter.<br />

The aim <strong>of</strong> the present study was to examine the interplay<br />

between the BDNF Val66Met polymorphism and stressful life<br />

events (SLEs) in bipolar disorder. Method: A total <strong>of</strong> 1026<br />

participants were recruited, 440 bipolar I cases and 585<br />

psychiatrically healthy controls. All participants completed<br />

the List <strong>of</strong> Threatening Life Events Questionnaire, bipolar<br />

subjects reported the events that occurred 6 months leading up<br />

to their worst manic episode and 6 months prior to their worst<br />

depressive episode; controls recorded events experienced 6<br />

months before their interview. The BDNF Val66Met<br />

polymorphism was genotyped for the entire sample. Results.<br />

Both BDNF genotype and SLEs were significantly associated<br />

with bipolar disorder. For the worst depressive episodes the<br />

effects <strong>of</strong> SLEs was significantly moderated by BDNF<br />

genotype, but only for women. Conclusions: The findings <strong>of</strong><br />

this study highlight the importance <strong>of</strong> the interplay between<br />

genes and the environment in bipolar disorder.<br />

103 WHY GENOTYPE-BY-ENVIRONMENT (GXE)<br />

INTERACTIONS DESERVE MORE ATTENTION: A<br />

LITERATURE REVIEW OF GXE INTERACTIONS IN<br />

PSYCHIATRY<br />

L. Duncan* (1), E. Willcutt (1), M. Keller (1)<br />

1. Institute for Behavioral Genetics, University <strong>of</strong> Colorado at<br />

Boulder<br />

*Laramie.Duncan@Colorado.edu<br />

Background: Considerable resources have been devoted to<br />

GxE studies and meta-analyses <strong>of</strong> GxE interactions. However,<br />

to our knowledge, no one has summarized the state <strong>of</strong><br />

empirical GxE research from a broader perspective. Method:<br />

Literature review. Results: Across the 46 empirical GxE<br />

studies that met criteria for inclusion in this review, 99<br />

interactions were examined. Fifty-nine percent <strong>of</strong> the<br />

empirical studies examined one <strong>of</strong> two interactions:<br />

‘Depression ~ 5-HTTLPR x Stressful Life Events’ OR<br />

‘Antisocial ~ MAOA x Maltreatment’. The former was<br />

generally refuted by meta-analyses, and the latter<br />

supported. Statistically significant interactions in the other<br />

41% <strong>of</strong> empirical studies involved a wide variety <strong>of</strong><br />

phenotypes and environments. However, across all studies,<br />

only ten unique genes were studied, and 96% <strong>of</strong> genes in<br />

interactions were related to serotonin and/or dopamine<br />

signaling. The pool <strong>of</strong> participants across studies was<br />

representative for sex, but was not representative <strong>of</strong> racial and<br />

ethnic diversity in the United States or the world.<br />

Conclusions: Very few unique interactions have been studied,<br />

rendering empirical evidence about the realm <strong>of</strong> potential GxE<br />

interactions almost non-existent. Therefore, we cannot<br />

conclude that GxE studies “aren’t working” based on<br />

available data. Research dollars would be well-spent<br />

conducting genome-wide by environment studies, attempting<br />

to replicate the most promising GxE studies already<br />

conducted, and analyzing putative GxE interactions in extant<br />

datasets. Attention must be paid to the problem <strong>of</strong> multiple<br />

testing in GxE research. Additionally, work is needed to<br />

conduct GxE research in genetically diverse samples because<br />

minority populations have been underrepresented.


104 LACK OF EFFECT BETWEEN CANNABIS AND<br />

BDNFVAL66MET POLYMORPHISM INTERACTION<br />

ON THE EMERGENCE OF PSYCHOSIS IN A<br />

POPULATION-BASED SAMPLE.<br />

L. Michelon* (1), G. Busatto (1), R. Murray (2), P. Menezes<br />

(3), M. Scazufca (3), H. Vallada (1)<br />

1. Department <strong>of</strong> Psychiatry, Universidade de São Paulo<br />

(USP), São Paulo (SP), Brazil 2. Institute <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> London, London, UK 3. Department <strong>of</strong><br />

Preventive Medicine, Universidade de São Paulo (USP), São<br />

Paulo (SP), Brazil<br />

*lmichelon2@uol.com.br<br />

Background: Brain-derived neurotrophic factor (BDNF) has a<br />

critical role in neurodevelopment, neuroprotection and<br />

cognition. The rs6265 (Val66Met) polymorphism has been<br />

consistently associated with clinical features <strong>of</strong><br />

neuropsychiatric disorders. Exposure to cannabis has also<br />

been associated with cognitive deficits and psychosis.<br />

Cannabinoids are known to modulate BDNF serum levels<br />

suggesting associated pathophysiological mechanism<br />

underlying psychosis. Accordingly, this gene-environment<br />

(GE) interaction may be <strong>of</strong> particular importance for the<br />

emergence <strong>of</strong> psychosis. Materials and Methods: We<br />

investigated the interaction between cannabis exposure and<br />

BDNF Val66Met polymorphism as a risk for psychosis in an<br />

epidemiological-based series <strong>of</strong> patients with first onset<br />

psychosis (n=134) recruited in the city <strong>of</strong> São Paulo/Brazil.<br />

Non-psychotic control subjects (n=191) were randomly<br />

selected from the same geographical areas. Results: Allelic<br />

frequencies for BDNF variants were in Hardy–Weinberg<br />

equilibrium (P>0.05). Psychotic subjects and controls did not<br />

differ in terms <strong>of</strong> sex, age and ethnic background. We could<br />

not demonstrate any association in this population between<br />

psychosis and BDNF variants. Logistic regression analyzes<br />

showed no GE interactions between cannabis use and<br />

genotype distributions or allele frequencies. The analyses were<br />

controlled for sex, age, ethnic background, scholarship and<br />

partnership. Instead, psychosis was significantly associated<br />

with cannabis exposure (X2 =5.18, p=0.02). Conclusions: The<br />

altered levels <strong>of</strong> BDNF may reflect the brain damaged caused<br />

by cannabis and not the result <strong>of</strong> a GE interaction. Despite the<br />

negative findings for rs6265 polymorphism in this population,<br />

gene-cannabis interactions seem to be a reasonable<br />

explanation to its connection with psychosis. Thus, the<br />

mechanisms underlying the association between cannabis and<br />

psychosis may be dependent <strong>of</strong> other genes variants.<br />

105 ASSOCIATION OF A HAPLOTYPE OF COMT<br />

WITH CANNABIS USE IN A FIRST EPISODE<br />

PSYCHOSIS COHORT<br />

S. Luzi* (1), F. Ducci (1), C. Iyegbe (1), J. Powell (1), R.<br />

Murray (1), M. Di Forti (1)<br />

1. Adult Psychiatry, Institute <strong>of</strong> Psychiatry, London, United<br />

Kingdom<br />

*sonija.luzi@iop.kcl.ac.uk<br />

Background: Cannabis intoxication can cause brief psychotic<br />

symptoms, whilst chronic abuse has features similar to the<br />

negative symptoms <strong>of</strong> schizophrenia. An interaction between<br />

cannabis use and the Val458Met polymorphism <strong>of</strong> COMT has<br />

been reported. The COMT gene is a key enzyme in<br />

catecholamine degradation and the Val458Met polymorphism<br />

has been reported to influence enzymatic activity (Lachman et<br />

al. 1996). Other polymorphisms within the COMT gene have<br />

also been studied. We hypothesized that a haplotype study<br />

may be a more sensitive measure <strong>of</strong> genetic variation.<br />

Methods: A total <strong>of</strong> 381 first episode psychosis subjects were<br />

genotyped for four polymorphisms using TaqMan SNP<br />

genotyping assays. Statistical analysis was performed applying<br />

a case only design. S<strong>of</strong>tware used: Hapstat, Haploview and<br />

PHASE. Results: Results show an association between a<br />

COMT haplotype and use <strong>of</strong> cannabis OR 2.02 (± 0.61);<br />

P-Value 0.01. When age <strong>of</strong> first cannabis use (first use before<br />

or after 16 years <strong>of</strong> age) was analysed, a more significant<br />

association was found: OR 2.65 (± 1.00); P-Value 0.009. After<br />

the sample was stratified by ethnicity, an association was<br />

observed in the Caucasian group: P-Value 0.04, but not in the<br />

Black Caribbean and African group (P-Value 0.21). No<br />

association between a COMT haplotype and cannabis use was<br />

found in a control group (P-Value 0.78). Conclusions: Our<br />

results suggest an interaction between a COMT haplotype and<br />

cannabis use, particularly when age <strong>of</strong> first use is during<br />

adolescence. Haplotype studies may capture genetic variations<br />

in the COMT gene more robustly.


GENETIC COUNSELING<br />

106 FAMILIARITY WITH MENTAL ILLNESS AND<br />

FEMALE PATIENTS’ INTEREST IN OBTAINING<br />

RISK ESTIMATES FOR THEIR CHILDREN TO<br />

DEVELOP SEVERE MENTAL ILLNESS<br />

T. Lepard* (1), C. Thrush (1), N. Agan (1), S. Jackson (1)<br />

1. University <strong>of</strong> Arkansas for <strong>Medical</strong> Sciences<br />

*tlepard@uams.edu<br />

It has been suggested that as the genetic component <strong>of</strong> mental<br />

illness (MI) becomes better understood, individuals with a<br />

personal or family history <strong>of</strong> MI will increasingly become<br />

interested in psychiatric risk assessment for themselves and<br />

their <strong>of</strong>fspring. Several studies have shown a high interest for<br />

psychiatric genetic testing, should it become available.<br />

However, no studies have assessed interest in receiving<br />

empiric risk estimates for <strong>of</strong>fspring to develop MI, which is<br />

available, and who may be interested in learning this<br />

information. This study assessed female participants’ interest<br />

in learning empiric risk estimates for their <strong>of</strong>fspring to develop<br />

MI, and explored associations between this interest and<br />

familiarity with severe MI and other participant<br />

characteristics. A total <strong>of</strong> 174 women receiving care at a<br />

university based obstetrical/gynecological (Ob/Gyn) clinic<br />

completed the study survey. Most participants (75%) were<br />

somewhat-to-very interested in learning empiric risk estimates<br />

for their <strong>of</strong>fspring, while 25% were not-at-all or only slightly<br />

interested. Overall, a positive correlation existed between<br />

level <strong>of</strong> familiarity with MI and the desire to learn empiric risk<br />

estimates [rs(174)=0.19, p=0.01]. This correlation was<br />

stronger among participants who were pregnant [rs(114)=0.31,<br />

p=0.001]. Among pregnant participants with a high school<br />

education, there was a large positive association between<br />

familiarity with MI and interest in risk estimates [rs(35)=0.59,<br />

p=0.001], but this correlation was negligible for pregnant<br />

participants who had attended college [rs(78) =0.107,<br />

p=0.35]. Ob/Gyn patients, particularly when pregnant, are<br />

interested in empiric risk estimates for MI in their <strong>of</strong>fspring.<br />

These results should encourage prenatal genetic counselors to<br />

engage their patients in discussion <strong>of</strong> these empiric risks.<br />

107 GENETIC COUNSELING FOR SCHIZOPHRENIA:<br />

A REVIEW OF REFERRALS TO A PROVINCIAL<br />

MEDICAL GENETICS PROGRAM FROM 1968-2007<br />

M. Hunter (1), C. Hippman (1), W. Honer (1), J. Austin* (1)<br />

1. University <strong>of</strong> British Columbia<br />

*jehannine.austin@ubc.ca<br />

Introduction: Recent studies have shown that individuals with<br />

schizophrenia and their family members are interested in<br />

genetic counseling, but few have received this service. We<br />

conducted an exploratory, retrospective study to describe (a)<br />

the population <strong>of</strong> individuals who were referred to the<br />

provincial program for genetic counseling for a primary<br />

indication <strong>of</strong> schizophrenia, and (b) trends in number <strong>of</strong><br />

referrals between 1968 and 2007. Methods: Referrals for a<br />

primary indication <strong>of</strong> schizophrenia were identified through<br />

the provincial program database. Charts were reviewed and<br />

the following information was recorded: discipline <strong>of</strong> referring<br />

physician, demographics, psychiatric diagnosis, referred<br />

individual’s and partner’s (if applicable) family history, and<br />

any current pregnancy history. Data was characterized using<br />

descriptive statistics. Results: Between 1968 and 2007, 288<br />

referrals were made for a primary indication <strong>of</strong> schizophrenia.<br />

Most referrals were made: (a) for individuals who had a<br />

first-degree family member with schizophrenia, rather than for<br />

affected individuals, (b) for preconception counseling, and (c)<br />

by family physicians (69%), with only 2% by psychiatrists.<br />

Conclusions: In British Columbia, individuals affected with<br />

schizophrenia and their family members are rarely referred for<br />

psychiatric genetic counseling. There is a need to identify<br />

barriers to psychiatric genetic counseling and develop<br />

strategies to improve access.


GENETIC EPIDEMIOLOGY<br />

108 FAMILY BASED-ASSOCIATION STUDY<br />

BETWEEN EATING DISORDER AND<br />

SEROTONINERGIC GENES.<br />

B. Camarena* (1), S. Hernández (1), L. Gonzalez (1), A.<br />

Caballero (1)<br />

1. Instituto Nacional de Psiquiatría Ramón de la Fuente<br />

*camare@imp.edu.mx<br />

Evidence from family and twin studies suggests a genetic<br />

contribution in the etiology <strong>of</strong> eating disorders (ED). The<br />

efficacy <strong>of</strong> serotonin reuptake inhibitors in the treatment <strong>of</strong><br />

ED suggests the serotonin transporter gene (SLC6A4) as a<br />

good candidate for genetic studies. A functional<br />

polymorphism (5-HTTLPR) located on the promoter region<br />

has been associated with basal transcriptional activity levels.<br />

In addition, the 5-HT1Dβ is involved in the regulation <strong>of</strong> 5-HT<br />

release from serotoninergic neuron terminals in the brain.<br />

Therefore, abnormal function <strong>of</strong> this auto-receptor could be<br />

involved in the serotonin dysfunction suggested in ED. We<br />

studied two polymorphisms <strong>of</strong> SLC6A4 and 5-HT1Dβ genes<br />

using a family-based association method. We analyzed 100<br />

families. This sample included 46% with bulimia nervosa,<br />

26% restricting anorexia nervosa and 21% purging anorexia<br />

nervosa subtypes, and 7% EDNOs. The patients showed high<br />

scores <strong>of</strong> HAM-D and HAM-A. TCI scores showed higher<br />

scores <strong>of</strong> harm avoidance and lower scores <strong>of</strong><br />

self-directedness compared with a control group. Plutchiks<br />

Impulsivity Scale showed high scores in ED patients.<br />

We analyzed the 5-HTTLPR and G861C polymorphisms 100<br />

family trios. Statistical analysis was performed using the<br />

FBAT method. Also, we analyzed clinical phenotypes with<br />

evidence <strong>of</strong> heritability and relevance to ED, such as BMI,<br />

impulsivity, aggressivity and anxiety. FBAT analysis showed<br />

linkage disequilibrium between 5-HTTLPR and eating<br />

disorders (z=3.01, p=0.0025). Also, FBAT quantitative<br />

analysis showed statistical differences between 5-HTTLPR<br />

and impulsivity (z=2.36, p=0.018) and 5-HT1Dβand anxiety<br />

(z=2.68, p=0.0073). Analysis <strong>of</strong> alternative phenotypes in<br />

larger samples <strong>of</strong> informative is necessary to try to find the<br />

gene involves in ED. Acknowledgements: The study was<br />

supported by Pfizer Scientific Institute Grant 2006, México<br />

City.<br />

109 DOWN SYNDROME IN LITHUANIA: SURVIVAL<br />

AND DISABILITY<br />

A. Sinkus* (1), L. Jurkeniene (1), I. Andriuskeviciute (1), G.<br />

Sinkute (1), L. Salomskiene (1)<br />

1. Kaunas Univ. <strong>of</strong> Medicine<br />

*sinkus@vision.kmu.lt<br />

Every year the medical boards in Lithuania (with the<br />

population 3.4 million people) ascribe about 800<br />

sixteen-year-olds to disablement groups, which makes 1.6% <strong>of</strong><br />

the population. Half <strong>of</strong> these individuals, about 400 people,<br />

are disabled because <strong>of</strong> severe mental retardation. Each year in<br />

Lithuania 70 babies suffering from Down syndrome are born<br />

but at the age <strong>of</strong> 16 Down syndrome is diagnosed only for<br />

12-16 individuals (1.8% <strong>of</strong> disabled patients, or 3.5% <strong>of</strong><br />

mentally retarded patients). The specified reasons <strong>of</strong> death are<br />

usually non-informative in medical histories (“Down<br />

syndrome”, “chromosomal disease”). In other 100 certificates<br />

the following reasons <strong>of</strong> death were enumerated:<br />

bronchopneumony in 60 patients, acute respirative infection –<br />

in 4, staphylococcous infection -1, tuberculosis -2, congenital<br />

vitium cordis -20, congenital defects <strong>of</strong> other organs -4,<br />

leukemy -4, accidents -5. In our country we have registered<br />

641 alive Down syndrome patients: 374 children under<br />

16 years <strong>of</strong> age and 267 adults (two oldest patients were 48<br />

years <strong>of</strong> age). We have made karyotype analysis in<br />

lymphocyte culture for 393 <strong>of</strong> them (61.3%). The<br />

chromosomal investigation has confirmed the diagnosis <strong>of</strong><br />

Down syndrome in 372 (94.6%) patients. Normal karyotype<br />

was found in 19 (4.8%) patients. In two patients nearby<br />

trisomy-21 an additionally reciprocal translocations <strong>of</strong> other<br />

chromosomes were present. In two patients chromosome<br />

anomalies other than trisomy-21 were found. The karyotype in<br />

one 3-year-old boy was 49,XXXXY. The boy was deeply<br />

mentally retarded and had mongolian eyeslit, epicanthus,<br />

hypertelorism, palmar simian crease, clinodaktily, arches in<br />

papilar patterns on all ten fingers, and other anomalies typical<br />

<strong>of</strong> Down syndrome patients. The other chromosome patient<br />

without trisomy-21 was 29-year-old mild mentally retarded<br />

woman in whose karyotype ring chromosome 9 was found.<br />

For both latter patients the physicians suspected Down<br />

syndrome. Therefore, in Lithuanian population false-positive<br />

diagnosis <strong>of</strong> Down syndrome was found in 5.3% <strong>of</strong> patients,<br />

i.e. two-three times rarely as compared with analogical<br />

investigations in other countries. We suppose that one <strong>of</strong> the<br />

reasons <strong>of</strong> such situation is the racial homogeneity <strong>of</strong><br />

Lithuanian population presented almost exclusively by<br />

Caucasians. But in newborn Lithuanian babies the frequency<br />

<strong>of</strong> false-positive diagnoses arises up to 32.1%: among 134<br />

patients with clinical diagnosis <strong>of</strong> Down syndrome the normal<br />

karyotype was found in 43. The main clinical feature – mental<br />

retardation – cannot be recognized in infants, and the<br />

neonatologists send the patients for karyotype analysis due to<br />

isolated microanomalies.


110 ASSOCIATION BETWEEN COMT, PTSD, AND<br />

INCREASED SMOKING FOLLOWING HURRICANE<br />

EXPOSURE IN AN EPIDEMIOLOGIC SAMPLE<br />

A. Amstadter* (1), N. Nugent (2), K. Koenen (3), K.<br />

Ruggiero (1), R. Acierno (1), D. Kilpatrick (1), S. Galea (4), J.<br />

Gelernter (5)<br />

1. <strong>Medical</strong> University <strong>of</strong> South Carolina 2. Brown <strong>Medical</strong><br />

School 3. Harvard University 4. University <strong>of</strong> Michigan 5.<br />

Yale University<br />

*amstadt@musc.edu<br />

Tobacco smoking has been found to increase after the<br />

experience <strong>of</strong> a traumatic event and has been associated with<br />

posttraumatic stress disorder (PTSD). Initiation and<br />

persistence <strong>of</strong> cigarette smoking is moderately heritable; two<br />

recent investigations have implicated the COMT Val158Met<br />

(also known as rs4680) polymorphism in smoking age <strong>of</strong><br />

initiation, dependence, as well as in quantity and frequency <strong>of</strong><br />

smoking. To examine a possible association <strong>of</strong> COMT<br />

Val158Met and post trauma increases in cigarette smoking, we<br />

studied 614 adults from the 2004 Florida Hurricane Study who<br />

returned saliva DNA samples via mail. PTSD was strongly<br />

associated with increased smoking. Moreover, each COMT<br />

Val158Met ‘Met’ allele predicted a 2.10 fold risk <strong>of</strong> smoking<br />

post-hurricane independent <strong>of</strong> PTSD; follow-up analyses<br />

revealed that this finding was primarily driven by<br />

European-American males. This study represents the first<br />

genetic association study (to our knowledge) <strong>of</strong> smoking<br />

behavior following an acute stressor.<br />

111 IDENTIFICATION OF LEPTIN, CREB1 AND TPH1<br />

AS CANDIDATE GENES FOR DEPRESSION, OBESITY<br />

AND RELATED COMORBIDITIES IN CASE<br />

CONTROL STUDY AMONG NORTH INDIANS<br />

M. Kapoor* (1), S. Kapur (1), S. Sidhu (2)<br />

1. Birla Institute <strong>of</strong> Technology and Sciences 2. Guru Nanak<br />

Dev University<br />

*kapoor.manav@gmail.com<br />

Aim: Epidemiological data suggests that there is an<br />

association between obesity and depression, as both disorders<br />

are associated with less control over appetite and increased<br />

risk for metabolic disorders. Present study attempts to find the<br />

common genetic link between these two diseases by looking<br />

into polymorphisms inthe Tryptophan Hydroxylase (TPH),<br />

Leptin (LEP) and CREB1 gene. Methods: This<br />

Exploratory case-control study comprised <strong>of</strong> 150 clinically<br />

diagnosed depressed individuals and 150 age matched<br />

controls. DNA from all 300 subjects was genotyped for repeat<br />

polymorphisms in leptin gene and CREB1 gene using<br />

PCR-SSLP method, while PCR-RFLP based method was used<br />

for the intronic polymorphism (A218C) in TPH gene. Results:<br />

A significant difference (p


112 A GENETIC ASSOCIATION STUDY OF THE<br />

FUNCTIONAL A118G POLYMORPHISM OF THE<br />

HUMAN OPIOID RECEPTOR GENE AND<br />

IMMUNO-MODULATION IN OPIATE DEPENDENT<br />

SUBJECTS<br />

S. Kapur* (1), S. Sharad (1)<br />

1. Birla Institute <strong>of</strong> Technology and Science, Pilani,<br />

Rajasthan, India<br />

*mssuman@gmail.com<br />

Purpose: Immunosuppressive effect <strong>of</strong> opiates is poorly<br />

described and poses a problem in assessing the true spectrum<br />

<strong>of</strong> these drugs. In this prospective, observational study we<br />

have explored the relationship <strong>of</strong> variation at the A118G locus<br />

in the Exon 1 <strong>of</strong> OPRM1 gene, with the amount <strong>of</strong> opiates<br />

consumed and correlate the immunosuppressive effects <strong>of</strong><br />

exogenous opiates with the mu-opiate receptor genotype <strong>of</strong> the<br />

opiate-dependent and control subjects from northern India.<br />

Methods: Frequency <strong>of</strong> the wild-type A118 and variant 118G<br />

alleles in dependent subjects (n=73) and age, sex and ethnicity<br />

matched opiate-naïve volunteers (n=73) were examined.<br />

The relationship among the A118G genotype, opiate<br />

consumption and circulating levels <strong>of</strong> immunoglobilins, IgG<br />

and IgA, were analyzed using linear regression and parametric<br />

tests. Results: Frequency <strong>of</strong> 118G allele was significantly<br />

lower in control subjects when compared to that observed in<br />

dependent subjects (X2=26.28, p


114 META-ANALYSIS OF GWAS FOR HANDEDNESS:<br />

RESULTS FROM THE ENGAGE CONSORTIUM<br />

S. Medland* (1), C. Lindgren (2), R. Magi (2), B. Neale (3),<br />

E. Albrecht (4), T. Esko (5), D. Evans (6), J. Hottenga (7), M.<br />

Ikram (8), M. Mangino (9), S. Ripatti (10), F. van Rooij (8),<br />

D. Boomsma (7), G. Davey Smith (6), I. Rückert (4), T.<br />

Spector (9), C. van Duijn (8), N. Martin (1), L. Peltonen<br />

(10), M. McCarthy (2). The ENGAGE Handedness<br />

Consortium<br />

1. Genetic Epidemiology, Queensland Institute <strong>of</strong> <strong>Medical</strong><br />

Research, Brisbane, Queensland, Australia 2. Wellcome Trust<br />

Centre for Human Genetics, University <strong>of</strong> Oxford, Oxford,<br />

UK 3. Center for Human Genetic Research, Massachusetts<br />

General Hospital, Boston, MA USA; 4. Institute <strong>of</strong><br />

Epidemiology, Helmholtz Zentrum München, German<br />

Research Center for Environmental Health, Neuherberg,<br />

Germany 5. Institute <strong>of</strong> Molecular and Cell Biology,<br />

University <strong>of</strong> Tartu, Tartu, Estonia 6. MRC Centre for Causal<br />

Analyses in Translational Epidemiology, Department <strong>of</strong> Social<br />

Medicine, University <strong>of</strong> Bristol, Bristol, UK 7. Department <strong>of</strong><br />

Biological Psychology, VU University Amsterdam,<br />

Amsterdam, The Netherlands 8. Dept <strong>of</strong> Epidemiology,<br />

Erasmus MC, Rotterdam, The Netherlands 9. Dept <strong>of</strong> Twin<br />

Research & Genetic Epidemiology, King's College, London,<br />

UK 10. National Institute <strong>of</strong> Health and Welfare, Public<br />

Health Genomics Unit and FIMM, Institute for Molecular<br />

Medicine Finland, Helsinki, Finland.<br />

*sarahMe@qimr.edu.au<br />

Background: Handedness, a consistent asymmetry in skill or<br />

preferential use between the hands, is a moderately heritable<br />

trait which is related to the lateralization <strong>of</strong> language functions<br />

within the brain. The prevalence <strong>of</strong> left-handedness (~10% in<br />

the general population) is increased in a number <strong>of</strong><br />

psychiatric, neurologic, and learning disorders suggesting that<br />

cerebral asymmetry may play a role in these disorders.<br />

Previous linkage and association studies have had only limited<br />

success. However these studies have used unselected samples<br />

resulting in low numbers <strong>of</strong> cases and low power. Here we<br />

present the results <strong>of</strong> a meta-analysis <strong>of</strong> 12 genome wide<br />

association studies, comprising 23,443 samples (2350 cases,<br />

21093 controls). Methods: Writing hand was used to classify<br />

participants as left- or right-handers, and sex and year <strong>of</strong> birth<br />

were used as covariates. All samples were derived from<br />

European descent populations. We tested for trait associations<br />

under an additive model for a total <strong>of</strong> ~2.5 million common<br />

HapMap SNPs (directly genotyped and imputed). Weighted<br />

Z-score based meta-analysis was conducted using METAL<br />

with weights defined by effective sample size. Results: We<br />

observed a number <strong>of</strong> signals approaching genome-wide<br />

significance (5x10-8) all <strong>of</strong> which were novel. Promising<br />

results were found on chromosome 5 (p = 2.455e-07) and 13<br />

(p = 6.149e-07), in regions that encompasses SLIT3 (which<br />

plays a role in the axon-guidance-pathways which guide motor<br />

neuronal development during fetal development), MAB21L1<br />

(involved in cerebellum development) and NBEA (which<br />

encodes a neuron-specific multidomain protein implicated in<br />

membrane trafficking that is predominantly expressed in the<br />

brain and during development). Conclusions: No evidence <strong>of</strong><br />

association was found for any the traditional asymmetry<br />

candidate genes (NODAL, LEFTY). Large-scale replication<br />

efforts are currently underway. These results vastly extend the<br />

number <strong>of</strong> loci implicated in the development <strong>of</strong> handedness<br />

and lateralization in general.


115 THE MGS2 CONTROL SAMPLE: COLLECTION,<br />

CHARACTERIZATION, AND VALIDITY OF A LARGE<br />

SAMPLE OF EUROPEAN-ANCESTRY AND AFRICAN<br />

AMERICAN INDIVIDUALS FOR GENETIC STUDIES<br />

A. Sanders* (1), D. Levinson (2), J. Duan (1), J. Dennis (3),<br />

R. Li (3), K. Kendler (4), J. Rice (5), J. Shi (2), B. Mowry (6),<br />

F. Amin (7), J. Silverman (8), N. Buccola (9), W. Byerley<br />

(10), D. Black (11), R. Freedman (12), C. Cloninger (5), P.<br />

Gejman (1)<br />

1. Center for Psychiatric Genetics, Department <strong>of</strong> Psychiatry<br />

and Behavioral Sciences, NorthShore University Health<br />

System, Evanston, IL 2. Department <strong>of</strong> Psychiatry and<br />

Behavioral Sciences, Stanford University, Stanford, CA<br />

3. Government & Academic Research, Knowledge Networks,<br />

Menlo Park, CA 4. Departments <strong>of</strong> Psychiatry, and Human<br />

Genetics, Virginia Commonwealth University, Richmond, VA<br />

5. Department <strong>of</strong> Psychiatry, Washington University, St.<br />

Louis, MO 6. Queensland Centre for Mental Health Research,<br />

and Queensland Institute for <strong>Medical</strong> Research, Brisbane,<br />

Queensland, Australia 7. Department <strong>of</strong> Psychiatry and<br />

Behavioral Sciences, Atlanta Veterans Affairs <strong>Medical</strong> Center,<br />

and Emory University, Atlanta, GA<br />

8. Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong> Medicine,<br />

New York, NY 9. School <strong>of</strong> Nursing, Louisiana State<br />

University Health Sciences Center, New Orleans, LA<br />

10. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California at San<br />

Francisco, San Francisco, CA 11. Mental Health Clinical<br />

Research Center, and Department <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Iowa Carver College <strong>of</strong> Medicine, Iowa City, IA 12.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Colorado Denver,<br />

Aurora, CO<br />

*alan.sanders.md@gmail.com<br />

Background: The Molecular Genetics <strong>of</strong> Schizophrenia<br />

(MGS2) internet-ascertained an adult control sample <strong>of</strong><br />

non-Hispanic European-ancestry (EA, 3,364) and African<br />

American (AA, 1,301) subjects. Methods: By an internet<br />

administered questionnaire (allowing control sample users to<br />

exclude subjects diagnostically similar to their case group), we<br />

collected phenotypic data and then blood samples, both<br />

repository-deposited for general research use. All subjects<br />

were anonymized, and 3,626 comprised the control portion <strong>of</strong><br />

the MGS2 genome-wide association study (GWAS) report.<br />

Results: Comparing to the national census, we find the MGS2<br />

control sample is generally representative. The sample is<br />

more female, and somewhat older, more educated and<br />

affluent, but all strata are represented. Self-reported ancestry<br />

matches well to genotypic and census data. Lifetime<br />

diagnostic prevalence estimates for substance, mood, and<br />

anxiety were higher than previous population-based surveys <strong>of</strong><br />

psychiatric disorders, probably due to factors such as omission<br />

<strong>of</strong> data collection for “organic” and other important exclusion<br />

criteria and the anonymity <strong>of</strong> the survey method. However,<br />

patterns such as sex ratios, comorbidity, relationships to<br />

neuroticism and extraversion, and demographic associations<br />

matched previous reports well. GWAS data cleaning resulted<br />

in equivalent proportions <strong>of</strong> exclusions for the internet<br />

collected/evaluated control sample compared to the<br />

face-to-face case sample. Conclusions: The results validate<br />

the use <strong>of</strong> the MGS2 control sample as being generally<br />

representative <strong>of</strong> the EA and AA U.S. population – for many<br />

aspects <strong>of</strong> demography, ancestry, and morbidity. The<br />

internet-based methods facilitated data-sharing and sample<br />

utility by enabling rapid collection <strong>of</strong> a large and<br />

representative sample and anonymization there<strong>of</strong>.<br />

116 GENETIC INFLUENCES ON THE NON-MEDICAL<br />

USE OF PRESCRIPTION DRUGS<br />

S. Medland* (1), M. Neale (2)<br />

1. Queensland Institute <strong>of</strong> <strong>Medical</strong> Research, Brisbane<br />

Australia<br />

2. Virginia Institute <strong>of</strong> Psychiatric and Behavioral Genetics,<br />

Richmond USA<br />

*sarahMe@qimr.edu.au<br />

Non-medical use <strong>of</strong> prescription drugs (PD) is the second most<br />

common form <strong>of</strong> drug use in teens and young adults, however<br />

relatively little is known about the aetiology <strong>of</strong> this type <strong>of</strong><br />

drug use and the influence <strong>of</strong> genetic risk factors. We used<br />

family level data collected from twins, sibling and half-sibling<br />

pairs within the Add Health study to examine the magnitude<br />

and relative importance <strong>of</strong> genetic and environmental<br />

influences on the initiation <strong>of</strong> PD misuse. The results from this<br />

study indicated that magnitude <strong>of</strong> genetic influences differed<br />

by drug type. Tranquilizer, Sedative and Stimulant initiation<br />

were moderately heritable ( 40-50%) suggesting that initiation<br />

<strong>of</strong> these types <strong>of</strong> prescription drugs are influenced by genetic<br />

risk factors to a similar extent as smoking initiation<br />

(heritability .58 within this sample). However, initiation <strong>of</strong><br />

analgesic misuse was found to be predominantly due to shared<br />

and unique environmental influences. Following, from the<br />

initial study, a second series <strong>of</strong> analyses were conducted using<br />

community level data from the Add Health study. These<br />

multi-level analyses made use <strong>of</strong> the rich data frame work <strong>of</strong><br />

the Add Health study combining data from friendship<br />

networks with the family level data to allow a more detailed<br />

exploration <strong>of</strong> the environmental influences affecting drug<br />

initiation. These analyses supported the role <strong>of</strong> genetic<br />

influences on the initiation <strong>of</strong> Tranquilizers, Sedatives and<br />

Stimulants and showed that friend and community level<br />

environmental influences were more important than within<br />

family influences on the initiation <strong>of</strong> analgesic misuse. As<br />

analgesic misuse is most common among females, these<br />

results have important implication for the targeting and design<br />

<strong>of</strong> public health campaigns and interventions.


117 GENOMEWIDE ASSOCIATION STUDY OF<br />

EXTERNALIZING DISORDERS<br />

F. Aliev* (1), M. Ozturk (1), D. Dick (1)<br />

1. Virginia Commonwealth University, Ankara University,<br />

Turkey<br />

*falievfaliev@vcu.edu<br />

Alcohol dependence is commonly comorbid with other<br />

externalizing disorders, including other forms <strong>of</strong> drug<br />

dependence, antisocial personality disorder, and childhood<br />

conduct disorder. Twin studies indicate that this overlap is due<br />

largely to shared genetic factors that predispose broadly to a<br />

variety <strong>of</strong> form <strong>of</strong> externalizing psychopathology (e.g.,<br />

Kendler et al., 2003; Krueger et al., 2000). This has<br />

implications for gene identification efforts, and suggests that<br />

some genes will predispose broadly to a variety <strong>of</strong> forms <strong>of</strong><br />

externalizing problems, rather than being disorder specific.<br />

We have previously shown that analyzing factor scores,<br />

comprised <strong>of</strong> symptoms <strong>of</strong> different externalizing disorders<br />

(alcohol dependence, drug dependence, conduct disorder,<br />

antisocial personality disorder), is useful in gene identification<br />

efforts (Dick et al., 2008). Here, we report analyses from the<br />

Collaborative Study on the Genetics <strong>of</strong> Alcoholism (COGA),<br />

in which a genome-wide association panel was genotyped by<br />

the Center for Inherited Disease Research using the Illumina 1<br />

million SNPchip platforms. Genome wide data was generated<br />

on 2000 individuals, 1905 <strong>of</strong> which remained after all data<br />

cleaning. All 1205 cases met criteria for DSM-IV Alcohol<br />

Dependence, as assessed by the SSAGA, and all 700 controls<br />

were screened against Alcohol Dependence and related<br />

substance use disorders. We created factor scores indexing<br />

overall externalizing psychopathology, based on<br />

log-transformed symptom counts for alcohol dependence, drug<br />

dependence, conduct disorder, and antisocial personality<br />

disorder. We generated results for overall externalizing factor<br />

scores, as well as for the symptom counts <strong>of</strong> the individual<br />

disorders used to create the factor scores. We find evidence for<br />

several genes and genomic regions that appear to confer risk<br />

to a variety <strong>of</strong> forms <strong>of</strong> externalizing psychopathology.<br />

118 USING LINKAGE INFORMATION TO WEIGHT A<br />

GENOME-WIDE ASSOCIATION OF BIPOLAR<br />

DISORDER<br />

D. Howrigan* (1), J. Smoller (2), P. Sklar (2), S. Purcell (2),<br />

V. Nimgaonkar (3), S. Faraone (4), B. Devlin (3), N. Laird (5),<br />

M. McQueen (1)<br />

1. University <strong>of</strong> Colorado, Boulder 2. Broad Institute <strong>of</strong><br />

Harvard and MIT 3.University <strong>of</strong> Pittsburgh School <strong>of</strong><br />

Medicine 4. SUNY Upstate <strong>Medical</strong> University, Syracuse,<br />

New York 5. Harvard School <strong>of</strong> Public Health<br />

*daniel.howrigan@gmail.com<br />

Multiple testing issues in genome-wide association studies<br />

(GWAS) have prompted statistical methods utilizing prior<br />

information to increase the power <strong>of</strong> association results.<br />

Using prior information derived from genome-wide linkage<br />

studies on Bipolar Disorder, we employed a weighted false<br />

discovery rate (wFDR; Roeder et al., 2006) approach to a<br />

GWAS drawn from the Systematic Treatment Enhancement<br />

Program for Bipolar Disorder (STEP-BD). Using this method,<br />

association signals are up or down-weighted as a function <strong>of</strong><br />

the linkage score in that genomic region. Although no SNPs<br />

reached genome-wide significance through the wFDR<br />

approach, the strongest single SNP result from the original<br />

GWAS results, rs4939921 in MYO5B (myosin VB), is<br />

up-weighted as it resides in a region on chromosome 18 that<br />

has a modest linkage signal. We also identify regions on<br />

chromosomes 9, 17, and 18 where clusters <strong>of</strong> association<br />

signals coincide with linkage evidence, implicating them as<br />

possible candidate regions for further analysis.


EARLY CAREER INVESTIGATOR TRACK:<br />

GENOMEWIDE AND META-ANALYTIC<br />

APPROACHES<br />

ECI 13 GENOME-WIDE ASSOCIATION ANALYSIS OF<br />

THE BECK DEPRESSION INVENTORY LOCALIZES<br />

A QTL INFLUENCING RISK OF MAJOR<br />

DEPRESSIVE DISORDER NEAR THE RND3 GENE<br />

M. Carless* (1), D. Glahn (2), R. Olvera (3), J. Curran (4), H.<br />

Göring (4), T. Dyer (5), E. Moses (5), L. Almasy (5), R.<br />

Duggirala (5), J. Blangero (5)<br />

1. Southwest Foundation for Biomedical Research 2. Yale<br />

University School <strong>of</strong> Medicine 3. University <strong>of</strong> Texas Health<br />

Science Center, San Antonio 4. UT Health Science Center,<br />

San Antonio 5. Southwest Foundation for Biomedical<br />

Research<br />

* mcarless@sfbrgenetics.org<br />

Major depressive disorder (MDD) is a common heritable<br />

mental disease for which few susceptibility loci have been<br />

identified. In this study, we describe the first genome-wide<br />

association study to search for genes influencing quantitative<br />

variation in the Beck Depression Inventory (BDI), an<br />

endophenotype measuring depressive severity. Extensive data<br />

has been obtained on 597 individuals from 40 Mexican<br />

American families. Based on a consensus diagnosis following<br />

standardized psychiatric examinations, lifetime prevalence <strong>of</strong><br />

MDD was 35% in this population. Heritability <strong>of</strong> total BDI<br />

(controlling for age and sex effects) was estimated to be 0.27<br />

(p=3.4×10-4), suggesting that approximately one third <strong>of</strong> the<br />

phenotypic variation in BDI scores has a genetic basis. In<br />

order to localize genomic regions harboring quantitative trait<br />

loci (QTLs) influencing BDI score, we genotyped individuals<br />

using Illumina HumanHap 550K BeadChips. Using a variance<br />

component model incorporating a “measured genotype” fixed<br />

effect, we performed genome-wide association analysis on<br />

BDI, localizing a QTL to chromosome 2q23. One SNP<br />

(rs289943), located between RND3 and FABP5L10 genes,<br />

was strongly associated with BDI (p = 2.1×10-8). This<br />

relatively rare SNP (MAF = 0.05) is also strongly associated<br />

with lifetime MDD affection status (nominal p-value =<br />

8.7×10-4). The RND3 gene may represent an interesting<br />

candidate for risk <strong>of</strong> depression, coding for a small GTPase<br />

known to be involved neuronal migration and postnatal<br />

development/maturation <strong>of</strong> the central nervous system and<br />

thought to be involved in the inflammatory response <strong>of</strong> brain<br />

astrocytes. This result now requires deep sequencing to<br />

identify causal functional variants within this genomic region.<br />

ECI 14 SLEEPLESSNESS SUBPHENOTYPE OF<br />

BIPOLAR I DISORDER HERALDS MORE SEVERE<br />

COURSE OF ILLNESS AND GWAS DATA SUGGESTS<br />

THREE ASSOCIATED GENE VARIANTS<br />

V. Coleman* (1), D. Koller (2), T. Foroud (2), X. Xuei (3), H.<br />

Edenberg (3), X. Cai (4), E. Brizendine (4), M. Erpe (5),<br />

Bipolar Genome Study (BiGS), J. Nurnberger, Jr. (5)<br />

1. Indiana University School <strong>of</strong> Medicine 2. IUSM,<br />

Department <strong>of</strong> <strong>Medical</strong> and Molecular Genetics 3. IUSM,<br />

Department <strong>of</strong> Biochemical and Molecular Biology 4. IUSM,<br />

Department <strong>of</strong> Medicine 5. IUSM, Department <strong>of</strong> Psychiatry<br />

* vecolema@iupui.edu<br />

One strategy for elucidating the genetics and pathophysiology<br />

<strong>of</strong> bipolar disorder (BPI) involves identification <strong>of</strong><br />

subphenotypes; we studied sleeplessness at the onset <strong>of</strong> mania<br />

as a possible index <strong>of</strong> circadian disruption. Using diagnostic<br />

data from NIMH Genetics Initiative Bipolar sample (BiGS<br />

Consortium), we performed a case-control study <strong>of</strong> 2213 BPI<br />

subjects retrospectively reporting sleeplessness preceded their<br />

first manic episode (SLN+; cases) and those who denied such<br />

(SLN-; controls). Genetic analysis using genotypic data <strong>of</strong> a<br />

SLN+ subset (n=221) versus normal controls (n=1034) was<br />

performed (see Smith, et al, 2009 for detailed methods).<br />

Chi-square tests were performed for categorical data, and<br />

t-tests or Mann-Whitney tests for quantitative variables.<br />

Compared to the SLN- controls, the SLN+ group had more<br />

manic episodes, hospitalizations for mania, suicide attempts,<br />

rapid mood switching and psychosis with mania. Association<br />

was found with two SNPs near KIAA0143, which codes for a<br />

plasma membrane binding protein. The SNPs have a<br />

frequency <strong>of</strong> ~4% in cases and ~1% in controls. The p-values<br />

for association were 8x10-9 and 2x10-7. Our second area <strong>of</strong><br />

interest comprised 3 SNPs near NAP5 (all p=2-4x10-6); this is<br />

designated as a peripheral clock protein and there is a possible<br />

association with multiple sclerosis. A third area <strong>of</strong> interest<br />

comprised 3 SNPs in MEP1B, the most significant showing a<br />

p-value <strong>of</strong> 5x10-7. This codes for a zinc metalloprotease.<br />

These results are now being tested in a separate replication<br />

sample. Conclusion: Sleeplessness prior to the first manic<br />

episode may identify a distinct and functional subphenotype<br />

for future investigations.


ECI 15 IDENTIFICATION OF TRAPPC9 AS A CAUSE<br />

OF NON-SYNDROMIC AUTOSOMAL RECESSIVE<br />

MENTAL RETARDATION<br />

L. Kaufman* (1), A. Mir (2), A. Noor (1), M. Ayub (3), J.<br />

Vincent (1)<br />

1. Centre for Addiction and Mental Health (CAMH),<br />

University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

2. Dept. <strong>of</strong> Bioscience, COMSATS Institute <strong>of</strong> Information<br />

Technology, Islamabad, Pakistan<br />

3. St. Luke's Hospital, Middlesborough, United Kingdom<br />

* liana_kaufman@camh.net<br />

In this study, we identify TRAPPC9 as the sixth causal gene<br />

for non-syndromic autosomal recessive mental retardation<br />

(NS-ARMR). Mental retardation is an intellectual disability<br />

characterized by an IQ below 70 and deficits in adaptive<br />

behaviours. Currently, there are only 5 genes known to cause<br />

NS-ARMR. We identified a mutation in TRAPPC9 in a large<br />

consanguineous family from Pakistan with NS-ARMR. Using<br />

5.0 Affymetrix genome wide SNP chips we analyzed the DNA<br />

<strong>of</strong> 20 family members. We used dChip s<strong>of</strong>tware to complete<br />

homozygosity mapping, and identified the disease locus at<br />

8q24. Consanguinity makes is possible to identify long<br />

stretches <strong>of</strong> autozygosity where the disease gene lies. We then<br />

used sequencing to elucidate the disease-causing gene. We<br />

identified a C>T mutation at the end <strong>of</strong> exon 7 <strong>of</strong> this 23 exon<br />

gene. The resulting protein contains the nonsense mutation<br />

R377X. Using qPCR we were able to show that the mRNA<br />

levels <strong>of</strong> TRAPPC9 are markedly decreased in affected family<br />

members. The protein product <strong>of</strong> TRAPPC9, NIBP, is<br />

involved in the activation <strong>of</strong> the NF-β signaling pathway. It is<br />

expressed ubiquitously, but at higher levels in the muscle,<br />

heart, brain, kidney, and placenta. It may be involved in<br />

neuronal differentiation and cell survival, as well as in golgi<br />

trafficking. Additional clinical features in affected family<br />

members include cerebellar white matter hypoplasia and<br />

tendency towards small head size. Understanding the genetic<br />

causes for mental retardation is a vital component <strong>of</strong><br />

elucidating pathological mechanisms, and other potentially<br />

pathological genes.<br />

ECI 16 COMPREHENSIVE META-ANALYSES BASED<br />

ON ASSOCIATION STUDIES OF ALCOHOL,<br />

COCAINE, HEROIN AND METHAMPHETAMINE<br />

DEPENDENCE<br />

D. Li* (1), H. Zhao (2)<br />

1. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, Yale<br />

University, New Haven, USA 2. Department <strong>of</strong> Epidemiology<br />

and Public Health and Department <strong>of</strong> Genetics, School <strong>of</strong><br />

Medicine, Yale University, New Haven, USA<br />

* dawei.li@yale.edu<br />

Background: Genetic studies have identified associations <strong>of</strong><br />

many susceptibility genes with alcohol, heroin, cocaine,<br />

ormethamphetamine dependence. However, a proportion <strong>of</strong><br />

alternative studies have produced contradictory results in<br />

terms <strong>of</strong> positive and negative findings, possibly reflecting<br />

inadequate statistical power and use <strong>of</strong> different ethnic<br />

populations. Methods: Using the cumulative case-control and<br />

family-based samples <strong>of</strong> European, Asian, African, and<br />

Hispanic origins published in recent two decades, the<br />

meta-analyses <strong>of</strong> 25 commonly-reported candidate genes seek<br />

to examine whether the aggregate data provide evidence <strong>of</strong><br />

statistical significance and to clarify the inconsistent findings<br />

using systematic methods and models (eg. allelic vs.<br />

genotypic; dominant vs. recessive; overall vs. sub-grouped;<br />

sensitivity and retrospective analyses; and random effects<br />

model). Results: The serotonin transporter gene (SLC6A4)<br />

was strongly associated with alcohol, heroin, cocaine, and<br />

methamphetamine dependence (P= 0.0002; OR = 0.79 (0.7 -<br />

0.9); Very strong associations were replicated for the aldehyde<br />

dehydrogenase 2 gene (ALDH2) (P = 5 × 10-37; OR = 0.23<br />

(0.18 - 0.29)) and alcohol dehydrogenase genes (P = 2 ×<br />

10-21; OR = 1.9 (1.66 - 2.17) for ADH1B and P = 4 × 10-33;<br />

OR = 2.14 (1.89 - 2.43) for ADH1C) with alcoholism and with<br />

alcohol-induced medical diseases; Strong associations were<br />

detected for the dopamine receptor genes (P = 1 × 10-8for<br />

DRD2 and P = 0.001 for DRD4) and gamma-aminobutyric<br />

acid subunit genes (P = 0.0005 for GABRG2; P = 0.00007 for<br />

GABRA2; and P = 0.00006 for GABRA6) with alcohol and<br />

drug abuse; Significant associations were also found with<br />

5-hydroxytryptamine receptor genes (HTR1B and HTR2A)<br />

and cannabinoid receptor 1 gene (CNR1); Significant<br />

association was also found between the cytochrome P450 2E1<br />

gene (CYP2E1) and alcoholic liver disease (P = 0.007); Strong<br />

association were revealed with the monoamine oxidase A gene<br />

(MAOA, P = 9 × 10-5) and tryptophan hydroxylase gene<br />

(TPH, P = 0.0002) in Caucasians; Weak association was found<br />

with the opioid receptor delta 1 gene (OPRD1); And weak<br />

associations were also revealed with the<br />

catechol-O-methyltransferase gene (COMT) and dopamine<br />

transporter gene (DAT1) in Asians. However, no evidence <strong>of</strong><br />

association was found with the DRD1, DRD3, neuropeptide Y<br />

gene (NPY), and OPRM1 genes. No strong evidence <strong>of</strong><br />

publication bias was found.Conclusion: The findings in this<br />

study, which may be the most comprehensive meta-analysis<br />

study for substance abuse up to now, may provide the<br />

strongest evidence <strong>of</strong> associations for these candidate genes.


ECI 17 A GENOME WIDE ASSOCIATION STUDY OF<br />

COMORBID ALCOHOLISM AND BIPOLAR<br />

DISORDER<br />

G. Lydall* (1), N. Bass (1), A. McQuillin (1), M. Robinson<br />

(1), J. Lawrence (1), A. Anjorin (1), R. Kandaswamy (1), A.<br />

Pereira (1), I. Guerrini (1), D. Curtis (2), A. Vine (2), P. Sklar<br />

(3), E. Scolnick (4), S. Purcell (5), H. Gurling (1)<br />

1. Molecular Psychiatry Laboratory, Department <strong>of</strong> Mental<br />

Health Sciences, Windeyer Institute <strong>of</strong> <strong>Medical</strong> Sciences,<br />

University College London, London, UK 2. Centre for<br />

Psychiatry, Barts and the London School <strong>of</strong> Medicine and<br />

Dentistry, Queen Mary, University <strong>of</strong> London, UK 3. 6Broad<br />

Institute <strong>of</strong> Harvard and MIT, Cambridge, MA; 5Departments<br />

<strong>of</strong> Genetics, Psychiatry or Medicine, Harvard <strong>Medical</strong> School,<br />

Boston, MA, USA 4. Broad Institute <strong>of</strong> Harvard and MIT,<br />

Cambridge, MA, USA 5. Departments <strong>of</strong> Genetics, Psychiatry<br />

or Medicine, Harvard <strong>Medical</strong> School, Boston, MA; Broad<br />

Institute <strong>of</strong> Harvard and MIT, Cambridge, MA, USA<br />

* g.lydall@ucl.ac.uk<br />

We have investigated the comorbidity between bipolar<br />

affective disorder and alcoholism with a genome-wide allelic<br />

association study <strong>of</strong> 506 patients from the University College<br />

London (UCL) bipolar disorder case control sample. In this<br />

group, there were 143 bipolar research subjects who also<br />

had a research diagnostic criteria (RDC) diagnosis <strong>of</strong><br />

alcoholism. These were compared to 510 ancestrally-matched<br />

supernormal controls. No marker reached conventional criteria<br />

for genome-wide significance. Genes previously shown to be<br />

associated with alcoholism and addiction phenotypes were<br />

tested for association in the bipolar alcoholic sample using<br />

gene wise permutation tests <strong>of</strong> all SNPs genotyped within a<br />

50kb region flanking each gene. Several CNS genes showed<br />

significant (p


ECI 19 GENOME-WIDE ASSOCIATION STUDY<br />

IDENTIFIES A RISK LOCUS FOR PEDIATRIC-ONSET<br />

BIPOLAR DISORDER ON CHROMOSOME 5Q<br />

D. Chen* (1), N. Akula (1), C. Steele (1), Bipolar Genome<br />

Study (BiGS), F. McMahon (1)<br />

1. Unit on the Genetic Basis <strong>of</strong> Mood & Anxiety Disorder,<br />

National Institute <strong>of</strong> Mental health, National Institutes <strong>of</strong><br />

Health, U.S. Dept <strong>of</strong> Health and Human Services<br />

* chend4@mail.nih.gov<br />

Pediatric-onset bipolar disorder (PBPD) is a strongly familial,<br />

<strong>of</strong>ten chronic and disabling condition. The American<br />

Academy <strong>of</strong> Child & Adolescent Psychiatry (AACAP) defines<br />

PBPD as onset <strong>of</strong> mania


GENOMICS<br />

119 GENOME-WIDE SCREENING FOR COMMON<br />

AND RARE COPY NUMBER VARIANTS IN A<br />

GERMAN SAMPLE OF BIPOLAR DISORDER<br />

L. Priebe (1), F. Degenhardt (1), S. Herms (1), M. Mattheisen<br />

(1), V. Nieratschker (2), S. Witt (2), R. Breuer (2), M. Alblas<br />

(1), S. Moebus (3), S. Schreiber (4), M. Rietschel (5), M.<br />

Nöthen (6), S. Cichon (6), T. Mühleisen (1)<br />

1. Institute <strong>of</strong> Human Genetics, Department <strong>of</strong> Genomics, Life<br />

& Brain Center, University <strong>of</strong> Bonn, Bonn, Germany 2.<br />

Department <strong>of</strong> Genetic Epidemiology, Central Institute <strong>of</strong><br />

Mental Health, Mannheim, Germany 3. Institute for <strong>Medical</strong><br />

Informatics, Biometry and Epidemiology, University Hospital<br />

<strong>of</strong> Essen, University Duisburg-Essen, Essen, Germany 4.<br />

Institute <strong>of</strong> Clinical Molecular Biology, University Hospital<br />

Schleswig-Holstein, Kiel, Germany 5. Department <strong>of</strong> Genetic<br />

Epidemiology, Central Institute <strong>of</strong> Mental Health, Mannheim,<br />

Germany 6. Institute <strong>of</strong> Human Genetics, University <strong>of</strong> Bonn,<br />

Bonn, Germany<br />

Only few studies have investigated the influence <strong>of</strong> copy<br />

number variants (CNVs) on susceptibility to bipolar<br />

disorder(BP). We conducted a systematic screening for<br />

common and rare CNVs in a sample <strong>of</strong> 882 patients (~25%<br />

early-onset <strong>of</strong> BP and/or positive family history) with a<br />

DSM-IV diagnosis <strong>of</strong> BP I and 872 controls, all <strong>of</strong> German<br />

descent. They were either genotyped on Illumina's<br />

HumanHap550 or human610-quad arrays, sharing a consensus<br />

set <strong>of</strong> ~550,000 SNPs. To identify potential CNVs, we used<br />

QuantiSNP and applied a series <strong>of</strong> quality control and CNV<br />

selection filters. CNVs were compared between patients and<br />

controls by permutation-based tests <strong>of</strong> association, as<br />

implemented in PLINK. Adopting a global burden approach,<br />

we found no difference in total number <strong>of</strong> CNVs in patients<br />

vs. controls. However, we observed that patients on average<br />

present larger singleton deletions than controls (patients vs.<br />

controls = 472.6kb vs. 249.3kb; P=0.0139). Furthermore, we<br />

identified three specific CNVs that were nominally associated<br />

with BP: a 224kb deletion on 9q (patients vs. controls = 0.57<br />

% vs. 0%; P=0.032; OR=4.96) and a 160kb duplication on 10q<br />

(patients vs.controls = 6% vs. 3.7%; P=0.035; OR=1.68), and<br />

a 265kb duplication on 6q overrepresented in the early onset<br />

BP disorder subgroup (12/196 patients vs. 22/872 controls =<br />

6.1% vs. 2.5%; P=0.00972; OR=2.52). Our results provide<br />

suggestive evidence for an involvement <strong>of</strong> large singleton<br />

deletions as well as CNVs on 6q, 9q, and 10q in BP. However,<br />

they need to be followed-up in independent samples before<br />

firm conclusions can be drawn.<br />

120 QUANTITATIVE TRAIT LOCI CONTRIBUTING<br />

TO PHYSIOLOGICAL AND BEHAVIORAL ETHANOL<br />

RESPONSES<br />

E. Drews* (1), I. Racz (1), A. Barth (1), A. Diaz Lacava (2),<br />

A. Bilkei-Gorzo (1), T. Wienker (2), A. Zimmer (1)<br />

1. Institute <strong>of</strong> Molecular Psychiatry, University <strong>of</strong> Bonn,<br />

Germany 2. Institute <strong>of</strong> <strong>Medical</strong> Biometry, Informatics and<br />

Epidemiology, University <strong>of</strong> Bonn, Germany<br />

* edrews@uni-bonn.de<br />

The aim <strong>of</strong> the present study was the identification <strong>of</strong> gene<br />

loci or quantitative traits that contribute to the development<br />

and manifestation <strong>of</strong> alcohol addiction and related behaviors.<br />

For a QTL study design the second filial (F2) generation <strong>of</strong> a<br />

C57BL/6J and C3H/HeJ mice intercross was first phenotyped<br />

in paradigms related to alcohol addiction. We examined<br />

behaviors including ethanol preference, stress-induced<br />

changes in ethanol preference, ethanol-induced hypothermia,<br />

tolerance, somatic withdrawal symptoms, withdrawal-induced<br />

anxiety and locomotor effects in 534 genetically<br />

heterogeneous mice from our intercross. The genotype <strong>of</strong><br />

these mice was subsequently assessed by microsatellite marker<br />

mapping accounting fragment lengths polymorphisms between<br />

the alleles <strong>of</strong> the <strong>of</strong>fspring. QTL contributing to alcohol<br />

dependence and related traits were subsequently identified<br />

using univariate and bivariate analyses. These analyses were<br />

performed with the R/qtl tool, which is an extensible,<br />

interactive environment for mapping QTL in experimental<br />

crosses. To determine individually-acting QTL, a genome scan<br />

using the multiple imputation algorithm was conducted, with a<br />

two-QTL genome scan for the full model (two QTL plus<br />

interaction) and for the additive model (two QTL but no<br />

interaction) interacting loci were analyzed. We detected<br />

chromosomal regions for future exploration where no obvious<br />

candidate gene has yet been mapped. Additionally, we found<br />

QTL that have already been published, thus confirming the<br />

impact <strong>of</strong> these loci on the development and manifestation <strong>of</strong><br />

alcoholism.


121 IDENTIFICATION OF MITOCHONDRIAL DNA<br />

MUTATIONS IN THREE PATIENTS EXHIBITING<br />

NEUROPSYCHIATRIC SYMPTOMS<br />

M. Nakamura* (1), M. Kato (1), M. Ichiba (1), A. Tomiyasu<br />

(1), H. Shimo (1), T. Higuchi (1), S. Ueno (2), A. Sano (1)<br />

1. Departments <strong>of</strong> Psychiatry, Kagoshima University Graduate<br />

School <strong>of</strong> <strong>Medical</strong> and Dental 2. Department <strong>of</strong> Psychiatry,<br />

Ehime University Graduate School <strong>of</strong> Medicine, Ehime<br />

University<br />

* nakamu36@m.kufm.kagoshima-u.ac.jp<br />

It has been suggested that mitochondrial dysfunction is an<br />

important factor in the pathogenesis <strong>of</strong> psychiatric disorders<br />

including depression, schizophrenia and dementia.Recently,<br />

we identified 3 adult patients exhibiting psychiatric symptoms<br />

and mitochondrial encephalopathy who were also shown to<br />

possess mitochondrial DNA (mtDNA) deletion mutations in<br />

their skeletal muscle, but not leukocytes. One patient showing<br />

schizophrenia-like symptoms and dementia was found to carry<br />

a novel polymorphism termed A5127G, which was shown not<br />

to be disease- or schizophrenia-specific. Although<br />

homoplasmic A8291T, which have been reported to be<br />

specific to limb-girdle type mitochondrial myopathy, was also<br />

detected in the patient, the mutation was shown not to be<br />

disease- or schizophrenia-specific. We next investigated<br />

whether mutations or copy number variants existed in the<br />

nuclear genes POLG, POLG2, C10orf2,SLC25A4, ECGF1,<br />

OPA1 and WFS1, as mutations in these genes are known to<br />

cause deletions in mtDNA. We failed to identify any<br />

pathogenic mutations or copy number variants in the 3 patients<br />

observed; however, A587C in SLC25A4was shown to<br />

represent a novel polymorphism not specific to the disease.<br />

Although further studies are required to determine the precise<br />

relationships between novel nuclear gene mutations and<br />

mtDNA mutations, it appears that themolecular pathways<br />

inducing mitochondrial abnormalities may be implicated in a<br />

variety <strong>of</strong> psychiatric conditions.<br />

122 CONTRIBUTION OF COPY NUMBER<br />

VARIATIONS IN DISC1 AND INTERACTION<br />

PARTNERS TO SCHIZOPHRENIA,<br />

SCHIZO-AFFECTIVE AND BIPOLAR DISORDER IN<br />

THE NORTHERN SWEDISH POPULATION<br />

M. Alaerts (1, 2), L. Moens (1, 2), S. Ceulemans (1, 2), D.<br />

Forero (1, 2), A. Lenaerts (1, 2), K. Norrback (3), D. Goossens<br />

(1, 2), R. Adolfsson (3), J. Del-Favero* (1, 2)<br />

1. 1Applied Molecular Genomics Group, Department <strong>of</strong><br />

Molecular Genetics, VIB, Belgium<br />

2. University <strong>of</strong> Antwerp (UA), Antwerp, Belgium<br />

3. Department <strong>of</strong> Clinical Sciences, Psychiatry, Umeå<br />

University, Sweden<br />

* jurgen.delfavero@molgen.vib-ua.be<br />

Accumulating genetic and functional evidence support DISC1<br />

as one <strong>of</strong> the most likely genes to be implicated in the etiology<br />

<strong>of</strong> schizophrenia(SZ), schizoaffective (SA) and bipolar (BP)<br />

disorders. Furthermore, many <strong>of</strong> its protein binding partners<br />

show evidence <strong>of</strong> involvement in the development <strong>of</strong> these<br />

psychiatric disorders. Since copy-number variation (CNV) is<br />

suspected to play an important causal role in these disorders,<br />

we explored DISC1 and its binding partners PAFAH1B1,<br />

NDE1, NDEL1, FEZ1, MAP1A, CIT and PDE4B for CNVs in<br />

1062 SZ, SA and BPpatients, 227 relatives and 512 unrelated<br />

control individuals from a Northern Swedish isolated<br />

population. This analysis was performed with the<br />

multiplexamplicon quantification (MAQ) method that allows<br />

high resolution CNV detection and resulted in the detection <strong>of</strong><br />

three duplications. A rare duplication covering NDEL1 and<br />

part <strong>of</strong> MYH10 detected in one SZ patientpotentially is a<br />

pathogenic variant with low frequency but high penetrance. A<br />

duplication encompassing NDE1 and adjacent genes might be<br />

a risk factor with small effect for SA disorder. The third is a<br />

relatively common duplication encompassing the first exon <strong>of</strong><br />

DISC1, and is not likely to be a susceptibility factor for these<br />

disorders. However, the presence <strong>of</strong> four copies <strong>of</strong> this DISC1<br />

CNV, observed exclusively in one patient with SZ, might<br />

nevertheless play a role. Our findings provide further evidence<br />

for involvement <strong>of</strong> DISC1 and some <strong>of</strong> its interaction partners<br />

in psychiatric disorders and also for a role <strong>of</strong> structural<br />

variants in the etiology <strong>of</strong> these devastating diseases.


123 RESEQUENCING AND COPY NUMBER<br />

VARIATIONS ANALYSES FOR GENES<br />

RESPONSIBLE FOR NEUROACANTHOCYTOSIS IN<br />

MOOD DISORDERS<br />

H. Shimo* (1), M. Nakamura (1), A. Tomiyasu (1), N.<br />

Shiokawa (1), M. Ichiba (1), S. Ueno (2), A. Sano (1)<br />

1. Department <strong>of</strong> Psychiatry, Kagoshima University Graduate<br />

School <strong>of</strong> <strong>Medical</strong> and Dental Sciences, Kagoshima<br />

University 6. Ehime University Graduate School <strong>of</strong> Medicine<br />

* hishimo7@m2.kufm.kagoshima-u.ac.jp<br />

Neuroacanthocytosis is an inclusive term for a genetically<br />

heterogeneous group <strong>of</strong> disorders characterized by the<br />

association <strong>of</strong> neurological and neuropsychiatric abnormalities<br />

with red cell acanthocytosis.The core neuroacanthocytosis<br />

syndromes mainly comprise <strong>of</strong> the two diseases,<br />

chorea-acanthocytosis(ChAc) and the McLeod<br />

syndrome(MLS). A high incidence <strong>of</strong> psychiatric disorders<br />

such as mood disorder is known in the patients with ChAc or<br />

MLS and also in carriers with heterozygous pathogenic<br />

mutation <strong>of</strong> VPS13A, the gene responsible for ChAc, in the<br />

patients’ pedigrees. Thus, we hypothesized that VPS13A<br />

andXK, the gene responsible for MLS, maybe associated with<br />

susceptibility to mood disorder. In the present study, we<br />

performed a comprehensive mutation screen <strong>of</strong> VPS13A and<br />

XK in 85 patients with mood disorder. We also performed<br />

copy number variations (CNVs) analysis by quantitative PCR<br />

and long range PCR in 72 patients with mood disorder. We<br />

identified four nonsynonymous and eight synonymous single<br />

nucleotide variants that were absent in 50 or more controls.<br />

We identified heterozygous exon60 deletion <strong>of</strong> VPS13A in<br />

one patient with mood disorder by CNVs analysis. Although<br />

further studies with larger sample size and functional analysis<br />

are needed, present study suggests that<br />

neuroacanthocytosis-related-genes may be associated with<br />

susceptibility to mood disorder.<br />

124 NOVEL PATHOGENIC MUTATIONS AND COPY<br />

NUMBER VARIATIONS OF VPS13A IN PATIENTS<br />

WITH CHOREA-ACANTHOCYTOSIS<br />

A. Tomiyasu* (1), M. Nakamura (1), H. Shimo (1), N.<br />

Shiokawa (1), S. Ueno (2), A. Sano (3)<br />

1. Department <strong>of</strong> Psychiatry, Kagoshima University Graduate<br />

School <strong>of</strong> <strong>Medical</strong> and Dental Sciences, Kagoshima<br />

University, Kagoshima 2. Ehime University Graduate School<br />

<strong>of</strong> Medicine, Ehime 3. Kagoshima University Graduate School<br />

<strong>of</strong> <strong>Medical</strong> and Dental Sciences, Kagoshima University,<br />

Kagoshima<br />

* aki-tomy@m2.kufm.kagoshima-u.ac.jp<br />

Chorea-acanthocytosis(ChAc) is a rare autosomal recessive<br />

neurodegenerative disorder caused by Loss <strong>of</strong> function<br />

mutations in the VPS13Agene encoding chorein, and<br />

characterized by adult-onset chorea and acanthocytosis in<br />

erythrocytes, psychiatric symptoms such as progressive<br />

cognitive dysfunction. In the present study, we performed<br />

mutational screenincluding sequencing and copy number<br />

variation (CNV) analyses <strong>of</strong> the VPS13A gene in ChAc<br />

patients. Sequencing analysis was performed for all 73 exons<br />

and flanking regions <strong>of</strong> VPS13A in 36 patients clinically<br />

diagnosed as ChAc. Since there is a possibility <strong>of</strong> the<br />

existence <strong>of</strong> CNVs which fail to be identified by only using<br />

sequencing analysis, we also performed real-time quantitative<br />

PCR for VPS13Agene on patients in whom only one<br />

heterozygous mutation was detected by the sequencing<br />

analysis. We identified 36 different mutations including<br />

disease causing novel CNVs, 27 <strong>of</strong> which have been<br />

unreported previously. In addition,we investigated the<br />

expression <strong>of</strong> chorein by the western blot analysis using red<br />

cell ghost samples from some <strong>of</strong> the patients. Western blot<br />

analysis perfectly demonstrated the absence <strong>of</strong> chorein in the<br />

patients. We provided the necessity <strong>of</strong> the real-time PCR as an<br />

accurate, simple, and reproducible method <strong>of</strong> detecting CNVs<br />

and confirmed an importance <strong>of</strong> western blot analysis as a<br />

diagnostic method <strong>of</strong> ChAc.


125 DEVELOPMENT OF NEXT GENERATION<br />

SEQUENCING METHODS FOR TARGETED<br />

SCREENING OF NUMEROUS CANDIDATE GENES IN<br />

MULTIPLE INDEXED DNA SAMPLES TO DETECT<br />

SNPS AND CNVS<br />

D. Morris* (1), E. Kenny (1), A. Gates (1), L. Cochrane (1),<br />

W. Gilks (1), P. Cormican (1), M. Gill (1), A. Corvin (1)<br />

1. Trinity College Dublin<br />

* morrisdw@tcd.ie<br />

Next-generation sequencing technology has allowed<br />

sequencing <strong>of</strong> whole genomes to be carried out in standard<br />

molecular genetics laboratories. However, an important<br />

application <strong>of</strong> this technology is sequencing <strong>of</strong> specific<br />

genomic regions, for example disease genes in patient<br />

samples. In order to sequence parts <strong>of</strong> the genome <strong>of</strong> interest,<br />

a number <strong>of</strong> methods have been developed including long<br />

range PCR and microarray capture. Both methods have been<br />

shown to work but are costly and expensive. Agilent<br />

Technologies have developed the SureSelect Target<br />

Enrichment System. This method allows targeting <strong>of</strong> 3.3Mb <strong>of</strong><br />

the genome by using cRNA baits. Indexing methods have<br />

been developed for next generation sequencing that allow<br />

multiplexing <strong>of</strong> samples in one sequencing library. We have<br />

combined the SureSelect Target Enrichment System with an<br />

indexing protocol to develop a cost-efficient method for<br />

targeting smaller regions <strong>of</strong> the genome (e.g. whole genes or<br />

just exons) in multiple DNA samples. We evaluated this<br />

method by sequencing target genes and known copy number<br />

polymorphic regions in HapMap CEU DNA samples and<br />

comparing the results to available online genetic variation<br />

data. For the SureSelect method, unique baits were designed to<br />

capture genomic fragments in the target regions and 3ug <strong>of</strong><br />

genomic DNA was used as input for the Illumina genomic<br />

DNA library prep method. The Illumina genomic DNA library<br />

prep method was modified to allow indexing <strong>of</strong> more than one<br />

sample in each sequencing library according to Craig et al<br />

(2008; PMID: 18794863). Analysis <strong>of</strong> 9 indexed DNA<br />

samples shows similar and even coverage across target genes<br />

indicating that an indexing method can be combined with the<br />

SureSelect method. We will present data on the performance<br />

<strong>of</strong> this method in detecting known sequence and structural<br />

variation in HapMap samples and present a protocol that will<br />

be extremely useful in the rapid sequencing <strong>of</strong> target genomic<br />

regions for SNPs and CNVs in patient and control samples.<br />

126 DETECTION OF STABLE REFERENCE GENES<br />

FOR REAL-TIME PCR ANALYSIS IN<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER<br />

G. Silberberg (1), K. Baruch (1), R. Navon* (1)<br />

1. Department <strong>of</strong> Human Genetics and Biochemistry, Sackler<br />

School <strong>of</strong> Medicine, Tel Aviv University, Tel Aviv, Israel<br />

* rnavon@post.tau.ac.il<br />

Gene expression studies using postmortem human brain tissue<br />

are a common tool for studying the etiology <strong>of</strong> psychiatric<br />

disorders. Quantitative real-time PCR (qPCR) is an accurate<br />

and sensitive technique used for gene expression analysis in<br />

which the expression level is quantified by normalization to<br />

one or more reference genes. Therefore, accurate data<br />

normalization is critical for validating results obtained by<br />

qPCR. This study aimed to identify genes that may serve as<br />

reference in postmortem dorsolateral–prefrontal cortices<br />

(Brodmann’s area 46) <strong>of</strong> schizophrenics, bipolar disorder<br />

(BPD) patients, and control subjects. In the exploratory stage<br />

<strong>of</strong> the analysis, samples <strong>of</strong> four BPD patients, two<br />

schizophrenics, and two controls were quantified using the<br />

TaqMan Low Density Array endogenous control panel,<br />

containing assays for 16 commonly used reference genes. In<br />

the next stage, six <strong>of</strong> these genes (TFRC, RPLP0, ACTB,<br />

POLR2a, B2M, and GAPDH) were quantified by qPCR in 12<br />

samples <strong>of</strong> each clinical group. Expressional stability <strong>of</strong> the<br />

genes was determined by GeNorm and NormFinder. TFRC<br />

and RPLP0 were the most stably expressed genes, whereas the<br />

commonly used 18S, POLR2a, and GAPDH were the least<br />

stable. This report stresses the importance <strong>of</strong> examining<br />

expressional stability <strong>of</strong> candidate reference genes in the<br />

specific sample collection to be analyzed.


127 COMPARISON OF FOUR CNV DETECTION<br />

PROGRAMS BASED ON GWAS DATA<br />

D. Zhang* (1), Y. Qian, N. Akula, F. McMahon , C. Liu, E.<br />

Gershon, Bipolar Genome Study (BiGS)<br />

1. University <strong>of</strong> Chicago<br />

* dandanz@uchicago.edu<br />

Methods for detecting Copy Number Variations (CNVs) using<br />

genome wide SNP arrays are frequently not consistent. We<br />

compared four CNV detection methods including Birdsuite,<br />

Partek, HelixTree, and PennCNV-Affy. We systematically<br />

assessed both rare and common CNVs in 1001 Bipolar cases<br />

and 1033 controls <strong>of</strong> European Ancestry from the Bipolar<br />

Genome Study (BiGS), and common CNVs in 270 HapMap<br />

samples. Quantitative real-time PCR (qPCR) was used to<br />

evaluate a subset <strong>of</strong> rare and common CNVs identified by the<br />

Affymetrix SNP 6.0 array in BiGs. For singleton deletions,<br />

Birdsuite and Partek had no false positive calls in our tested<br />

regions. At default parameters, HelixTree called twice as<br />

many singletons relative to the other methods, with the highest<br />

predictive error rate (>70%) by qPCR. For two common<br />

duplication regions, each program had a substantial predictive<br />

error rate (>=25%). For one common deletion region, qPCR<br />

validated all deletions called by Canary. However, the other<br />

three programs all had an unusually high error rate (>70%).<br />

We assessed the true detection rate <strong>of</strong> 893 independently<br />

discovered and validated CNVs from HapMap samples for<br />

each algorithm. Birdsuite is superior to the other three<br />

programs in the detection rate <strong>of</strong> known CNVs from HapMap<br />

samples. In conclusion, Birdsuite out-performed the other<br />

programs for singletons and common deletions. For common<br />

duplications, all programs had a substantial error rate.<br />

128 HISTONE GENE EXPRESSION PATTERNS IN<br />

DISCORDANT BIPOLAR SIBLINGS<br />

H. Chen (1), M. Burmeister (1), M. McInnis* (1)<br />

1. University <strong>of</strong> Michigan<br />

* mmcinnis@umich.edu<br />

Histones pr<strong>of</strong>oundly influence the structure <strong>of</strong> the<br />

chromosome and are subsequently likely to significantly affect<br />

the transcriptional activity <strong>of</strong> regional genes. We<br />

hypothesized that genes coding for histone proteins contribute<br />

to the genetic complexity <strong>of</strong> bipolar disorder. We studied the<br />

expression pattern <strong>of</strong> 20,589 gene transcripts in<br />

lymphoblastoid cell lines (LCL) from a sample <strong>of</strong> 24<br />

discordant bipolar sibling pairs, using the Illumina Bead chip<br />

technology (Human Refseq 8 V2). A paired t-statistic analysis<br />

identified a total <strong>of</strong> 91 genes with changed expression patterns<br />

in LCLs from bipolar individuals compared to their unaffected<br />

corresponding siblings (false discovery rate controlled at ~<br />

5%). It is striking that among the genes with significant<br />

expression changes, a systematic down regulation was<br />

observed in histone genes located on chromosomes 6p21 and<br />

1q21 in samples from patients compared to that from<br />

corresponding discordant sibling controls. Real time<br />

quantitative reverse transcriptional polymerase chain reaction<br />

assays confirmed the changes in five histone genes selected<br />

for validation. Genetic studies have suggested a bipolar<br />

disorder locus on 6p21. Our results suggest that down<br />

regulation <strong>of</strong> histone genes on 6p21 may contribute to the<br />

molecular biology <strong>of</strong> bipolar disorder.


130 INTEGRATED NEXT-GENERATION<br />

SEQUENCING AND ARRAY BASED ANALYSIS OF<br />

GENETIC MODIFIERS IN VELOCARDIOFACIAL<br />

SYNDROME (VCFS)<br />

A. Urban* (1), Y. Zhang (1), D. Palejev (1), R. Haraksingh<br />

(2), J. Korbel (3), M. Snyder (2), S. Weissman (1)<br />

1. Yale University Department <strong>of</strong> Genetics 2. Yale University<br />

MCDB Department 3. EMBL Heidelberg<br />

* alexander.e.urban@yale.edu<br />

Schizophrenia is a complex disease with a strong genetic<br />

component. Velocardi<strong>of</strong>acial Syndrome (VCFS) is typically<br />

caused by a 3 Mbp heterozygous deletion on chromosome<br />

22q11 and up to 30% <strong>of</strong> patients develop schizophrenia or<br />

similar symptoms. This makes VCFS a prime point <strong>of</strong> entry to<br />

understand the complex genetic etiology <strong>of</strong> schizophrenia.<br />

However, the search for the modifying genetic variants needed<br />

to differentiate the 30% <strong>of</strong> the patients with mental disease<br />

from the remaining 70%, with both groups carrying the 3 Mbp<br />

main deletion, has so far stayed incomplete. Earlier we used<br />

High-Resolution CGH (HR-CGH) on NimbleGen 385K<br />

custom arrays to show that the main deletion can substantially<br />

vary in size between patients [Urban, Korbel et al., PNAS 06]<br />

and that there are additional, smaller, CNVs to be found<br />

within the 3 Mbp deletion interval on the non-deleted<br />

chromosome [Korbel, Urban et al., PNAS 07]. Then we<br />

developed HR-PEM (paired-end mapping) [Korbel, Urban,<br />

Affourtit et al., Science 07] (also [Korbel et al., Genome Biol.,<br />

09]) for sequencing based high-resolution CNV analysis, and<br />

identified 22q11 as a hotspot for small CNV/SV. Here we<br />

report on the integrated genome-wide search for modifiers in<br />

VCFS using HR-CGH and HR-PEM in conjunction with array<br />

capture resequencing (ACRes). We are using a custom<br />

designed 385K NimbleGen longmer array to capture a 5 Mbp<br />

locus containing the VCFS region on chromosome 22q11 and<br />

a NimbleGen 2.1M longmer array to capture all exons in the<br />

human genome. We sequence the captured fractions on the<br />

Illumina Genome Analyzer 2, but using the paired-end<br />

protocol to enhance the power <strong>of</strong> detection and to reach out<br />

from the exons into their regulatory regions. Through this we<br />

can detect any type <strong>of</strong> genetic variant in the target regions,<br />

from single nucleotide changes to large CNV/SV, and we<br />

correlate and validate the results with HR-CGH data for the<br />

same samples. We have applied this approach to a panel <strong>of</strong><br />

VCFS patients, unaffected relatives and controls and have<br />

detected several hundred genetic variants. We also analyze<br />

genome-wide transcriptional activity in cell lines from the<br />

same VCFS patients, using Illumina GA2 based RNA-Seq to<br />

be able to detect intensity <strong>of</strong> expression as well as variable<br />

splicing, allele specific transcription and novel and non-exonic<br />

transcripts, and can now correlate the RNA-Seq data with the<br />

ACRes data on genetic variation.<br />

131 A COMBINED GENOME-WIDE ASSOCIATION<br />

ANALYSIS OF SCHIZOPHRENIA AND BIPOLAR<br />

DISORDER IN 20,000 INDIVIDUALS<br />

Douglas Ruderfer & Colm O’Dushlaine on behalf <strong>of</strong> the<br />

International Schizophrenia Consortium<br />

D. Ruderfer (1), C. O'dushlaine* (1), P. Sklar (1), S. Purcell<br />

(1)<br />

1. MGH<br />

* druderfer@chgr.mgh.harvard.edu<br />

Recent family and molecular genetic studies have provided<br />

strong evidence for a substantially shared genetic basis to<br />

schizophrenia (SCZ) and bipolar disorder (BP), the two major<br />

psychotic illnesses. This overlap suggests that a strategy <strong>of</strong><br />

combining genome-wide association study (GWAS) data from<br />

studies <strong>of</strong> both disorders, along with matched controls, could<br />

increase power to detect the shared common variants <strong>of</strong> small<br />

effect. We combined previously reported GWAS data on<br />

5,854 patients with schizophrenia, 4,737 with bipolar disorder<br />

and 9,303 controls from matching populations. We sought to<br />

identify genetic factors that are shared, and those that are<br />

unique to one <strong>of</strong> these two diseases, both at specific loci and at<br />

aggregate levels, including genes, pathways and genome-wide.<br />

We report results from a single SNP analysis for the combined<br />

phenotype (SCZ and BP versus controls) and a parallel<br />

analysis <strong>of</strong> SCZ versus BP, to search for disease-specific and<br />

modifier variants. A second set <strong>of</strong> analyses looked for<br />

co-occurrence <strong>of</strong> potentially distinct risk variants in similar<br />

genes and pathways, investigating the possibility that different<br />

risk alleles in the same gene or pathway may predispose to<br />

either SCZ or BP.


132 GENOME-WIDE SCREEN OF COPY NUMBER<br />

VARIANTS (CNVS) IN THE ADNI SAMPLE<br />

G. Guffanti* (1), F. Torri (2), J. Rasmussen (1), F. Kruggel<br />

(1), J. Turner (1), A. Lakatos (1), S. Potkin (1), F. Macciardi<br />

(1)<br />

1. University <strong>of</strong> California, Irvine 2. University <strong>of</strong> Milan<br />

* gguffant@uci.edu<br />

Alzheimer’s disease (AD) is the most common<br />

neurodegenerative disorder affecting more than 15 million<br />

people over the age <strong>of</strong> 65 worldwide. We present the results<br />

from a whole-genome Copy Number Variants (CNVs) study<br />

on the 818 subjects collected in the ADNI project. Our goal<br />

was to detect genomic structural copy-number variants that<br />

may increase the genetic susceptibility to developing AD.<br />

The ADNI sample was genotyped with more than 600,000<br />

SNPs using the Illumina Human610-Quad BeadChip. We<br />

identified CNVs by using Nexus, a CNV calling program that<br />

relies on SNP genotyping signal intensities. Identification <strong>of</strong><br />

events overlapping with previously reported CNVs was<br />

performed using the Database <strong>of</strong> Genomic Variants (Toronto).<br />

After both genotyping and CNV quality control procedures,<br />

we detected 11,694 CNVs in 638 subjects. The range <strong>of</strong><br />

frequencies <strong>of</strong> the CNVs we detected is in accordance with the<br />

trend (rarely more than 10%) reported in worldwide control<br />

populations (Jacobsson et al., 2008). We identified 10,135<br />

deletions, 1,647 <strong>of</strong> which are homozygous deletions, and<br />

1,559 duplications, including 6 high copies gain CNVs. We<br />

found only one region <strong>of</strong> overlapping segments homozygously<br />

deleted exclusively in MCI subjects. The gene affected by the<br />

deletion is ME3 that encodes the malic enzyme 3,<br />

NADP(+)-dependent, involved in the pathway <strong>of</strong> carbon<br />

fixation in the mitochondria metabolism. We previously<br />

identified another mitochondrial gene, TOMM40 associated<br />

with AD risk (Potkin et al., 2009). We present a<br />

comprehensive analysis <strong>of</strong> the structural variants that may<br />

contribute to the genetic susceptibility to AD.<br />

133 GENETIC POLYMORPHISM OF<br />

N-ACETYLTRANSFERASE 2 GENE IN KOREAN AND<br />

FOUR OTHER POPULATIONS<br />

H. Park* (1), T. Kand (1), H. Shin (1), H. Na (1), M. Chung<br />

(1)<br />

1. Korea Food and Drug Administration<br />

* hjpark@kfda.go.kr<br />

Background and objective : N-acetyltransferase 2 (NAT2) is<br />

responsible for inter-individual variations in the acetylation <strong>of</strong><br />

numerous drugs and in the transformation <strong>of</strong> aromatic and<br />

heterocyclic amines into carcinogenic intermediates. The aim<br />

<strong>of</strong> this study was to determine the allele frequencies <strong>of</strong><br />

polymorphisms <strong>of</strong> the NAT2 gene, analyze Linkage<br />

Disequilibrium (LD) block and haplotypes in Koreans and<br />

compare them with those <strong>of</strong> other ethnic groups. Methods :<br />

We analyzed genetic polymorphism in all functional promoter<br />

and exons <strong>of</strong> the NAT2 gene by direct sequencing <strong>of</strong> genomic<br />

DNA from 192 healthy Korean subjects; the LD and<br />

haplotypes blocks <strong>of</strong> these subjects were constructed from<br />

genotype data using an expectation-maximization algorithm.<br />

We compared these allele frequencies, LD block and<br />

haplotype structure with those <strong>of</strong> other ethnic groups<br />

registered on the International HapMap database. Results and<br />

discussion : We identified 33 polymorphisms including six<br />

novel single nucleotide polymorphisms(SNPs), -10778 T>C,<br />

-10777 A>G, -10351 A>G, -10199 C>T and -10104 G>T in<br />

promoter and 578 C>T in exon2(T193M) in the Korean<br />

subjects tested. All allele frequencies reported in the Koreans<br />

were similar to those <strong>of</strong> Asians except for one allele<br />

(rs4345600, -9306 A>G), whereas African and European<br />

Groups had different frequencies in exon2. The haplotype<br />

structure and LD block among the five groups also revealed<br />

significant differences. Conclusion : Ethnic differences in the<br />

NAT2 genotype frequencies may be one <strong>of</strong> the important<br />

factors that affect the cancer incidence and drug toxicity. Our<br />

current observations could be useful in assessing the<br />

susceptibility <strong>of</strong> different populations to the risk <strong>of</strong> cancer, as<br />

well as contribute to predicting the pharmacokinetics and<br />

pharmacodynamics <strong>of</strong> drugs that are metabolized by NAT2.


134 THE ASSOCIATIONS OF NONSYNONYMOUS<br />

SNPS IN CYP4A GENES WITH KOREAN STROKE<br />

PATIENTS<br />

J. Park* (1), Y. Bu (1), S. Park (1), J. Kim (1), S. Ko (1)<br />

1. Kyung-Hee University<br />

* suyahpark@gmail.com<br />

Stroke is affected by genetic factors as well as other diseases<br />

including hypertension and arteriosclerosis. However, the<br />

distinctive association with genetic variations is not<br />

discovered. Recently, 20-hydroxyeicosatetraenoic acid<br />

(20-HETE), which is produced from arachidonic acid by<br />

CYP4A family, has been reported to be associated with<br />

cerebral blood flow. Therefore, to investigate the association<br />

between genetic variations <strong>of</strong> CYP4A genes and stroke, this<br />

study genotyped five common nonsynonymous variants <strong>of</strong><br />

CYP4A11 and CYP4A22 in 674 stroke patients and 267<br />

controls. Each variant showed no significant association with<br />

stroke and its subtypes, such as hemorrhagic, ischemic and<br />

TOAST classification. However, a haplotype <strong>of</strong> CGC, which<br />

is composed <strong>of</strong> rs12564525C/T, rs2056900G/A,<br />

rs10789501T/C SNPs in CYP4A22, showed a significant high<br />

frequency in hemorrhagic group. In addition, it was found that<br />

the frequency <strong>of</strong> rs2056899A/T and rs10789501T/C <strong>of</strong><br />

CYP4A22 was lower than public database. Our findings<br />

suggest a possible association between the haplotype CGC <strong>of</strong><br />

CYP4A22 and hemorrhagic stroke. These results need further<br />

studies based on larger cohort group with stroke, and its<br />

functional study for the activity <strong>of</strong> haplotype CGC. This work<br />

was supported by the Second stage <strong>of</strong> Brain Korea 21 project<br />

in 2009.<br />

135 OLIGONUCLEOTIDE ARRAY FOR DETECTING<br />

LONG TANDEM REPEATS IN THE HUMAN GENOME<br />

L. Yu* (1), H. Bruce (1), G. Benson (2), R. Bannen (3), P.<br />

Warburton (4), T. Richmond (3), L. DeLisi (5), R. Margolis<br />

(1)<br />

1. Department <strong>of</strong> Psychiatry, Johns Hopkins University School<br />

<strong>of</strong> Medicine 2. Departments <strong>of</strong> Biology and Computer<br />

Science,Program in Bioinformatics,Boston University 3.<br />

Reseach Informatics ar NimbleGen Systems Inc. 4.<br />

Department <strong>of</strong> Genetics and Genomic Sciences,Mount Sinai<br />

School <strong>of</strong> Medicine 5. Department <strong>of</strong> Psychiatry, New York<br />

University<br />

* lyu17@jhmi.edu<br />

At least 12% <strong>of</strong> the human genome is subject to variations in<br />

copy number. The scope <strong>of</strong> variation ranges from single bp<br />

insertions and deletions to duplications and deletions <strong>of</strong> whole<br />

chromosomes. Our emphasis here is on tandem repeats. Short<br />

tandem repeats with repeating units <strong>of</strong> about 2-12 bp in length<br />

have emerged both as powerful markers for linkage studies<br />

and as mutations causing a number <strong>of</strong> human diseases. We<br />

hypothesize that polymorphisms <strong>of</strong> longer tandem repeats<br />

(unit length <strong>of</strong> 50 bp to >150,000 bp), relatively unexplored<br />

features <strong>of</strong> the human genome, may also contribute to normal<br />

human variation and to disease, including schizophrenia. To<br />

systematically address this issue, we developed an<br />

oligonucleotide array specifically designed to detect changes<br />

in repeating units in 2977 tandem repeats with a mean length<br />

<strong>of</strong> 4143 bp, using the Roche Nimblegen singleplex<br />

Comparative Genomic Hybridization (CGH) oligonucleotide<br />

platform. Validation by PCR confirmed the capacity <strong>of</strong> this<br />

array to detect repeat length variation. We now report the<br />

development <strong>of</strong> a second generation array, using the Roche<br />

Nimblegen 12X135K platform, designed to increase the<br />

number <strong>of</strong> repeats assessed and improve hybridization<br />

efficiency. This new array examines 4064 repeats with units a<br />

minimum <strong>of</strong> 50 bp in length and <strong>of</strong> at least 95% identity.<br />

Once validated, this array will provide a new tool for the<br />

discovery <strong>of</strong> genetic factors contributing to the etiology <strong>of</strong><br />

schizophrenia and other complex diseases.


HIGH THROUGHOUT SEQUENCING<br />

136 454 BASED SEQUENCING ANALYSIS OF THE<br />

DISC1 PATHWAY IN SCHIZOPHRENIA PATIENTS<br />

L. Moens (1,2), P. De Rijk (1,2), K. Norrback (3), D.<br />

Goossens (1,2), R. Adolfsson (3), J. Del-Favero* (1,2)<br />

1. 1Applied Molecular Genomics Group, VIB Department <strong>of</strong><br />

Molecular Genetics, Belgium 2. University <strong>of</strong> Antwerp (UA),<br />

Antwerpen, Belgium 3. Department <strong>of</strong> Clinical Sciences,<br />

Division <strong>of</strong> Psychiatry, University <strong>of</strong> Umeå, Sweden<br />

* jurgen.delfavero@molgen.vib-ua.be<br />

Schizophrenia(SZ) is among the most common disabling brain<br />

diseases worldwide, and its predisposition is influenced by a<br />

complex interplay between genetic and environmental factors.<br />

Despite major efforts during the last decade to identify risk<br />

genes for SZ, only a handful <strong>of</strong> candidate genes could be<br />

replicated in independent studies, and even a smaller number<br />

demonstrated a clear biological support. A recent and<br />

promising exception is Disrupted in Schizophrenia 1 (DISC1),<br />

originally identified via a balanced chromosomal translocation<br />

in a Scottish pedigree. Since then, compelling genetic and<br />

functional evidence has been accumulating, and several<br />

DISC1 interactors have been identified as independent genetic<br />

susceptibility factors for psychiatric illness, indicating that<br />

a‘DISC1 pathway’ is involved in the etiology <strong>of</strong> psychiatric<br />

diseases. To investigate the involvement <strong>of</strong> the ‘DISC1<br />

pathway’ in the etiology <strong>of</strong> SZ, we conducted an extensive<br />

sequence analysis <strong>of</strong> DISC1and 10 known interaction partners.<br />

Hereto, we applied a multiplex PCR based targeted pooled<br />

sample sequencing strategy, using the 454 platform. 4 DNA<br />

pools were sequenced (2 patient pools and 2 control pools),<br />

each consisting <strong>of</strong> 40 DNA samples. Variant detection and<br />

annotation was done using an in-house developed s<strong>of</strong>tware<br />

program designed to handle 454 data in pooled samples.<br />

Obtained variants were verified and their frequency<br />

determined by MassArray based genotyping. Although no<br />

obvious difference in (rare) variation load between<br />

schizophrenia patients and control individuals was observed,<br />

we did identify some rare potentially etiological variants.<br />

Furthermore, our results show that sequencing <strong>of</strong> pooled<br />

samples is a valuable strategy for simultaneous rare variant<br />

discovery and frequency estimation.<br />

MOOD DISORDERS<br />

137 ASSOCIATION OF MANIA AND<br />

HYPOTHYROIDISM<br />

D. Khemka* (1), J. Ali (1)<br />

1. University Of Mississippi <strong>Medical</strong> Center<br />

* dkhemka@psychiatry.umsmed.edu<br />

In the differential diagnosis <strong>of</strong> mood disorders thyroid<br />

abnormalities rank high on the list <strong>of</strong> medical conditions to be<br />

ruled out in clinical practice. The classic associations are<br />

bipolar disorder with hyperthyroidism and depression with<br />

hypothyroidism. Recently we encountered two patients who<br />

presented with mania in the presence <strong>of</strong> persisting<br />

hypothyroidism. This prompted us to conduct a literature<br />

review using Pub Med and Medline on a possible association.<br />

Nine case reports were identified in the period from 1980<br />

-2007. Analyses <strong>of</strong> these show the following associations:<br />

1) Mania and hypothyroidism were reported mostly in<br />

females; there is only one case report <strong>of</strong> a male with<br />

hypothyroidism and co-occurring mania; 2) Rapid cycling and<br />

hypothyroidism were reported more in females (consistent<br />

with established data); 3) Treatment generally involved using<br />

thyroid replacement (usually Levothyroxine).<br />

No specific mechanism(s) underlying the association <strong>of</strong><br />

thyroid dysfunction and bipolar disorder were clearly<br />

identified. In addition, no data is available on ethnic and<br />

cultural variation. Further investigations to better understand<br />

this association both from an etiological and therapeutic<br />

perspectives are needed.


138 COMMON VARIANTS IN THE B3 NICOTINIC<br />

RECEPTOR ARE ASSOCIATED WITH BIPOLAR<br />

DISORDER, SCHIZOPHRENIA, AND NICOTINE<br />

DEPENDENCE<br />

S. Hartz* (1), P. Lin (2), N. Rochberg (1), S. Saccone (1), W.<br />

Berrettini (3), H. Edenberg (4), E. Nelson (1), J. Nurnberger<br />

(4), L. Bierut (1), J. Rice (1)<br />

1. Department <strong>of</strong> Psychiatry, Washington University 2.<br />

Biology & Biomedical Sciences, Washington University 3.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Pennsylvania 4.<br />

Department <strong>of</strong> Psychiatry, Indiana University School <strong>of</strong><br />

Medicine<br />

* hartzs@wustl.edu<br />

Objective: Based on published associations between nicotine<br />

dependence and nicotinic receptor subunits, we investigated<br />

the association <strong>of</strong> schizophrenia and bipolar disorder with<br />

nicotinic receptor subunits. Method: Our primary sample<br />

consisted <strong>of</strong> 916 European American (EA) cases with bipolar<br />

disorder, and 1028 EA controls. Based on known associations<br />

with nicotine dependence, we genotyped 8 SNPs on<br />

chromosome 8 (3 bins) in the regions <strong>of</strong> CHRNB3 and<br />

CHRNA6, and 6 SNPs on chromosome 15 (3 bins) in the<br />

regions <strong>of</strong> CHRNA5 and CHRNA3. Two African American<br />

(AA) samples were used for validation: 389 subjects with<br />

bipolar disorder and 671 controls, and 921 subjects with<br />

schizophrenia and 941 controls. Results: Two highly<br />

correlated synonymous SNPs in CHRNB3, rs4952 and rs4953,<br />

were significantly associated with bipolar disorder (OR 1.74,<br />

95% CI (1.2, 2.4), p=0.001). This association remained<br />

significant both after adjusting for a smoking covariate and<br />

analyzing the association in non-smokers only. Two SNP bins<br />

(tagged by rs13439479 and rs7824160, r2=0.4 between the<br />

two bins in Africans) in high LD (r2=1) with rs4952 in EA<br />

were imputed in AA for both the bipolar and schizophrenia<br />

samples. The association with bipolar disorder was validated<br />

with rs13439479 (OR 1.78, p=0.007). An association with<br />

schizophrenia was found with rs7824160 (OR 1.26, p=0.046).<br />

Conclusions: Our results suggest that the β3 nicotinic receptor<br />

subunit is associated with schizophrenia and bipolar disorder<br />

in addition to its known association with nicotine dependence.<br />

139 PREVALENCE OF DEPRESSION AND SOME<br />

RELATED FACTORS AMONG DIABETIC PATIENTS<br />

AT PHC/ DUBAI-UAE<br />

H. Hussain* (1), Z. Kerem (1)<br />

1. Dubai Department <strong>of</strong> Health and <strong>Medical</strong> Services<br />

*hyHussain@dha.gov.ae<br />

Recognition <strong>of</strong> depression may be troublesome among patients<br />

with diabetes due to the overlap <strong>of</strong> symptoms, Co morbid<br />

diabetes and depression may result in poor clinical outcomes,<br />

impaired health status, economic burden, increased health care<br />

utilization, and poor self-care behaviors .Objective: To study<br />

the prevalence rate <strong>of</strong> depression among adults with diabetes<br />

attending Jabal Ali Health center & assess some associated<br />

factors. Materials & Methods: A cross sectional study has<br />

been carried out to identify the prevalence <strong>of</strong> Depression<br />

among DM patients, Systematic Random sample has been<br />

used for this study, no <strong>of</strong> patients selected for the study was<br />

114 out <strong>of</strong> total no <strong>of</strong> diabetic patients in JAC (341), Interview<br />

questionnaire using Patient health questionnaire ((PHQ9)) was<br />

used from the period 5th November 2007 – 3d march 2008 in<br />

Dubai/UAE. Results: (12) patients found to have depression<br />

out <strong>of</strong> total sample (114) examined, with a prevalence rate <strong>of</strong><br />

(11%), all case were mild to moderate depression with score<br />

ranging from (5-14/27), all cases were middle age group<br />

persons Age (20 – 55) years old, depression was significantly<br />

associated with glycemic control factor, the mean <strong>of</strong> glycemic<br />

control (HbA1C > .8.8%) among depressed in comparison<br />

with non depressed patient (HbA1C .7%) P


140 CYTOCHROME P450 CYP1A2, CYP2C9, CYP2C19<br />

AND CYP2D6 GENESARE NOT ASSOCIATED WITH<br />

RESPONSE AND REMISSION IN A SAMPLE OF<br />

DEPRESSIVE PATIENTS<br />

R. Calati* (1), I. Massat (2), S. Linotte (2), S. Kasper (3), Y.<br />

Lecrubier (4), R. Sens-Espel (5), J. Bollen (6), J. Zohar (7), J.<br />

Berlo (5), P. Lienard (5), D. De Ronchi (8), J. Mendlewicz (2),<br />

D. Souery (9), A. Serretti (1)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna, Bologna,<br />

Italy 2. Université Libre de Bruxelles, Brussels, Belgium<br />

3. Department <strong>of</strong> General Psychiatry, <strong>Medical</strong> University<br />

Vienna, Austria 4. Hôpital la Salpetriere, INSERM U302,<br />

Paris, France 5. DNA Vision sa, Charleroi, Belgium<br />

6. Sint-Truiden, Psychiatric center, Sint-Truiden, Belgium<br />

7. Chaim Sheba <strong>Medical</strong> Center, Tel-Hashomer, Israel<br />

8. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna, Bologna,<br />

Italy 9. Laboratoire de Psychologie Médicale, Université Libre<br />

de Bruxelles and Psy Pluriel, Centre Européen de Psychologie<br />

Médicale, Brussels, Belgium<br />

* raffaella.calati@unibo.it<br />

Cytochrome P450 genes are involved in the oxidative<br />

metabolism <strong>of</strong> antidepressants and have been hypothesized to<br />

be implicated in treatment response. The aim <strong>of</strong> the present<br />

study was to investigate the role <strong>of</strong> the allelic variations <strong>of</strong> the<br />

cytochrome P450 CYP1A2, CYP2C9, CYP2C19 and<br />

CYP2D6 genes in antidepressant treatment response and<br />

remission rates. Two hundred and seventy-eight patients<br />

(mean age: 58.77±14.67 years; female/male: 206/72) affected<br />

by major depression, responders (N=81) and non-responders<br />

(N=197) to at least one adequate antidepressant treatment,<br />

were recruited with a multicenter design for resistant<br />

depression and genotyped for all relevant variations.<br />

Genotypes were also pooled in activity pr<strong>of</strong>iles according to<br />

the literature (as an example, for the CYP2D6 gene pr<strong>of</strong>iles<br />

were poor, intermediate, extensive, and ultra-rapid). Using the<br />

General Linear Model and covariating for age and gender, the<br />

metabolic pr<strong>of</strong>iles <strong>of</strong> the genes were not associated with<br />

response and remission in the whole sample. However<br />

metabolic pr<strong>of</strong>ile <strong>of</strong> antidepressants differ, therefore, we<br />

repeated the analysis including as covariant the classes <strong>of</strong><br />

antidepressant with evidence <strong>of</strong> being metabolized by each<br />

specific cytochrome but no association was found as well. The<br />

investigated cytochrome genes do not seem to play a major<br />

role in antidepressant response in the present sample <strong>of</strong><br />

depressive patients. Nevertheless, methodological and sample<br />

size limitations <strong>of</strong> this study do not allow to provide definitive<br />

conclusions.<br />

141 HERITABILITY OF BIG FIVE PERSONALITY<br />

FACTORS IN MULTIPLEX FAMILIES WITH<br />

BIPOLAR DISORDER<br />

E. Hare* (1), J. Contreras (2), H. Raventos (2), R. Mendoza<br />

(3), D. Flores (4), A. Jerez (4), H. Nicolini (5), A. Ontiveros<br />

(6)<br />

1. University <strong>of</strong> Texas Health Science Center, San Antonio<br />

2. School <strong>of</strong> Biology and Center for Cellular and Molecular<br />

Biological Studies, University <strong>of</strong> Costa Rica 3. LABIOMED,<br />

UCLA 4. Centro Internacional de Trastornos Afectivos y<br />

Comportamiento Adictivo (CITACA),Guatemala City<br />

5. Grupo de Estudios Medicos y Familiares Carracci,<br />

S.C.,Mexico City 6. INFOSAME,Monterrey, Mexico.<br />

* hare@uthscsa.edu<br />

Associations have been observed between bipolar disorder and<br />

personality traits such as openness to experience and<br />

extraversion. In this study, 136 families including 351<br />

individuals with bipolar disorder I and their family members<br />

(1148 total subjects) took the short form <strong>of</strong> the NEO-PI-R.<br />

Best-estimate diagnosis <strong>of</strong> bipolar I was made using the<br />

Diagnostic Interview for Genetic Studies, Family Interview<br />

for Genetic Studies, and the patients' medical records.<br />

Heritabilities were 0.28, 0.23, 0.24, 0.20, and 0.35 for<br />

Agreeableness, Conscientiousness, Extroversion, Neuroticism,<br />

and Openness to Experience, respectively. Genetic<br />

correlations between Big Five factors ranged from -0.81 for<br />

Conscientiousness and Neuroticism to 0.77 for<br />

Conscientiousness and Extraversion. The genetic correlation<br />

between affectation with bipolar I and Extraversion was -0.32,<br />

suggesting that common genetic factors affect the<br />

development <strong>of</strong> bipolar I and introverted personality. Future<br />

linkage and association analyses in this population are likely<br />

to locate genomic regions associated with these traits,<br />

ultimately leading to the identification <strong>of</strong> genes and<br />

biochemical pathways involved in the development <strong>of</strong> bipolar<br />

disorder and introverted personality.


142 CNV-DISCOVERY IN LINKAGE REGIONS FOR<br />

BIPOLAR AFFECTIVE DISORDER<br />

M. Lekman* (1), R. Karlsson (2), L. Graae (2), A. Carmine<br />

Belin (2), D. Galter (2), I. Kockum (1), S. Paddock (2)<br />

1. Dept. <strong>of</strong> Clinical Neuroscience 2. Dept. <strong>of</strong> Neuroscience<br />

* magnus.lekman@ki.se<br />

The aim <strong>of</strong> the present study was to analyze whether linkage<br />

regions may harbor copy number variants with high<br />

penetrance for the susceptibility <strong>of</strong> bipolar affective disorder<br />

(BPAD). We carried out a combined whole-genome SNP and<br />

CNV-analysis to correlate linkage scores with CNV contents.<br />

Families were selected from 648 pedigrees (NIMH waves<br />

I-IV) based on genome-wide parametric linkage analysis using<br />

microsatellites. 277 individuals from 48 families were<br />

included in our study and genotyped on the Human610-Quad<br />

BeadChip-platform. Quality-control was performed to remove<br />

poorly performing SNPs or SNPs located in regions <strong>of</strong> CNVs<br />

(for calculating linkage scores). Only markers in linkage<br />

equilibrium were included in the linkage analysis. CNV<br />

detection was carried out using all 620,000 markers. Samples<br />

with a standard deviation for the LogR ratio <strong>of</strong> more than 0.3<br />

were re-genotyped. For the final analysis, all samples had a<br />

standard deviation <strong>of</strong> the LogR-ratio less than 0.3. Our<br />

preliminary CNV-discovery identified 12,935 CNVs<br />

(PennCNV). Obvious cell-culture artifacts (e.g., trisomies)<br />

were excluded from further analyses. Multipoint linkage<br />

analysis (NPL-all) was performed under three different<br />

affection status models (Merlin). Before weighted for CNVs,<br />

maximum linkage regions were identified on chromosome 3p<br />

(non-parametric LOD>2.75) and 18q (non-parametric<br />

LOD>2.3), with several additional, lower peaks in other<br />

regions. CNVs passing the calling-filter are currently<br />

correlated with the sum <strong>of</strong> family-wise linkage scores and<br />

further analyzed by functional studies regarding their possible<br />

significance in BPAD susceptibility.<br />

143 GENETIC OVERLAP BETWEEN BODY MASS<br />

INDEX (BMI) AND RECURRENT DEPRESSION<br />

M. Rivera* (1), S. Cohen-Woods (1), A. Schosser (1,2), K.<br />

Aitchison (1), G. Breen (1), A. Butler (1), N. Craddock (3), M.<br />

Gill (4), A. Korszun (5), C. Lewis (1,6), W. Maier (7), O.<br />

Mors (8), M. Ng (1), M. Owen (3), K. Pirlo (1), M. Preisig (9),<br />

J. Rice (10), M. Rietschel (11), R. Uher (1), I. Craig (1), A.<br />

Farmer (1), P. McGuffin (1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London. 2. Psychotherapy, <strong>Medical</strong> University Vienna,<br />

Austria. 3. Department <strong>of</strong> Psychological Medicine, School <strong>of</strong><br />

Medicine, Cardiff University, Cardiff CF10 3XQ, UK. 4.<br />

Department <strong>of</strong> Psychiatry, Trinity Centre for Health Sciences,<br />

Dublin 8, Ireland. 5. Barts and The London, Queen Mary’s<br />

School <strong>of</strong> Medicine and Dentistry, London E1 4NS, UK. 6.<br />

Molecular Genetics, School <strong>of</strong> Medicine, King’s College<br />

London. 7. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Bonn,<br />

53127 Bonn, Germany. 8. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Aarhus, DK-8000 Aarhus, Denmark. 9.<br />

Department <strong>of</strong> Adult Psychiatry, University Hospital <strong>of</strong><br />

Lausanne, 1008 Prilly-Lausanne, Switzerland. 10. Department<br />

<strong>of</strong> Psychiatry, Washington University, St Louis, MO 63130,<br />

USA. 11. Central Institute <strong>of</strong> Mental Health, 68159<br />

Mannheim, Germany.<br />

* margarita.rivera_sanchez@kcl.ac.uk<br />

There is growing evidence suggesting that rates <strong>of</strong> physical<br />

disease, such as obesity, are much higher among people with<br />

depression. Some recent studies support the hypothesis that<br />

there may be shared aetiological factors between recurrent<br />

unipolar depression, obesity and physical disorders. We aimed<br />

to investigate genetic overlap between Body Mass Index<br />

(BMI) and recurrent unipolar depression (UPD). The sample<br />

consists <strong>of</strong> 2628 depression cases and 1023 controls<br />

from several studies; Depression Case-Control (DeCC) study,<br />

Bipolar Association Case-Control (BACC) study, Depression<br />

Network (DeNT) study and the Genome Based Therapeutic<br />

Drugs for Depression (GENDEP) study. The sample is part <strong>of</strong><br />

a genome-wide association study (GWAS) <strong>of</strong> unipolar<br />

depression carried out by The Depression Consortium at the<br />

SGDP Centre, Institute <strong>of</strong> Psychiatry. DSM-IV/ICD-10<br />

depression phenotypes were ascertained using SCAN<br />

interview. Controls individuals were screened for absence <strong>of</strong><br />

psychiatric disorder. Furthermore, a wealth <strong>of</strong> detailed data on<br />

chronic, co-morbid physical diseases such as obesity, diabetes,<br />

heart disease and migraine in both the cases and the controls<br />

has been collected. These individuals were genotyped by CNG<br />

(Paris) using Illumina 610 Quad array platform. We also<br />

examined in detail known “obesity genes” FTO and MC4R in<br />

depression cases and controls. Statistical analyses <strong>of</strong><br />

association between these SNPs and obesity and depression<br />

was undertaken using PLINK.


144 LONGITUDINAL ASSOCIATIONS BETWEEN<br />

ANXIETY SENSITIVITY, ANXIETY AND<br />

DEPRESSION USING A GENETICALLY SENSITIVE<br />

DESIGN<br />

H. Zavos* (1), F. Rijsdijk (1), T. Eley (1)<br />

1. Social, Genetic, & Developmental Psychiatry Centre,<br />

Institute <strong>of</strong> Psychiatry, King’s College London<br />

*helena.zavos@kcl.ac.uk<br />

Anxiety and depression have a high incidence <strong>of</strong> co-morbidity<br />

and are commonly occurring problems in childhood and<br />

adolescence. Research has shown that the short variant <strong>of</strong> the<br />

5HTTLPR reduces the transcriptional efficiency <strong>of</strong> the<br />

serotonin transporter gene and is associated with both anxiety<br />

and depression. Anxiety sensitivity, the fear <strong>of</strong> anxiety related<br />

sensations, is a cognitive bias which has been associated with,<br />

and is predictive <strong>of</strong> anxiety in childhood, adolescence and<br />

adulthood. Previous research has also shown that anxiety<br />

sensitivity is both phenotypically and genetically correlated<br />

with depression. Our aim was to look at the relationships<br />

between these variables developmentally to better understand<br />

the direction <strong>of</strong> effects between them over the adolescent<br />

period and given the high genetic correlation between<br />

variables, look at associations between 5HTTLPR and a factor<br />

score <strong>of</strong> the 3 variables. Longitudinal associations between<br />

anxiety sensitivity, anxiety and depression were examined<br />

using a genetic cross-lagged model. This model estimates<br />

phenotypic causal effects <strong>of</strong> all variables on one another,<br />

while controlling for concurrent (genetic and environmental)<br />

associates between them. Analysis was conducted on 2-wave<br />

sibling and twin data from the G1219 study (mean age 15<br />

years at wave 1 and 17 years at wave 2). Genotyping <strong>of</strong><br />

5HTTLPR was performed <strong>of</strong> 1,467 individuals from the same<br />

sample. Our data suggests that early depression leads to later<br />

anxiety but that there is bi-directional relationship between<br />

anxiety sensitivity and anxiety and depression. Effects <strong>of</strong><br />

genes were more stable over time whereas environmental<br />

effects were more time specific.<br />

145 NEW CANDIDATE GENES FOR BIPOLAR<br />

DISORDER ARE REPLICATED IN SCANDINAVIAN<br />

SAMPLES<br />

M. Tesli* (1)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Oslo, Department <strong>of</strong><br />

Psychiatry, Oslo University Hospital<br />

*m.s.tesli@medisin.uio.no<br />

Bipolar disorder (BD) has a complex and important genetic<br />

component. Many genes, each with a limited effect, are<br />

believed to interact with each other and with environmental<br />

factors, to give rise to the disorder. Recent Genome-wide<br />

association studies (GWAS) have found association between<br />

variants <strong>of</strong> PALB2, ANK3 and BD. We wanted to replicate<br />

these findings in Scandinavian samples. In a replication study<br />

<strong>of</strong> PALB2 (n=787 cases/2862 controls), the preliminary<br />

results showed a significant association with BD in our<br />

Scandinavian sample (P=0.0058 after 100000 permutations).<br />

This was followed by investigations <strong>of</strong> BRCA2, a gene which<br />

function is closely related to that <strong>of</strong> PALB2, where the<br />

preliminary results showed a significant association with BD<br />

(P=0.0088 after Bonferroni correction). These findings<br />

support PALB2 and BRCA2 as risk genes specifically for BD,<br />

and suggest that dysfunctional DNA repair mechanism could<br />

be involved in BD pathophysiology. The preliminary analysis<br />

<strong>of</strong> ANK3 showed evidence <strong>of</strong> nominally significant<br />

association between 14 <strong>of</strong> 111 ANK3 SNPs and BD in a<br />

Norwegian case-control sample (n=182 cases/353 controls),<br />

with a lowest nominal P-value <strong>of</strong> 0.003. Due to increasing<br />

evidence <strong>of</strong> genetic overlap between BD and schizophrenia<br />

(SCZ), we also genotyped these ANK3 SNPs in 230 SCZ<br />

cases, and found 22 SNPs to be nominally significantly<br />

associated with SCZ (lowest nominal P-value=0.0004). These<br />

findings give further support to ANK3 being a BD<br />

susceptibility gene and to the hypothesis <strong>of</strong> overlapping risk<br />

genes between BD and SCZ.


146 COMT, INHIBITION, AND FUNCTIONAL<br />

ABILITY IN MANIC BIPOLAR DISORDER<br />

A. Minassian* (1), J. Kelsoe (1), M. Geyer (1), E. Twamley<br />

(1), W. Perry (1)<br />

1. U.C. San Diego Department <strong>of</strong> Psychiatry<br />

*aminassian@ucsd.edu<br />

Dysregulation <strong>of</strong> the dopaminergic (DA) system causes<br />

impairments in the adaptive function <strong>of</strong> inhibition and is<br />

thought to contribute to Bipolar Disorder (BD). The Valine<br />

(Val) allele <strong>of</strong> the catechol-O-methyltransferase (COMT)<br />

Val158Met polymorphism putatively reflects a genetic<br />

propensity for DA underactivity in the frontal cortex and is<br />

associated with the most pr<strong>of</strong>ound inhibitory and daily<br />

functioning deficits in schizophrenia patients. In BD,<br />

however, little is known about whether a genetic<br />

predisposition towards altered frontal DA activity may<br />

influence the ability to complete daily activities. The presence<br />

<strong>of</strong> inhibitory deficits may even mediate the relationship<br />

between genes and functional ability. We obtained COMT<br />

genotypes on acutely ill manic BD patients who were<br />

administered the <strong>UCSD</strong> Performance-Based Skills Assessment<br />

(UPSA-2), a role-play-based measure <strong>of</strong> functioning. Subjects<br />

underwent testing in the human Behavioral Pattern Monitor<br />

(hBPM), which quantifies exploratory activity. Val<br />

homozygote manic BD patients had lower total UPSA scores<br />

than did Methionine (Met) homozygotes, as well as trends<br />

towards lower UPSA Comprehension/Planning and<br />

Communication subscale scores. Moreover, worse UPSA-2<br />

performance was associated with restricted, perseverative<br />

exploration in the hBPM (r = -.87, p < .01). These preliminary<br />

findings are consistent with our hypothesis that inhibitory<br />

deficits such as perseverative behavior may influence the<br />

relationship between genes and real-world functional ability.<br />

The simultaneous study <strong>of</strong> genotypes and cognition might<br />

yield a compelling model to explain variations in functional<br />

ability in the BD population and identify the factors that make<br />

these individuals most vulnerable to impairments in<br />

day-to-day functioning.<br />

147 DEPRESSION IS INFLUENCED BY MANY<br />

COMMON GENETIC VARIANTS WITH SMALL<br />

EFFECT PARTLY SHARED WITH ANXIETY<br />

C. Middeldorp* (1), A. demirkan (2), N. Wray (3), P.<br />

Sullivan (4), J. Hottenga (5), C. Janssens (2), B. Oostra (2), G.<br />

Willemsen (5), E. de Geus (5), R. van Dyck (6), Y. Aulchenko<br />

(2), W. Nolen (6), F. Zitman (7), D. Boomsma (5), B. Penninx<br />

(8), C. van Duijn (2)<br />

1. Biological Psychology, VU University Amsterdam, De<br />

Bascule, Academic Center for Child and Adolescent<br />

Psychiatry 2. Departments <strong>of</strong> Epidemiology and Clinical<br />

Genetics, Erasmus University <strong>Medical</strong> Center Rotterdam 3.<br />

Genetic Epidemiology, Queensland Institute <strong>of</strong> <strong>Medical</strong><br />

Research 4. Departments <strong>of</strong> Genetics and Psychiatry,<br />

University <strong>of</strong> North Carolina 5. Biological Psychology, VU<br />

University Amsterdam 6. Department <strong>of</strong> Psychiatry,<br />

University <strong>Medical</strong> Center Groningen 7. Department <strong>of</strong><br />

Psychiatry, Leiden University <strong>Medical</strong> Center 8. Departments<br />

<strong>of</strong> Psychiatry, VU University <strong>Medical</strong> Center Amsterdam,<br />

University <strong>Medical</strong> Center Groningen, Leiden University<br />

<strong>Medical</strong> Center<br />

*cm.middeldorp@psy.vu.nl<br />

Recently, with the use <strong>of</strong> Genome Wide Association Study<br />

(GWAS) data, it has been shown that a substantial polygenic<br />

component contributes to the risk for schizophrenia (ISC,<br />

Nature, 2009). In addition, bipolar disorder appears to be<br />

influenced by the same polygenic component. The genetic<br />

architecture <strong>of</strong> depression and anxiety was investigated in a<br />

similar way using genetic scores, based on GWAS results in<br />

one set, to predict depression and anxiety in another set. The<br />

Dutch discovery set consisted <strong>of</strong> 1738 subjects with<br />

depression (MDD) and 1803 controls with over 400,000 SNPs<br />

genotyped (Sullivan et al., Molecular Psychiatry, 2008). The<br />

prediction set was another Dutch sample (ERF) consisting <strong>of</strong><br />

~1890 subjects who completed the Hospital Anxiety and<br />

Depression Scale (HADS) and the Center for Epidemiological<br />

Studies – Depression scale (CES-D) questionnaires. SNPs<br />

were selected based on the p-values


148 COMMINGLING ANALYSIS OF THE<br />

AGE-OF-ONSET IN BIPOLAR I DISORDER: TWO OR<br />

THREE CURVE MIXTURE?<br />

M. Grigoroiu-Serbanescu* (1), M. Rietschel (2), T. Paul (2),<br />

T. Schulze (3), M. Noethen (4), S. Cichon (4), R. Elston (5)<br />

1. Biometric Psychiatric Genetics Research Unit, Alexandru<br />

Obregia Psychiatric Hospital, Bucharest, Romania 2. Dept.<br />

Genetic Epidemiology in Psychiatry, Central Institute <strong>of</strong><br />

Mental Health, Mannheim, Germany 3. Unit on the Genetic<br />

Basis <strong>of</strong> Mood and Anxiety Disorders, National Institute <strong>of</strong><br />

Mental Health, National Institutes <strong>of</strong> Health, US Department<br />

<strong>of</strong> Health and Human Services, Bethesda, MD, USA 4.<br />

Institute <strong>of</strong> Human Genetics and Dept. <strong>of</strong> Genomics, Life and<br />

Brain Center, University <strong>of</strong> Bonn, Bonn, Germany 5. Dept.<br />

Epidemiology and Biostatistics, Case WesternReserve<br />

University School <strong>of</strong> Medicine, Cleveland, Ohio, USA<br />

*maria.serbanescu@web.de<br />

Age-<strong>of</strong>-onset (AO) seems to be a phenotypic variable with a<br />

strong genetic component and therefore useful in molecular<br />

analysis <strong>of</strong> bipolar disorder (BP). A debate about the cut-<strong>of</strong>f<br />

point for defining early AO has developed over the last few<br />

years. Using an Expectation-Maximization algorithm Bellivier<br />

et al (2001) found the best fit for a model with three<br />

onset-groups, proposing the age 20-21 as cut-<strong>of</strong>f for early<br />

onset, while using finite mixture models Kennedy et al (2005)<br />

found the best fit for a two onset-group model with age 40 as<br />

cut-<strong>of</strong>f and an incidence peak for mania in the age-band 21-25.<br />

Based on segregation analysis, we proposed a two AO-group<br />

model with cut-<strong>of</strong>f age 25 for early onset<br />

(Grigoroiu-Serbanescu et al. 2001). The present study aimed<br />

at investigating the best AO-model in 468 Romanian BPI and<br />

1458 German BPI patients using commingling analysis<br />

(SAGEv6.01-s<strong>of</strong>tware) (Elston et al, 2009). The best model<br />

was selected according to Akaike’s A Information Criterion<br />

(AIC). Results: The two and three AO-group models provided<br />

similar AIC-values both in the Romanian and the German<br />

sample. The means were 18 years (SD=7) in the Romanian<br />

early-onset group (40% <strong>of</strong> cases) and 20 years (SD=9) in the<br />

German early-onset group (50% <strong>of</strong> cases). Thus the cut-<strong>of</strong>f for<br />

early-onset (X + 1 SD) was different.Conclusion: Our results<br />

overlapped with the findings <strong>of</strong> Kennedy et al (2005) showing<br />

that two-curve and three-curve AO mixtures similarly fit the<br />

AO-distribution in BPI disorder and the cut-<strong>of</strong>fs for<br />

early-onset differ by sample.<br />

149 NON-PSYCHOTIC BIPOLAR I DISORDER<br />

ASSOCIATED WITH G72 GENE IN A ROMANIAN<br />

SAMPLE<br />

M. Grigoroiu-Serbanescu* (1), C. Diaconu (2), S. Herms<br />

(3), C. Bleotu (2), A. Neagu (2), R. Abou Jamra (3), M.<br />

Noethen (3), S. Cichon (3)<br />

1. Biometric Psychiatric Genetics Research Unit, Alexandru<br />

Obregia Psychiatric Hospital, Bucharest, Romania 2. Stefan S<br />

Nicolau Institute <strong>of</strong> Virology, Bucharest, Romania 3. Institute<br />

<strong>of</strong> Human Genetics, Department <strong>of</strong> Genomics, Life and Brain<br />

Center, University <strong>of</strong> Bonn, Germany<br />

*maria.serbanescu@web.de<br />

Objective: Since the complex G72/G30 was considered one <strong>of</strong><br />

the most replicated candidate gene for bipolar I disorder (BPI),<br />

we investigated the association between this complex and BPI<br />

in the Romanian population. Special attention was paid to the<br />

association <strong>of</strong> G72/G30 with psychotic BPI in view <strong>of</strong><br />

previous contradictory results concerning this association<br />

(positive findings: Schulze et al, 2005; Goes et al, 2007;<br />

negative: Williams et al, 2006; Maheshwari et al, 2009).<br />

Method: Fourteen G72-SNPs were genotyped at the Institute<br />

<strong>of</strong> Human Genetics, Bonn in a Romanian sample <strong>of</strong> 198 BPI<br />

patients and 180 controls screened for psychiatric disorders.<br />

Statistical analysis was performed with FAMHAP and<br />

Haploview v.3.32. The sample power to detect allelic<br />

association was 84%. Results: All genotyped SNPs were in<br />

HWE in controls and patients. The comparison <strong>of</strong> MAF in<br />

patients and controls showed no association <strong>of</strong> the fourteen<br />

SNPs with BPI in our total patient sample. But when<br />

subdividing the sample according to the presence/absence <strong>of</strong><br />

psychosis (incongruent plus congruent) in at least one illness<br />

episode, four SNPs reached nominal significance in the<br />

non-psychotic BPI subgroup [rs3916965 (M12) (P=0.044),<br />

rs1935057 (P=0.037), rs3916967 (M14) (P=0.043), rs2391191<br />

(M15, non-synonymous) (P=0.043)]. In the same subgroup, a<br />

two-SNP haploblock (GA) including M14 and M15 was<br />

significantly associated with BPI (P=0.0524 for global ?2<br />

corrected through permutations; OR=1.82).<br />

Conclusion: Similarly to Williams et al. (2006) and Hattori et<br />

al. (2003) we found the same G72-SNPs M12(G) and M15(A)<br />

associated with non-psychotic BPI. No SNP was associated<br />

with psychotic BPI.


150 DNA SEQUENCE ANALYSIS OF ERBB4 IN<br />

BIPOLAR DISORDER WITH MOOD-INCONGRUENT<br />

PSYCHOSIS<br />

F. Goes* (1), S. Wheelan (2), J. Potash (1)<br />

1. Dept. <strong>of</strong> Psychiatry, Johns Hopkins University School <strong>of</strong><br />

Medicine 2. Center for Computational Genomics, Johns<br />

Hopkins School <strong>of</strong> Medicine<br />

*fgoes@jhmi.edu<br />

Introduction: Prior studies <strong>of</strong> bipolar disorder (BP) with<br />

mood-incongruent psychosis (MICP) have shown that this<br />

subphenotype is familial and possibly linked to chromosomal<br />

regions implicated in susceptibility to schizophrenia. We<br />

recently performed a secondary GWAS analysis <strong>of</strong> BP-MICP<br />

in the Bipolar Genome Study (BiGS), where the strongest<br />

association was found in ERBB4 (P = 1.15 x 10-6), a<br />

neuregulin 1 receptor, also associated with schizophrenia. In<br />

this study, we hypothesized that common and rare variants<br />

might also be associated with BP-MICP, the form <strong>of</strong> BP with<br />

arguably the greatest phenotypic overlap with schizophrenia.<br />

Methods: Using the Sanger method, we have sequenced the<br />

promoter region, all 28 exons, and splice sites <strong>of</strong> ERBB4 in<br />

the 189 cases with BP-MICP from the BiGS study. Traces<br />

were analyzed using Phred/Phrap for base calling and<br />

sequence assembly, and bioinformatic annotation was<br />

performed using a variety <strong>of</strong> publicly available tools.<br />

Results: We identified 37 sequence variants, <strong>of</strong> which 17<br />

were novel. Of these, four were in coding regions, including a<br />

non-conservative missense mutation (Cys -> Trp) in exon 15.<br />

Novel variants were also found in the UTRs (n=4) and in<br />

splice site sequences (n=9). We are currently comparing the<br />

burden <strong>of</strong> rare variants, both across the gene and within<br />

annotated functional categories, with data from European<br />

origin subjects <strong>of</strong> the 1000 Genomes Project. Conclusion: Our<br />

ongoing analyses suggest that ERBB4, a putative<br />

schizophrenia susceptibility gene, might harbor common and<br />

rare variants associated with susceptibility to a form <strong>of</strong> bipolar<br />

disorder with phenotypic similarities to schizophrenia.<br />

151 GENETIC ASSOCIATION BETWEEN G PROTEIN<br />

beta 3 SUBUNIT, INTERLEUKIN-10, TUMOR<br />

NECROSIS FACTOR-alpha GENE POLYMORPHISMS<br />

AND IRRITABLE BOWEL SYNDROME IN KOREANS<br />

S. Lee (1), H. Lee (2), J. Choi (2), I. Sung* (1), H. Park (1),<br />

C. Jin (1)<br />

1. Konkuk University College <strong>of</strong> Medicine 2. Korea<br />

University College <strong>of</strong> Medicine<br />

*leehjeong@korea.ac.kr<br />

The aim <strong>of</strong> this study was to determine the role <strong>of</strong> G protein<br />

β3 subunit (GNB3), interleukin (IL)-10, and tumor necrosis<br />

factor (TNF)-α gene polymorphisms in Korean patients with<br />

irritable bowel syndrome (IBS) as diagnosed based on Rome<br />

III criteria. Korean case and control subjects <strong>of</strong> the study were<br />

recruited between December 2006 and December 2008 from<br />

visitors to the Health Promotion Center and Digestive Disease<br />

Center for gastrointestinal endoscopy. All the subjects signed<br />

an informed consent for genetic analysis and completed<br />

questionnaires on Rome III criteria and the Beck’s Depressive<br />

Inventory. GNB3, IL-10, and TNF-α gene polymorphisms<br />

were genotyped using a polymerase-chain-reaction-based<br />

method. Multifactor dimensionality reduction (MDR) analysis<br />

was used to assess gene-gene interactions. Although genotype<br />

and allele frequencies for all polymorphisms did not differ<br />

between the 94 IBS patients and 88 healthy controls, genotype<br />

and allele frequencies <strong>of</strong> GNB3 showed marginal significance<br />

between two group (X2=5.92, p=0.052; X2=3.76, p=0.053).<br />

MDR analysis revealed that there were no significant<br />

interactions <strong>of</strong> GNB3, IL-10, and TNF-α gene variants with<br />

susceptibility to IBS (p > 0.05). According to the IBS<br />

subtypes, GNB3 T allele was more strongly correlated with<br />

IBS with constipation (12 <strong>of</strong> constipation-dominant type and<br />

31 <strong>of</strong> mixed type) than with 51 diarrhea-dominant type and 88<br />

normal subjects (X2 =13.91, P=0.008). The results suggest<br />

that GNB3 825T allele frequency might be associated with<br />

IBS with constipation in Koreans.


152 REPLICATION STUDY OF 3Q29 LINKAGE AND<br />

ASSOCIATION FINDINGS IN BIPOLAR AFFECTIVE<br />

DISORDER.<br />

A. Schosser* (1), K. Fuchs (2), M. Stojanovic (2), M.<br />

Schloegelh<strong>of</strong>er (3), S. Cohen-Woods (4), I. Craig (4), P.<br />

McGuffin (4), H. Aschauer (3)<br />

1. Dep. <strong>of</strong> Psychiatry and Psychotherapy, <strong>Medical</strong> University<br />

Vienna, Austria; MRC SGDP Centre, Institute <strong>of</strong> Psychiatry,<br />

King’s College London, UK 2. Dep. <strong>of</strong> Psychiatry and<br />

Psychotherapy, <strong>Medical</strong> University Vienna, Austria;Center for<br />

Brain Research, Div. <strong>of</strong> Biochemistry and Molecular Biology,<br />

<strong>Medical</strong> University Vienna, Austria 3. Dep. <strong>of</strong> Psychiatry and<br />

Psychotherapy, <strong>Medical</strong> University Vienna, Austria 4. MRC<br />

SGDP Centre, Institute <strong>of</strong> Psychiatry, King’s College London,<br />

London, UK<br />

*alexandra.schosser@iop.kcl.ac.uk<br />

We identified a bipolar disorder (BPD) susceptibility region<br />

on chromosome 3q29 in a genome-wide linkage scan (Bailer<br />

et al. 2002, NPL-score4.09 in BPD families and 2.19 in<br />

schizophrenia families) and follow-up linkage analysis<br />

(Schosser et al. 2004, NPL-scores >3 with five markers in<br />

BPD families and 2.3 with one marker in schizophrenia<br />

families) performed in Austria. These results were supported<br />

by fine-mapping <strong>of</strong> 3q29 (Schosser et al.2007), finding<br />

NPL-scores >3.9 with SNPs (single<br />

nucleotidepolymorphisms) spanning a region <strong>of</strong> 3.46 Mbp in<br />

BPD families, and >1.7 in schizophrenia families. These<br />

results support the hypothesis <strong>of</strong> common genetic<br />

contributions to both disorders. Since genetic association<br />

studies are more powerful than linkage studies for detecting<br />

susceptibility genes <strong>of</strong> small effect size, the aim <strong>of</strong> the current<br />

study is to investigate the 3q29 region in a BPD case-control<br />

sample (BACCS) from UK and replicate the linkage findings<br />

from Austria.The BACCS sample consists <strong>of</strong> 522 cases<br />

(DSM-IV, Diagnostic and Statistical Manual, 4 th Edition; and<br />

ICD-10, International Classification <strong>of</strong> Diseases, 10thEdition)<br />

and 512 controls. Cases were interviewed using SCAN<br />

(Schedules forClinical Assessment in Neuropsychiatry) and<br />

controls screened for a lifetime absence <strong>of</strong> psychiatric<br />

disorders by a modified version <strong>of</strong> the Past History Schedule.<br />

Genotyping <strong>of</strong> 96 SNPs in the identified 3q29 linkage region<br />

is performed using the SNPlex Genotyping System (Applied<br />

Biosystems, Foster City,CA, USA), and raw data are analysed<br />

using the GeneMapper S<strong>of</strong>tware v3.7. The results <strong>of</strong> this<br />

currently ongoing genetic association study will be presented.<br />

153 FUNCTIONAL CONSEQUENCES OF THE ANK3<br />

GENE POLYMORPHISM ON SUSTAINED<br />

ATTENTION: INFLUENCES IN PATIENTS WITH<br />

BIPOLAR DISORDER, THEIR FIRST DEGREE<br />

RELATIVES AND HEALTHY CONTROLS.<br />

G. Ruberto* (1,4), E. Vassos (2), C. Lewis (2,3), R. Tatarelli<br />

(4), P. Girardi (4), D. Collier (2), S. Frangou (1)<br />

1. Section <strong>of</strong> Neurobiology <strong>of</strong> Psychosis, Institute <strong>of</strong><br />

Psychiatry, King’s College London, UK 2. Social, Genetic &<br />

Developmental Psychiatry Centre, Institute <strong>of</strong> Psychiatry,<br />

King’s College London, UK 3. Division <strong>of</strong> Genetics and<br />

Molecular Medicine, King’s College London School <strong>of</strong><br />

Medicine ,UK 4. Department <strong>of</strong> Psychiatry, Sant'Andrea<br />

Hospital, Second <strong>Medical</strong> School, La Sapienza University,<br />

Italy<br />

*gaia.ruberto@kcl.ac.uk<br />

Recent genome-wide genetic studies have reported a strong<br />

association with Bipolar Disorder (BD) in a region<br />

(rs10994336) within ANK3. Ankyrin 3, the protein coded by<br />

this gene, is involved in facilitating the propagation <strong>of</strong> action<br />

potentials by regulating the assembly <strong>of</strong> sodium gated ion<br />

channels. Both these processes are crucially involved in<br />

sequential signal detection which influences performance<br />

during sustained attention tasks. Abnormalities in sustained<br />

attention are prominent trait features <strong>of</strong> BD and may be<br />

associated with genetic risk. We examined the relationship <strong>of</strong><br />

allelic variation in the ANK3 gene and sustained attention,<br />

measured with the Degraded Symbol Continuous Performance<br />

Test (DS-CPT) in sample (n=189) individuals comprising BD<br />

patients (n=47), their unaffected first-degree relatives (n=75)<br />

and healthy controls (n=67). We found that the risk allele T<br />

was associated with reduced sensitivity and increased errors <strong>of</strong><br />

commission regardless <strong>of</strong> diagnosis. This genotype effect<br />

appeared relatively specific to sustained attention and was not<br />

present in the domains <strong>of</strong> general intellectual ability, memory,<br />

decision making and response inhibition. Our results suggest<br />

that the allelic variation in ANK3 impacts on sodium-channel<br />

dependent cognitive processes relating to signal detection and<br />

this mechanism may relate to risk for BD.


154 METAMOODICS: AN INTEGRATED WEB<br />

RESOURCE FOR SYSTEMATIC META-ANALYSIS OF<br />

GENETIC ASSOCIATION STUDIES IN MOOD<br />

DISORDERS<br />

M. Pirooznia* (1), F. Seifuddin (1), J. Judy (1), P. Belmonte<br />

(1), J. Potash (1), P. Zandi (1)<br />

1. The Johns Hopkins University School <strong>of</strong> Medicine<br />

*mpirooz1@jhmi.edu<br />

Nearly 15% <strong>of</strong> females and about 8% <strong>of</strong> males suffer from<br />

mood disorders at some point in life. Recent advances from<br />

the Human Genome Project and high-throughput technologies<br />

like single nucleotide polymorphisms (SNPs) genotyping<br />

platform have made it possible to study the complex genetic<br />

architecture <strong>of</strong> mood disorders at an unprecedented scale.<br />

However, these studies generate huge amounts <strong>of</strong> data and<br />

their interpretation presents an imposing challenge. We have<br />

developed a database with a web front application resource,<br />

called Metamoodics, to integrate the existing data from<br />

published genomic experiments <strong>of</strong> mood disorders. These<br />

include data from three different classes <strong>of</strong> genomic<br />

experiments: a) association studies <strong>of</strong> sequence variation, b)<br />

association studies <strong>of</strong> copy number variation, and c) gene<br />

expression studies. Data from these three classes <strong>of</strong> genomic<br />

experiments are meta-analyzed and the synthesized results are<br />

visualized within the context <strong>of</strong> relevant genomic annotations.<br />

Metamoodics provides additional functionality for users to<br />

conduct customized gene set enrichment analyses <strong>of</strong> the<br />

meta-analyzed results in order to test various hypotheses about<br />

the relevance <strong>of</strong> particular molecular pathways in the risk for<br />

mood disorders. The first release <strong>of</strong> the database will include<br />

data from three major depression genome-wide association<br />

studies (GWAS), six bipolar disorder GWAS, over 500<br />

candidate gene association studies, and sixteen genome-wide<br />

expression studies. Metamoodics will act as a virtual hub<br />

where the scientific community can explore the current state<br />

<strong>of</strong> knowledge about which SNPs, genes, and pathways may<br />

contribute to susceptibility to mood disorders. Current<br />

developments in the project are available at<br />

http://psychiatry.igm.jhmi.edu/metamoodics/.<br />

155 GENOME-WIDE ASSOCIATION STUDY OF<br />

PEDIATRIC DSM-IV BIPOLAR-1 DISORDER<br />

E. Mick* (1)<br />

1. Massachusetts General Hospital<br />

*emick1@partners.org<br />

We conducted a preliminary genome-wide association study<br />

<strong>of</strong> pediatric Bipolar-I disorder (BP1) nested within an ADHD<br />

GWAS <strong>of</strong> 735 affected-<strong>of</strong>fspring trios genotyped on the<br />

Illumina Human1M BeadChip. After applying quality control<br />

filters association with pediatric BP1 was tested with 835,136<br />

SNPs. ADHD children with psychiatric comorbidity were not<br />

excluded from participation and 126 <strong>of</strong> trios had comorbid<br />

BP1 disorder with an average age at onset <strong>of</strong> 7.3±4.8 years <strong>of</strong><br />

age. We observed one genome-wide statistically significant<br />

association (p=2.7E-8), Bonferonnip-value=0.025) with a SNP<br />

(rs2978024 MAF=0.24) located on chromosome 8q24.22 in<br />

3rd intron <strong>of</strong> ST3GAL1 (ST3<br />

beta-galactosidealpha-2,3-sialyltransferase 1). Two additional<br />

SNPs in ST3GAL1 were statistically significant gene-wide<br />

(rs2945787 at p=6.5E-6 and rs2978008 at p=7.0E-5) both also<br />

located in the 3rd intron and in moderate LD with the<br />

genome-wide significant finding (r2 = 0.6-0.78). ST3GAL1<br />

encodes an enzyme that transfers sialic acid to<br />

oligosaccharides; the resulting sialic acid rich glycoproteins<br />

bind to the selectin family <strong>of</strong> cell adhesion molecules.<br />

Although additional work is needed to replicate this finding is<br />

noteworthy that ST3GAL1 has been associated with adult BP1<br />

in two independent samples and is located under one <strong>of</strong> two<br />

genome-wide significant linkage peaks from a meta-analysis<br />

<strong>of</strong> 5,179 individuals (1,067 families) with adult BP-I disorder.


156 ANK3 SEQUENCING IN BIPOLAR DISORDER<br />

CASES<br />

A. McQuillin* (1), C. Her (1), K. Schlegel (2), A. Dedman<br />

(1), S. Sharp (1), R. Kandaswamy (1), A. Anjorin (1), N. Bass<br />

(1), J. Lawrence (1), H. Gurling (1)<br />

1. University College London 2. University <strong>of</strong> Nottingham<br />

*a.mcquillin@ucl.ac.uk<br />

Collaborative genome-wide association analysis in 4,387 cases<br />

<strong>of</strong> bipolar disorder and 6,209 controls found strong evidence<br />

for association in a region encompassing the ANK3 (ankyrin<br />

G) gene. Since then further evidence for association between<br />

ANK3 and bipolar disorder has been reported. ANK3 is an<br />

adaptor protein found at axon initial segments that has been<br />

shown to regulate the assembly <strong>of</strong> voltage-gated sodium<br />

channels. Sequence analysis <strong>of</strong> the ANK3 gene in bipolar<br />

disorder cases has been performed to identify potentially<br />

aetiological changes in the gene. Several novel potentially<br />

functional changes have been detected in the gene. Full<br />

genotyping <strong>of</strong> these potential aetiological changes will be<br />

done in our sample <strong>of</strong> 1000 cases and 1000 controls.<br />

157 GENOME-WIDE ASSOCIATION OF BIPOLAR<br />

DISORDER<br />

E. Smith* (1), J. Badner (2), T. Barrett (3), P. Belmonte (4),<br />

W. Berrettini (5), C. Bloss (1), W. Byerley (6), W. Coryell (7),<br />

D. Craig (8), H. Edenberg (9, 10), E. Eskin (11), T. Foroud<br />

(10), E. Gershon (2), T. Greenwood (12), M. Hipolito (13), D.<br />

Koller (14), W. Lawson (13), C. Liu (2), F. Loh<strong>of</strong>f (5), M.<br />

McInnis (15), F. McMahon (14), M. Sarah (1), C. Nievergelt<br />

(12), J. Nurnberger (14), E. Nwulia (13), J. Paschall (16), J.<br />

Potash (4), J. Rice (17), T. Schulze (18), W. Scheftner (19), C.<br />

Panganiban (20), N. Zaitlen (21), P. Zandi (4), P. Zhang (22),<br />

S. Zollner (15), J. Kelsoe (12,23), N. Schork (1,24,25)<br />

1. Scripps Translational Science Institute 2. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> Chicago 3. Department <strong>of</strong><br />

Psychiatry, Portland VA <strong>Medical</strong> Center 4. Department <strong>of</strong><br />

Psychiatry, Johns Hopkins School <strong>of</strong> Medicine 5. Department<br />

<strong>of</strong> Psychiatry, University <strong>of</strong> Pennsylvania 6. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> California, San Francisco 7.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Iowa 8.<br />

Neurogenomics Division, The Translational Genomics<br />

Research Institute 9. Department <strong>of</strong> Biochemistry and<br />

Molecular Biology, Indiana University School <strong>of</strong> Medicine<br />

10. Department <strong>of</strong> <strong>Medical</strong> and Molecular Genetics, Indiana<br />

University School <strong>of</strong> Medicine 11. Department <strong>of</strong> Computer<br />

Science, University <strong>of</strong> California, Los Angeles 12.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> California, San Diego<br />

13. Department <strong>of</strong> Psychiatry, Howard University 14.<br />

Department <strong>of</strong> Psychiatry, Indiana University School <strong>of</strong><br />

Medicine 15. Department <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Michigan 16. National Center for Biotechnology Information,<br />

National Library <strong>of</strong> Medicine, National Institutes <strong>of</strong> Health 17.<br />

Division <strong>of</strong> Biostatistics, Washington University 18. Genetic<br />

Basis <strong>of</strong> Mood and Anxiety Disorders Unit, National Institute<br />

<strong>of</strong> Mental Health Intramural Research Program, National<br />

Institutes <strong>of</strong> Health, US Dept <strong>of</strong> Health and Human Services<br />

19. Department <strong>of</strong> Psychiatry, Rush University 20.<br />

Neurogenomics Division, The Translational Genomics<br />

Research Institute 21. Department <strong>of</strong> Computer Science,<br />

University <strong>of</strong> California, Los Angeles 22. Biostatistics<br />

Department, University <strong>of</strong> Michigan 23. Department <strong>of</strong><br />

Psychiatry, VA San Diego Healthcare System 24. Department<br />

<strong>of</strong> Molecular and Experimental Medicine, The Scripps<br />

Research Institute 25. Scripps Health<br />

*ensmith@scripps.edu<br />

To identify BD genetic susceptibility factors, we have<br />

conducted genome-wide association in two populations: one<br />

<strong>of</strong> European Ancestry (EA, n = 2,200 cases; n = 1,436<br />

genotyped controls) and one <strong>of</strong> African Ancestry (AA; n =<br />

345 cases; n = 670 controls). We have previously reported<br />

results for a subset <strong>of</strong> these individuals that were genotyped<br />

through the GAIN initiative. Here we expand the study with<br />

an additional 1,199 EA cases and 403 EA controls genotyped<br />

on the Affymetrix 6.0 chip. In the full EA dataset, the<br />

strongest association did not reach genome-wide significance<br />

(rs17498753 in C2orf34, P = 2 x 10-6), and previously<br />

characterized associations at ANK3 and CACNA1C did not


significantly replicate, although the GAIN subset had shown<br />

replication <strong>of</strong> rs1938526 in ANK3 (p = 0.036). This is<br />

consistent with a marked heterogeneity in the genetic basis <strong>of</strong><br />

BD. We have partitioned this heterogeneity through more<br />

precise characterization <strong>of</strong> clinical subphenotypes and by<br />

performing analyses designed to detect alternative genetic<br />

architectures. Preliminary analyses include an examination <strong>of</strong><br />

polygenetic effects from additive and epistatic models; rare<br />

variants using population based identity-by-descent mapping;<br />

and allelic heterogeneity using haplotype analyses.<br />

Identity-by-descent mapping in the GAIN sample has yielded<br />

loci with intriguing candidates including the locus around the<br />

neuronal calcium sensor FREQ on chromosome 9 (P = 1 x<br />

10-6), which will be further followed up in the full dataset.<br />

These results suggest that although common variants may be<br />

challenging to reproducibly identify in BD, alternative<br />

analysis methods may be fruitful in identifying genetic factors<br />

for BD.<br />

158 MAPPING LOCI AND GENES FOR BIPOLAR<br />

AFFECTIVE DISORDER IN 34 CONSANGUINEOUS<br />

FAMILIES FROM IRAN USING SNP MICROARRAYS<br />

N. Moghimi (1), A. Noor (1), A. Lionel (2), A. Ziaolhagh (3),<br />

M. Ghadiri (3), M. Ohadi (4), J. Vincent* (1)<br />

1. CAMH 2. The Hospital for Sick Children 3. Iran University<br />

<strong>of</strong> <strong>Medical</strong> Sciences 4. The Social Welfare and Rehabilitation<br />

Sciences University,Tehran<br />

*john_vincent@camh.net<br />

Homozygosity mapping is a powerful method for finding rare<br />

recessive disease genes in monogenic disorders and may also<br />

be useful for locating risk genes in complex disorders. We<br />

have used Affymetrix 5.0 Single Nucleotide Polymorphism<br />

micorarrays to screen 72 individuals from 34 consanguineous<br />

families from Iran that are multiplex for bipolar affective<br />

disorder (BPAD), genome-wide for homozygosity-by-descent<br />

(HBD), or autozygosity, under the hypothesis that, in some<br />

rare instances, there are autosomal recessive mutations that are<br />

major risk factors for disease, and that autozygosity mapping<br />

<strong>of</strong> consanguineous multiplex BPAD families will enable the<br />

mapping <strong>of</strong> such genes. In addition, we looked for the<br />

presence <strong>of</strong> copy number variants (CNVs) that may also be<br />

risk factors for BPAD in these families. We identified 43 large<br />

HBD regions. For instance, in one family with 4 affected<br />

individuals we identified a 56Mb common HBD region on<br />

chromosome 8 (including candidate genes such as IMPA1,<br />

IMPAD1), and a 10Mb region on chromosome 17 (including<br />

SLC6A4). In a second family with four affected sisters, we<br />

identified a 7Mb HBD region on 5q35.2-q35.3, including the<br />

genes DRD1 and GRM6. We plan to screen selected candidate<br />

genes within these regions for homozygous mutations. We<br />

have also identified a number <strong>of</strong> large runs <strong>of</strong> homozygosity<br />

that appear frequently among the BPAD probands, including a<br />

400Kb run that spans the glutamate receptor gene, GRIK6. We<br />

also have found 48 CNVs <strong>of</strong> interest that potentially disrupt<br />

candidate genes such as SYN3, SLC39A11, and S100A10.


159 EVIDENCE FOR MOLECULAR PATHWAYS IN<br />

BIPOLAR DISORDER USING INDEPENDENT<br />

GENOME-WIDE ASSOCIATION DATASETS<br />

C. O'Dushlaine* (1), D. Ruderfer (1), S. Purcell (1), P. Sklar<br />

(1)<br />

1. MGH<br />

*odushlaine@chgr.mgh.harvard.edu<br />

The etiology <strong>of</strong> biopolar disorder (BP), particularly with<br />

regards to enrichment <strong>of</strong> genetic association within specific<br />

molecular pathways, remains poorly described. Here we apply<br />

a recently described pathway analysis method to 325,690<br />

SNPs common to two independent BP datasets (1,829 cases,<br />

2,935 controls (WTCCC); 1,498 cases, 954 controls (Sklar et<br />

al.)) and report pathways that are significantly enriched in<br />

both. Among those pathways implicated in BP<br />

pathophysiology were pathways relating to Glycan structures<br />

and cell-adhesion molecules. Compelling evidence exists in<br />

the literature supporting the putative effects <strong>of</strong> disturbance to<br />

these pathways and processes in effecting the BP phenotype.<br />

These pathways and their underlying genetic loci represent<br />

new and promising targets for additional studies <strong>of</strong> BP.<br />

160 INDEPENDENT EVIDENCE THAT REFINED<br />

PHENOTYPING OF BIPOLAR PATIENTS REVEALS<br />

SIGNIFICANT ASSOCIATION FOR GABAA<br />

RECEPTOR GENES<br />

R. Breuer* (1), M. Hamshere (2), J. Strohmaier (1), M.<br />

Mattheisen (3), F. Degenhardt (4), S. Meier (1), T. Paul (1),<br />

M. O’Donovan (2), T. Mühleisen (4), T. Schulze (5), M.<br />

Nöthen (4), S. Cichon (4), N. Craddock (2), M. Rietschel (1)<br />

1. Department <strong>of</strong> Genetic Epidemiology in Psychiatry, Central<br />

Institute <strong>of</strong> Mental Health, University <strong>of</strong> Heidelberg,<br />

Mannheim, Germany 2. Department <strong>of</strong> Psychological<br />

Medicine, School <strong>of</strong> Medicine, Cardiff University, UK 3.<br />

Institute for <strong>Medical</strong> Biometry, Informatics, and<br />

Epidemiology (IMBIE), University <strong>of</strong> Bonn, Germany 4.<br />

Institute <strong>of</strong> Human Genetics, Department <strong>of</strong> Genomics, Life &<br />

Brain Center, University <strong>of</strong> Bonn, Germany 5. Unit on the<br />

Genetic Basis <strong>of</strong> Mood and Anxiety Disorders, National<br />

Institute <strong>of</strong> Mental Health, National Institutes <strong>of</strong> Health, US<br />

Department <strong>of</strong> Health and Human Services, Bethesda, MD,<br />

USA<br />

*rene.breuer@zi-mannheim.de<br />

There is increasing evidence that refinement <strong>of</strong> phenotype <strong>of</strong><br />

complex psychiatric disorders can produce stronger<br />

association signals. Craddock et al.1 recently reported that the<br />

index association signal obtained using a set <strong>of</strong> GABAA<br />

receptor genes in bipolar disorder was enriched and became an<br />

order <strong>of</strong> magnitude stronger in a much smaller subsample <strong>of</strong><br />

DSM IV bipolar patients who had been assigned a refined<br />

RDC diagnosis <strong>of</strong> “schizoaffective disorder, bipolar (SABP).”<br />

In order to replicate this finding, we re-diagnosed n=682 DSM<br />

IV bipolar patients included in our previous GWA study<br />

according to RDC criteria based on OPCRIT data<br />

(RDC-SABP, n=352, and, RDC-nonSABP, n=330). We<br />

applied a gene-based approach using all available SNPs<br />

(n=362) in the 19 GABAA receptor genes. While no<br />

association with a DSM IV diagnosis <strong>of</strong> bipolar disorder was<br />

observed, significant association (p=8.8x10-3) was obtained<br />

when only SABP patients were included. The genes making a<br />

major contribution to these results were GABRB2<br />

(p=1.9x10-4), GABRG2 (p=1.2x10-2) and GABRA1<br />

(p=1.7x10-2). The strongest SNP association was obtained for<br />

rs7714930 (p=1.1x10-5) within GABRB2. Our results thus<br />

replicate the association findings for RDC-SABP with<br />

GABAA receptor genes, and show that the refining <strong>of</strong> major<br />

phenotypes may be a prerequisite to the identification <strong>of</strong><br />

genetic risk factors for complex psychiatric disorders.<br />

1Craddock et.al. Mol Psychiatry, 2008 Jul 1. [Epub ahead <strong>of</strong><br />

print]


161 HIGHER LEVELS OF SUICIDAL BEHAVIOR ARE<br />

ASSOCIATED WITH ELEVATED SCORES OF<br />

NEUROTICISM AND SUBSTANCE USE IN A SAMPLE<br />

OF BIPOLAR I AND BIPOLAR II SUBJECTS.<br />

M. Kamali* (1), S. Langenecker (1), A. Prossin (1), A.<br />

Vederman (1), M. McInnis (1)<br />

1. University <strong>of</strong> Michigan, Department <strong>of</strong> Psychiatry and<br />

Depression Center<br />

*masoud@med.umich.edu<br />

Introduction: Further differentiating the clinical phenotypes <strong>of</strong><br />

bipolar disorder will assist in the search for biomarkers.<br />

Suicidal behavior is a common and dangerous complication <strong>of</strong><br />

bipolar disorder. Factors such as high impulsivity have been<br />

associated with elevated risk <strong>of</strong> suicidal behavior. Higher<br />

impulsivity has also been associated with substance use<br />

disorder and violent and aggressive behavior. The subtype <strong>of</strong><br />

bipolar disorder with higher levels <strong>of</strong> impulsivity as indicated<br />

by higher rates <strong>of</strong> aggression, suicidal behavior and substance<br />

use could be categorized as a distinct phenotype and an<br />

appropriate candidate for biomarker investigations. Methods:<br />

The studied sample is part <strong>of</strong> the larger cohort <strong>of</strong> bipolar<br />

subjects from the Heinz C. Prechter Longitudinal Project.<br />

This is a longitudinal study <strong>of</strong> bipolar disorder with more than<br />

400 subjects enrolled. Data from over 200 subjects with<br />

bipolar I and II were used in this analysis. Information<br />

regarding diagnosis, substance use and suicidal behavior was<br />

collected using the Diagnostic Interview for Genetic Studies<br />

(DIGS) and confirmed by a best estimate process involving at<br />

least one <strong>of</strong> the authors. The NEO-PI personality inventory<br />

was collected for all subjects. Results: The subjects with<br />

higher levels <strong>of</strong> suicidal behavior showed elevated scores in<br />

the N domain and N1-6 facets <strong>of</strong> the NEO-PI. Subjects with<br />

higher rates <strong>of</strong> suicidal behavior also had higher levels <strong>of</strong><br />

substance use disorders compared to the non-suicidal bipolar<br />

subjects. Conclusion: Higher scores on the six N facets <strong>of</strong> the<br />

NEO-PI, which measure personality traits such as anxiety,<br />

angry hostility, depression, self-consciousness, impulsivity<br />

and vulnerability, along with substance use disorders are<br />

correlated with higher rates <strong>of</strong> suicidal behavior in this<br />

population <strong>of</strong> bipolar subjects. This phenotype <strong>of</strong> the disorder<br />

may be a candidate for further biomarker studies.<br />

162 A GENE EXPRESSION STUDY OF<br />

SPERMIDINE/SPERMINE<br />

N(1)-ACETYLTRANSFERASE (SSAT) IN BIPOLAR<br />

PATIENTS CHARACTERIZED FOR THE RISK OF<br />

SUICIDE: PRELIMINARY FINDINGS<br />

A. Squassina* (1), D. Congiu (1), M. Manchia (1), P. Piccardi<br />

(1), G. Severino (1), C. Chillotti (2), R. Ardau (2), M. Alda<br />

(3), M. Del Zompo (1, 2)<br />

1. Laboratory <strong>of</strong> Molecular Genetics, Department <strong>of</strong><br />

Neurosciences “B.B. Brodie”, University <strong>of</strong> Cagliari,<br />

Monserrato, (Ca), Italy 2. Unit <strong>of</strong> Clinical Pharmacology <strong>of</strong><br />

the University Hospital <strong>of</strong> Cagliari, Italy 3. Department <strong>of</strong><br />

Psychiatry, Dalhousie University, Nova Scotia, Canada<br />

*squassina@unica.it<br />

It is well known that chronic lithium treatment significantly<br />

reduces the risk <strong>of</strong> suicide in subjects with major affective<br />

disorders. A number <strong>of</strong> studies reported an underexpression <strong>of</strong><br />

spermidine/spermine N(1)-acetyltransferase (SSAT) gene, a<br />

key enzyme <strong>of</strong> the polyamine system, in several brain regions<br />

<strong>of</strong> suicide victims (Sequeira et al. 2006; Guipponi et al.,<br />

2008). Interestingly, some evidences shows that lithium in<br />

vitro induces an upregulation in SSAT expression in<br />

lymphoblasts from bipolar patients (BD). In this abstract we<br />

present data <strong>of</strong> a preliminary study in which SSAT expression<br />

was measured in a group <strong>of</strong> lymphoblastoid cell lines (LCL)<br />

from 7 Sardinian BD subjects that were part <strong>of</strong> a sample <strong>of</strong> 29<br />

BD patients characterized for the risk <strong>of</strong> suicide. LCLs from<br />

each subject were divided into two lines: one was treated with<br />

lithium 1mM for 1 week while the other was not treated. The<br />

fold changes in SSAT relative expression levels were<br />

calculated using the ΔΔCt method. The analysis <strong>of</strong> paired<br />

SSAT expression values showed that lithium significantly<br />

increases SSAT expression (fold difference = 1.20; paired t<br />

test, p = 0.006). The limited number <strong>of</strong> LCLs included in this<br />

preliminary study did not allow us to perform comparisons<br />

among the groups <strong>of</strong> patients with different risk <strong>of</strong> suicide.<br />

Our preliminary findings provide strong support for an<br />

upregulating effect <strong>of</strong> lithium on SSAT expression in<br />

lymphoblasts. Further analyses <strong>of</strong> the entire sample will<br />

provide more insights into the role <strong>of</strong> SSAT in lithium<br />

mechanism <strong>of</strong> action and in the risk <strong>of</strong> suicide.


163 A GENOME-WIDE ASSOCIATION STUDY OF<br />

AMYGDALA ACTIVATION IN YOUTHS WITH AND<br />

WITHOUT BIPOLAR DISORDER<br />

X. Liu*, N. Akula, M. Skup, M. Brotman, E. Leibenluft, F.<br />

Mcmahon<br />

*liuxinmin@mail.nih.gov<br />

OBJECTIVE: Functional magnetic resonance imaging (fMRI)<br />

is commonly used to characterize brain activity that may<br />

underlie a variety <strong>of</strong> psychiatric disorders. Previously, fMRI<br />

uncovered differences, compared to healthy controls, in<br />

amygdala activation during a face processing task in pediatric<br />

patients with bipolar disorder. In order to explore the genetic<br />

architecture <strong>of</strong> this neuroimaging phenotype, we undertook a<br />

genome-wide association study. METHOD: 39 patients with<br />

bipolar disorder and 29 healthy controls, who had previously<br />

undergone fMRI when viewing a neutral face, were genotyped<br />

with a genome-wide single nucleotide polymorphism (SNP)<br />

array. After quality control, a total <strong>of</strong> 104,043 SNPs were<br />

tested against normalized amygdala activation scores obtained<br />

from the right and left hemispheres. Genetic association was<br />

tested with covariates to control for race and ethnicity.<br />

RESULTS: Right amygdala activation under the hostility<br />

contrast was most strongly associated with a SNP in the gene<br />

DOK5 (rs2023454, P=4.88x10-7, FDR=0.05), a substrate <strong>of</strong><br />

TrkB/C receptors that is involved in neurotrophin signaling.<br />

This SNP accounted for about 32% <strong>of</strong> the variance among<br />

youth with bipolar disorder and 12% <strong>of</strong> the variance among<br />

healthy youth in this sample. Interesting results (FDR


165 A FAMILY BASED STUDY OF DENOVO COPY<br />

NUMBER VARIANTS IN BIPOLAR DISORDER USING<br />

A 2.1 MILLION PROBE MICROARRAY CGH<br />

PLATFORM<br />

D. Malhotra* (1), V. Makarov (1), S. McCarthy (1), A.<br />

Malhotra (2), J. Potash (3), F. McMahon (4), T. Schulze (4),<br />

E. Leibenluft (5)<br />

1. 1 Bungtown Road, Cold Spring Harbor, New York 2.<br />

Department <strong>of</strong> Psychiatry Research, The Zucker Hillside<br />

Hospital, Glen Oaks, Long Island, New York 3. Department <strong>of</strong><br />

Psychiatry and Behavioral Sciences Johns Hopkins Hospital,<br />

Baltimore, Maryland 4. Genetic Basis <strong>of</strong> Mood and Anxiety<br />

Disorders Unit; Institute for Mental Health, National Institutes<br />

<strong>of</strong> Health, Bethesda, Maryland 5. Genetic Basis <strong>of</strong> Mood and<br />

Anxiety Disorders Unit; Institute for Mental Health, National<br />

Institutes <strong>of</strong> Health, Bethesda, Maryland 6. Mood and Anxiety<br />

Disorders Program, National Institute for Mental Health,<br />

National Institutes <strong>of</strong> Health, Bethesda, Maryland<br />

*malhotra@cshl.edu<br />

The role <strong>of</strong> copy number variants in the etiology <strong>of</strong> bipolar<br />

disorder has been much less studied. Recent studies show that<br />

rare denovo CNVs occur with significantly increased<br />

frequency in children with neurodevelopmental disorders like<br />

autism and schizophrenia. However, the role <strong>of</strong> denovo cnvs<br />

in patients with mood disorders, including bipolar disorder is<br />

not known. To investigate the role <strong>of</strong> denovo cnvs in bipolar<br />

disorder, we initiated a family based genetic study called<br />

GEM (Genetics <strong>of</strong> early onset Mania) on 198 bipolar trios.<br />

The GEM bipolar sample collection includes a significant<br />

fraction <strong>of</strong> bipolar cases that are sporadic and also,<br />

approximately half <strong>of</strong> patients in the collection have an early<br />

age-at-onset (age at first mania


167 AKT1 AND NEUROCOGNITION IN BIPOLAR<br />

DISORDER<br />

C. Nnadi* (1), K. Burdick (1), C. Hodgkinson (2), B. Buzas<br />

(2), P. DeRosse (1), T. Lencz (1), D. Goldman (2), A.<br />

Malhotra (1)<br />

1. Psychiatry Research, The Zucker Hillside Hospital, North<br />

Shore-Long 2. Laboratory <strong>of</strong> Neurogenetics, National Institute<br />

<strong>of</strong> Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.<br />

*cnnadi@lij.edu<br />

Background: The AKT1, a gene encoding a serine/threonine<br />

kinase, modulates gene expression, protein synthesis and cell<br />

cycle apparatus and is involved in the pathways for<br />

lithium-sensitive cell survival and proliferation. Recent data<br />

appear to implicate a haplotype <strong>of</strong> AKT1 in susceptibility to<br />

schizophrenia and in decreased AKT1 protein levels in the<br />

brains <strong>of</strong> individuals with schizophrenia (Emamian et al<br />

2004). Interestingly, a linkage study <strong>of</strong> bipolar disorder<br />

reported a modest signal in chromosome 14q 22-32 in the<br />

region <strong>of</strong> AKT1. Thus, in the current study, we tested for<br />

association <strong>of</strong> AKT1 gene and bipolar diagnosis and also<br />

analyzed our data for the role <strong>of</strong> AKT1 in neurocognition<br />

Methodology/Principal findings: Following an informed<br />

consent process, we recruited and characterized 206 unrelated<br />

Non-Hispanic White participants. Participants with bipolar<br />

disorder (n=81) had mean age 37.2 ± 12.3 years; female<br />

49.9%. Study controls (n=125) had mean age 36.9 ± 14.1<br />

years, female 46.4%. Diagnosis used the Structured Clinical<br />

Interview for DSM-IV (SCID) version 2.0, clinical notes and a<br />

consensus opinion <strong>of</strong> at least 3 experts. Each participant gave<br />

30cc <strong>of</strong> whole blood for DNA extraction and to establish<br />

immortalized cell lines. The three single nucleotide<br />

polymorphisms (SNPs) from the Emamian haplotype, rs<br />

22498799, rs 3730358 and rs1130214, were genotyped and<br />

examined for association with bipolar disorder and<br />

neurocognition.None <strong>of</strong> the three SNPs was associated with<br />

bipolar disorder (all p-values >0.05). However, AKT1<br />

rs2498799, that was previously associated with schizophrenia<br />

risk (risk allele =G), significantly influenced cognitive<br />

performance among the bipolar sample but not among the<br />

control subjects. Specifically, bipolar patients who carry two<br />

copies <strong>of</strong> the risk allele (G) performed significantly worse on<br />

measures <strong>of</strong> pre-morbid IQ and working memory than did<br />

A-allele carriers. Conclusions/Significance: We found no<br />

association between AKT1 gene polymorphisms and bipolar<br />

disorder in our cohort. However, AKT1 gene appears to be<br />

associated with neurocognition in our sample <strong>of</strong> Caucasian<br />

bipolar patients.<br />

168 FUNCTIONAL ASSESSMENT OF S100B AS A<br />

SUSCEPTIBILITY GENE FOR BIPOLAR DISORDER<br />

E. Dagdan* (1), M. Hill (2), S. Roche (1, 3), P. McKeon (3,<br />

4)<br />

1. Smurfit Institute <strong>of</strong> Genetics, Trinity College, Dublin,<br />

Ireland 2. Neuropsychiatric Genetics Research Group,<br />

Institute <strong>of</strong> Molecular Medicine, Trinity Centre for Health<br />

Sciences, St. James' Hospital, Dublin, Ireland 3. Department<br />

<strong>of</strong> Psychiatry, Trinity College, Dublin, Ireland 4.St. Patrick's<br />

Hospital, Dublin, Ireland<br />

*elif.dagdan@yahoo.ie<br />

Introduction: The glial cell-derived neurotrophic factor,<br />

S100B, is implicated in the pathology <strong>of</strong> bipolar affective<br />

disorder (BPAD) and schizophrenia. S100B protein levels are<br />

elevated in serum <strong>of</strong> patients with both disorders and S100B<br />

variants are associated with schizophrenia. We previously<br />

reported association <strong>of</strong> a SNP in the promoter <strong>of</strong>S100B,<br />

rs3788266, with a psychotic form <strong>of</strong> BPAD (P=0.0088). The<br />

disease-associated C allele disrupts a Trex/MEF3 consensus<br />

recognition, which is bound by Six-family transcription<br />

factors, suggesting that it could affectS100B expression.<br />

Methods: The functional effect <strong>of</strong> rs3788266 onS100B<br />

promoter activity was determined using the luciferase reporter<br />

system. Promoter fragments containing the T or C alleles <strong>of</strong><br />

rs3788266 were subcloned into the pGL4.23 minimal<br />

promoter-luciferase vector and were assayed for activity in<br />

U373MG glioblastoma cells. S100B RNA and protein levels<br />

were measured in post-mortem brain tissue and serum,<br />

respectively, to test for possible genotypic effects in vivo. The<br />

allelic expression imbalance (AEI) <strong>of</strong> rs3788266 was also<br />

investigated in 41heterozygote post-mortem brain RNA<br />

samples. Finally, allelic effects on protein complex formation<br />

at theS100B promoter were investigated by an electrophoretic<br />

mobility shift assay (EMSA). Results: Luciferase reporter<br />

gene expression was significantly increased in the presence <strong>of</strong><br />

the T- compared to C-allele (t= 4.151, P=0.001). However,<br />

lower mean serumS100B levels were observed in BPAD<br />

individuals with the TT genotype compared to those with the<br />

TC or CC genotypes (ANOVA: F=13.44, P=0.001). A similar<br />

pattern was observed at the RNA level but was not significant.<br />

The binding affinity <strong>of</strong> a U373MG protein complex that binds<br />

to the S100B promoter was 40 % stronger on the T- compared<br />

C-allele promoter fragments. Allelic expression imbalance<br />

was not observed with rs3788266, but we are investigating<br />

other variants. Discussion: The disease-associated C allele <strong>of</strong><br />

rs3788266 is associated with reduced promoter activity in<br />

U373MG glial cells and increased protein levels in serum.<br />

SNP rs3788266 may represent a functional susceptibility<br />

variant that contributes to the increased S100B levels observed<br />

in BPAD patients. We are investigating the effect <strong>of</strong> other<br />

SNPs on S100B RNA and serum levels.


169 GENOME-WIDE ASSOCIATION OF<br />

SELF-REPORTED CO-MORBID SEIZURES IN<br />

INDIVIDUALS WITH BIPOLAR DISORDER<br />

A. Schork* (1, 2), C. Bloss (2), E. Smith (2), J. Kelsoe (1), N.<br />

Schork (2, 3)<br />

1. <strong>UCSD</strong> 2. Scripps Translational Science Institute 3. The<br />

Scripps Research Institute<br />

*aschork@ucsd.edu<br />

Individuals diagnosed with affective disorders exhibit a higher<br />

prevalence <strong>of</strong> seizures than the general population. Further,<br />

there is considerable overlap in drug treatments for both<br />

conditions, with anti-convulsants <strong>of</strong>ten used to prevent mood<br />

swings in individuals with bipolar disorder (BD). These<br />

observations, together with evidence that both seizures and<br />

BD are closely related to noradrenergic and serotonergic<br />

pathways in the human brain, led us to conduct a<br />

genome-wide association study (GWAS) <strong>of</strong> co-morbid<br />

seizures in two samples <strong>of</strong> individuals with BD. Our initial test<br />

<strong>of</strong> association was performed in a sample <strong>of</strong> 1,001 individuals<br />

with BD (n=82 with co-morbid seizures) genotyped through<br />

the Genetic Association Information Network (GAIN) Bipolar<br />

Genetics Study (BiGS). This sample was then combined with<br />

1,199 additional cases (n=93 with co-morbid seizures) from<br />

the Translational Genetics subsample <strong>of</strong> BiGS. Standard<br />

univariate association analyses were performed using PLINK.<br />

The strongest statistical signal from the combined sample in<br />

this BD case-only association analysis was found for a SNP<br />

between PHF14 and THSD7A on chromosome 7 (4.01 x 10-9;<br />

OR = 5.276), a region that has been previously implicated in<br />

cerebellar ataxia. Secondary analyses have provided further<br />

support for this association, although further replication is<br />

needed, particularly in cohorts where the seizure phenotype<br />

has been more rigorously defined. Overall, our findings<br />

suggest that co-morbid seizures and BD may represent a more<br />

homogenous form <strong>of</strong> neuropsychiatric illness, with identifiable<br />

genetic underpinnings. It is possible that GWAS applied to<br />

clinically homogenous sub-groups may have greater utility for<br />

gene identification in complex disorders.<br />

170 ASSOCIATION OF THE SEROTONIN<br />

TRANSPORTER GENE (SLC6A4) POLYMORPHISMS<br />

WITH COMMON MENTAL DISORDERS IN A<br />

BRAZILIAN POPULATION COHORT<br />

U. Paredes* (1, 2), M. Diniz (1, 2, 3), A. de Saloma (1, 2, 3),<br />

X. Xu (1, 2), C. Guindallini (3), J. Mari (3), R.<br />

Affonseca-Bressan (3), P. McKeigue (4), . Prince (2), C.<br />

Lewis (2), G. Breen (1, 2)<br />

1. SGDP 2. IoP King's college London 3. UNIFESP 4.<br />

University <strong>of</strong> Edinburgh<br />

*ursula.paredes@kcl.ac.uk<br />

We present preliminary results from genetic study using a<br />

unique Brazilian cohort (from Sao Paulo [SP] and Rio de<br />

Janeiro [RJ]) to unveil the complex genetic and environmental<br />

aetiology <strong>of</strong> Common Mental Disorders (CMDs). These states<br />

were selected as it has been demonstrated previously that<br />

SP&RJ are stressful environments with high violent crime and<br />

inequality and that the populations are highly admixed. For<br />

this purpose we collected DNA from a cohort <strong>of</strong> 2200 subjects<br />

selected via a cross-sectional survey with a multistage<br />

probability sample design used to draw a representative<br />

sample in the cities <strong>of</strong> SP and RJ. The cohort was then<br />

assessed with the Composite International Diagnostic<br />

Interview version 2.1 and 9 other scales including the General<br />

Health Questionaire, Social Capital scale, and the Risk and<br />

Resilience Factor Questionnaire. We conducted a preliminary<br />

analysis <strong>of</strong> the prevalence <strong>of</strong> serotonin transporter gene<br />

(SLC6A4) promoter, intron 2 and Ile 425 polymorphisms as<br />

variants <strong>of</strong> these markers has been previously shown to act as<br />

a risk factors for a variety <strong>of</strong> CMDs e.g. depression, PTSD,<br />

etc.


283 CLASSIFICATION OF BEHAVIORS DURING<br />

MANIA: A NOVEL APPROACH TO BIPOLAR<br />

GENETICS<br />

R. McKinney* (1), T. Greenwood (1), J. Head (1), J. Cadigan<br />

(1), B. Study (2), Bipolar Genome Study (BiGS), J. Kelsoe (1)<br />

1. <strong>UCSD</strong> Dept <strong>of</strong> Psychiatry<br />

* rmckinney@ucsd.edu<br />

Although the high rate <strong>of</strong> heritability in bipolar disorder<br />

(BPD) is well demonstrated, the search for specific<br />

susceptibility genes has been challenging. It is likely these<br />

mapping difficulties are in large part the result <strong>of</strong> a high level<br />

<strong>of</strong> genetic and allelic heterogeneity. BPD may result from<br />

genetic variants in a variety <strong>of</strong> different brain signaling<br />

systems. Therefore, any clinical or other trait that could<br />

distinguish these variants would be invaluable in reducing<br />

heterogeneity. We describe a novel, strictly behavioral<br />

approach based on impairing actions displayed during manic<br />

and mixed states. Information was collected regarding these<br />

behaviors using DIGS ratings from our bipolar GWAS study<br />

(BiGS). Clinicians independently coded behaviors for 935<br />

subjects to determine whether each <strong>of</strong> 22 behavioral categories<br />

were present. Multiple correspondence analysis was used with<br />

564 unrelated individuals to explore and reduce the<br />

dimensionality <strong>of</strong> the data, with six dimensions explaining<br />

roughly 39% <strong>of</strong> the variance. We then used SOLAR to assess<br />

the heritability <strong>of</strong> both individual behaviors and the<br />

dimensions in a set <strong>of</strong> 356 affected sibling groups. Of<br />

individual behaviors, several displayed significance, including<br />

compulsive (h2=0.85, p=2x10-6) and sexual (h2=0.89,<br />

p=5x10-9) behaviors. In addition, we have taken a<br />

preliminary look at genetic association as well as developed a<br />

new self report questionnaire assessing manic behaviors.<br />

These data support the concept that the types <strong>of</strong> behaviors<br />

displayed during mania may be heritable and index different<br />

forms <strong>of</strong> BPD. This approach may then be useful in<br />

identifying susceptibility genes, underlying pathophysiology<br />

and clinical applications.<br />

284 BDNF MET-CARRIERS SHOW WHITE MATTER<br />

ABNORMALITIES IN RECURRENT DEPRESSION<br />

J. Cole* (1), C. Fu (1), A. Farmer (1), S. Cohen-Woods (1),<br />

D. Gaysina (1), N. Craddock (2), J. Gray (1), C. Gunasinghe<br />

(1), F. Hoda (1), L. Jones (3), J. Knight (1), A. Korszun (4),<br />

M. Owen (2), A. Sterne (1), I. Craig (1), S. Williams (5), A.<br />

Simmons (5), P. McGuffin (1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London 2. Department <strong>of</strong> Psychological Medicine, Cardiff<br />

University, School <strong>of</strong> Medicine 3. Division <strong>of</strong> Neuroscience,<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Birmingham 4.<br />

Centre for Psychiatry, Wolfson Institute <strong>of</strong> Preventive<br />

Medicine, Barts and The London 5. Centre for Neuroimaging<br />

Sciences, Institute <strong>of</strong> Psychiatry, King's College London<br />

*james.cole@iop.kcl.ac.uk<br />

Background: White matter abnormalities have previously been<br />

reported in both young and elderly patients with major<br />

depressive disorder (MDD), however little is known about<br />

genetic influences on white matter in MDD. Brain derived<br />

neurotrophic factor is known to play a role in neuronal<br />

survival and differentiation and carrying the met allele <strong>of</strong> the<br />

val66met SNP has been shown to confer greater number <strong>of</strong><br />

white matter hyperintensities in MDD.<br />

Objective: To investigate whether a plausible risk genotype,<br />

BNDF val66met, has an effect on white matter integrity in<br />

depression, using diffusion-tensor imaging techniques.<br />

Methods: Diffusion-weighted MR scans were obtained from<br />

49 recurrent MDD patients (mean age = 50.1 yrs, 31 females,<br />

18 males). These scans were then grouped by val66met<br />

genotype (19 met carriers versus 30 val/val homozygotes) and<br />

underwent a voxel-based group mapping analysis <strong>of</strong> fractional<br />

anisotropy (FA). The two comparison groups were matched<br />

for age, sex, IQ and current depression levels.<br />

Results: Met-allele carriers have significantly decreased FA<br />

scores in four large voxel clusters throughout the brain,<br />

compared to val/val homozygotes. These clusters were located<br />

in bilateral corona radiata regions and the corticospinal tract.<br />

Conclusions: The study indicates that having at least one met<br />

allele in codon 66 <strong>of</strong> the BDNF gene is associated with widely<br />

distributed decreases in white matter microstructural integrity<br />

in patients with major depressive disorder.


OTHER CHILDHOOD DISORDERS<br />

171 READING DISABILITIES AND ADHD:<br />

CANDIDATE GENES ON CHR.6<br />

A. Elbert* (1), L. Gomez, T. Cate-Carter (2), K. Wigg, Y.<br />

Feng, A. Pitch, A. Ickovicz, M. Malone, E. Kerr, M. Lovett<br />

(2), C. Barr (3)<br />

1. University <strong>of</strong> Toronto: Institute <strong>of</strong> <strong>Medical</strong> Sciences 2.<br />

Sickkids Hospital 3. University <strong>of</strong> Toronto: Dept <strong>of</strong> Psychiatry<br />

*adrienne.elbert@ymail.com<br />

Reading Disabilities (RD) is the most common learning<br />

disability, affecting 5-12% <strong>of</strong> children. ADHD is <strong>of</strong>ten<br />

comorbid with RD and twin studies indicate common genetic<br />

influences. Previous linkage studies have identified<br />

chromosome 6p as a risk locus, and two genes in this region,<br />

KIAA0319 and DCDC2, have been associated to RD in<br />

multiple samples. In a prior study, we mapped the putative<br />

regulatory elements <strong>of</strong> KIAA0319 and DCDC2 using ChIP on<br />

chip. The purpose <strong>of</strong> the current study was to investigate<br />

polymorphisms within these regulatory elements in order to<br />

determine whether they contribute to risk for RD and ADHD.<br />

Eight polymorphisms in the regulatory element <strong>of</strong> KIAA0319<br />

were tested for association in a sample <strong>of</strong> 291 nuclear families<br />

with RD-affected children. Of these polymorphisms, a<br />

microsatellite ([GT]n repeat) in the 1st exon <strong>of</strong> KIAA0319<br />

showed our most significant result for association to RD in<br />

this gene (p=0.009). It is established that longer (GT)n alleles<br />

can decrease gene transcription by forming z-DNA, however it<br />

is unclear how the microsatellite in KIAA0319 may be<br />

contributing to RD risk. We previously found no evidence for<br />

association <strong>of</strong> DCDC2 markers to our RD sample. However,<br />

we recently found that these markers in DCDC2 were<br />

associated with measures <strong>of</strong> hyperactivity and inattention in<br />

the RD and ADHD samples. Therefore, I investigated the only<br />

polymorphism in the putative regulatory element <strong>of</strong> DCDC2<br />

in our RD and ADHD samples. The results did not provide<br />

significant evidence for association to ADHD (p=0.07) or RD<br />

(p=0.89).<br />

172 STUDY OF THE<br />

CATECHOL-O-METHYL-TRANSFERASE (COMT)<br />

GENE WITH HIGH AGGRESSION IN CHILDREN<br />

Y. Hirata* (1), N. King (1), B. Nowrouzi (2), J. Beitchman<br />

(2), J. Kennedy (1)<br />

1. Neurogenetics Section, Centre for Addiction and Mental<br />

Health, the Departments <strong>of</strong> Psychiatry, the University <strong>of</strong><br />

Toronto, Toronto, ON, Canada 2. Child Psychiatry Section,<br />

Centre for Addiction and Mental Health, the Departments <strong>of</strong><br />

Psychiatry, the University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

*yuko_hirata@camh.net<br />

Background: The etiology <strong>of</strong> Childhood Onset Aggression<br />

(COA) is still poorly understood, but early COA can be<br />

considered as a strong risk factor for adult delinquency or<br />

criminal behaviour. The COMT gene has been suggested to be<br />

associated with aggression, as well as other psychiatric<br />

disturbances such as schizophrenia, depression, suicide, and<br />

attention deficit/hyperactivity disorder (ADHD). Methods:<br />

168 children participated, who were required to have scores<br />

ator exceeding the 90th percentile on the aggression subscale<br />

<strong>of</strong> the parent-reported Child Behavior Checklist and also on<br />

the Teacher's Report Form. The subjects must have shown a<br />

minimum two-year history <strong>of</strong> aggressive behaviour. Four<br />

COMT markers were genotyped for cases and controls<br />

matched for gender and ethnicity. The cases were also divided<br />

into subgroups using the Callous-Unemotional (CU) scale, as<br />

well as ADHD status. Results: The four SNPs defined one LD<br />

block. The genotype analysis <strong>of</strong> rs6269 (Val158Met) showed<br />

nominally significant association (p=0.025) and rs4818<br />

showed a trend (p=0.091). Haplotype analyses showed no<br />

significant association. High versus low CU score groups<br />

showed nominally significant association with rs6269<br />

(p=0.019) and a trend with rs4818 (p=0.086). ADHD groups<br />

showed no significant results. Conclusions: This is the first<br />

report to our knowledge evaluating COMT SNPs with the<br />

phenotype <strong>of</strong> high aggression in children, and we found a<br />

possible role for the Val158Met marker in COA. Given the<br />

importance <strong>of</strong> COA in future behaviour problems, further<br />

investigation <strong>of</strong> COMT in this disorder is warranted.


173 A NOVEL LOCUS FOR AUTOSOMAL RECESSIVE<br />

NON-SYNDROMIC MENTAL RENTARDATION<br />

(NSMR) MAPS TO 13Q12.3-Q13.2.<br />

A. Noor* (1), A. Mir (2), A. Mikhailov (1), A. Fennell (1), M.<br />

Ayub (3), J. Vincent (1)<br />

1. CAMH 2. COMSATS Inst, Islamabad 3. St. Lukes<br />

Hospital, UK<br />

*abdul_noor@camh.net<br />

Mental retardation (MR) is defined by an intelligence quotient<br />

(IQ) <strong>of</strong> less than 70 associated with functional deficits in<br />

adaptive behavior, and has a prevalence <strong>of</strong> 1–3% in the<br />

population. Although non syndromal autosomal recessive<br />

forms <strong>of</strong> MR (NS-ARMR) are believed to be relatively more<br />

common, only five genes have been reported so far. The<br />

objective <strong>of</strong> the present study was to identify a new locus\gene<br />

for ARMR. We ascertained a consanguineous family affected<br />

with non-syndromic autosomal recessive mental retardation.<br />

The phenotype was present in 4 individuals from two branches<br />

<strong>of</strong> the family. To map the chromosomal location <strong>of</strong> the<br />

causative gene we undertook Affymetrix 5.0 gene chip SNP<br />

analyses <strong>of</strong> all affected individuals and two unaffected<br />

individuals, assuming that a founder mutation was<br />

responsible. All affected individuals shared a 4.9 Mb<br />

haplo-identical region <strong>of</strong> homozygosity located on<br />

chromosome 13q12.3-q13.2, defined by SNP markers<br />

rs9506126 and rs9598929. The physical location <strong>of</strong> the critical<br />

region is 28,727 Mb to 33,621 Mb (UCSC, March 2006) and it<br />

contains ~30 Refseq genes. We have identified several<br />

candidate genes within this locus including UBL3,<br />

KATNAL1, FRY, PDS5B and KIAA0774. Sequencing <strong>of</strong><br />

candidate genes to identify the causative mutation is in<br />

process. In summary, we have mapped the chromosomal<br />

location <strong>of</strong> a novel gene responsible for autosomal recessive<br />

non-syndromic mental retardation.<br />

174 FIRST GENOME-SCAN ON<br />

NEUROPHYSIOLOGICAL ENDOPHENOTYPES<br />

POINTS TO TRANS-REGULATION EFFECTS ON<br />

SLC2A3 IN DYSLEXIC CHILDREN<br />

D. Roeske* (1), K. Ludwig (2), N. Neuh<strong>of</strong>f (3), J. Becker (2),<br />

J. Bartling (3), J. Bruder (3), F. Brockschmidt (2), A. Warnke<br />

(4), H. Remschmidt (5), P. H<strong>of</strong>fmann (6), M. Nöthen (6), G.<br />

Schulte-Körne (3), B. Müller-Myhsok (1)<br />

1. Max-Planck Institute <strong>of</strong> Psychiatry, Munich, Germany 2.<br />

Department <strong>of</strong> Genomics, Life & Brain Center, University <strong>of</strong><br />

Bonn, Germany 3. Department <strong>of</strong> Child and Adolescent<br />

Psychiatry, Psychosomatics and Psychotherapy,<br />

Ludwig-Maximilians-University Munich, Germany 4.<br />

Department <strong>of</strong> Child and Adolescent Psychiatry and<br />

Psychotherapy, University <strong>of</strong> Würzburg, Germany 5.<br />

Department <strong>of</strong> Child and Adolescent Psychiatry and<br />

Psychotherapy, University <strong>of</strong> Marburg, Germany 6. Institute<br />

<strong>of</strong> Human Genetics, University <strong>of</strong> Bonn, Germany<br />

*darina@mpipsykl.mpg.de<br />

We present the results <strong>of</strong> a genome-wide association study on<br />

EEG-measurements in dyslexic children. In 200 dyslexic<br />

children, genotypes <strong>of</strong> 300.000 SNPs were associated with<br />

two mismatch negativity (MMN) phenotypes, an early and a<br />

late MMN component which have been shown to be<br />

endophenotypes for dyslexia. 19 top SNPs were taken into<br />

replication in a second German sample <strong>of</strong> 186 dyslexic<br />

children. Rs4234898 could be replicated in this sample, with a<br />

genome-wide significant result in the combined sample. This<br />

SNP is located in an intergenic region on chromosome 4. In a<br />

public available database (Dixon 2007) rs4234898 is<br />

significantly associated with gene expression levels <strong>of</strong><br />

SLC2A3 with lower expression levels being present in<br />

individuals carrying the T-allele. We confirmed this<br />

trans-regulational effect in a cell line experiment using<br />

lymphoblastoid cell lines from dyslexic children. This is in<br />

concordance to our association results where carriers <strong>of</strong> the<br />

T-allele showed an attenuated MMN. SLC2A3 is the main<br />

glucose transporter in neurons. Our findings suggest that an<br />

attenuated MMN in dyslexic children might be due to reduced<br />

glucose levels. This is strengthened by several studies where<br />

hypoglycaemia has been linked to deficits in learning and<br />

memory. We find no evidence for a trans-regulation <strong>of</strong><br />

rs4234898 on SLC2A3 in gene expression data from adults.<br />

This suggests that the identified effect might be restricted to<br />

human childhood when a higher glucose demand is needed,<br />

e.g. for cell migration processes such as elongating <strong>of</strong> axons or<br />

branching <strong>of</strong> synapses. Acknowledgements GSK, BMM,<br />

MMN were funded by the EU in the Sixth Framework<br />

Program, LifeScienceHealth, project title Dyslexia genes and<br />

neurobiological pathways (Neurodys, 018696)


PERSONALITY AND TEMPERAMENT<br />

175 TRUE HEALING ART OF ALZHEIMER'S<br />

DISEASE BY HOLISTIC HOMOEOPATHY<br />

D. Patel*<br />

*dr_darshanapatel@hotmail.com<br />

Homoeopathic theory: The highest ideal <strong>of</strong> cure is rapid,<br />

gentle and permanent restoration <strong>of</strong> health, or removal and<br />

annihilation <strong>of</strong> the disease in its whole extent, in the shortest,<br />

most reliable, and harmless way, on easily comprehensible<br />

principle. Homoeopathy <strong>of</strong>fers reasonably positive treatment<br />

not cure but control and relief. Timely Administered<br />

homoeopathy medicines can prevent further progress <strong>of</strong><br />

disease. The homoeopathic remedies work by stimulating the<br />

body's own healing power. Homoeopathy the holistic<br />

approach believes in treating the patient who is diseased and<br />

not merely diseased parts <strong>of</strong> the patients. Every person is<br />

unique and dementia affects people differently no two people<br />

will follow exactly the same course and the homoeopathy the<br />

ultimate pathy in the world believes in individuality <strong>of</strong> the<br />

person has been shown to have a positive impact on the<br />

progress <strong>of</strong> the disease. Homoeopathy undertakes study <strong>of</strong><br />

Each Alzheimer's as separate entity. The study also involves<br />

deep evaluation <strong>of</strong> the mind and emotions & after that a<br />

suitable medicine, called as constitutional medicine is given.<br />

The medication brings deeper level healing. It should be noted<br />

that there is no single specific remedy for all the cases <strong>of</strong><br />

Alzheimer's. The exact treatment is determined only on<br />

in-depth evaluation <strong>of</strong> individual case.<br />

176 INFLUENCE OF HTR2A POLYMORPHISMS AND<br />

PARENTAL REARING ON THE PERSONALITY<br />

TRAITS IN HEALTHY JAPANESE<br />

Y. Nakamura* (1), Y. Ito (1), B. Aleksic (1), I. Kushima (1),<br />

N. Yasui-Furukori (2), T. Inada (3), Y. Ono (4), N. Ozaki (1)<br />

1. Department <strong>of</strong> Psychiatry, Nagoya University Graduate<br />

School <strong>of</strong> Medicine, Nagoya, Japan 2. Department <strong>of</strong><br />

Neuropsychiatry, Graduate School <strong>of</strong> Medicine, Hirosaki<br />

University, Hirosaki, Japan 3. Seiwa Hospital, Institute <strong>of</strong><br />

Neuropsychiatry,Tokyo, Japan 4. Health Center, Keio<br />

University, Tokyo, Japan<br />

*y-nakamu@med.nagoya-u.ac.jp<br />

Introduction: Serotonin plays a significant role in the<br />

biological basis <strong>of</strong> human behavior, psychiatric diseases and<br />

temperamental predisposition. Genetic factors and<br />

environmental influences significantly contribute to the<br />

determination <strong>of</strong> human personality traits. The main purpose<br />

<strong>of</strong> this study was to examine an influence <strong>of</strong> HTR2A<br />

polymorphisms and parental rearing on temperament in<br />

healthy Japanese. Methods: The subjects were 1,254 Japanese<br />

volunteers (male=597, female=657), and 3 SNPs (rs6311,<br />

rs6313, rs643627) were selected for genotyping. Out <strong>of</strong> 1,254<br />

subjects, 1,245 completed the 125 items <strong>of</strong> the Japanese short<br />

version <strong>of</strong> the Temperament and Character Inventory, and 572<br />

completed the Japanese version <strong>of</strong> the Parental Bonding<br />

Instrument. Result: All SNPs were in Hardy-Weinberg<br />

equilibrium. As for female subjects a significant association<br />

(p=0.003) was observed between rs643627 and novelty<br />

seeking (NS). On the other hand, maternal overprotection had<br />

significant effect on the score <strong>of</strong> harm avoidance (HA) in male<br />

subjects only (p=0.049). Conclusion: Our result indicated the<br />

relationship between the HTR2A rs643627 polymorphism and<br />

NS only in females. The interplay between HTR2A receptor<br />

and sex hormone might be responsible for the differences in<br />

temperament between males and females. The results <strong>of</strong> our<br />

study indicated that that maternal overprotection has more<br />

influence than genetic factors on HA in male subjects. We<br />

speculate that in males, the effects <strong>of</strong> genetic polymorphisms<br />

on personality traits might be nullified by parental rearing.


177 ASSOCIATION BETWEEN OXYTOCIN<br />

RECEPTOR GENE (OXTR) AND AFFECTIVE<br />

TEMPERAMENTS<br />

Y. Kawamura* (1), X. Liu (2), T. Akiyama (3), T. Shimada<br />

(1), T. Otowa (1), Y. Sakai (4), C. Kakiuchi (1), T. Umekage<br />

(5), T. Sasaki (6)<br />

1. Department <strong>of</strong> Neuropsychiatry, Graduate School <strong>of</strong><br />

Medicine, University <strong>of</strong> Tokyo 2. Department <strong>of</strong> Human<br />

Genetics, Graduate School <strong>of</strong> Medicine, University <strong>of</strong> Tokyo<br />

3. Department <strong>of</strong> Psychiatry, Kanto <strong>Medical</strong> Center NTT EC<br />

4. Department <strong>of</strong> Psychiatry, Juntendo University School <strong>of</strong><br />

Medicine 5. Division for Environment, Health and Safety,<br />

University <strong>of</strong> Tokyo 6. Office for Mental Health Support,<br />

University <strong>of</strong> Tokyo<br />

*yoshiya-tky@umin.ac.jp<br />

Background: Oxytocin is known to be related to social<br />

interaction, trust and affectivity. Affective temperaments are<br />

traits based on Kraepelin’s typological definition <strong>of</strong><br />

‘‘fundamental states’’ <strong>of</strong> manic depressive illness. They can<br />

be measured by the Temperament Evaluation <strong>of</strong> Memphis,<br />

Pisa, Paris and San Diego-Autoquestionnaire version<br />

(TEMPS-A), and it was confirmed that they had a good<br />

enough long-term stability to be considered as<br />

endophenotypes. The objective is to assess the association<br />

between oxytocin receptor gene (OXTR) polymorphism and<br />

affective temperaments. Methods: Firstly, fifteen OXTR tag<br />

single nucleotide polymorphisms (SNPs) were genotyped<br />

using TaqMan® or direct sequencing. Secondly, haplotype<br />

block structure was determined by Haploview 4.1. Lastly, the<br />

association between SNPs and affective temperaments<br />

(depressive, cyclothymic, hyperthymic, irritable and anxious<br />

temperaments) was assessed by quantitative trait association<br />

analysis permuted 10,000 times, and the association between<br />

haplotypes and the temperaments was assessed by<br />

haplotype-based quantitative trait association analysis with<br />

Bonferroni correction, using PLINK 1.06 for 493 Japanese as<br />

screened to exclude those who had lifetime diagnoses <strong>of</strong><br />

schizophrenia and other psychotic disorders by the<br />

Mini-International Neuropsychiatric Interview (MINI).<br />

Results: Two haplotype blocks were identified on OXTR.<br />

Depressive temperament was significantly associated with<br />

SNP rs11131149, rs13316193 (empirical p < 0.05) and one<br />

common haplotype CTGTGGG<br />

(rs11131149/rs2243370/rs2243369/rs13316193/rs2254298/rs2<br />

268493/rs2268491) (adjusted p < 0.01). Cyclothymic<br />

temperament was significantly associated with rs2268495<br />

(empirical p < 0.05). Irritable temperament was significantly<br />

associated with rs2268495 (empirical p < 0.01) and rs1042778<br />

(empirical p < 0.05). Conclusions: The findings suggest that<br />

OXTR polymorphisms or its haplotype are associated with<br />

affective temperaments.<br />

178 POPULATION GENETIC ANALYSIS AND<br />

ASSOCIATION STUDY OF THE<br />

CHOLECYSTOKININ-B RECEPTOR GENE WITH<br />

PERSONALITY TRAITS<br />

P. Zoë* (1), J. Bradwejn (2), J. Kennedy (3), X. Xia (4), D.<br />

Koszycki (5)<br />

1. Institute <strong>of</strong> Mental Health Research, Royal Ottawa Mental<br />

Health Centre, Ottawa, Canada 2. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Ottawa, Ottawa, Canada 3. Neurogenetics<br />

Section, Clarke Division, Centre for Addiction and Mental<br />

Health, University <strong>of</strong> Toronto, Toronto, Ontario, Canada 4.<br />

Centre for Advanced Research in Environmental Genomics,<br />

University <strong>of</strong> Ottawa, Ottawa, Canada. 5. University <strong>of</strong><br />

Ottawa, Institute <strong>of</strong> Mental Health Research, Royal Ottawa<br />

Mental Health Centre, Ottawa, Canada<br />

*zoe.prichard@rohcg.on.ca<br />

A polymorphism in the cholecystokinin-receptor gene<br />

(CCKBR) has been associated with panic disorder (PD). This<br />

study attempted to associate the same polymorphism with<br />

personality traits that are postulated to increase vulnerability<br />

to PD. Healthy volunteers completed the Anxiety Sensitivity<br />

Index (ASI) and the NEO-Personality Inventory-Revised<br />

(NEO-PI-R). No association was observed between anxiety<br />

sensitivity or any personality trait with alleles previously<br />

implicated in PD. Unexpectedly, Population genetics analysis<br />

revealed the Caucasian sample (n = 103) to be in<br />

Hardy-Weinberg Disequilibrium (HWD) in a sex-specific<br />

pattern. Although there were no significant differences in<br />

allele frequencies between the sexes, significant differences in<br />

genotype frequencies were observed between the sexes. The<br />

sample was also found to deviate from predictions under the<br />

neutral-equilibrium and stepwise mutation models <strong>of</strong><br />

molecular evolution. The same genotypes implicated in the<br />

deviation from Hardy Weinberg Equilibrium (HWE) were also<br />

found to be associated with scoring low for the personality<br />

traits <strong>of</strong> Openness and Conscientiousness. Interestingly,<br />

Openness and Conscientiousness are not personality traits<br />

predicted to play a role in the etiology <strong>of</strong> PD. Although this<br />

study failed to detect the predicted relationship between<br />

polymorphism and personality traits, the results are interesting<br />

because the findings <strong>of</strong>fer preliminary evidence for selection<br />

at the CCKBR gene. The method <strong>of</strong> conducting association<br />

analysis by first partitioning genotypes according to departure<br />

from HWE is novel, and is a possible method for<br />

understanding the dynamics <strong>of</strong> a population subjected to<br />

forces that affect assumptions <strong>of</strong> evolutionary neutrality.


179 CANDIDATE GENE STUDY OF SCHIZOTYPY IN<br />

NONPSYCHOTIC RELATIVES IN 270 IRISH<br />

HIGH-DENSITY SCHIZOPHRENIA FAMILIES<br />

A. Fanous* (1), D. Thiselton (2), V. Vladimirov (2), R.<br />

Amdur (1), F. O'Neill (3), D. Walsh (4), B. Riley (2), X. Chen<br />

(2), K. Kendler (2)<br />

1. Washington VA <strong>Medical</strong> Center 2. Virginia Commonwealth<br />

University 3. Queen's University, Belfast 4. Health Research<br />

Board, Dublin<br />

*ahfanous@vcu.edu<br />

Background: It has long been observed that the relatives <strong>of</strong><br />

schizophrenics, even if not psychotic themselves, have odd<br />

personality traits. These have been termed schizotypy, which<br />

may be an endophenotype. It may include magical thinking,<br />

ideas <strong>of</strong> reference, odd speech and behavior, and social<br />

isolation. Candidate gene studies <strong>of</strong> schizotypy have only<br />

been published since 2004. Furthermore, all <strong>of</strong> these previous<br />

studies have used population, rather than family-based<br />

samples. Methods: In our previous work in the Irish Study <strong>of</strong><br />

High-Density Schizophrenia families (n=270 families and<br />

1408 individuals), we have reported positive associations in<br />

DTNBP1, COMT, RGS4, Akt1, TAAR6, SPEC2, PDZ-GEF2,<br />

ACSL6, IL3, CSFR2B, MEGF10, and FBXL21. We<br />

hypothesized that at least some <strong>of</strong> these genes would contain<br />

SNPs that influence schizotypy in non-psychotic relatives in<br />

this sample. We tested schizotypy as a quantitative trait using<br />

122 SNPs in these genes, previously tested for association<br />

with schizophrenia, using the UNPHASED package. The<br />

schizotypy trait consisted <strong>of</strong> the sum <strong>of</strong> ratings on all <strong>of</strong> the<br />

individual items <strong>of</strong> the DSM-III-R criteria for Schizotypal<br />

Personality Disorder. Results: We observed 4 contiguous<br />

SNPs in MEGF10 with P


181 RELATIONSHIP BETWEEN D2 and D4<br />

DOPAMINE RECEPTOR POLYMORPHISMS, STRESS<br />

AND THE PERSONALITY TRAITS OF<br />

EXTRAVERSION AND PSYCHOTICISM IN SONS OF<br />

ALCOHOLICS<br />

T. Ozkaragoz* (1), E. Noble (1)<br />

1. UCLA<br />

*tulino@ucla.edu<br />

The influence <strong>of</strong> both molecular genetic (D2 and D4 dopamine<br />

receptor polymorphisms) and environmental factors (i.e.<br />

stress) on the personality traits <strong>of</strong> Psychoticism, Extraversion<br />

and Neuroticism as measured by the Jr. Eysenck Personality<br />

Questionnaire was investigated. High scorers on the<br />

Psychoticism scale are described as impulsive, unempathic,<br />

aggressive and sensation-seeking. Participants were 49 12-17<br />

year old sons <strong>of</strong> <strong>of</strong> either active or recovering alcoholic<br />

parents. It was found that sons <strong>of</strong> alcoholics (SAs) with the<br />

DRD2 A1+ allele (A1/A1 and A1/A2 genotypes) showed<br />

higher Psychoticism and Extraversion scores than SAs with<br />

the DRD2 A1- allele (A2/A2 genotype). There also was a<br />

positive relationship between stress and Psychoticism.<br />

Furthermore, an interaction between the D4 dopamine<br />

receptor gene and stress was found. Specifically, the<br />

Psychoticism scores <strong>of</strong> SAs with the DRD4 7R+ allele (7R/7R<br />

and 7R/Non7R genotypes) were more strongly influenced by<br />

stress than the scores <strong>of</strong> SAs' with the DRD4 7R- allele<br />

(Non7R/Non7R genotype) on this scale. A negative<br />

relationship between age and Neuroticism and a positive<br />

relationship between stress and Neuroticism also was found.<br />

The results <strong>of</strong> this study suggest that the A1+ allele <strong>of</strong> the D2<br />

dopamine receptor gene influences the expression <strong>of</strong><br />

Psychoticism and Extraversion in adolescent sons <strong>of</strong><br />

alcoholics. In addition, both the 7R+ allele <strong>of</strong> the D4<br />

dopamine receptor gene and level <strong>of</strong> stress also appear to<br />

contribute to the expression <strong>of</strong> Psychoticism.


PHARMACOGENETICS<br />

182 AMINE RESISTANCE" IN BRAIN REWARD<br />

CIRCUITRY AS A FUNCTION OF DRD2 GENE<br />

POLYMORPHISMS IN REWARD DEFICIENCY<br />

SYNDROME (RDS): SYNAPTAMINE COMPLEX<br />

VARIANT INDUCED "DOPAMINE SENSITIVITY"<br />

AND ENHANCEMENT OF HAPPINESS<br />

K. Blum* (1, 2, 3), E. Stice (4), B. Downs (2), R. Waite (2),<br />

E. Braverman (3, 5), M. Kerner (3), M. Oscar-Berman (6)<br />

1. Department <strong>of</strong> Physiology and Pharmacology, Wake Forest<br />

Unversity; Winston-Salem, North Carolina 2. Department <strong>of</strong><br />

Nutrigenomics, LifeGen, Inc.; San Diego, California 3. PATH<br />

Clinics, New York, New York 4. Oregon Research Institute;<br />

Eugene, Oregon 5. Department <strong>of</strong> Neurological Surgery, Weill<br />

Cornell College <strong>of</strong> Medicine; New York, N.Y. 6. Departments<br />

<strong>of</strong> Psychiatry and Anatomy & Neurobiology, Boston<br />

University School <strong>of</strong> Medicine; and Boston VA Healthcare<br />

System, Boston, MA<br />

*drd2gene@aol.com<br />

Since the discovery <strong>of</strong> the double helix, brain function in<br />

terms <strong>of</strong> both physiology and behavioral traits have resulted in<br />

a plethora <strong>of</strong> studies linking these activities to<br />

neurotransmitter functions having a genetic basis. The<br />

mechanisms underlining gene expression and the potential<br />

impairments due to polygenic inheritance -- and as such,<br />

predisposition to addiction and self-destructive behaviors --<br />

have been amply identified. Human genetic information<br />

derived from these scientific explorations may have important<br />

links to feelings <strong>of</strong> well-being and potentially phenomena<br />

related to “happiness.” While non-genetic oriented biological<br />

explorations <strong>of</strong> social and political research have addressed<br />

the impact <strong>of</strong> social and institutional environments on human<br />

attitudes and behaviors, there is a paucity <strong>of</strong> research<br />

exploring gene-environmental influences on these parameters.<br />

Moreover, traditional fields such as Psychology and Molecular<br />

Biology are subject to inherent limitations that may be<br />

resolved only through collaboration across disciplines. By<br />

contrast, attempts to identify key "vector influences" that link<br />

genes, the brain, and social behaviors to a so-called state <strong>of</strong><br />

"happiness" are important areas for developing a new science<br />

<strong>of</strong> human nature. Two disciplines, one relating to<br />

spirituality("Genospirtuality") and the other to Political<br />

Science, are beginning to emerge as fruitful grounds for<br />

identification <strong>of</strong> specific polymorphic gene associations and<br />

may pave the way to advance this new science. We address<br />

the age-old question <strong>of</strong> "Nature vs. Nurture" as it relates to the<br />

question <strong>of</strong> happiness and to the larger question relating to<br />

human nature as an emerging science. It is well established<br />

that in both food addicted and obese individuals there<br />

is "dopamine resistance” due to an association with the<br />

DRD2gene A1 allele. We propose a course <strong>of</strong> action that will<br />

impact globesity, i.e., that a positive effect on body<br />

composition/body mass index (BMI) will increase not only<br />

self-image but also wellness, including a state <strong>of</strong> happiness.<br />

Further, we propose a genetic map <strong>of</strong> happiness. We provide<br />

preliminary evidence that utilization <strong>of</strong> a customized DNA<br />

directed nutraceutical, LG839 [SYNATAMINECOMPLEX<br />

VARIANT], significantly increased happiness (p < 0.05) in<br />

obese subjects. Synaptamine is a putative dopaminergic<br />

agonist, and it also is being investigated by using fMRI to<br />

measure food induced blunted response in reward circuitry<br />

(e.g., dorsal striatum and orbit<strong>of</strong>rontal cortex) in DRD2 A1<br />

allele humans. We cautiously suggest that long-term activation<br />

<strong>of</strong> dopaminergic receptors (i.e., DRD2 receptors) will<br />

proliferate D2 receptors leading to enhanced "dopamine<br />

sensitivity" and an increased sense <strong>of</strong> happiness.<br />

183 SMOKING CESSATION MOLECULAR GENETICS<br />

G. Uhl* (1), J. Rose (2), T. Drgon (1), F. Behm (2), D.<br />

Walther (1), C. Johnson (1), Q. Liu (1)<br />

1. NIH 2. Duke University<br />

*guhl@intra.nida.nih.gov<br />

Twin data support substantial heritable contributions to<br />

individual differences in ability to quit smoking. Genome wide<br />

association approaches identify small chromosomal regions<br />

that are reproducibly tagged by clusters <strong>of</strong> SNPs whose allele<br />

frequencies distinguish successful vs unsuccessful quitters in<br />

clinical trial, general practice and community quitter settings<br />

in the US and UK. Genes identified by these<br />

clustered, reproducibly-positive results include numbers <strong>of</strong><br />

cell adhesion related genes, genes implicated in indivdual<br />

differences in memory, and "druggable" genes. Some markers<br />

display stronger association with quitting in trials <strong>of</strong><br />

bupropion, some with nicotine replacement (NRT) but most<br />

with both. Data from retrospective analysis provided a v1.0<br />

"quit success" genetic score that has provided significant<br />

prospective results. This v1.0 score prospectively provides a<br />

significant interaction with FTND assessments <strong>of</strong> nicotine<br />

dependence to predict dose-related responses in individual<br />

differences in ability to quit smoking. Taken together, these<br />

data support polygenic contributions to smoking cessation<br />

success from a number <strong>of</strong> allelic variants, including those that<br />

we have validated in v1.0 quit success genotype scores.


184 GENE REGULATION BY DRUGS USED TO<br />

TREAT MOOD DISORDERS<br />

X. Deng* (1), P. McHugh (1), K. Doudney (1), P. Joyce (2),<br />

M. Kennedy (1)<br />

1. Christchurch, New Zealand 2. Department <strong>of</strong> Psychological<br />

Medicine, University <strong>of</strong> Otago, Christchurch, New Zealand<br />

*sarah.deng@otago.ac.nz<br />

Antidepressant (AD) and mood stabilizer drugs are the main<br />

treatments for mood disorders including major depressive<br />

disorder and bipolar disorder. Although widely used, the<br />

mechanisms <strong>of</strong> action <strong>of</strong> these drugs are not well understood.<br />

Our first attempts to establish a model cell culture system to<br />

study these drugs involved exposure <strong>of</strong> RN46A cells for 14<br />

days, with various drugs, followed by transfection with<br />

luciferase reporter constructs, in order to monitor gene<br />

expression effects.Of the 19 promoter constructs tested,<br />

occasional significant differences in expression were noted<br />

between drug-exposed and control cell cultures. However,<br />

these proved not to be robust or reproducible despite<br />

considerable care in experimental design and execution. One<br />

outcome <strong>of</strong> this work, however, was the finding that 72 hours<br />

exposure <strong>of</strong> cells with the AD paroxetine appeared to give<br />

optimal expression differences, which was chosen for future<br />

experiments. Our second approach was to use quantitative<br />

PCR analysis <strong>of</strong> candidate genes after exposure <strong>of</strong> RN46A<br />

cells to various drugs for 72 hours. Our most striking results to<br />

date are: (1) a gene called sepiapterin reductase (SPR), which<br />

encodes a key enzyme in neurotransmitter synthesis, is<br />

massively (>1700-fold, p=0.001) and specifically up-regulated<br />

by sodium valproate; (2) another gene in the same pathway<br />

(QDPR) is also significantly up-regulated; (3) the serotonin<br />

receptor 2A (HTR2A) is significantly down-regulated by<br />

paroxetine and citalopram, the two selective serotonin<br />

reuptake inhibitor antidepressants (p=.001 to .003); and (4) the<br />

histone deacetylase HDAC2 gene is reduced significantly by<br />

all <strong>of</strong> the drug treatments except sodium valproate.<br />

185 INFLUENCE OF TAAR6 POLYMORPHISMS ON<br />

RESPONSE TO ARIPIPRAZOLE<br />

A. Serretti (1), C. Pae (2), A. Chiesa* (1), L. Mandelli (1), D.<br />

De Ronchi (1)<br />

1. Institute <strong>of</strong> <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna 2. The<br />

Catholic University <strong>of</strong> Korea College <strong>of</strong> Medicine, Bucheon,<br />

Kyounggi-Do, Republic <strong>of</strong> Korea<br />

*albertopnl@yahoo.it<br />

Background: There is some evidence suggesting a role <strong>of</strong><br />

TAAR6 in schizophrenia. The aim <strong>of</strong> the present study is to<br />

investigate possible influences <strong>of</strong> a panel <strong>of</strong> markers in<br />

TAAR6 (rs8192625, rs4305745, rs4305746, rs6903874,<br />

rs6937506) on clinical outcomes and side effects in a sample<br />

<strong>of</strong> Korean schizophrenic aripiprazole treated patients.<br />

Methods: Efficacy was assessed at baseline and weeks 1, 2, 4,<br />

6, 8 using CGI-S, CGI-I, BPRS and SANS. Side effects were<br />

evaluated through SAS, BAS and AIMS. Multivariate analysis<br />

<strong>of</strong> covariance (MANCOVA) was used to test possible<br />

influences <strong>of</strong> single SNPs on clinical and safety scores. Tests<br />

for associations using multi-marker haplotypes were<br />

performed using the statistics environment “R”. Results: A<br />

significant time per genotype interaction was found between<br />

rs4305746 and repeated measures <strong>of</strong> ANOVA on BPRS scores<br />

(F=2.45, d.f.=10,365, p=0.008). In particular G/A and A/A<br />

genotype patients were more likely to improve over time.<br />

Permutation analysis confirmed a significant effect <strong>of</strong><br />

rs4305746 on course <strong>of</strong> BPRS scores over time (p=0.007).<br />

Haplotype analysis did not revealed any significant association<br />

with clinical and safety scores at any time. Conclusion: A<br />

possible association could exist between some genotypes in<br />

TAAR6 and response to aripiprazole. However, several<br />

limitations as the small sample size, the finding related to a<br />

single scale and the possibility <strong>of</strong> false positive findings<br />

require further investigations.


186 GRIA1, GRIA4, GRIA3 AND GRIK4 GENE<br />

VARIANTS INFLUENCE HALOPERIDOL EFFICACY<br />

AND SIDE EFFECTS IN A SAMPLE OF<br />

SCHIZOPHRENIC PATIENTS<br />

A. Serretti* (1), A. Drago (1), I. Giegling (2), A. Hartmann<br />

(2), M. Schafer (2), H. Moller (2), D. De Ronchi (1), D.<br />

Rujescu (2)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna, Italy 2.<br />

Department <strong>of</strong> Psychiatry, Ludwig Maximilians University,<br />

Munich, Germany<br />

*alessandro.serretti@unibo.it<br />

The unbalanced orchestration <strong>of</strong> the glutamate system is<br />

thought to underlie the pathophysiology <strong>of</strong> schizophrenic<br />

disorders. Consistently, there is evidence that the glutamate<br />

system is pivotal to the effects <strong>of</strong> the antipsychotic treatments.<br />

We investigated a set <strong>of</strong> 50 SNPs located in 11 genes coding<br />

for subunits <strong>of</strong> glutamatergic receptors, as modulators <strong>of</strong> the<br />

efficacy <strong>of</strong> haloperidol in a sample <strong>of</strong> 101 schizophrenic<br />

patients. Furthermore, we investigated the possible impact on<br />

the motor side effect pr<strong>of</strong>ile associated with haloperidol<br />

treatment. Patients were administered the PANSS and ESRS<br />

tests at baseline and at day 3, 7, 14, 21 and 28. MANCOVA<br />

analysis for repeated measures was applied along with the<br />

FDR correction for multiple tests. T/T genotype at rs472792<br />

(GRIA1) was found to be associated with a better response to<br />

haloperidol treatment than G/T at PANSS positive at week 2<br />

(f=6.18, p=0.001). C/C genotype at rs1461231 (GRIA1) was<br />

found to be associated with a better response to treatment<br />

compared to G/G genotype at PANSS positive scores at week<br />

1 (f=7.32, p=0.00032). Those findings provide further support<br />

to the glutamatergic theory <strong>of</strong> schizophrenia identifying<br />

putative modulators <strong>of</strong> the antipsychotic effect <strong>of</strong> haloperidol.<br />

187 CLINICAL PHARMACOGENETICS, REASONS<br />

FOR THE SLOW APPLICATION AND REALISTIC<br />

EXPECTATIONS<br />

A. Serretti* (1)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna, Bologna,<br />

Italy<br />

*alessandro.serretti@unibo.it<br />

The practical application <strong>of</strong> pharmacogenetics is not as<br />

straightforward as expected, particularly in psychiatric<br />

disorders. Despite the large benefit expected, only a very small<br />

numbers <strong>of</strong> clinical applications are available. The reasons for<br />

this are many, in particular the key questions are: Is science<br />

robust enough for routine application? Are physicians and<br />

patients ready? Are stakeholders (e.g. Government, insurance<br />

industry, etc) ready to pay and implementing? What is the<br />

position <strong>of</strong> the pharmaceutical industry? The presentation will<br />

cover such aspects <strong>of</strong>fering a coverage <strong>of</strong> present status and<br />

realistic expectations in the field.<br />

188 FURTHER EVIDENCE SUPPORTING THE<br />

INFLUENCE OF BDNF ON THE OUTCOME OF<br />

DEPRESSION. INDEPENDENT EFFECT OF BDNF<br />

AND HARM AVOIDANCE<br />

L. Mandelli* (1), M. Mazza (2), D. Marco (2), G. Martinotti<br />

(2), D. Tavian (3), E. Colombo (3), S. Missaglia (3), G. Negri<br />

(3), D. De Ronchi (1), R. Colombo (2, 3), L. Janiri (2)<br />

1. University <strong>of</strong> Bologna, Italy 2. Catholic University <strong>of</strong><br />

Rome, Italy 3. Catholic University <strong>of</strong> Milan, Italy<br />

*laura.mandelli@unibo.it<br />

Brain derived neurotrophic factor (BDNF) is a candidate gene<br />

for response to antidepressant treatment, as its product exerts<br />

trophic effects on hippocampus and serotoninergic neurons.<br />

Moreover, BDNF levels are increased by chronic<br />

administration <strong>of</strong> antidepressants. However, response to<br />

pharmacological treatments is moderated by both genetic and<br />

individuals factors. In previous studies, we confirmed the<br />

influence <strong>of</strong> the temperamental trait <strong>of</strong> Harm avoidance (HA)<br />

on outcome from depression and we observed a significant<br />

interaction with the promoter polymorphism <strong>of</strong> serotonin<br />

transporter (5-HTTLPR). In the present study we aimed to<br />

investigate the effect <strong>of</strong> 2 BDNF polymorphisms (rs6265 –<br />

also called Val66Met– and rs11030104) on medium-term<br />

outcome in a naturalistic sample <strong>of</strong> 86 depressed bipolar (BP)<br />

spectrum patients, taking into account a number <strong>of</strong> clinical<br />

features including HA. Rs6265 and rs11030104 were in full<br />

linkage disequilibrium. Both single marker and haplotypes<br />

analysis showed a significant association with severity <strong>of</strong><br />

depression at 6 months after intake. In particular, the<br />

haplotype composed by the less frequent alleles A-C was<br />

associated with a poorer outcome. HA maintained a significant<br />

effect on depressive outcome, independently from BDNF<br />

genotypes. However, HA’s influence appeared to be more<br />

consistent in patients carrying the protective G-T combination<br />

<strong>of</strong> alleles.


189 NDE1 VARIANT MODULATES OUTCOME FOR<br />

DEPRESSION TREATED WITH ESCITALOPRAM<br />

W. Hennah* (1), L. Muhonen (2), S. Saarikoski (2), J. Lahti<br />

(3), L. Peltonen (1), J. Lönnqvist (2), H. Alho (2)<br />

1. Institute for Molecular Medicine Finland FIMM, Nordic<br />

EMBL Partnership for Molecular Medicine, Helsinki, Finland<br />

2. National Institute for Health and Welfare, Department <strong>of</strong><br />

Mental Health and Substance Abuse Services, Helsinki,<br />

Finland 3. University <strong>of</strong> Helsinki, Department <strong>of</strong> Psychology,<br />

Helsinki, Finland<br />

*william.hennah@thl.fi<br />

In an analysis <strong>of</strong> public datasets it was demonstrated that there<br />

are correlations between variants in the NDE1 gene and levels<br />

<strong>of</strong> gene expression for existing targets for drugs developed for<br />

depression and psychosis. One such target was HTR3A, which<br />

is targeted by the drug memantine. In a clinical trial studying<br />

major depressive disorder comorbid with alcohol dependence,<br />

the effectiveness <strong>of</strong> memantine and escitalopram as treatments<br />

were evaluated. This demonstrated that both treatments were<br />

able to significantly reduce the baseline levels <strong>of</strong> depression<br />

and anxiety. Here we have tested if the NDE1 variant<br />

rs4781678 correlates with treatment outcome in depression.<br />

We further tested for correlation dependent on drug treatment.<br />

Outcome in depression was significantly improved in males<br />

homozygous for the A allele <strong>of</strong> rs4781678 (p = 0.022).<br />

However, this was dependent on treatment by escitalopram,<br />

not the hypothesised memantine. Further bioinformatics and<br />

studies <strong>of</strong> public datasets revealed that although NDE1<br />

modulates expression levels <strong>of</strong> one <strong>of</strong> the nine targets for<br />

memantine, it also modulates the expression level <strong>of</strong> the<br />

principal metabolizing enzyme <strong>of</strong> escitalopram, CYP2C19. A<br />

variant in strong LD with rs4781678 demonstrated that, in<br />

human post-mortem brain tissue, carriers <strong>of</strong> the A allele have<br />

significantly decreased amounts <strong>of</strong> CYP2C19 gene expression<br />

(p = 0.029). We provide pro<strong>of</strong> <strong>of</strong> concept that the DISC1<br />

pathway could be used as a means to target treatment for<br />

major mental illnesses, as hypothesised by Hennah and<br />

Porteous, specifically by demonstrating that the NDE1 variant<br />

rs4781678 modulates outcome for depression when treated<br />

using escitalopram.<br />

190 CIRCADIAN CLOCK GENES AND RESPONSE TO<br />

ANTIDEPRESSANTS IN GENDEP<br />

R. Keers* (1), R. Smith (1), R. Uher (1), P. Huezo-Diaz (1),<br />

A. Elkin (1), M. Rietschel (2), T. Schulze (2), N. Henigsberg<br />

(3), Z. Kovacic (3), A. Marusic (4), D. Kozel (5), O. Mors (6),<br />

E. Larsen (5), J. Hauser (7), P. Czerski (7), W. Maier (8), J.<br />

Perez (9), A. Placentino (9), J. Mendlewicz (10), D. Souery<br />

(10), A. Farmer (11), P. McGuffin (12), I. Craig (11), K.<br />

Aitchison (11)<br />

1. Institute <strong>of</strong> Psychiatry at King’s College London, MRC<br />

SGDP Centre, London, UK 2. Zentralinstitut für Seelische<br />

Gesundheit, Division <strong>of</strong> Genetic Epidemiology in Psychiatry,<br />

Mannheim, Germany 3. University <strong>of</strong> Zagreb, Croatian<br />

Institute for Brain Research, Zagreb, Croatia 4. Institute <strong>of</strong><br />

Public Health, Ljubljana, Slovenia 5. University Hospital,<br />

Department <strong>of</strong> Psychiatry, Risskov, Denmark 6. Aarhus<br />

University Hospital, Department <strong>of</strong> Psychiatry, Risskov,<br />

Denmark 7. Academic Dept <strong>of</strong> Medicine, Poznan, Poland 8.<br />

University <strong>of</strong> Bonn, Anstalt des öffentlichen Rechts, für den<br />

Fachbereich Medizin, Bonn, Germany 9. IRCCS, Centro San<br />

Giovanni di Dio, FBF, Biological Psychiatry Unit and Dual<br />

Diagnosis Ward, Brescia, Italy 10. Free University <strong>of</strong><br />

Brussels, Department <strong>of</strong> Psychiatry, Brussels, Belgium 11.<br />

Farmer A.E.1, McGuffin P,1 Craig I.1, Aitchison K.J.1<br />

Institute <strong>of</strong> Psychiatry at King’s College London, MRC SGDP<br />

Centre, London, UK 12. Aitchison K.J.1 Institute <strong>of</strong><br />

Psychiatry at King’s College London, MRC SGDP Centre,<br />

London, UK<br />

*robert.keers@kcl.ac.uk<br />

There is substantial individual variation in response to<br />

antidepressants, and genetic variation may, in part, explain<br />

these differences. A number <strong>of</strong> successful treatments for<br />

depression have downstream effects on the circadian rhythm.<br />

For example, the selective serotonin-reuptake inhibitor (SSRI)<br />

fluoxetine has been shown to induce a phase shift in cell<br />

culture studies. Functional polymorphisms in genes implicated<br />

in the circadian rhythm may therefore be predictive <strong>of</strong><br />

response to antidepressants. GENDEP, a part-randomised drug<br />

trial collected longitudinal data on the response <strong>of</strong> more than<br />

800 patients undergoing treatment with either an SSRI<br />

(escitalopram) or a tricyclic antidepressant (nortriptyline).<br />

Two functional polymorphisms in genes associated with the<br />

circadian clock were genotyped in this sample including a<br />

variable number tandem repeat in PER3 and a SNP (rs180260)<br />

in CLOCK. Both polymorphisms moderated response to both<br />

drugs and these effects were age-dependent. This suggests that<br />

circadian clock genes may not only be important in the<br />

aetiology <strong>of</strong> mood disorders but also in the prediction <strong>of</strong><br />

response to antidepressants.


191 GENETIC STUDY OF<br />

CATECHOL-O-METHYL-TRANSFERASE IN<br />

TARDIVE DYSKINESIA<br />

C. Zai* (1), D. Mueller (1), D. Mueller (1), V. de Luca (1), X.<br />

Ni (1), D. Sibony (1), A. Aristotle (1), H. Meltzer (2), J.<br />

Lieberman (3), S. Potkin (4), G. Remington (1), J. Kennedy<br />

(1)<br />

1. Centre for Addiction and Mental Health 2. Psychiatric<br />

Hospital at Vanderbilt University 3. New York State<br />

Psychiatric Institute, Columbia University <strong>Medical</strong> Centre 4.<br />

University <strong>of</strong> California, Irvine<br />

*clement_zai@camh.net<br />

Tardive dyskinesia (TD) is a severe motor side effect <strong>of</strong><br />

long-term antipsychotic medication. Changes in dopamine<br />

neurotransmission have been implicated in the etiology <strong>of</strong><br />

TD. Catechol-O-Methyl-Transferase (COMT) is an enzyme<br />

that metabolizes dopamine. The functional Val158Met<br />

polymorphism has been associated with TD; however, the<br />

small effect size reported in a recent meta-analysis suggests<br />

that additional genetic factors in COMT may contribute to TD<br />

development. We investigated four single-nucleotide<br />

polymorphisms in addition to the functional Val158Met<br />

variant in the COMT gene for association with TD. Three <strong>of</strong><br />

the polymorphisms have been associated with pain sensitivity<br />

and COMT enzymatic activity (Nackley et al, 2006). We<br />

genotyped the five COMT polymorphisms in our sample <strong>of</strong><br />

schizophrenia/schizoaffective disorder patients (n=196<br />

Caucasians and 30 African Americans) using commercially<br />

available assays and performed statistical analyses, which<br />

included comparing the severity <strong>of</strong> TD among patients<br />

carrying different COMT variants. We found the rs165599<br />

marker in the 3’ untranslated region <strong>of</strong> the COMT gene to be<br />

marginally associated with TD occurrence. We did not find a<br />

significant association <strong>of</strong> the other four tested polymorphisms<br />

or their combinations with TD in our samples. When we<br />

included age and sex as covariates in the analyses, the results<br />

were not significant. We will be performing an updated<br />

meta-analysis <strong>of</strong> the Val158Met polymorphism, including<br />

additional studies. The results with our sample suggest that<br />

the COMT gene is not a major factor in TD, but further studies<br />

with additional markers in larger clinical samples are required.<br />

192 ASSOCIATION OF OPRM1 DNA VARIANTS<br />

GENETIC CORRELATIONS IN HEROIN ADDICTED<br />

PATIENTS UNDERGOING NALTREXONE IMPLANT<br />

TREATMENT.<br />

A. Scott* (1, 2), N. Morris (1), G. Hulse (1), D. Wildenauer<br />

(1), S. Schwab (1, 2, 3)<br />

1. School <strong>of</strong> Psychiatry and Clinical Neurosciences,<br />

University <strong>of</strong> Western Australia 2. Centre for <strong>Medical</strong><br />

Research and Western Australian Institute for <strong>Medical</strong><br />

Research, University <strong>of</strong> Western Australia 3. School <strong>of</strong><br />

Medicine and Pharmacology, University <strong>of</strong> Western Australia<br />

* ascott@cyllene.uwa.edu.au<br />

While environmental factors are likely to contribute to<br />

different degrees <strong>of</strong> addiction, it is becoming clear that<br />

susceptibility is strongly influenced by an individual’s genetic<br />

makeup. Heroin has one <strong>of</strong> the highest levels <strong>of</strong> genetic<br />

variance compared to other illicit drugs, in particular<br />

marijuana, stimulants, sedatives and psychedelics. Much<br />

research into heroin addiction genetics has focussed on the<br />

µ-opioid receptor (OPRM1), which is the primary target for<br />

heroin action. The coding region polymorphism A118G<br />

(Asn40Asp) has been the primary focus <strong>of</strong> interest. This<br />

genetic variant has recently been associated with treatment<br />

outcome in alcoholism using naltrexone. We have genotyped<br />

this polymorphism in a large sample <strong>of</strong> heroin addicted<br />

patients who have been treated with naltrexone implants. In<br />

total a cohort <strong>of</strong> 750 heroin addicted Caucasian patients have<br />

been collected. Phenotypic data such as naltrexone serum<br />

levels after implant and treatment outcome as measured by<br />

repeated contact with the clinic was established. High<br />

throughput Taqman assays available on demand from Applied<br />

Biosystems have been used for genotyping, and association <strong>of</strong><br />

this polymorphism with treatment outcome in heroin addiction<br />

was attempted. Due to the large number <strong>of</strong> patients, our study<br />

has a high power for showing association with genetic<br />

variants. The unique phenotypic characterization <strong>of</strong> our study<br />

allows conclusion on treatment outcome using naltrexone<br />

implants in heroin addiction.


193 ALTERATION IN THE RATIO OF BAX/BCL-2<br />

EXPRESSION LEVEL IN MODIFIED<br />

ELECTROCONVULSIVE THERAPY<br />

A. Tsutsumi*, S. Kawashige, T. Kanazawa, H. Kikuyama, J.<br />

Koh, H. Yoneda<br />

* psy063@poh.osaka-med.ac.jp<br />

Schizophrenia is generally considered as a<br />

neurodevelopmental disorder. Evidences for progressive<br />

clinical deterioration and subtle neurostructural changes<br />

following the onset <strong>of</strong> psychosis has led to the hypothesis that<br />

apoptosis may contribute to the pathophysiology <strong>of</strong><br />

schizophrenia. Among the antipsychoticdrugs, olanzapine may<br />

have neuroprotective effects, whereas haloperidol was<br />

apparently neurotoxic through some mechanism apoptosis<br />

(Kim NR et al: 2008). Modified-electroconvulsive therapy<br />

(m-ECT) isa widely used and efficient treatment in psychiatry,<br />

although the basis for its therapeutic effect is still unknown.<br />

There are some reports which indicate therelation with m-ECT<br />

and TNF-alpha (HestadKA et al: 2003). We had also<br />

investigated the mRNA expression levels<strong>of</strong> TNF-alpha, and<br />

found out the relationship between TNF-alpha and efficacy in<br />

the schizophrenia. TNF-alpha serves as potent inductors <strong>of</strong><br />

apoptosis and changes the ratio <strong>of</strong> Bax-/Bcl-Xl expression in<br />

human (Szuster-CiesielskaA et al: 2008). Therefore, we<br />

hypothesized that therapeutic mechanism <strong>of</strong> m-ECT might be<br />

associated with apoptosis. We investigated mRNA expression<br />

levels <strong>of</strong> Bax and Bcl-2 to see if there is relationship with<br />

efficacy in the schizophrenia treated with m-ECT.<br />

194 ALTERATION IN GSK-3β MRNA EXPRESSION<br />

ON THE TREATMENT BOTH BY SDAS AND BY<br />

M-ECT FOR SCHIZOPHRENIA<br />

S. Kawashige*, A. Tsutsumi, T. Kanazawa, H. Kikuyama, J.<br />

Koh, H. Yoneda<br />

* seiyart@gmail.com<br />

Akt-GSK-3β signaling has been recently reported as<br />

associated not only with bipolar disorder, but also with<br />

schizophrenia. Glycogen synthase kinase-3β (GSK-3β) is<br />

considered as a target <strong>of</strong> lithium. Emamian et al. reported a<br />

decrease in AKT1 protein levels and Ser9 phosphorylation <strong>of</strong><br />

GSK-3β in the peripheral lymphocytes and brains <strong>of</strong><br />

schizophrenia patients. Moreover, increased serine<br />

phosphorylation <strong>of</strong> GSK-3β in brain <strong>of</strong> mouse was also<br />

induced by electroconvulsive seizure treatment. (Roh MS et<br />

al. 2003). We investigate the alteration <strong>of</strong> GSK-3β in<br />

schizophrenia with gene expression analysis (RT-PCR). With<br />

employing the mRNA derived from peripheral blood <strong>of</strong><br />

schizophrenia treated by SDAs (n=10), by m-ECT (n=10) and<br />

control (n=3), expression levels are determined at the points <strong>of</strong><br />

every four weeks (0W, 4W, 8W). The role <strong>of</strong> GSK-3β will be<br />

discussed in the conference.<br />

195 CANNABINOID TYPE-1 RECEPTOR GENE<br />

POLYMORPHISMS ARE NOT ASSOCIATED WITH<br />

OLANZAPINE-INDUCED WEIGHT GAIN<br />

Y. Park (1), J. Choi (2), S. Kang (1), L. Kim (1), H. Lee* (1)<br />

1. Inje University College <strong>of</strong> Medicine 2. Korea University<br />

College <strong>of</strong> Medicine<br />

* leehjeong@korea.ac.kr<br />

Objective: Olanzapine is an atypical antipsychotic known to<br />

cause considerable weight gain. The CNR1 cannabinoid<br />

receptor is involved in energy balance control, stimulating<br />

appetite and increasing body weight. In the present study, we<br />

investigated the 1359G/A (rs1049353), 4895A/G (rs806368),<br />

and rs4707436 polymorphisms in the CNR1 gene and weight<br />

gain in a Korean schizophrenic patients receiving olanzapine<br />

treatment. Methods: Weight and height measurements were<br />

obtained prior to starting olanzapine and measured again after<br />

long-term treatment in 79 schizophrenic patients. Genotyping<br />

for the CNR1 polymorphisms were performed using<br />

PCR-RFLP methods. Results: There were no association<br />

between three CNR1 gene polymorphism and body weight<br />

change from baseline to the endpoint after olanzapine<br />

treatment (p>0.05). There were also no significant differences<br />

in genotype or allele frequencies between the severe<br />

weight-gain (more than 7%) and less weight-gain (less than<br />

7%) groups. The haplotype analyses did not show the<br />

significant differences between two groups. Conclusion:<br />

Within the limitations imposed by the size <strong>of</strong> the clinical<br />

sample, our findings suggest that CNR1 gene polymorphisms<br />

may be not associated with olanzapine-induced weight gain.


196 NO ASSOCIATION BETWEEN IL-10 AND<br />

TNF-alpha GENES POLYMORPHISMS AND TARDIVE<br />

DYSKINESIA IN SCHIZOPHRENIA<br />

S. Kang (1), J. Choi (1), H. Lee* (1), L. Kim (1)<br />

1. Korea University College <strong>of</strong> Medicine<br />

* leehjeong@korea.ac.kr<br />

Objective: Cytokines has been reported to be associated with<br />

several psychiatric illnesses including schizophrenia.<br />

Antipsychotic treatment has also been involved in the altered<br />

levels <strong>of</strong> some cytokines. There are emerging evidences<br />

suggest that the development <strong>of</strong> TD is related to the oxidative<br />

stress, excitotoxicity, and immune activation. The purpose <strong>of</strong><br />

this study is to investigate whether single-nucleotide<br />

polymorphisms (SNPs) <strong>of</strong> interleukin(IL)-10 and tumor<br />

necrosis factor(TNF)-α genes are associated with the<br />

susceptibility <strong>of</strong> TD and schizophrenia. Methods: Two<br />

hundred and nine unrelated Korean schizophrenic patients (83<br />

TD and 126 non-TD patients) and two hundred and twelve<br />

normal healthy controls participated in the present study. The<br />

schizophrenic participants consisted <strong>of</strong> patients with (n=83)<br />

and without (n=126) TD who were matched for antipsychotic<br />

drug exposure and other relevant variables. All the<br />

schizophrenic patients met the criteria for a diagnosis <strong>of</strong><br />

schizophrenia as determined by board-certified psychiatrists<br />

using the Korean version <strong>of</strong> the Structured Clinical Interview<br />

for the fourth edition <strong>of</strong> the Diagnostic and Statistical Manual<br />

<strong>of</strong> Mental Disorders. TD was diagnosed based on the<br />

Abnormal Involuntary Movement Scale (AIMS). The IL-10<br />

gene -1082G/A and TNF-α gene -308G/A SNPs were<br />

analyzed by polymerase chain reaction (PCR)-based methods.<br />

Results: The frequencies <strong>of</strong> genotypes (?2=2.65, p=0.263) <strong>of</strong><br />

the IL-10 gene -1082G/A SNP and the frequencies <strong>of</strong><br />

genotypes (?2=0.13, p=0.715) <strong>of</strong> the TNF-α gene -308G/A<br />

SNP did not differ significantly between schizophrenic<br />

patients with and without TD. The distribution <strong>of</strong> genotype at<br />

two polymorphisms between the schizophrenic patients and<br />

controls was not significantly different, either (p>0.05). MDR<br />

analysis did not show a significant interaction between the<br />

IL-10 gene and TNF-α gene and susceptibility to TD and<br />

schizophrenia (p>0.05) Conclusions: This is the first study on<br />

the association <strong>of</strong> antipsychotics-induced TD with gene<br />

polymorphisms in cytokine production. These results suggest<br />

that the IL-10 gene -1082G/A and TNF-α gene -308G/A SNPs<br />

are not associated with TD and schizophrenia in a Korean<br />

population. Further association studies <strong>of</strong> TD with other<br />

candidate genes for cytokines would help us understand the<br />

pathophysiological mechanisms <strong>of</strong> TD.<br />

197 INVOLVEMENT OF THE ATRIAL NATRIURETIC<br />

PEPTIDE TRANSCRIPTION FACTOR GATA4 IN<br />

ALCOHOL DEPENDENCE, RELAPSE RISK, AND<br />

TREATMENT RESPONSE TO ACAMPROSATE<br />

S. Witt (1), F. Kiefer (2), J. Frank (1), A. Richter (2), J.<br />

Treutlein (1), T. Lemenager (2), M. Nöthen (3), S. Cichon (3),<br />

A. Batra (4), M. Berner (5), N. Wodarz (6), U. Zimmermann<br />

(7), R. Spanagel (8), K. Wiedemann (9), M. Smolka (10), A.<br />

Heinz (10), K. Mann (2), M. Rietschel* (1)<br />

1. Dept. <strong>of</strong> Genetic Epidemiology in Psychiatry, Central<br />

Institute <strong>of</strong> Mental Health, Mannheim 2. Dept. <strong>of</strong> Addictive<br />

Behavior and Addiction Medicine, Central Institute <strong>of</strong> Mental<br />

Health, Mannheim 3. Department <strong>of</strong> Genomics, Life & Brain<br />

Center, University <strong>of</strong> Bonn 4. Dept. <strong>of</strong> Psychiatry and<br />

Psychotherapy, University Göttingen 5. Dept. <strong>of</strong> Psychiatry<br />

and Psychotherapy, University Freiburg 6. Dept. <strong>of</strong> Stem Cell<br />

Biology, University Göttingen 7. Dept. <strong>of</strong> Psychiatry and<br />

Psychotherapy, Technical University Dresden 8. Dept. <strong>of</strong><br />

Psychopharmacology, Central Institute <strong>of</strong> Mental Health,<br />

Mannheim 9. Dept. <strong>of</strong> Psychiatry and Psychotherapy,<br />

University <strong>Medical</strong> Center Hamburg-Eppendorf 10. Dept. <strong>of</strong><br />

Psychiatry and Psychotherapy, Charite Berlin<br />

* marcella.rietschel@zi-mannheim.de<br />

Context: Identifying genes contributing to relapse and<br />

treatment response will improve our understanding <strong>of</strong> the<br />

mechanisms underlying alcohol dependence and will yield<br />

treatment targets for personalized medicine. Objective: To<br />

determine whether 15 single nucleotide polymorphisms<br />

(SNPs), linked to alcohol dependence by a recent<br />

genome-wide association (GWA) and replication study, are<br />

associated with relapse behavior. Secondary objective: To<br />

examine whether those associations can be linked to<br />

pharmacological treatment response. Design:<br />

Genotype-phenotype association study and pharmacogenetics<br />

in alcohol dependent patients: Fifteen SNPs were genotyped<br />

and tested for an association with relapse during twelve weeks<br />

<strong>of</strong> treatment. Setting: Five academic medical centers in<br />

Germany Patients: 374 participants with a DSM-IV diagnosis<br />

<strong>of</strong> alcohol dependence that underwent a randomized,<br />

double-blind, placebo-controlled trial with acamprosate,<br />

naltrexone or placebo. Main Outcome Measures: Primary:<br />

Association between 15 SNPs identified by a recent GWA<br />

study on alcohol dependence and relapse. Secondary:<br />

Treatment response with naltrexone and acamprosate in<br />

association with these genetic variations. Post-hoc:<br />

Measurement <strong>of</strong> a trial natruiuretic peptide (ANP) in plasma.<br />

Results: SNP rs13273672, an intronic SNP in GATA4, was<br />

significantly associated with relapse within a 90-days medical<br />

treatment period (P < .01). Subsequent pharmacogenetic<br />

analysis showed that this association was mainly based on<br />

patients treated with acamprosate (P


198 CYP2D6 AND CYP2C19 IN A BRAZILIAN<br />

POPULATION: POTENTIAL ROLE IN IMPROVING<br />

TREATMENT OUTCOMES IN MAJOR DEPRESSION.<br />

C. Prado (1), R. Moreno (2), W. Gattaz (1), E. Ojopi* (1)<br />

1. Laboratório de Neurociências (LIM 27), Institute <strong>of</strong><br />

Psychiatry, University <strong>of</strong> São Paulo (USP), São Paulo, Brazil.<br />

2. Grupo de Estudos de Doenças Afetivas (GRUDA), Institute<br />

<strong>of</strong> Psychiatry, University <strong>of</strong> São Paulo (USP), São Paulo,<br />

Brazil.<br />

* elida@usp.br<br />

The enzymes <strong>of</strong> cytochrome P450 (CYP), mainly CYP2D6<br />

and CYP2C19 are known for metabolizing approximately half<br />

<strong>of</strong> the 200 more <strong>of</strong>ten prescript medications. The CYP2D6<br />

enzyme by itself is responsible for the metabolism <strong>of</strong><br />

approximately 25% <strong>of</strong> these medications, especially<br />

antidepressants (SSRIs e TCAs). At present, more than 90<br />

different allelic variants and sub variants in the CYP2D6 gene<br />

are known and there are only four major phenotypes: poor,<br />

intermediate, extensive, and ultrarapid metabolizers, while for<br />

the CYP2C19 gene, the possible phenotypes are: PMs, EMs<br />

and UMs. The enzymes <strong>of</strong> cytochrome P450 (CYP), mainly<br />

CYP2D6 and CYP2C19 are known for metabolizing<br />

approximately half <strong>of</strong> the 200 more <strong>of</strong>ten prescript<br />

medications. The CYP2D6 enzyme by itself is responsible for<br />

the metabolism <strong>of</strong> approximately 25% <strong>of</strong> these medications,<br />

especially antidepressants (SSRIs e TCAs). This study aimed<br />

to explore the influence <strong>of</strong> variation in CYP2D6 and<br />

CYP2C19 genes on treatment response to antidepressants in a<br />

Brazilian sample <strong>of</strong> patients with Major Depression. Up to<br />

now 73 samples <strong>of</strong> patients with Major Depression and 90<br />

control samples were genotyped for the following alleles:<br />

CYP2D6*1, *2, *3, *4, *6, *9, *10, *15, *17, *29, *34, *35,<br />

*40, *41, and CYP2C19*1, *2, *3, and *17. Genotyping was<br />

performed by real-time PCR allelic discrimination. The<br />

CYP2D6*5 allele and the number <strong>of</strong> copies <strong>of</strong> the gene<br />

CYP2D6 were identified by Real-Time PCR. The Hamilton<br />

scale <strong>of</strong> evaluation for depression (HAM-D) was used to<br />

determine the clinic course <strong>of</strong> response to the treatment and<br />

the adverse affects were also registered. As described in other<br />

studies in different populations alleles CYP2D6*1 and<br />

CYP2D6*2 were the most frequent in our sample (42,8 e<br />

24.31%, respectively), followed by the alleles CYP2D6*4<br />

(11,64%) and CYP2D6*41 (5,82%). For the CYP2C19 gene,<br />

our the allele CYP2C19*1 is the most frequent (60%),<br />

followed by the allele CYP2C19*17 (24%) and the<br />

CYP2C19*2 (16%). To our knowledge the CYP2C19*17<br />

allele was evaluated in a Brazilian population for the first<br />

time. We have found positive correlations genotype-phenotype<br />

although possible jeopardized because there was heterogeneity<br />

among medications. The future <strong>of</strong> ‘personalized prescription’<br />

in psychiatry requires consideration <strong>of</strong> pharmacogenomic<br />

testing and environmental and personal variables that<br />

influence pharmacokinetic and pharmacodynamic drug<br />

response for each individual drug used by each patient.<br />

Financial support: CNPq and ABADHS.<br />

199 GENETIC VARIATION IN ERBB4 GENE<br />

ASSOCIATED WITH RESPONSE TO PALIPERIDONE<br />

TREATMENT IN PATIENTS WITH SCHIZOPHRENIA<br />

D. Fu* (1), D. Wang (1), Q. Li (1), J. Berwaerts (1), X. Wu<br />

(1), R. Favis (1), M. Haas (1), N. Turner (2), H. Chung (2), L.<br />

Alphs (2), H. Manji (1), R. Twyman (1), N. Cohen (1)<br />

1. J&J PRD 2. Ortho-McNeil Janssen Scientific Affairs<br />

* DFu@its.jnj.com<br />

Background: Schizophrenia is a heterogeneous disorder with<br />

inter-individual differences in its onset, symptoms, and<br />

treatment responses. For these reasons, response to the novel<br />

antipsychotic compound, paliperidone, is likely to be<br />

influenced by individual genetic variation. We therefore<br />

conducted a study to identify genes that may contribute to the<br />

variation in the treatment response to paliperidone. Method: A<br />

CNS chip containing over 25,000 single nucleotide<br />

polymorphisms in 1,204 candidate genes related to<br />

pathophysiology and treatment response <strong>of</strong> a wide range <strong>of</strong><br />

psychiatric disorders was developed. 684 patients<br />

experiencing an acute exacerbation <strong>of</strong> schizophrenia from the<br />

paliperidone treatment arms <strong>of</strong> 3 paliperidone-ER pivotal trials<br />

and 1 paliperidone palmitate phase 2 trial were genotyped<br />

using the CNS chip. Genetic associations were tested with a<br />

general linear model using the absolute changes from baseline<br />

to end point in their PANSS total score, and in PANSS factor<br />

scores, as outcome variables. Results: SNP rs6435681 in the<br />

ErbB4 gene was associated with PANSS total score<br />

improvement from baseline to end point (Bonferroni adjusted<br />

p-value: 0.02). Patients carrying the GG or GA genotypes<br />

(N=659) had a significantly greater average reduction in<br />

PANSS total score than AA carriers (N=25). Significant<br />

association <strong>of</strong> the ErbB4 SNP in the same direction was not<br />

observed in the placebo arms (N=258) or the olanzapine active<br />

control arms (N=235) across trials. Conclusion: These<br />

findings provide the first evidence that genetic variation in the<br />

ErbB4 gene (associated with neuregulin regulation) may<br />

differentially affect treatment response to paliperidone in<br />

acutely exacerbated schizophrenic patients.


200 GAMMA-AMINOBUTYRIC ACID RECEPTOR<br />

GENE POLYMORPHISMS AND<br />

ANTIPSYCHOTICS-INDUCED TARDIVE<br />

DYSKINESIA<br />

J. Choi (1), S. Kang (1), H. Lee* (1)<br />

1. Korea University<br />

* leehjeong@korea.ac.kr<br />

Background: Gamma-aminobutyric acid (GABA)<br />

insufficiency has been reported to be related with the TD<br />

susceptibility. Arinami et al. reported that the GABA receptor<br />

signaling pathway may be involved in the pathophysiology <strong>of</strong><br />

TD by genome wide association study. We conducted a<br />

genetic association analysis between GABA related genes and<br />

TD in Korean schizophrenic patients. Methods: Applying<br />

gene-based association analysis, we selected two<br />

polymorphisms in GABRG3 (c-3 subunit <strong>of</strong> GABA-A<br />

receptor) and SLC6A11 (GABA transporter 3). Two SNP<br />

were analyzed by PCR-based methods in 99 schizophrenic<br />

patients with TD and 164 schizophrenic without TD who were<br />

matched for antipsychotic drug exposure and other relevant<br />

variables. Results: Among the two variants, SCL6A11<br />

genotypes distribution showed a significant difference<br />

between TD and non-TD (X2=12.59, p=0.002), however,<br />

GABRG3 genotype distribution was not associated with TD<br />

(X2=2.65, p=0.27). Conclusions: These results suggest that<br />

SLC6A11 gene polymorphism was associated with the<br />

increased susceptibility to TD in patients with schizophrenia<br />

in the Korean populations.<br />

201 THE CIRCADIAN CLOCK INFLUENCES<br />

LITHIUM TREATMENT RESPONSE IN BIPOLAR<br />

DISORDER<br />

M. McCarthy* (1), T. Shekhtman (2), R. McKinney (1), D.<br />

Kripke (3), J. Kelsoe (1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California San<br />

Diego, La Jolla, CA 2. VA San Diego Healthcare System, San<br />

Diego, CA 3. Scripps Clinic Sleep Center, La Jolla, CA<br />

* mmccarthy@ucsd.edu<br />

Several circadian clock genes are lithium responsive, and<br />

lithium treatment for bipolar disorder is effective in part<br />

because <strong>of</strong> its ability to correct disturbances in circadian<br />

rhythms. However, treatment response to lithium varies. We<br />

hypothesized that variants in circadian clock genes may<br />

contribute to lithium treatment outcomes in bipolar disorder.<br />

Similarly, we hypothesized that differences in daily<br />

sleep/activity cycles may predict lithium response. We<br />

identified 164 bipolar disorder patients who responded well to<br />

lithium, and 154 who responded poorly based on retrospective<br />

assessment. These subjects were genotyped for 29 SNPs<br />

across 13 core clock genes. In a subset <strong>of</strong> these patients, we<br />

administered the basic language morningness questionnaire<br />

(BALM) to 50 lithium responders and 46 lithium<br />

non-responders. Subjects who scored in the phase delayed<br />

range on the BALM had a better response to lithium than<br />

subjects in the normal or phase advanced ranges. Genetic<br />

analysis revealed the rare variant <strong>of</strong> the intronic SNP<br />

rs2071427 in the Rev-Erbα gene was associated with a<br />

favorable lithium response. The SNPs rs1801260 (3111 T/C)<br />

and rs34897046 in the gene Clock were also significantly<br />

associated with response to lithium. Allele-specific gene<br />

expression at the Clock locus in heterozygotes for rs1801260<br />

was measured in lymphoblastoid cells. This SNP did not<br />

confer an allelic expression imbalance either under baseline or<br />

lithium treated conditions. From these data a link between<br />

lithium response and genetic variation within the circadian<br />

clock is supported but the precise biological mechanism<br />

remains unknown.


202 OXYTOCIN AND PROLACTIN SYSTEM<br />

POLYMORPHISMS IN CHILDHOOD-ONSET MOOD<br />

DISORDERS<br />

J. Strauss* (1), N. Freeman (1), S. Shaikh (1), A. Vetro (2),<br />

E. Kiss (2), K. Kapornai (2), C. George (3), E. Dempster (4),<br />

C. Barr (5), M. Kovacs (3), J. Kennedy (1)<br />

1. Centre for Addiction and Mental Health 2. Szeged<br />

University <strong>Medical</strong> Faculty 3. Western Psychiatric Institute<br />

and Clinic 4. Institute <strong>of</strong> Psychiatry 5. Toronto Western<br />

Research Institute<br />

* john_strauss@camh.net<br />

Background: Oxytocin (OXT) and prolactin (PRL) are<br />

neuropeptide hormones that are involved in the stress response<br />

and social affiliation. They interact with the serotonin system.<br />

Reports in the literature indicate that human serum OXT and<br />

PRL levels are associated with depressive symptoms and<br />

related phenotypes. In a genetic association study with a<br />

family-based design, we examined single nucleotide<br />

polymorphisms (SNPs) at the loci for OXT, PRL and their<br />

receptors (OXTR and PRLR) for association with<br />

childhood-onset mood disorders (COMD). Methods: Using<br />

lymphocyte DNA from 678 families recruited from Hungary,<br />

we genotyped sixteen SNPs at OXT, PRL and their receptors<br />

to test for association with COMD. Results: We observed no<br />

statistically significant associations for OXTR, PRL, and<br />

PRLR SNPs. Two <strong>of</strong> three SNPs 3' <strong>of</strong> the OXT gene were<br />

associated with COMD (p < 0.02), were significant after<br />

spectral decomposition, but were not significant after<br />

additionally adjusting for the number <strong>of</strong> genes tested.<br />

Supplementary analyses demonstrated significant transmission<br />

bias to daughters, but not sons, for two OXT SNPs -<br />

rs2740210 (p = 0.04); rs2770378 (p = 0.02). Conclusions: We<br />

have examined OXT and PRL system gene variants, with no<br />

evidence <strong>of</strong> statistically significant association after multiple<br />

testing corrections. We found a nonsignificant trend between<br />

OXT SNPs and COMD. Additional analyses suggest two<br />

OXT SNPs have transmission bias to daughters, consistent<br />

with evidence in the literature that OXT is regulated by<br />

estrogen.<br />

SCHIZOPHRENIA<br />

203 NO ASSOCIATION BETWEEN THE PPARG GENE<br />

AND SCHIZOPHRENIA IN A BRITISH POPULATION<br />

A. Mathur*, M. Law, I. Megson, D. Shaw, J. Wei<br />

* aditi.mathur@uhi.ac.uk<br />

It has consistently been reported that patients with<br />

schizophrenia have an increased risk <strong>of</strong> type-2 diabetes. To<br />

investigate a genetic link between these 2 diseases, the<br />

combined effects <strong>of</strong> the PLA2G4A, PTGS2 and PPARG genes<br />

were tested among 221 British nuclear families consisting <strong>of</strong><br />

fathers, mothers and affected <strong>of</strong>fspring with schizophrenia. A<br />

total <strong>of</strong> 10 single nucleotide polymorphisms (SNPs) were<br />

tested and the likelihood-based association analysis for nuclear<br />

families was used to analyse genotyping data. Eight SNPs<br />

detected across the PPARG gene did not show allelic<br />

association with schizophrenia; a weak association was<br />

detected at rs2745557 in the PTGS2 locus (c2=4.19, p=0.041)<br />

and rs10798059 in the PLA2G4A locus (c2=4.28, p=0.039)<br />

but these associations did not survive after 10000<br />

permutations to correct the p-value (global p=0.246). The<br />

gene-gene interaction test did not show any evidence <strong>of</strong> either<br />

cis-phase interactions for the PLA2G4A and PTGS2<br />

combinations or a trans-phase interaction for the PLA2G4A<br />

and PPARG combinations. The PPARG gene has been<br />

reported to be strongly associated with type-2 diabetes, but the<br />

present study did not support the hypothesis that the PPARG<br />

gene may also play an important role in the development <strong>of</strong><br />

schizophrenia.


204 ASSOCIATION STUDY OF EFNB2 GENE AND<br />

SCHIZOPHRENIA IN THE CHINESE POPULATION<br />

R. Zhang (1), N. Zhong (1), X. Liu (2), H. Yan (2), C. Qiu (2),<br />

Y. Han (1), W. Wang (1), W. Hou (1), Y. Liu (1), C. Gao (3),<br />

T. Guo (4), S. Lu (1), H. Deng (2), J. Ma* (1)<br />

1. Department <strong>of</strong> Genetics and Molecular Biology, Xi’an<br />

Jiaotong University School <strong>of</strong> Medicine 2. School <strong>of</strong> Life<br />

Science & Technology, Xi’an Jiaotong University 3. First<br />

Affiliated Hospital, Xi’an Jiaotong University School <strong>of</strong><br />

Medicine 4. National Institutes <strong>of</strong> Diabetes and Digestive and<br />

Kidney Diseases, National Institutes <strong>of</strong> Health<br />

* tigermajie@yahoo.com.cn<br />

Recently, evidence <strong>of</strong> linkage <strong>of</strong> schizophrenia to<br />

chromosome 13q22-q34 has been demonstrated in multiple<br />

studies. Based on structure and function, EFNB2 may be<br />

considered as a compelling candidate gene for schizophrenia<br />

on chromosome13q33. We genotyped three single-nucleotide<br />

polymorphisms (SNPs: rs9520087, rs11069646, and<br />

rs8000078) in this region in 846 Han Chinese subjects (477<br />

cases and 369 controls). Significant association between an<br />

allele <strong>of</strong> marker rs9520087 and schizophrenia was found<br />

(P=0.0044). Furthermore, since no LD was observed in the<br />

three SNPs linkage disequilibrium estimation, all three SNPs<br />

were used in multiple haplotype analysis, and a strongly<br />

significant difference was found for the common haplotype<br />

TTC (P=0.004). Overall our findings indicate that EFNB2<br />

gene may be a candidate susceptibility gene for schizophrenia<br />

in the Han Chinese population, and also provide further<br />

support for the potential importance <strong>of</strong> the NMDA receptor<br />

pathway in the etiology <strong>of</strong> schizophrenia.<br />

205 THE SIGNIFICANT GENETIC REGIONS OF<br />

SCHIZOPHRENIA SUSCEPTIBILITY GENE,<br />

CALCIUM CHANNEL, VOLTAGE-DEPENDENT,<br />

GAMMA SUBUNIT (CACNG2)<br />

Y. Liu* (1), C. Liu (2), C. Fann (3), T. Liu (4), S. Tsai (5), U.<br />

Yang (6), P. Hsu (6), W. Yang (3), C. Wen (2), H. Hwu (2)<br />

1. Division <strong>of</strong> Mental Health and Addiction Medicine,<br />

Institute <strong>of</strong> Population Health Sciences, National Health<br />

Research Institutes, Taiwan 2. Department <strong>of</strong> Psychiatry,<br />

National Taiwan University Hospital and National Taiwan<br />

University College <strong>of</strong> Medicine, Taiwan 3. Institute <strong>of</strong><br />

Biomedical Sciences, Academia Sinica, Taiwan 4. Genome<br />

Research Center, National Yang-Ming University, Taiwan 5.<br />

Division <strong>of</strong> Molecular and Genomic Medicine, National<br />

Health Research Institutes, Taiwan 6. Institute <strong>of</strong><br />

Bioinformatics, National Yang-Ming University, Taiwan<br />

* ylliou@nhri.org.tw<br />

Calcium Channel, Voltage-Dependent, Gamma Subunit<br />

(CACNG2) is a schizophrenia susceptibility gene. In our<br />

previous study, the single nucleotide polymorphism (SNP)<br />

fine mapping in the region <strong>of</strong> chromosome 22 showed a<br />

statistically significant association at the intron 1 region <strong>of</strong><br />

CACNG2. In a later direct sequencing study for risk SNP <strong>of</strong><br />

CACNG2 in schizophrenia, a total <strong>of</strong> 4 SNPs were found<br />

fitting the criteria <strong>of</strong> the least minor allele frequency<br />

differences above 2% between normal controls and<br />

schizophrenia. These four SNPs (rsCACNG2-3,<br />

rsCACNG2-16, rsCACNG2-18, and rsCACNG2-20) were<br />

further genotyped on 600 normal controls and 912<br />

schizophrenic patients. A risk SNP <strong>of</strong> rsCACNG2-3 located at<br />

intron 2 showed statistically significant association with<br />

schizophrenia (odds ratio = 1.2476, p=0.008). The normal<br />

controls and the schizophrenic patients had the same<br />

haplotypes structure. Furthermore, the risk SNP demonstrated<br />

a statistically significant association with the subgroups <strong>of</strong><br />

deficit endophenotypes <strong>of</strong> sustained attention, assessed by<br />

continuous performance test (CPT; p=0.0015 for degraded and<br />

p=0.003 for undegraded CPT), and executive function,<br />

assessed by the Wisconsin Card Sorting Test (WCST;<br />

p=0.0143 for categorical achievement and p=0.0133 for<br />

perseverative error). We conclude that the intron 2 genetic<br />

region at CACNG2 may be a true susceptibility region risk for<br />

schizophrenia.


206 THE OXYTOCIN AND VASOPRESSIN GENES AS<br />

CANDIDATES FOR SCHIZOPHRENIA IN A LARGE<br />

INBRED ISRAELI ARAB PEDIGREE<br />

T. Omri* (1), K. Kyra (1), K. Osnat (1), K. Mira (2), L.<br />

Bernard (1), A. Hamdan (3), K. Yoav (1)<br />

1. Biological Psychiatry Laboratory, Department <strong>of</strong><br />

Psychiatry, Hadassah-Hebrew University <strong>Medical</strong> Center,<br />

Jerusalem, Israel 2. The Center for Genomic Technologies,<br />

Institute <strong>of</strong> Life Sciences, Hebrew University, Jerusalem,<br />

Israel 3. Regional Mental Health Center, Taibe, Israel<br />

* omri.teltsh@mail.huji.ac.il<br />

We study the genetics <strong>of</strong> schizophrenia in a large, multiplex<br />

Israeli Arab pedigree. The use <strong>of</strong> rare pedigrees, in which risk<br />

genetic variants have a major effect on the disorder<br />

occurrence, has been proven as a beneficial strategy in studies<br />

<strong>of</strong> other complex disorders. By performing a genome wide<br />

linkage analysis for schizophrenia in this pedigree, we have<br />

previously demonstrated evidence for linkage on chromosome<br />

20p13. A haplotype <strong>of</strong> 3 markers in this region, 1.8Mb in<br />

length and containing strong candidate genes for<br />

schizophrenia, was found to be shared by most affected<br />

pedigree members. More recently, we have sequenced the<br />

coding domain and flanking regions <strong>of</strong> four <strong>of</strong> these genes,<br />

among both affected and healthy individuals from the<br />

pedigree. Our sequencing results led us to concentrate on the<br />

genes OXT and AVP. The hormones encoded by these genes,<br />

Vasopressin and Oxytocin, are known to be involved in social<br />

behavior. Evidences exist for their association with OCD and<br />

autism. In the current study we completed sequencing <strong>of</strong> the<br />

intronic regions <strong>of</strong> OXT and AVP, as well as four additional<br />

regulatory sites <strong>of</strong> these genes. 7 rare SNPs were identified<br />

and studied in the whole pedigree. Four SNPs were<br />

significantly associated with schizophrenia. A 7-SNP<br />

haplotype was also significantly associated with disease<br />

status. We also study these genes contribution to<br />

schizophrenia, by genotyping the same SNPs in other samples,<br />

in order to replicate our findings. In addition, the possible<br />

effect <strong>of</strong> these SNPs on expression <strong>of</strong> the genes will also be<br />

studied.<br />

207 PRIORITIZING CANDIDATE GENES FOR<br />

SCHIZOPHRENIA: CONVERGENCE OF<br />

ASSOCIATION AND GENE EXPRESSION ANALYSES<br />

P. D. Shilling* (1), T. A. Greenwood (1), D. L. Braff (1), G.<br />

L. Light (1), K. S. Cadenhead (1), M. A. Geyer (1), J. Sprock<br />

(1), N. R. Swerdlow (1)<br />

1. University <strong>of</strong> California, San Diego<br />

* pshilling@ucsd.edu<br />

We had previously developed a database (Shilling, et al., Biol<br />

Psych 2008) composed <strong>of</strong> gene expression values in the<br />

nucleus accumbens (NAC) for two rat strains that exhibit<br />

differential prepulse inhibition (PPI) responsiveness to the<br />

dopamine agonist, apomorphine (APO). We used this database<br />

to prioritize 22 candidate genes, identified using a custom<br />

COGS/<strong>UCSD</strong> SNP chip, that are significantly associated with<br />

schizophrenia and/or at least one <strong>of</strong> the PPI-related<br />

endophenotypes examined: % PPI, startle magnitude, and<br />

habituation (Greenwood et al. 2008), for further analysis.<br />

Gene expression was measured with Affymetrix 230 2.0 chips.<br />

Twenty <strong>of</strong> the 22 genes identified by association analysis were<br />

represented on this gene chip. Gene expression <strong>of</strong> these 20<br />

genes in the NAC was analyzed in an animal model <strong>of</strong><br />

differential PPI sensitivity to APO in Long Evans vs. Sprague<br />

Dawley rats. Statistical analysis <strong>of</strong> microarrays (SAM) was<br />

employed to identify significant between strain differences in<br />

expression using a false discovery rate < 5 %. SAM analysis<br />

revealed that 10 (50%) <strong>of</strong> the significantly associated genes<br />

also exhibited significant gene expression differences. These<br />

genes include COMT, NRG1, DAT and GRIN2B. In addition,<br />

Ingenuity Pathway Analysis indicated that glutamate and<br />

dopamine receptor signaling pathways were significantly<br />

overrepresented. Many <strong>of</strong> the genes that were associated with<br />

schizophrenia and/or PPI-related endophenotypes were also<br />

differentially expressed across rat strains exhibiting<br />

differences in PPI sensitivity to a dopamine agonist. This<br />

convergent translational approach could facilitate the<br />

identification <strong>of</strong> genes that contribute to the development <strong>of</strong><br />

schizophrenia.Supported by MH42228 and MH68366


208 NEXT GENERATION SEQUENCING OF THE<br />

DISC1 LOCUS<br />

P. Thomson* (1, 2), M. De La Bastide (3), A. McRae (4), D.<br />

Soares (1, 2), J. Parla (3), K. Ramakrishnan (1, 2), W. Hennah<br />

(1, 5), K. Evans (1, 2), K. Millar (1, 2), S. Harris (1, 2, 6), S.<br />

Macgregor (4), S. Brown (1, 2), D. Rebolini (3), L. Cardone<br />

(3), M. Bell (3), J. Watson (3), J. Starr (7), I. Deary (6), D.<br />

Blackwood (1, 2, 8), W. Muir (1, 2, 8), P. Visscher (4), W.<br />

McCombie (3), D. Porteous (1, 2, 6)<br />

1. <strong>Medical</strong> Genetics Section, University <strong>of</strong> Edinburgh,<br />

Edinburgh, EH4 2XU, UK 2. Molecular Medicine Centre,<br />

Western General Hospital, Crewe Road, Edinburgh, EH4<br />

2XU, UK 3. Cold Spring Harbor Laboratory, Cold Spring<br />

Harbor, New York 11724, USA 4. Queensland Statistical<br />

Genetics, Queensland Institute <strong>of</strong> <strong>Medical</strong> Research, 300<br />

Herston Road, Brisbane, QLD, 4029, Australia 5. Institute for<br />

Molecular Medicine Finland FIMM, Nordic EMBL<br />

Partnership 6. Centre for Cognitive Ageing and Cognitive<br />

Epidemiology, Department <strong>of</strong> Psychology, University <strong>of</strong><br />

Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK 7.<br />

Centre for Cognitive Ageing and Cognitive Epidemiology,<br />

Geriatric Medicine Unit, University <strong>of</strong> Edinburgh, Royal<br />

Victoria, Craigleith Road, Edinburgh, EH4 2DN, UK 8.<br />

Department <strong>of</strong> Psychiatry, Royal Edinburgh Hospital,<br />

Morningside Park, Edinburgh, EH10 5HF, UK<br />

* pippa.thomson@ed.ac.uk<br />

DISC1 was first identified as a candidate for psychiatric<br />

illness through segregation <strong>of</strong> a t(1:11) translocation in a large<br />

Scottish family. This was confirmed by linkage <strong>of</strong> variants in<br />

DISC1 to psychiatric illness in further families from Scotland<br />

and Finland. In addition, many, although not all, association<br />

studies <strong>of</strong> DISC1 and the upstream gene, TSNAX, have<br />

detected association to a range <strong>of</strong> mental illnesses and<br />

cognitive phenotypes in multiple populations worldwide.<br />

Results from genome-wide association studies performed to<br />

date have been more equivocal. Only weakly significant<br />

p-values have been detected in these large scale studies for<br />

DISC1 and other strong candidate genes for schizophrenia,<br />

most likely due to genetic heterogeneity. In line with this a<br />

recent publication by Song et al. suggested that DISC1 exons<br />

may harbour multiple rare structural variants conferring<br />

susceptibility to schizophrenia.In order to examine the full<br />

range <strong>of</strong> variants in the 550 kb TSNAX/DISC1 region, we<br />

have sequenced up to 300 subjects for each <strong>of</strong> schizophrenia,<br />

bipolar disorder, depression, and healthy controls by high<br />

throughput sequencing on the Illumina Genome Analyzer<br />

platform. In contrast to previous studies, we have sequenced<br />

the entire genomic locus including exons, introns, promoters<br />

and intergenic regions.The results identify multiple new<br />

variants and confirm some <strong>of</strong> those identified in patient<br />

samples by Song et al. Bioinformatic analysis allows us to<br />

make structure-function predictions that support a causal role<br />

for these rare variants in major mental illness.<br />

209 ANALYSES OF GENE EXPRESSION PROFILE<br />

AND ABERRANT SPLICING IN SCHIZOPHRENIA<br />

USING EXON ARRAY<br />

Y. Ito* (1), S. Yamada (2), B. Aleksic (1), I. Kushima (1), Y.<br />

Nakamura (1), A. Yoshimi (3), T. Nagai (3), Y. Noda (2), K.<br />

Ohno (5), N. Ozaki (1)<br />

1. Department <strong>of</strong> Psychiatry, Nagoya University Graduate<br />

School <strong>of</strong> Medicine 2. Division <strong>of</strong> Clinical Sciences and<br />

Neuropsychopharmacology, Graduate School <strong>of</strong><br />

Pharmaceutical Sciences, Meijo University 3. Department <strong>of</strong><br />

Neuropsychopharmacology and Hospital Pharmacy, Nagoya<br />

University Graduate School <strong>of</strong> Medicine 4. Department <strong>of</strong><br />

Neurogenetics, Nagoya University Graduate School <strong>of</strong><br />

Medicine<br />

* y-ito@med.nagoya-u.ac.jp<br />

The contribution <strong>of</strong> genetic factors to schizophrenia is well<br />

established because recent studies have identified a number <strong>of</strong><br />

strong candidate genes. However, the pathophysiology <strong>of</strong><br />

schizophrenia has not been totally elucidated yet. Alternative<br />

pre-mRNA splicing increases proteomic diversity and<br />

provides a potential mechanism underlying both phenotypic<br />

diversity and susceptibility to genetic disorders in human<br />

populations. To identify gene/pathway dysregulation<br />

underpinning the neurobiological defects associated with<br />

schizophrenia, we compared the transcriptomes <strong>of</strong> the<br />

lymphoblastoid cell lines (LCLs) derived from 30<br />

schizophrenic patients and 30 healthy controls. LCLs were<br />

adopted to avoid the confounding factors related to the<br />

postmortem brain such as agonal-pH factors, long-term<br />

medication, and cause <strong>of</strong> death. We used the Affymetrix<br />

GeneChip Human Exon 1.0 ST Array, which has 1.4 million<br />

probe sets covering over one million known and predicted<br />

human exons. In the gene level analysis with relaxed<br />

parameters (p≤0.05, without multiple testing correction), 11<br />

genes <strong>of</strong> schizophrenia group significantly changed ≥1.5-fold<br />

or ≤0.67-fold compared to control group. In order to validate<br />

the aforementioned finding, we conducted quantitative<br />

real-time PCR using TaqMan Gene Expression Assays.<br />

Moreover, we are going to combine the genotype data <strong>of</strong> the<br />

same samples using the Genome-Wide Human SNP Array 5.0<br />

to investigate whether single nucleotide polymorphisms<br />

(SNPs) or copy number variations (CNVs) influence both<br />

gene expression and alternative splicing (AS). The exon level<br />

analysis and correlation analysis between SNPs/CNVs and<br />

expression/AS is now on going.


210 LARGE SCALE RE-SEQUENCING OF THE<br />

PSYCHOSIS SUSCEPTIBILITY GENE ZNF804A IN<br />

SCHIZOPHRENIA<br />

S. Dwyer* (1), M. Owen (1), N. Craddock (1), H. Williams<br />

(1), M. O'Donovan (1)<br />

1. Cardiff University<br />

* dwyersl@cf.ac.uk<br />

Recently, we undertook a GWAS <strong>of</strong> 479 UK schizophrenia<br />

cases and 3000 controls (O'Donovan et al. 2008), with the<br />

strongest 12 GWAS findings (P


212 MCHR1 SHOWS ASSOCIATION WITH<br />

SCHIZOPHRENIA, AND POSSIBLY TREATMENT<br />

RESPONSE AMONG DANISH CHRONIC<br />

SCHIZOPHRENIA PATIENTS<br />

A. Hedemand* (1), D. Demontis (1), M. Nyegaard (1), J.<br />

Christensen (1), J. Severinsen (1), T. Hansen (2), T. Werge<br />

(2), O. Mors (3), A. Børglum (1)<br />

1. Institute <strong>of</strong> Human Genetics, Aarhus University, DK-8000<br />

Århus C, Denmark 2. Research Institute <strong>of</strong> Biological<br />

Psychiatry, Mental Health Centre Sct. Hans, Copenhagen<br />

University Hospital, Roskilde, Denmark 3. Centre for<br />

Psychiatric Research, Aarhus University Hospital, DK-8240<br />

Risskov, Denmark<br />

* anneh@humgen.au.dk<br />

The gene MCHR1 is located on chromosome 22q13.2 and has<br />

previously been associated with schizophrenia (SZ) in a study<br />

<strong>of</strong> cases and controls from the Faeroe Islands and Scotland.<br />

Here we report support <strong>of</strong> association between MCHR1 and<br />

SZ based on analyses <strong>of</strong> a larger sample and increased number<br />

<strong>of</strong> SNPs than used in the previous study. We analyzed a<br />

case-control sample from Denmark consisting <strong>of</strong> 390 patients<br />

with SZ and 814 control individuals, genotyped with 16<br />

tag-SNPs. We performed gender-specific analysis as well as<br />

analysis <strong>of</strong> association with antipsychotic treatment. We found<br />

significant associations <strong>of</strong> independent SNPs in the proximal<br />

region <strong>of</strong> GPR24 with SZ. The associations seemed to be<br />

gender specific, coming from the males, and could be related<br />

to medical treatment response. After correction for multiple<br />

testing a single SNP remained significant (P=0.003), in the<br />

association analysis <strong>of</strong> the males. A gene-wide p-value <strong>of</strong><br />

0.006 was obtained after correction for multiple testing. The<br />

results suggest that MCHR1 may influence SZ susceptibility<br />

in the Danish population and may possibly be involved in<br />

treatment response among SZ patients.<br />

213 UBIQUITIN PROTEASOME SYSTEM GENE<br />

EXPRESSION CORRELATES FOR POSITIVE AND<br />

NEGATIVE SYMPTOMS IN PSYCHOSIS<br />

C. Bousman* (1), E. Tatro (1), G. Chana (1), S. Glatt (2), S.<br />

Chandler (1), T. May (1), J. Lohr (1), W. Kremen (1), M.<br />

Tsuang (1), I. Everall (1)<br />

1. <strong>UCSD</strong> 2. SUNY Upstate <strong>Medical</strong> University<br />

* cbousman@ucsd.edu<br />

Several gene expression studies have identified genes in the<br />

ubiquitin proteasome system (UPS) pathway as putative<br />

biomarkers in psychotic disorders (e.g. schizophrenia).<br />

However, to date an in-depth examination <strong>of</strong> the UPS pathway<br />

in the context <strong>of</strong> psychosis has not been conducted. The<br />

purpose <strong>of</strong> this study was to explore blood-based expression<br />

correlates <strong>of</strong> 43 well characterized genes that operate within<br />

the UPS pathway among a sample <strong>of</strong> 19 psychotic patients<br />

diagnosed with schizophrenia (n = 13) or bipolar disorder (n =<br />

6). Specifically, we sought to estimate the association <strong>of</strong> each<br />

gene’s expression with clinical scores on the scales for the<br />

assessment <strong>of</strong> positive and negative symptoms (SAPS-SANS).<br />

Pearson’s partial correlations, adjusting for gender, ethnicity,<br />

age, education, medication, smoking, and past six-month<br />

substance use, were performed between each <strong>of</strong> the selected<br />

UPS genes and both scales. SAPS results revealed significant<br />

positive associations (Bonferroni p < 0.05) for two ubiquitin<br />

conjugation genes (i.e. UBE2K, SIAH2) and a negative<br />

association with one deubiquitination gene (i.e. USP2). No<br />

gene expression levels were significantly associated with<br />

scores on the SANS after multiple testing correction, albeit a<br />

positive trend was observed for UBE2K. Findings suggest<br />

that the UPS pathway, specifically ubiquitin conjugation and<br />

deubiquitination may point to a possible underlying biological<br />

mechanisms by which the severity <strong>of</strong> positive but not negative<br />

symptoms clinically manifest.


214 CHRONIC RESTRAINT STRESS RESULTS IN<br />

DIFFERENTIAL EXPRESSION OF SPLICE VARIANTS<br />

OF THE SCHIZOPHRENIA AND BIPOLAR<br />

DISORDER ASSOCIATED BRD1 GENE IN RAT<br />

HIPPOCAMPUS<br />

J. Christensen* (1), T. Fryland (1), B. Elfving (2), M.<br />

Nyegaard (1), T. Corydon (1), A. Nielsen (1), O. Mors (2), G.<br />

Wegener (2), A. Børglum (1)<br />

1. Department <strong>of</strong> Human Genetics, Aarhus University,<br />

Denmark 2. Centre for Psychiatric Research, Aarhus<br />

University Hospital, Risskov, Denmark<br />

* jhc@humgen.au.dk<br />

Recent evidence has implicated the bromodomain containing 1<br />

gene (BRD1) with brain development and susceptibility to<br />

schizophrenia (SZ) and bipolar affective disorder (BD). The<br />

Brd1 protein has been identified in the MOZ/MORF complex<br />

responsible for H3 histone acetylation. Although indicating a<br />

role <strong>of</strong> Brd1 in transcriptional regulation much remains to be<br />

clarified with regard to its function and regulation. The aim <strong>of</strong><br />

the present study was to investigate the regulation and splicing<br />

<strong>of</strong> the Brd1 gene in rats subjected to chronic restraint stress<br />

(CRS). Male Sprague-Dawley rats (n=36) were subjected to<br />

either half an hour or 6 hours <strong>of</strong> CRS once a day for 21 days,<br />

or no CRS (controls). Relative transcript levels <strong>of</strong> the long<br />

exon 7 splice variant <strong>of</strong> Brd1, total Brd1, and Mchr1 were<br />

determined by quantitative RT-PCR on hippocampus RNA<br />

using two stably expressed genes for normalization. We found<br />

a significant 2-fold upregulation <strong>of</strong> the long splice variant <strong>of</strong><br />

Brd1 in both groups <strong>of</strong> CRS rats compared to controls. The<br />

transcript levels <strong>of</strong> total Brd1 was not changed during CRS<br />

indicating an equivalent downregulation <strong>of</strong> any other splice<br />

variants. The amount <strong>of</strong> the Mchr1 transcript, another gene<br />

associated with SZ, was unchanged. In conclusion, we<br />

demonstrate that CRS in rats results in differential expression<br />

<strong>of</strong> splice variants <strong>of</strong> the Brd1 gene in the hippocampus. As<br />

CRS causes decreased hippocampal neurogenesis and adaptive<br />

structural plasticity, it is tempting to speculate that regulation<br />

and splicing <strong>of</strong> the Brd1 gene plays an important role in these<br />

processes that are well-known to be disturbed in SZ and BD.<br />

215 IDENTIFICATION OF NOVEL CANDIDATE<br />

GENES FOR TREATMENT RESPONSE TO<br />

RISPERIDONE AND SUSCEPTIBILITY FOR<br />

SCHIZOPHRENIA<br />

M. Ikeda* (1, 2), Y. Tomita (3), A. Mouri (4), M. Koga (5, 6),<br />

T. Okochi (2), R. Yoshimura (7), Y. Yamanouchi (2), Y.<br />

Kinoshita (2), R. Hashimoto (6, 8), H. Williams (1), M.<br />

Takeda (8), J. Nakamura (7), T. Nabeshima (4), M. Owen (1),<br />

M. O'Donovan (1), H. Honda (3), T. Arinami (5, 6), N. Ozaki<br />

(3, 6), N. Iwata (2, 6)<br />

1. Cardiff University 2. Fujita Health University 3. Nagoya<br />

University 4. Meijo University 5. University <strong>of</strong> Tsukuba<br />

6. CREST 7. University <strong>of</strong> Occupational and Environmental<br />

Health 8. Osaka University<br />

* ikedam@cardiff.ac.uk<br />

Background: Pharmacogenomic approaches based upon<br />

genome-wide sets <strong>of</strong> single nucleotide polymorphisms (SNPs)<br />

are now feasible, and <strong>of</strong>fer the potential to uncover variants<br />

that influence drug response. Methods: To detect potential<br />

predictor gene variants for risperidone response in<br />

schizophrenic subjects, we performed a convergent analysis<br />

based upon (1) a genome-wide (100K SNP) SNP<br />

pharmacogenetic study <strong>of</strong> risperidone response and (2) a<br />

global transcriptome study <strong>of</strong> genes whose mRNA levels are<br />

influenced by risperidone exposure in mouse prefrontal cortex.<br />

Results: Fourteen genes were highlighted as <strong>of</strong> potential<br />

relevance to risperidone activity in both studies; ATP2B2,<br />

HS3ST2, UNC5C, BAG3,PDE7B, PAICS, PTGFRN, NR3C2,<br />

ZBTB20, ST6GAL2, PIP5K1B, EPHA6, KCNH5 and AJAP1.<br />

The SNPs related to these genes that were associated in the<br />

pharmacogenetic study were further assessed for evidence for<br />

association with schizophrenia in up to 3 case control series<br />

comprising 1564 cases and 3862 controls in total (Japanese<br />

(JPN) 1st and 2nd samples and UK sample). Of 14 SNPs<br />

tested, one (rs9389370) in PDE7B showed significant<br />

evidence for association with schizophrenia in a discovery<br />

sample (Pallele=0.026 in JPN1st, 2-tailed). This finding<br />

replicated in a joint analysis <strong>of</strong> two independent case-control<br />

samples (PJPN2nd+UK=0.008, 1-tailed, uncorrected) and in<br />

all combined datasets (Pall=0.0014, 2-tailed, uncorrected and<br />

Pall=0.018, 2-tailed, Bonferroni correction). Conclusions: We<br />

identify novel candidate genes for treatment response to<br />

risperidone, and provide evidence that one <strong>of</strong> these<br />

additionally may confer susceptibility to schizophrenia.<br />

Specifically, PDE7B is an attractive candidate gene though<br />

evidence from integrated methodology including<br />

pharmacogenomics, pharmacotranscriptomic, and case-control<br />

association approaches.


216 NPAS3 AND SOX FAMILY GENES:<br />

TRANSCRIPTIONAL CONTROL OF NEURONAL<br />

PROLIFERATION AND DIFFERENTIATION AND ITS<br />

RELATION TO PSYCHIATRIC ILLNESS<br />

L. Sha* (1), M. Malloy (2), W. Muir (3), D. Blackwod (3), B.<br />

Pickard (4)<br />

1. University <strong>of</strong> Edinburgh, Western General Hospital,<br />

Einburgh, EH4 2XU, UK 2. University <strong>of</strong> Edinburgh,<br />

Einburgh, EH4 2XU, UK 3. Division <strong>of</strong> Psychiatry, University<br />

<strong>of</strong> Edinburgh, EH10 5HF, UK 4. University <strong>of</strong> Strathclyde,<br />

Glasgow, G4 0NR, UK<br />

* s0676701@sms.ed.ac.uk<br />

Neuronal basic helix-loop-helix PAS domain transcription<br />

factor 3, NPAS3, has been implicated in susceptibility to both<br />

schizophrenia and bipolar disorder by cytogenetics,<br />

genome-wide association and a knockout mouse model.<br />

Studying the established role <strong>of</strong> NPAS3 in adult hippocampal<br />

neurogenesis may provide a biological mechanism for the link<br />

between neurogenesis dysfunction and psychiatric disorders.<br />

SOX family transcription factors are also known to play a role<br />

in neural progenitor proliferation and differentiation. To<br />

explore the transcriptional network controlled by these genes<br />

we used overexpression in HEK293 cells coupled with<br />

microarray analysis to identify target genes and pathways.<br />

Four key observations were made. Firstly, NPAS3 strongly<br />

activates VGF, a gene that links defective neurogenesis with<br />

depression in a mouse model. Secondly, NPAS3 regulates<br />

several SOX genes, including SOX11, and displays a similar<br />

target activation pr<strong>of</strong>ile to SOX5/SOX6. Thirdly, NPAS3, like<br />

NPAS2, is implicated in energy homeostasis by repressing<br />

genes encoding the majority <strong>of</strong> enzymes involved in<br />

glycolysis. Lastly, SOX11, which plays a key role during<br />

neuronal differentiation activates expression <strong>of</strong> a cluster <strong>of</strong><br />

histone genes on chromosome 6 that lie within the principal<br />

region associated with schizophrenia in recent genome-wide<br />

association studies. Q-PCR confirmation studies will be<br />

presented along with immun<strong>of</strong>luorescence findings <strong>of</strong> the<br />

NPAS3, SOX11 and target proteins in the hippocampus. We<br />

will also detail ongoing work attempting to place these genes<br />

within the time-course <strong>of</strong> neural prescursor cell differentiation.<br />

These data begin to unravel the component pathways <strong>of</strong><br />

neurogenesis and how they might impact on the aetiological<br />

processes <strong>of</strong> psychiatric illness.<br />

217 ANALYSIS OF AN ASSOCIATION BETWEEN THE<br />

DRD3, TPH2 AND HTR-2A GENES POLYMORPHISM<br />

AND SCHIZOPHRENIA (SCHZ) IN MALAYSIAN<br />

INDIVIDUALS<br />

S. Tee*, T. Chow, H. Loh, P. Tang<br />

* teesf@utar.edu.my<br />

Molecular components <strong>of</strong> the dopaminergic and serotoninergic<br />

pathways may play an important role in the pathophysiology<br />

<strong>of</strong> schizophrenia. The roles <strong>of</strong> the dopaminergic and<br />

serotoninergic systems have been implicated in the etiology <strong>of</strong><br />

schizophrenia (SCHZ) and other behavioral disorders. In this<br />

study, we investigated the relationship <strong>of</strong> the single nucleotide<br />

polymorphisms (SNPs) <strong>of</strong> Ser9Gly DRD3, rs-1438G/A<br />

5-HTR2A and intronic rs1386494 TPH2 with Malaysia<br />

cases-control. A total <strong>of</strong> 201 patients with SCHZ and 111<br />

healthy controls were investigated. PCR-RFLP was performed<br />

to genotype these three SNPs and deviation from Hardy<br />

Weinberg Equilibrium was determined. Allele and genotype<br />

distributions <strong>of</strong> rs-1438G/A 5-HTR2A showed a significant<br />

difference for cases and controls. However, no associations<br />

were found between the Ser9Gly DRD3 and intronic<br />

rs1386494 TPH2 with SCHZ while rs-1438G/A 5-HTR2A<br />

could be a susceptible gene for SCHZ based our preliminary<br />

results. We believe that further studied are required to<br />

examine the relationship between other dopamine and<br />

serotonin-related genes and the behavioral phenotypes <strong>of</strong><br />

SCHZ in the Malaysia individuals.


218 CYTOGENETIC DAMAGE IN SCHIZOPHRENIA<br />

POPULATION, TAMIL NADU, INDIA.<br />

V. Balachandar* (1)<br />

1. Human Genetics Laboratory, Bharathiar UNiversity,<br />

Coimbatore, Tamil Nadu, India<br />

*geneticbala@yahoo.co.in<br />

Schizophrenia is a relatively common debilitating, chronic,<br />

psychotic disorder. The focal aim <strong>of</strong> the present investigation<br />

was to identify the foremost chromosomal aberrations (CA)<br />

like deletion, translocation, inversion and mosaic in<br />

schizophrenic subjects <strong>of</strong> Tamilnadu, Southern India. In the<br />

present investigation 54 schizophrenia patients from 15 to<br />

58yrs <strong>of</strong> age and equal number <strong>of</strong> normal,<br />

Healthy controls in the same age wise manner were selected.<br />

After signing a consent form, both cases (experimentals and<br />

controls) provided a blood sample (5 ml) to establish cell<br />

culture. In 41 patients random numerical and structural<br />

aberrations were identified. Structural aberrations<br />

predominated, consisted <strong>of</strong> deletions, translocation, inversion<br />

and mosaicism <strong>of</strong> various chromosomes. Our study has<br />

detected 1, 7, 9, 11, 13, 21, and X suggested that these<br />

chromosomal scratches are prevalent in schizophrenics. In<br />

comparison with experimental subjects, the control subjects<br />

exhibited very low levels <strong>of</strong> major CA (P


220 DENSE ASSOCIATION MAPPING OF<br />

DIHYDROPYRIMIDINASE-LIKE 2 (DPYSL2) GENE IN<br />

SUSCEPTIBILITY TO SCHIZOPHRENIA IN THE<br />

JAPANESE POPULATION<br />

T. Koide*, B. Aleksic, I. Kushima, Y. Nakamura, Y. Ito, T.<br />

Inada, N. Iwata, N. Ozaki<br />

*t-koide@med.nagoya-u.ac.jp<br />

Dihydropyrimidinase-like 2 (DPYSL2), also known as<br />

collapsin response mediator protein 2 (CRMP2), mediates the<br />

intracellular response to collapsin, a repulsive extracellular<br />

guidance cue or axonal outgrowth. DPYSL2 was reported as a<br />

schizophrenia susceptibility gene by Nakata et al, and several<br />

independent association studies have replicated this<br />

association signal. However, the results <strong>of</strong> replication studies<br />

using smaller sets <strong>of</strong> markers have been inconsistent.<br />

Therefore, in order to address this issue, we performed dense<br />

association mapping <strong>of</strong> DPYSL2 in a Japanese population.<br />

SNP Tagging criteria were based on minor allele frequency<br />

(>10 %) and correlation coefficient between loci (>0.8) as<br />

reported in HAPMAP database.After genotyping <strong>of</strong> 23 SNPs,<br />

we identified 2 association signals in a statistical analysis<br />

(rs2585458; allelic P value: 0.048, rs4733048; genotypic P<br />

value: 0.02 and allelic P value: 0.01, before multiple<br />

comparison correction). In order to confirm this finding, we<br />

are typing independent cohorts for these 2.Our data provided<br />

evidence that DPYSL2 might elevate the risk for<br />

schizophrenia in the Japanese population. However,<br />

replicating the finding using larger sample size would be<br />

necessary for conclusive results. We concluded that the results<br />

were suggestive <strong>of</strong> an association with schizophrenia at this<br />

locus, but that they could not be considered conclusive<br />

without further replications.<br />

221 FURTHER EVIDENCE FOR ASSOCIATION OF<br />

WNT PATHWAY RELATED GENES WITH<br />

SCHIZOPHRENIA<br />

I. Kushima* (1), B. Aleksic (1), Y. Ito (1), Y. Nakamura (1),<br />

N. Iwata (2), N. Ozaki (1)<br />

1.Nagoya University Department <strong>of</strong> Psychiatry 2. Fujita health<br />

University Department <strong>of</strong> Psychiatry<br />

*kushima.itaru@a.mbox.nagoya-u.ac.jp<br />

Abnormalities in brain development, such as faulty neuronal<br />

migration, altered spatial neuronal arrangement and the<br />

absence <strong>of</strong> significant gliosis, have been reported in<br />

schizophrenia. Recent advances in the understanding <strong>of</strong> genes<br />

that regulate brain development <strong>of</strong>fer insights into the<br />

mechanism that underlie such developmental brain changes.<br />

One such genetic components involves the Wnt signaling<br />

which is central to normal brain development and which<br />

mediates cell adhesion, proliferation and gene transcription.<br />

Both DKK1 and KREMEN1 are on the Wnt pathway. DKK1<br />

gene encodes a protein that is a member <strong>of</strong> the dickkopf<br />

family. It is a secreted protein with two cysteine rich regions<br />

and is involved in embryonic development through its<br />

inhibition <strong>of</strong> the Wnt signaling. KREMEN1 encodes a<br />

high-affinity DKK1 transmembrane receptor that functionally<br />

cooperates with DKK1 to block Wnt signaling. KREMEN1 is<br />

located in chromosome 22 q12.1, a region implicated in<br />

several schizophrenia linkage studies.The aim <strong>of</strong> the current<br />

study was to examine the association <strong>of</strong> KREMEN1 and<br />

DKK1 and schizophrenia in a Japanese population. We<br />

genotyped 16 genetic variants within the two genes and<br />

examined their association with schizophrenia. Our samples<br />

consisted <strong>of</strong> 1573 patients with schizophrenia and 1580<br />

healthy controls divided into two sets. All subjects were <strong>of</strong><br />

Japanese descent. One variant in KREMEN1, rs713526 was<br />

significantly associated with schizophrenia. We replicated this<br />

association at the second sample sets.


222 AN ASSOCIATION STUDY OF TAGGING SNPS OF<br />

RAB23 AND SCHIZOPHRENIA IN JAPANESE<br />

POPULATION<br />

B. Aleksic* (1), I. Kushima (1), Y. Ito (1), Y. Nakamura (1),<br />

T. Inada (1), N. Iwata (1), N. Ozaki (1)<br />

1. Department <strong>of</strong> Psychiatry, Graduate School <strong>of</strong> Medicine,<br />

Nagoya University, Japan<br />

*branko@med.nagoya-u.ac.jp<br />

RAB23, member RAS oncogene family, is a human gene. The<br />

protein encoded by this gene belongs to the small GTPase<br />

superfamily, Rab family. It is involved in small GTPase<br />

mediated signal transduction and intracellular protein<br />

transportation. Alternative splicing occurs at this locus and<br />

two transcript variants encoding the same protein have been<br />

identified. In the brain, RAB23 is found predominantly in cell<br />

bodies <strong>of</strong> neurons. This gene is attractive functional candidate<br />

gene for schizophrenia, and so far there was no association<br />

study focused on this locus. Therefore, the goal <strong>of</strong> this study<br />

was to assess the influence <strong>of</strong> genetic factors related to this<br />

locus on schizophrenia from point <strong>of</strong> common disease<br />

common variant hypothesis. Total <strong>of</strong> 11 SNPs were selected<br />

for comprehensive coverage <strong>of</strong> RAB23 locus. Tagging criteria<br />

are based on minor allele frequency (>5 %) and correlation<br />

coefficient between loci (>0.8) as reported in HAPMAP<br />

database. Our sample consisted <strong>of</strong> 754 schizophrenic patients<br />

and 783 healthy controls. We detected 3 association signals in<br />

3’ region <strong>of</strong> the RAB23.Our data provided evidence that<br />

common SNP within RAB23 locus might elevate the risk for<br />

schizophrenia in the Japanese population. However,<br />

replicating the finding using larger sample size would be<br />

necessary for conclusive results. We concluded that the results<br />

were suggestive <strong>of</strong> an association with schizophrenia at this<br />

locus, but that they could not be considered conclusive<br />

without further replications.<br />

223 TAG-SNP BASED ASSOCIATION STUDY OF 109<br />

CANDIDATE GENES FOR SCHIZOPHRENIA<br />

SELECTED USING AN EVIDENCE-BASED<br />

PRIORITIZATION ALGORITHM<br />

A. Fanous* (1), Z. Zhao (2), B. Webb (3), E. van den Oord<br />

(3), B. Maher (3), S. Bergen (3), R. Amdur (1), F. O'Neill (4),<br />

D. Walsh (5), B. Riley (3), K. Kendler (3)<br />

1. Washington VA <strong>Medical</strong> Center 2. Vanderbilt University<br />

3. Virginia Commonwealth University 4. Queen's University,<br />

Belfast 5. Health Research Board, Dublin<br />

*ahfanous@vcu.edu<br />

Integrating evidence from multiple domains may be useful in<br />

prioritizing genes for subsequent testing. We ranked all<br />

known genes under several linkage peaks in the Irish Study <strong>of</strong><br />

High-Density Schizophrenia Families using three different<br />

evidence domains; 1) a meta-analysis <strong>of</strong> microarray gene<br />

expression results using the Stanley Brain collection, 2) a<br />

schizophrenia protein-protein interaction network, and 3)<br />

systematic literature searching. Each gene was assigned a<br />

domain specific p-value and ranked after evaluating the<br />

evidence within each domain. For Protein-Protein Interaction,<br />

the steps from the schizophrenia susceptibility genes a)<br />

DTNBP1, b) NRG1, and c) Akt1 in Protein-Protein<br />

Interaction databases were determined, respectively. For<br />

Literature Searches, Pubmed was systematically searched<br />

using all genes with 40 schizophrenia-related search terms.<br />

For a final ranking <strong>of</strong> these genes, we summed the -log10 <strong>of</strong><br />

their P-values on each <strong>of</strong> these five variables (or Q-values in<br />

the case <strong>of</strong> Gene Expression). In addition, any gene with Gene<br />

Ontology terms that included “nervous system development”<br />

or “brain development” was also selected (n=65).<br />

Approximately 3000 SNPs from a custom Illumina iSelect<br />

genotyping array using the Infinium assay were allocated to<br />

evaluate selected genes. After assigning all LD tagging SNPs<br />

in ranked genes, these, and the top 44 genes were investigated<br />

using 2,849 SNPs. No individual SNPs were significant after<br />

correction for multiple testing. However, a number <strong>of</strong> genes<br />

contained clusters <strong>of</strong> SNPs with p


224 GENOME-WIDE ASSOCIATION STUDY OF<br />

SCHIZOPHRENIA IN A SWEDISH<br />

POPULATION-BASED SAMPLE<br />

Y. Liu* (1), C. Hultman (2), P. Lichtenstein (2), P. Sklar (3),<br />

P. Sullivan (1)<br />

1. The University North Carolina at Chapel Hill, Department<br />

<strong>of</strong> Genetics 2. Karolinska Institute 3. Massachusetts General<br />

Hospital<br />

*youfang@email.unc.edu<br />

Schizophrenia is a complex disease with multiple genes and<br />

environmental exposures likely involved. This study includes<br />

three sets <strong>of</strong> Swedish cases and controls totaling 1097 cases<br />

and 1303 controls with all individuals selected from<br />

population-based registries. Cases were hospitalized at least<br />

twice with a discharge diagnosis <strong>of</strong> schizophrenia or<br />

schizoaffective disorder (we have previously shown that this<br />

definition <strong>of</strong> illness corresponds closely to standard<br />

definitions). Controls were required never to have been<br />

hospitalized for a psychotic illness. GWAS genotyping was<br />

conducted at the Broad Institute using Affymatrix 5.0 and 6.0<br />

arrays. To ensure data quality, harsh quality control was<br />

performed: SNPs with missing rate > 0.01, MAF < 0.05 or<br />

HWE deviations were excluded. Subjects with missing rate ><br />

0.01, 2nd or 1st degree relationship with other individuals, and<br />

outlying empirical ancestries were removed. Imputation was<br />

accomplished by MACH for each data set. GWAS was<br />

performed by ProbABEL using logistic regression.<br />

Fixed-effects meta-analysis was applied using a weighted<br />

z-score method and its distribution was checked for inflation<br />

by (lambda = 1.00). Although no marker reached<br />

genome-wide significance, the top two genes were interesting.<br />

MAGI2 (Pmin= 1.3e-6) is a possible scaffold molecule at<br />

synaptic junctions and may assemble neurotransmitter<br />

receptors and cell adhesion proteins and regulate<br />

activin-mediated signaling in neuronal cells. The second gene,<br />

GABRB3 (Pmin=3.7e-6), is a ligand-gated ionic channel<br />

family and encodes a subunit <strong>of</strong> the receptor for<br />

gamma-aminobutyric acid which is the major inhibitory<br />

transmitter <strong>of</strong> the nervous system, and a prior candidate gene<br />

for schizophrenia.<br />

225 EXTENDED HAPLOTYPE SHARING IN<br />

SCHIZOPHRENIA<br />

Y. Liu* (1), K. Wilhelmsen (1), C. Hultman (2), P.<br />

Lichtenstein (2), P. Sklar (3), P. Sullivan (1)<br />

1. The University North Carolina at Chapel Hill, Department<br />

<strong>of</strong> Genetics 2. Karolinska Institutet 3. Massachusetts General<br />

Hospital<br />

*youfang@email.unc.edu<br />

Recently, a long segment with high LD containing hundreds<br />

SNPs and multiple genes in the MHC region on chromosome<br />

6p21.3-22.1 was identified by association analyses in<br />

European schizophrenia studies. Thus, long haplotype or long<br />

DNA segment based association methodology is worthy <strong>of</strong><br />

exploration for relevance to schizophrenia. Haplotype sharing<br />

methods have been shown by simulation to achieve higher<br />

power than single maker analysis. However, most current<br />

haplotype sharing methods can only handle short haplotypes.<br />

CHAT (Convergent Haplotype Association Tagging), a new<br />

long haplotype sharing method developed by Dr Kirk<br />

Wilhelmsen (unpublished), implements a computationally<br />

intensive heuristic algorithm that identifies long shared<br />

haplotypes for which subsets <strong>of</strong> the population are likely to<br />

have inherited from a recent common ancestor. CHAT is<br />

designed to detect mutations <strong>of</strong> moderate penetrance and is<br />

resistant to the confounding effects <strong>of</strong> population substructure<br />

and allelic and locus heterogeneity. We applied CHAT to an<br />

unpublished schizophrenia case-control data set from a<br />

population based sample <strong>of</strong> 538 cases and 904 controls<br />

genotyped at the Broad Institute using Affymatrix 6.0 arrays.<br />

Harsh quality control was performed before analysis. Two<br />

regions were identified by CHAT. Both are rare variants<br />

which were not detected by single marker analysis - a 350 Kb<br />

region located on chromosome 3 containing one possible gene<br />

and a 3.8 Mb region located on chromosome 15 containing<br />

more than 20 genes involved in FGFR signaling pathway and<br />

lipid and lipoprotein metabolism – although the MHC region<br />

was not detected.


226 ASSOCIATION BETWEEN COMT GENE<br />

MET158VAL POLYMORPHISM AND<br />

SCHIZOPHRENIA<br />

H. Loh* (1), S. Tee (2), T. Chow (3), P. Tang (4)<br />

1. Universiti Tunku Abdul Rahman<br />

*hcloh@utar.edu.my<br />

Catechol-O-methyltransferase (COMT) is an enzyme involved<br />

in the degradation <strong>of</strong> the neurotransmitter dopamine (DA). A<br />

single nucleotide polymorphism (SNP) within the<br />

humanCOMT gene dramatically affects the activity <strong>of</strong> the<br />

enzyme and as a result, directly influences DA availability in<br />

the brain. In this study, we examined the relationship <strong>of</strong> the<br />

single nucleotide polymorphism (SNP) between the COMT<br />

Met158Val polymorphism in 123 healthy individuals and 143<br />

schizophrenic patients (MINI criteria) in Malaysia.<br />

PCR-RFLP was performed to genotype the COMT Met158Val<br />

SNP. Allele and genotype distributions showed a significant<br />

difference between Malaysia cases and controls. Both <strong>of</strong> the<br />

groups are out <strong>of</strong> Hardy -Weinberg equilibrium for the<br />

analyzed genetic variability. Further studies are required to<br />

examine the relationship between dopamine-related genes and<br />

the behavioral phenotypes <strong>of</strong> schizophrenia in the Malaysia<br />

population.<br />

227 FIBROBLAST GROWTH FACTORS AND BRAIN<br />

VOLUME IN SCHIZOPHRENIA PATIENTS AND<br />

HEALTHY CONTROLS<br />

A. Terwisscha van Scheltinga* (1), S. Bakker (1), N. van<br />

Haren (1), H. Hulsh<strong>of</strong>f Pol (1), W. Cahn (1), R. Oph<strong>of</strong>f (1), R.<br />

Kahn (1)<br />

1. UMC Utrecht<br />

*aterwiss@umcutrecht.nl<br />

Background: There is convincing evidence for an involvement<br />

<strong>of</strong> fibroblast growth factors (FGFs) in schizophrenia 1. FGFs<br />

regulate growth, maintenance and repair <strong>of</strong> neuronal tissue.<br />

This makes the genes in the FGF system plausible candidate<br />

genes for the smaller brain volume seen in schizophrenia<br />

patients and their relatives relative to control subjects. We use<br />

total brain volume as an endophenotype <strong>of</strong> schizophrenia,<br />

since it has a 90% heritability, can be reliably measured,<br />

cosegregates with illness in families and is relatively<br />

independent <strong>of</strong> clinical state 2. Method: Using online genomic<br />

databases 915 SNPs in 22 FGFs, 5 FGF receptors and 56<br />

interacting genes were selected and genotyped using the<br />

Illumina HumanHap550 beadchip. A 1.5T MRI scan <strong>of</strong> the<br />

brain was acquired for 169 schizophrenia cases and 159<br />

healthy controls. Total brain volume was estimated and<br />

corrected for age, sex and intracranial volume. A linear<br />

regression analysis was performed, using disease status as an<br />

independent variable. Results: After correction for 83 genes 1<br />

SNP in fibroblast growth factor receptor substrate 2 (FRS2)<br />

showed suggestive association with total brain volume<br />

(corrected p = 0.056) both in patients and controls, explaining<br />

3% <strong>of</strong> the variation in total brain volume. Discussion: This<br />

SNP is associated with the normal variation in total brain<br />

volume rather than with the disease specific decline in total<br />

brain volume.


228 IDENTIFICATION OF THE SUBCELLULAR<br />

LOCALIZATION DISC1 ISOFORMS AND THE<br />

IMPACT OF A NOVEL DISC1 ISOFORM, DISC1-ESV,<br />

IN SCHIZOPHRENIA<br />

C. Wen* (1), C. Liu (1), Y. Liu (2), H. Hwu (1)<br />

1. Department <strong>of</strong> Psychiatry, National Taiwan University<br />

Hospital and National Taiwan University College <strong>of</strong><br />

Medicine, Taipei, Taiwan 2. Division <strong>of</strong> Mental Health and<br />

Substance Abuse Research, National Health Research<br />

Institutes, Miaoli, Taiwan<br />

*f90445111@ntu.edu.tw<br />

Schizophrenia is one <strong>of</strong> the neuropsychiatry disorders. Genetic<br />

studies have indicated that many genes are associated with<br />

schizophrenia. Disrupted-in-schizophrenia 1 (DISC1) was first<br />

identified in a large Scottish family which had been reported<br />

as one <strong>of</strong> the risk gene involved in the pathogenesis <strong>of</strong><br />

schizophrenia. Previous studies indicated that endogenous<br />

DISC1 could locate in nucleus, cytoplasm, centrosome and<br />

mitochondria, and DISC1-L form could locate in nucleus and<br />

cytoplasm. The functions <strong>of</strong> DISC1 might involve with neurite<br />

outgrowth and cell cycle regulation; however, the detail<br />

functions <strong>of</strong> DISC1 were still unclear. DISC1 included at least<br />

four is<strong>of</strong>orms but, few <strong>of</strong> studies described the functions <strong>of</strong> all<br />

DISC1 is<strong>of</strong>orms. In this paper, we wanted to identify the<br />

functions and cellular distributions <strong>of</strong> all DISC1 is<strong>of</strong>orms. It is<br />

the first time to study the difference between all DISC1<br />

is<strong>of</strong>orms. We cloned DISC1 is<strong>of</strong>orms expression constructs<br />

included DISC1-L, DISC1-Lv, DISC1-S and a novel is<strong>of</strong>orm,<br />

DISC1-Esv, from human hippocampus but we failed to clone<br />

DISC1-Es form. Immun<strong>of</strong>luorenscence assay indicated that<br />

majority <strong>of</strong> DISC1-L and Lv form could be observed in<br />

cytoplasm and DISC1-S could be detected both in nucleus and<br />

cytoplasm and DISC1-Esv also could be recognized both in<br />

nucleus and cytoplasm, but DISC1-Esv with dense stained in<br />

nucleus. The subcellular localization <strong>of</strong> DISC1-L, Lv and Esv<br />

could change after retinoic acid treatment. Furthermore, the<br />

RNA expression level <strong>of</strong> DISC1-Esv in schizophrenia patient<br />

was higher than that in control. Taken together, DISC1-L Lv<br />

and Esv might involve in neurite outgrowth regulation, and<br />

DISC1-Esv might be an candidate risk protein in Taiwanese<br />

schizophrenia population.<br />

229 NO ASSOCIATION OF THE SEROTONIN<br />

TRANSPORTER POLYMORPHISMS 5-HTTLPR AND<br />

RS25531 WITH SCHIZOPHRENIA OR<br />

NEUROCOGNITION<br />

T. Konneker* (1), J. Crowley (1), C. Quackenbush (1), R.<br />

Keefe (2), D. Perkins (3), T. Stroup (3), J. Lieberman (4), E.<br />

van den Oord (5), P. Sullivan (6)<br />

1. Department <strong>of</strong> Genetics, University <strong>of</strong> North Carolina at<br />

Chapel Hill 2. Department <strong>of</strong> Psychiatry, Duke University<br />

3. Department <strong>of</strong> Psychiatry, University <strong>of</strong> North Carolina at<br />

Chapel Hill 4. Department <strong>of</strong> Psychiatry, Columbia University<br />

5. Department <strong>of</strong> Pharmacology, Virginia Commonwealth<br />

University 6. Department <strong>of</strong> Genetics, University <strong>of</strong> North<br />

Carolina at Chapel Hill; Department <strong>of</strong> <strong>Medical</strong> Epidemiology<br />

& Biostatistics, Karolinska Institutet, Stockholm, Sweden<br />

*kneeker@med.unc.edu<br />

The 5-HTTLPR is a 44 base pair insertion-deletion<br />

polymorphism in the promoter region <strong>of</strong> the serotonin<br />

transporter (SLC6A4), and contributes to the regulation <strong>of</strong> the<br />

expression <strong>of</strong> SLC6A4. The serotonin transporter is a<br />

membrane bound protein which transports serotonin from<br />

synapses into pre-synaptic neurons. The 5-HTTLPR has been<br />

heavily studied in relation to many psychiatric diseases,<br />

endophenotypes, and drug response phenotypes. A single<br />

nucleotide polymorphism rs25531 (A/G) within the<br />

polymorphic 5-HTTLPR has been shown to alter expression<br />

<strong>of</strong> SLC6A4 . Many prior studies have thus incorrectly<br />

considered this triallelic polymorphism as biallelic (i.e.,<br />

5-HTTLPR*LA, LG, and S as opposed to 5-HTTLPR*L or S).<br />

In light <strong>of</strong> this discovery and the prevalence <strong>of</strong> rs25531 in<br />

certain ancestry groups, it is possible that previous studies<br />

may have been confounded by the previously unknown<br />

variant. Therefore, we tested 5-HTTLPR and rs25531 for<br />

association with chronic schizophrenia, cognitive phenotypes<br />

related to schizophrenia, and symptom status in a sample <strong>of</strong><br />

728 cases and 724 controls from the CATIE sample. We<br />

found no significant association <strong>of</strong> the 5-HTTLPR/rs25531<br />

composite genotype with the diagnosis <strong>of</strong> schizophrenia<br />

(p=0.72), an omnibus measure <strong>of</strong> neurocognition (p=0.21) or<br />

with working memory (p=0.32) at study baseline. Similarly,<br />

there was no association with PANSS positive (p=0.67) or<br />

negative symptoms (p=0.46) at study baseline (covariates age,<br />

sex, and 7 principal components). In conclusion, we were<br />

unable to identify association <strong>of</strong> the 5-HTTLPR/ rs25531 with<br />

schizophrenia, neurocognition, or core psychotic symptoms –<br />

even at levels <strong>of</strong> significance unadjusted for multiple<br />

comparisons.


230 GENETIC MARKERS FOR SUICIDE RISK IN<br />

SCHIZOPHRENIA<br />

E. Olsson* (1), D. Sudic (2), H. Druid (3), M. Schalling (2),<br />

U. Ösby (1)<br />

1. Department <strong>of</strong> Clinical Neuroscience, Karolinska Institutet,<br />

Stockholm, Sweden; Department <strong>of</strong> Psychiatry, Danderyd<br />

University Hospital, Stockholm, Sweden 2. Department <strong>of</strong><br />

Molecular Medicine and Surgery, Karolinska Institutet,<br />

Stockholm, Sweden 3. Department <strong>of</strong> Oncology-Pathology,<br />

Karolinska Institutet, Stockholm, Sweden<br />

*eric.olsson@sll.se<br />

Around 5% <strong>of</strong> patients with schizophrenia will die from<br />

suicide. Improved prediction <strong>of</strong> increased suicide risk is <strong>of</strong><br />

great importance in order to improve prognosis in<br />

schizophrenia. Molecular genetic association studies <strong>of</strong> suicide<br />

risk, also in schizophrenia, have focused on genes related to<br />

the serotonin system. There are several promising findings but<br />

also some contradictory results. This may be due to limitations<br />

<strong>of</strong> association studies, too small samples, diagnostic<br />

heterogeneity, and suicide attempt as phenotype rather than<br />

suicide per se. The purpose <strong>of</strong> this study is to identify genetic<br />

risk markers for suicide in schizophrenia. We use genetic<br />

association, comparing schizophrenic patients who have died<br />

from suicide with living schizophrenic patients, and also with<br />

a population-based control group. By linkage <strong>of</strong> diagnosis<br />

from the Patient Register with the cause <strong>of</strong> death from the<br />

Swedish Forensic database, 351 cases from three Counties in<br />

Sweden have been identified. The cases have a clinical<br />

schizophrenia diagnosis, have suicide as the certain, probable<br />

or possible cause <strong>of</strong> death and there is tissue available from<br />

the forensic autopsy. The control groups consist <strong>of</strong> patients<br />

with schizophrenia, and blood donor population controls from<br />

Stockholm County. This poster will describe the cases and the<br />

structure <strong>of</strong> the project.We will initially analyze SNPs based<br />

on previous studies in the genes TPH1, TPH2, 5-HTT,<br />

MAO-A, 5HT2A, all involved in the serotonin pathway. This<br />

study will in a favorable way address previous problems with<br />

identifying genetic markers for increased suicide risk in<br />

schizophrenia.<br />

231 PROTEOMIC ANALYSIS OF THE<br />

LYMPHOBLASTOID CELL LINE IN THE JAPANESE<br />

SCHIZOPHRENIC PATIENTS<br />

A. Yoshimi* (1), T. Nagai (1), S. Yamada (2), R. Ishihara (3),<br />

M. Ohashi (2), Y. Ito (3), Y. Noda (2), K. Yamada (1), N.<br />

Ozaki (3)<br />

1. Department <strong>of</strong> Neuropsychopharmacology and Hospital<br />

Pharmacy, Nagoya University Graduate School <strong>of</strong> Medicine<br />

2. Division <strong>of</strong> Clinical Sciences and<br />

Neuropsychopharmacology, Graduate School <strong>of</strong><br />

Pharmaceutical Sciences, Meijo University 3. Department <strong>of</strong><br />

Psychiatry, Nagoya University Graduate School <strong>of</strong> Medicine<br />

*akira-yoshimi@med.nagoya-u.ac.jp<br />

Schizophrenia is a chronic and disabling mental disorder with<br />

a lifetime prevalence <strong>of</strong> approximately 1% <strong>of</strong> the global<br />

population. There is no validated method based on the<br />

pathophysiology for diagnosis <strong>of</strong> the disease although a<br />

number <strong>of</strong> studies have shown several convincing candidate<br />

genes or molecules. Proteomic analyses have many<br />

advantages compared to gene expression analyses since they<br />

can provide further understandings for pathophysiology <strong>of</strong><br />

disease and the molecular biology based diagnostic tools. To<br />

explore the biomarkers <strong>of</strong> schizophrenia, we performed the<br />

florescence two-dimensional differential gel electrophoresis<br />

(2D–DIGE) analysis <strong>of</strong> lymphoblastoid cell line from 29<br />

schizophrenia and 29 control subjects. Differentially expressed<br />

proteins were subsequently identified by LC-MS/MS. A<br />

2D-DIGE analysis revealed that 1103±11.7 and 1103±13.3<br />

unique spots were identified in the schizophrenia and control<br />

group, respectively. No differences were observed between<br />

two groups in the total number <strong>of</strong> protein spots, and more than<br />

95% <strong>of</strong> all spots were matched to the reference gel. Changes<br />

in the relative abundance <strong>of</strong> 20 protein spots were identified in<br />

the schizophrenia compared to the control group. Seven spots<br />

exhibited an increase in the schizophrenia whereas 13 protein<br />

spots were decreased (Student’s t-test, p


232 ANALYSIS OF GENE EXPRESSION IN<br />

DORSOLATERAL PREFRONTAL CORTEX OF<br />

SCHIZOPHRENICS AND CONTROLS<br />

E. Shen* (1), A. Guillozet-Bongaarts (1), T. Hyde (2), B.<br />

Swanson (1), L. Kuan (1), J. Royall (1), J. Hohmann (1), A.<br />

Jones (1), J. Kleinman (2), H. Zeng (1)<br />

1. Allen Institute for Brain Science 2. Clinical Brain Disorders<br />

Branch, Genes Cognition and Psychosis Program, Intramural<br />

Research Program, NIMH, NIH<br />

*elaines@alleninstitute.org<br />

A dataset <strong>of</strong> gene expression information using colorimetric in<br />

situ hybridization (ISH) in postmortem human dorsolateral<br />

prefrontal cortex (DLFPC) <strong>of</strong> schizophrenic (SCZ; n = 15) and<br />

control subjects (n = 33) was generated for 60 genes<br />

comprising SCZ candidate genes, cell-type markers and<br />

cortical layer markers. An established high-throughput<br />

process and platform was used to perform ISH on frozen<br />

tissue that was sectioned, placed on slides and exposed to<br />

cDNA probes specifically targeted to each <strong>of</strong> the 60 genes <strong>of</strong><br />

interest. Nissl stained reference sections were generated every<br />

500 um. All Nissl and ISH slides were digitally scanned and<br />

all QC-passed data images were made publicly available via<br />

the Allen Institute’s data portal, www.brain-map.org.<br />

Brodmann areas 9 and 46 were delineated on all nissl stained<br />

sections. To characterize overall variability in DLPFC gene<br />

expression patterns, as well as differences in gene expression<br />

between SCZ and control subjects, a quantitative data analysis<br />

strategy was developed that includes informatics-based<br />

detection and analysis <strong>of</strong> four main ISH metrics: percentage<br />

<strong>of</strong> total area labeled, number <strong>of</strong> expressing objects<br />

(presumably cells) per mm2, object optical density (gene<br />

expression intensity in cells), and integrated optical density.<br />

Data pr<strong>of</strong>iles were generated by plotting each metric against<br />

cortical depth. Our initial analyses demonstrated that: (1) data<br />

pr<strong>of</strong>iles recapitulated gene expression patterns and (2) GRIK4<br />

gene expression differed between SCZ and control groups.<br />

Analysis <strong>of</strong> KIAA1576 indicated possible population<br />

differences in expression patterns.<br />

233 WHOLE GENOME SCAN ON SCHIZOPHRENIA<br />

IN 2 ARAB ISRAELI SAMPLES<br />

S. Lupoli* (1), P. Cozzi (2), A. Alkelai (3), A. Orro (2), F.<br />

Torri (4), E. Salvi (5), C. Cosentino (5), V. Tieran (5), L.<br />

Milanesi (2), B. Lerer (3), F. Macciardi (6)<br />

1. INSPE, San Raffaele Scientific Institute, Milan, Italy;<br />

University <strong>of</strong> Milan, Filarete Foundation, Milan, Italy<br />

2. ITB, CNR, Milan, Italy 3. Hadassah–Hebrew University<br />

<strong>Medical</strong> Center, Jerusalem, Israel 5. University <strong>of</strong> Milan,<br />

Filarete Foundation, Milan, Italy 6. University <strong>of</strong> Milan,<br />

Filarete Foundation, Milan, Italy; Department <strong>of</strong> Psychiatry<br />

and Human Behavior, University <strong>of</strong> California, Irvine, CA,<br />

USA<br />

*sara.lupoli@hsr.it<br />

Schizophrenia is a major public health problem with a 1%<br />

incidence worldwide, a high suicide rate, and enormous<br />

human and financial costs. It is widely considered to be a<br />

genetically mediated neurodevelopmental disorder and<br />

identification <strong>of</strong> potential candidate genes has accelerated in<br />

recent years. Whole-genome association (WGA) studies have<br />

identified multiple regions across the genome that are<br />

promising candidates <strong>of</strong> modest effect for schizophrenia. For 2<br />

Arab Israeli families with multiple cases <strong>of</strong> schizophrenia, we<br />

have performed a WGA family study examining<br />

approximately 370 000 markers (Illumina<br />

HumanCNV370-Duo). Plink 1.06 was used for quality<br />

control (QC) <strong>of</strong> genotype data, IBD (identity-by-descent)<br />

similarity matrix for checking pedigree errors, Transmission<br />

Disequilibrium Test (TDT) and permutations as correction<br />

methods for the TDT. Preliminary analysis shows 34 SNPs<br />

with p-value exceeding significance 10E-5 or less. Two SNPs<br />

bypass a p-value <strong>of</strong> 4.00E-06; one maps in SLIT (slit<br />

(Drosophila) homolog 1) gene and 1 in AHI1. Thirty-two<br />

SNPs reach 1e-05. Among our best results we detected<br />

associated SNPs in N-Methyl-D-Aspartate Receptor subunit<br />

2B (GRIN2B), Nerve Growth Factor Receptor (NGFR), and<br />

Glutamate Receptor Metabotropic 8 (GRM8), confirming<br />

previous studies on schizophrenia. Most <strong>of</strong> our associated<br />

genes are involved in neurodevelopment pathways.As recent<br />

papers (1,2) support association between chromosomal<br />

abnormalities and schizophrenia, analysis <strong>of</strong> Copy Number<br />

Variation (CNV), structural variations within the human<br />

genome, is on going on our datasets. 1 The International<br />

Schizophrenia Consortium: Rare chromosomal deletions and<br />

duplications increase risk <strong>of</strong> schizophrenia. Nature 455,<br />

237-241, 2008. 2Terry Vrijenhoek et al.: Recurrent CNVs<br />

Disrupt Three Candidate Genes in Schizophrenia Patients. The<br />

American Journal <strong>of</strong> Human Genetics 83, 504–510, 2008.


234 NRG1 AND DTNBP1 ARE ASSOCIATED WITH<br />

NON-DEFICIT SCHIZOPHRENIA AND SYMPTOM<br />

SEVERITY IN A HUNGARIAN SAMPLE<br />

J. Réthelyi* (1), S. Bakker (2), P. Polgár (1), P. Czobor (1), E.<br />

Strengman (3), P. Pásztor (4), R. Kahn (2), I. Bitter (1)<br />

1. Department <strong>of</strong> Psychiatry and Psychotherapy, Semmelweis<br />

University, Budapest, Hungary 2. The Rudolf Magnus<br />

Institute <strong>of</strong> Neuroscience, Department <strong>of</strong> Psychiatry,<br />

University <strong>Medical</strong> Centre Utrecht, Utrecht, The Netherlands<br />

3. Complex Genetics Section, Department <strong>of</strong> Biomedical<br />

Genetics, University <strong>Medical</strong> Center Utrecht, Utrecht, The<br />

Netherlands 4. Department <strong>of</strong> Psychiatry, St. John’s Hospital,<br />

Budapest, Hungary<br />

*rethelyi@psych.sote.hu<br />

Association studies have yielded extensive, but frequently<br />

contradictory data about the genetic risk factors <strong>of</strong><br />

schizophrenia. Clinical and genetic heterogeneity are possible<br />

factors explaining the inconclusive findings. The objective <strong>of</strong><br />

this study was to test the association <strong>of</strong> commonly<br />

incriminated candidate genes with two clinically divergent<br />

subgroups, non-deficit (SZ-ND) and deficit-schizophrenia<br />

(SZ-D), and with symptom severity. DNA samples from a<br />

homogeneous sample <strong>of</strong> 280 schizophrenia patients and 230<br />

healthy controls <strong>of</strong> Hungarian, Caucasian descent were<br />

genotyped for polymorphisms in schizophrenia candidate<br />

genes NRG1, DTNBP1, RGS4, G72/G30 and PIP5K2A.<br />

Patients were divided into the diagnostic subgroups <strong>of</strong> SZ-ND<br />

and SZ-D using the Schedule for Deficit Syndrome (SDS),<br />

and assessed clinically by the Positive and Negative Symptom<br />

Scale (PANSS). Polymorphisms in RGS4, G72/G30, and<br />

PIP5K2A were neither associated with SZ-ND, nor SZ-D.<br />

SNP8NRG241930 in NRG1 and rs1011313 in DTNBP1 were<br />

associated with SZ-ND (p=0.04 and p=0.03, respectively).<br />

SNP8NRG241930 showed association with the PANSS<br />

cognitive and hostility/excitability factor, rs1011313 with the<br />

negative factor and SDS total score, and rs10917670 in RGS4<br />

was associated with the depression factor. Our data seem to<br />

confirm previously reported association <strong>of</strong> NRG1 with<br />

schizophrenia without prominent negative symptoms. It was<br />

possible to detect association <strong>of</strong> small to medium effect sizes<br />

between the investigated candidate genes and symptom<br />

severity. Similar studies may unravel the possible connection<br />

between genetic and clinical heterogeneity in schizophrenia.<br />

235 SYSTEMATIC MUTATION SCREENING STUDY<br />

FOR ANXA7 AND PPP3CB IN SCHIZOPHRENIA<br />

C. Liu* (1), H. Hwu (1), U. Yang (2), P. Hsu (2), C. Fann (3),<br />

Y. Liu (4), C. Wen (1), C. Chang (3)<br />

1. Department <strong>of</strong> Psychiatry, National Taiwan University<br />

Hospital, Taipei, Taiwan 2. Institute <strong>of</strong> Bioinformatics,<br />

National YangMing University, Taipei, Taiwan 3. Institute <strong>of</strong><br />

Biomedical Science, Academia Sinica, Taipei, Taiwan 4.<br />

Division <strong>of</strong> Mental Health and Substance Abuse Research,<br />

National Health Research Institutes, Taipei, Taiwan<br />

* cmliu1968@ntu.edu.tw<br />

Annexin A7 (ANXA7) and protein phosphatase 3 (formerly<br />

2B), catalytic subunit, beta is<strong>of</strong>orm (PPP3CB) have been<br />

reported associated with schizophrenia in our previous<br />

association studies. The aim <strong>of</strong> the study is to search for the<br />

risk polymorphisms <strong>of</strong> the two genes for schizophrenia using<br />

systematic mutation screening method. We have sequenced<br />

the exons and the promoter regions <strong>of</strong> the two genes in 50<br />

controls and 42 schizophrenic patients. We have identified 16<br />

and five genetic variations <strong>of</strong> ANXA7 and PPP3CB,<br />

respectively. and 11 genetic variations <strong>of</strong> them have been<br />

genotyped in a large case-control sample, including 600<br />

supernormal controls and 912 schizophrenic patients. We<br />

found the C allele <strong>of</strong> a novel SNP (PPP3CB-1) in the promoter<br />

region, and the C allele <strong>of</strong> SNP rs58876184 in the 3’UTR<br />

region <strong>of</strong> PPP3CB, is significantly over-represented in the<br />

case groups (p = 0.038 and 0.018, respectively). The haplotype<br />

composed <strong>of</strong> the four genetic variations <strong>of</strong> PPP3CB also<br />

showed significant haplotype frequency difference between<br />

the cases and controls. Our data showed the genetic variations<br />

in the promoter region and 3’ UTR region <strong>of</strong> PPP3CB is<br />

probably related to genetic vulnerability <strong>of</strong> schizophrenia.<br />

Further clarification <strong>of</strong> the biological function <strong>of</strong> the two<br />

genetic variations is ongoing.


236 ASSOCIATION BETWEEN NRG-1 MARKERS AND<br />

REGIONAL BRAIN MORPHOMETRY IN PSYCHOTIC<br />

DISORDERS<br />

A. Dutt* (1,2), C. McDonald (1,2), E. Dempster (1,2), D.<br />

Prata (1,2), M. Shaikh (1,2), I. Williams (1,2), S. Amankwa<br />

(1,2), K. Schulze (1,2), N. Marshall (1,2), M. Walshe (1,2), M.<br />

Allin (1,2), D. Collier (1,2), R. Murray (1,2), E. Bramon (1,2)<br />

1. NIHR Biomedical Research Centre, Institute <strong>of</strong> Psychiatry<br />

(King’s College London)/South London 2. Maudsley NHS<br />

Foundation Trust, London, UK<br />

*Anirban.Dutt@kcl.ac.uk<br />

Introduction: Lateral ventricular enlargement and hippocampal<br />

volume deficits are commonly observed morphometric<br />

abnormalities in psychotic disorders such as schizophrenia and<br />

psychotic bipolar disorder. The neuregulin gene (NRG1) has<br />

been associated with both these disorders, and it is believed to<br />

regulate different neurodevelopmental processes including<br />

neuronal and glial cell survival, proliferation, migration and<br />

differentiation. In our study we investigated the effect <strong>of</strong><br />

NRG-1 markers on lateral ventricular and hippocampal<br />

volumes in psychosis. Methods: 360 participants comprising<br />

<strong>of</strong> patients with psychosis, their family members, and<br />

unrelated healthy controls <strong>of</strong> white European ancestry<br />

underwent structural MRI and genotyping. The<br />

NRG-1 markers consisted <strong>of</strong> one single nucleotide<br />

polymorphism marker (SNP8NGR22153) and two<br />

micro-satellites (478B14-848 and 420 M9-1395). The effect <strong>of</strong><br />

candidate genes on brain morphometry was examined using<br />

linear regression models adjusting for clinical group, age, sex<br />

and correlations between members <strong>of</strong> the same family.<br />

Results: There was no evidence <strong>of</strong> association between the<br />

NRG-1 markers and either ventricular or hippocampal<br />

volumes. Conclusion: Our study demonstrates that the<br />

neuregulin gene does not influence ventricular or hippocampal<br />

volumetric deficits in psychotic disorders. Although the<br />

neuregulin gene has been associated with both schizophrenia<br />

and bipolar disorder, it must have very modest influence on<br />

regional morphometric deviations on its own.<br />

237 IDENTIFYING CNV’S IN SCHIZOPHRENIA<br />

USING MULTIPLEX AMPLICON QUANTIFICATION<br />

(MAQ) AND THEIR VERIFICATION BY FISH<br />

M. Malloy* (1), J. Del-Favero (2), D. Goossens (2), L.<br />

Heyrman (2), A. Laenerts (2), D. St Clair (3), D. Blackwood<br />

(4), W. Muir (4), B. Pickard (5)<br />

1. Molecular Medicine Centre, University <strong>of</strong> Edinburgh<br />

2. University <strong>of</strong> Antwerp 3. University <strong>of</strong> Aberdeen<br />

4. University <strong>of</strong> Edinburgh 5. University <strong>of</strong> Strathclyde<br />

*Pat.Malloy@ed.ac.uk<br />

Schizophrenia is a chronic and disabling mental illness that<br />

affects around 1% <strong>of</strong> the world’s population. Although a<br />

genetic basis for the disorder is not disputed, our<br />

understanding <strong>of</strong> the risk factors involved until recently has<br />

been very limited. We have shown that chromosomal<br />

abnormalities such as reciprocal balanced translocations, can<br />

act as direct positional pointers by disruption, to schizophrenia<br />

risk genes. DISC1, PDE4b (Miller K., Pickard B. et al.<br />

Science 310 (5751): 1187-1191, 2005) and NDEL1<br />

(unpublished) are either directly disrupted, or immediately<br />

proximate to chromosomal breakpoints. Our hypothesis is that<br />

such breakpoints lie near LCRs which should also predispose<br />

to microdeletions and microduplications that comprise CNV.<br />

We are using Multiplex Amplicon Quantification (MAQ,<br />

Sutrala et al Scz Res 96 (1-3): 93-9), firstly to detect CNVs<br />

found by Walsh et al (Science 320 (5875): 539-43, 2008) and<br />

Kirov et al (Hum Mol Gen 17 (3): 458-65) in large<br />

case-control cohorts <strong>of</strong> schizophrenia (n=1000), co-morbid<br />

Learning Disability and Schizophrenia (n=200), bipolar<br />

disorder (n=400) and major depressive disorder (n=400).<br />

Secondly we have analysed regions around DISC1 pathway<br />

genes in a site directed approach to quantify the allelic<br />

variation in the same patients. Sex-matched controls (normal<br />

and learning disabled alone) are available for comparator<br />

purposes. FISH is used at the BAC or Fosmid level on<br />

metaphase or interphase preparations, or where applicable<br />

additional information from our GWAS data, to confirm any<br />

variation


238 THE REGULATION OF SUSCEPTIBILITY GENES<br />

FOR SCHIZOPHRENIA<br />

F. Macciardi* (1), E. Osimo (2), S. Potkin (1), F. Torri (2), J.<br />

Fallon (1), J. Turner (1), G. Guffanti (1), M. Vawter (1), B.<br />

Lerer (3), S. Gaudi (4)<br />

1. UCI 2. UNIMI 3. Hadassah 4. ISS (Roma)<br />

*fmacciar@uci.edu<br />

The complexity <strong>of</strong> the genome has little to do with the number<br />

and heterogeneity <strong>of</strong> its genes: only 2% <strong>of</strong> the human genome<br />

codes for proteins, while 45% consists <strong>of</strong> Transposable<br />

Elements (TEs). The relative amount <strong>of</strong> TEs increases<br />

consistently with organism complexity, the TEs domains <strong>of</strong><br />

the regulatory genome drive genome evolution, alter gene<br />

expression and can substantially contribute to the etiology and<br />

characteristics <strong>of</strong> complex diseases. The regulation <strong>of</strong> TE<br />

expression has been already associated with complex diseases<br />

and cancer cells are characterized by high expression levels <strong>of</strong><br />

LINE-1, correlating cancer cell to the reactivation <strong>of</strong> retroviral<br />

transcripts (RT) expression. Moreover, complex human<br />

diseases cannot be explained by genetic factors alone and are<br />

likely to be the result <strong>of</strong> gene-environment interactions with<br />

the contribution <strong>of</strong> TEs. The detection <strong>of</strong> retroviral transcripts<br />

in the brains <strong>of</strong> schizophrenics suggests that activation or<br />

upregulation <strong>of</strong> distinct human endogenous retroviruses<br />

(HERVs) may play a role in the etiopathogenesis <strong>of</strong><br />

neuropsychiatric diseases. Our aim is to propose a novel<br />

mechanism for gene dysregulation in schizophrenia by<br />

investigating the functional relationship between TE and genes<br />

identified as "best" candidate from GWAS, validating them<br />

with exon-expression arrays and reconstructing their<br />

regulatory network with computational methods. We found<br />

preliminary evidence <strong>of</strong> alternative regulation <strong>of</strong><br />

tissue-specific gene expression in schizophrenic patients from<br />

genes that we identified as associated with schizophrenia from<br />

our GWAS. We present results for both tissue specific<br />

expression and alternative Transcription Starting Sites (TSS)<br />

in pivotal disease-related genes.<br />

239 GENE EXPRESSION PROFILES OF CULTURED<br />

NEURAL PROGENITOR CELLS DEMONSTRATE<br />

DIFFERENTIAL EXPRESSION OF<br />

DEVELOPMENTAL-RELATED GENES IN<br />

SCHIZOPHRENIA<br />

O. Evgrafov* (1), B. Wrobel (1), X. Kang (1), G. Simpson<br />

(1), D. Malaspina (2), J. Knowles (1)<br />

1. USC 2. NYU<br />

*evgrafov@keck.usc.edu<br />

We investigated the expression <strong>of</strong> poly-A RNA isolated from<br />

cultured neuronal cells derived from olfactory<br />

neuroepithelium (CNON) <strong>of</strong> 8 patients with schizophrenia<br />

(SZ) and 8 control individuals. Human neuroepithelium<br />

constantly produces olfactory neurons from neural stem cells<br />

and this process is simulated in CNON cultures, which are<br />

primarily composed <strong>of</strong> neuronal progenitors. The tight<br />

environmental control provided by cell culture provided a high<br />

signal-to-noise ratio and yielded a clear separation <strong>of</strong> SZ and<br />

control samples, in a principle component analysis. In<br />

contrast to neurons from adult brain, CNON cells express<br />

genes that are specific for development <strong>of</strong> neural system. We<br />

measured gene expression using Human Exon 1.0 ST Arrays<br />

(Affymetrix) followed by ANOVA analysis using Partek GS<br />

(Partek). We have also performed RNA-Seq using an<br />

Illumina Genome Analyzer II, on a portion <strong>of</strong> these samples.<br />

The top most differentially expressed genes were validated by<br />

qPCR. The three most differentially expressed genes were<br />

CDH6, VCAM1 and S1PR1 (EDG1). Another cadherin,<br />

CDH11, was also differentially expressed, when validated by<br />

qPCR. Three <strong>of</strong> these are cell adhesion molecules, and the<br />

fourth, S1PR1, directly regulates cadherin expression and is<br />

involved in regulation <strong>of</strong> VCAM1. Cell adhesion molecules<br />

are known to play an important role in brain development and<br />

the observation <strong>of</strong> lower levels <strong>of</strong> these molecules in the SZ<br />

group may point to a possible mechanism for the altered<br />

neurodevelopment observed in individuals with schizophrenia.


240 SIGNIFICANT LINKAGE AT CHROMOSOME<br />

2p15 TO A SCHIZOPHRENIA ENDOPHENOTYPE IN A<br />

NEPALESE POPULATION GENETIC ISOLATE<br />

S. Santangelo* (1), T. Sitnikova (1), M. Hall (2), P. Shrestha<br />

(3), D. Yu (4), J. Subedi (5), S. Ojha (6), S. Tamang (3), R.<br />

Chapagain (3), S. Kamar (3), J. VandeBerg (7), J. Blangero<br />

(7), S. Williams-Blangero (7)<br />

1. Harvard <strong>Medical</strong> School, Dept Psychiatry, Center for<br />

Human Genetic Research, Massachusetts General Hospital,<br />

Boston, MA, USA 2. Psychology Research Laboratory,<br />

McLean Hospital, Belmont, MA, USA 3. Nepal Biomedical<br />

Research Center, Kathmandu, Nepal 4. Center for Human<br />

Genetic Research, Massachusetts General Hospital, Boston,<br />

MA, USA 5. Miami University, Dept. Sociology,<br />

Gerontology, Oxford, OH, USA 6. Tribhuvan University<br />

Institute <strong>of</strong> Medicine, Dept. Psychiatry, Kathmandu, Nepal<br />

7. Southwest Foundation for Biomedical Research, San<br />

Antonio, TX, USA<br />

*ssantangelo@pngu.mgh.harvard.edu<br />

Electrophysiological traits indexing schizophrenia<br />

endophenotypes were measured in over 600 members <strong>of</strong> an<br />

extended pedigree in Jiri, Nepal. In an auditory "oddball"<br />

paradigm, we recorded peak amplitude and latency <strong>of</strong> the<br />

P300 evoked response as well as power <strong>of</strong> oscillatory brain<br />

activity in the gamma-band range (35-45Hz) induced between<br />

250-550 msec after odd tones onset. To examine<br />

anterior-posterior and hemispheric differences, the data were<br />

examined at Fz, Pz, P3 and P4 scalp locations and evaluated in<br />

a quantitative trait locus genome-wide linkage analysis.<br />

Heritabilities for both P300 amplitude and induced gamma at<br />

Pz, P3, and P4, and for P300 latency at P4 were significant.<br />

For this Nepalese pedigree and marker set, evidence <strong>of</strong><br />

suggestive linkage is obtained for LOD scores ≥ 1.65, and<br />

significant linkage for LODs ≥ 2.83. Peak LODs for the ERPs<br />

were 2.93 at chromosome 2 (87 cM) for Pz amplitude, 2.07 at<br />

chromosome 13 (76 cM) for Fz latency, and 2.32 at<br />

chromosome 17 (61 cM) for P4 latency. QTL linkage results<br />

for gamma response centered on chromosome 2p13 (98 cM)<br />

with peak LOD scores <strong>of</strong> 1.31 for Pz, 1.42 for P3, and 1.84 for<br />

P4. Thus, we obtained significant linkage for P300 amplitude<br />

at 2p15 and suggestive linkage for induced gamma-band<br />

response in the P300 time-window at 2p13; these loci are only<br />

10 MB apart. Genes in this region include sepiapterin<br />

reductase, an enzyme that catalyzes the final step in the<br />

synthesis <strong>of</strong> tetrahydrobiopterin (BH4), an essential c<strong>of</strong>actor<br />

for synthesis <strong>of</strong> many neurotransmitters, including serotonin.<br />

242 COMT GENOTYPE, COGNITION, AND<br />

FUNCTIONAL CAPACITY IN SCHIZOPHRENIA<br />

E. Twamley* (1), C. Burton (1), V. Lea (1), G. Savla (1), J.<br />

Guidry (1), M. Abeles (1), A. Minassian (1), R. Bilder (2), J.<br />

Kelsoe (1)<br />

1. University <strong>of</strong> California, San Diego 2. University <strong>of</strong><br />

California, Los Angeles<br />

*etwamley@ucsd.edu<br />

The Val158Met single nucleotide polymorphism <strong>of</strong> the<br />

catechol-O-methyltransferase (COMT) gene on chromosome<br />

22 influences dopamine catabolism and is related to cognitive<br />

impairment in schizophrenia. Previous research has found that<br />

the Met allele is associated in a dose-response fashion with<br />

better performance on most neuropsychological tests. We<br />

assessed 116 outpatients with schizophrenia-spectrum<br />

disorders (62% men; 68% Caucasian; mean age=48; mean<br />

years <strong>of</strong> education=13) with a blood draw and a<br />

comprehensive neuropsychological, functional, and clinical<br />

battery. Participants with the Val/Val (n=38), Val/Met (n=53),<br />

and Met/Met (n=25) genotypes did not differ on demographic<br />

variables, diagnosis, type or dosage <strong>of</strong> antipsychotic<br />

medication, or duration <strong>of</strong> psychosis. Spearman correlations<br />

between the “dose” <strong>of</strong> the Met allele (0, 1, or 2 Met alleles)<br />

and the neuropsychological, functional, and clinical variables<br />

demonstrated that a higher number <strong>of</strong> Met alleles was<br />

associated with better estimated premorbid intellectual<br />

functioning (r=.21, p=.028). Correlations between the number<br />

<strong>of</strong> Met alleles and performance on the <strong>UCSD</strong> Performance<br />

Based Skills Assessment (Household Chores and Recreation<br />

Planning subtests) were >.15, but did not reach statistical<br />

significance. Met homozygotes significantly outperformed Val<br />

homozygotes on the UPSA Household Chores subtest (t=2.0,<br />

df=59, p=.049). The Met allele appears to be associated with<br />

better performance in selected neuropsychological domains<br />

and functional capacity tasks. Because the Val158Met<br />

polymorphism typically explains a small amount <strong>of</strong> variance<br />

in cognitive performance and given the size <strong>of</strong> the current<br />

sample, these results need to be replicated with a larger sample<br />

and other variants should be considered to further understand<br />

genetic contributions to cognition.


243 CONSANGUINITY AND INCREASED RISK FOR<br />

SCHIZOPHRENIA IN EGYPT<br />

H. Mansour* (1, 2), W. Fathi (2), L. Klei (1), J. Wood (1), K.<br />

Chowdari (1), A. Watson (1), A. Eissa (2), A. Yassin (2), H.<br />

Salah (2), S. Tobar (2), H. El-Boraie (2), H. Gaafar (2), M.<br />

Elassy (2), I. Ibrahim (2), N. Ibrahim (1), K. Kandil (1), W.<br />

El-Bahaei (2), M. Alatrouny (2), F. El-Chennawi (3), B.<br />

Devlin (4), V. Nimgaonkar (1, 5)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Pittsburgh School<br />

<strong>of</strong> Medicine, Western Psychiatric Institute and Clinic,<br />

Pittsburgh, Pennsylvania 2. Department <strong>of</strong> Psychiatry,<br />

Mansoura University School <strong>of</strong> Medicine, Mansoura, Egypt<br />

3. Department <strong>of</strong> Clinical Pathology, Mansoura University<br />

School <strong>of</strong> Medicine, Mansoura, Egypt 4. Department <strong>of</strong><br />

Human Genetics, Graduate School <strong>of</strong> Public Health,<br />

University <strong>of</strong> Pittsburgh, Pittsburgh, Pennsylvania<br />

5. Department <strong>of</strong> Human Genetics, Graduate School <strong>of</strong> Public<br />

Health, University <strong>of</strong> Pittsburgh, Pittsburgh, Pennsylvania<br />

*mansourha@upmc.edu<br />

Background: Consanguinity has been suggested as a risk<br />

factor for psychsoses in some Middle Eastern countries, but<br />

adequate control data are unavailable. Our recent studies in<br />

Egypt have shown elevated parental consanguinity rates<br />

among patients with bipolar I disorder (BP1), compared with<br />

controls. We have now extended our anlayses to<br />

Schizophrenia (SZ) in the same population. Methods: A<br />

case-control study was conducted at Mansoura University<br />

Hospital, Mansoura, Egypt (SZ, n = 75; controls, n = 126, and<br />

their available parents). The prevalence <strong>of</strong> consanguinity was<br />

estimated from family history data (‘self report’), followed by<br />

DNA analysis using short tandem repeat polymorphisms<br />

(STRPs, n = 64) (‘DNA-based’ rates). Results: Self reported<br />

consanguinity was significantly elevated among the patients<br />

(SZ: 45%, controls: 18%, OR 3.7, 95% CI 1.95, 7.05; p =<br />

0.0001, 1 d.f.). These differences were confirmed using DNA<br />

based estimates for coefficients <strong>of</strong> inbreeding (inbreeding<br />

coefficients, mean ± standard error, cases: 0.058 ± 0.007,<br />

controls: 0.022 ± 0.003, test statistic = -6.76, p = 6.6 e-12).<br />

Conclusion: Consanguinity rates are significantly elevated<br />

among Egyptian SZ patients in the Nile delta region. The<br />

associations are similar to those observed with BP1 in our<br />

earlier study. If replicated, the substantial risk associated with<br />

consanguinity raises public health concerns.<br />

244 ADMIXTURE ANALYSIS OF AGE AT ONSET IN<br />

SCHIZOPHRENIA: GENETIC ASSOCIATION STUDY<br />

OF 384 MARKERS ACROSS THE GENOME<br />

V. De Luca* (1), F. Panariello (1), C. Zai (1)<br />

1. University <strong>of</strong> Toronto<br />

*vincenzo_deluca@camh.net<br />

Despite the considerable amount <strong>of</strong> data from several studies,<br />

none <strong>of</strong> the thresholds used to define age at onset (AAO) in<br />

schizophrenia have been validated. The aim <strong>of</strong> this study is to<br />

assess the presence <strong>of</strong> different homogenous subgroups in<br />

schizophrenia patients based on the age at onset, by<br />

determining the cut-<strong>of</strong>f age <strong>of</strong> each subgroup. Admixture<br />

analysis was applied to identify model(s) <strong>of</strong> separate normal<br />

distributions <strong>of</strong> AAO characterized by different means,<br />

variances and population proportions. This helped us in<br />

identifying different subgroups in our sample <strong>of</strong> 466 patients<br />

with schizophrenia. We apply now a candidate gene<br />

association strategy aimed to detect genetic susceptibility for<br />

early onset in patients’ schizophrenia by using a custom<br />

Illumina Bread Chips comprising 384 SNPs selected among<br />

candidate genes implicated in the neurobiology <strong>of</strong><br />

schizophrenia. We analyzed 154 schizophrenic patients from<br />

White European ancestry recruited in Toronto at the Centre for<br />

Addiction and Mental Health. Thirty-four patients who had a<br />

psychotic onset when they were younger than 20 (optimal<br />

cut-<strong>of</strong>f from admixture analysis) were considered early onset<br />

schizophrenics. By using a GWA-like approach, we screened<br />

all 384 markers for the nominal significance level and then<br />

corrected for multiple test. The most significant SNP was the<br />

rs460401 (p=0.0003) however after the False Discovery Rate<br />

correction the p-value was not significant (p=0.113). Our<br />

analysis <strong>of</strong> 384 markets across candidate genes revealed that<br />

the SNP rs460401 located in the Ionotropic Kainate 1<br />

Glutamate receptor (GRIK1) is associated with early onset in<br />

schizophrenia. This preliminary result pointed out the<br />

importance <strong>of</strong> the kainate receptors in determining the onset <strong>of</strong><br />

schizophrenia therefore further work on these genes regarding<br />

this phenotype is required.


245 GENOME-WIDE ASSOCIATION STUDY OF<br />

MULTIPLEX SCHIZOPHRENIA PEDIGREES<br />

SUPPORTS A ROLE FOR CONTACTIN PROTEINS<br />

D. Levinson* (1), J. Shi (1), B. Riley (2), A. Pulver (3), M.<br />

Owen (4), D. Wildenauer (5), P. Gejman (6), B. Mowry (7), C.<br />

Laurent (8), S. Oh (1), K. Kendler (2), G. Nestadt (3), N.<br />

Williams (4), S. Schwab (5), A. Sanders (6), D. Nertney (7), J.<br />

Mallet (9), B. Wormley (2), V. Lasseter (3), M. O'Donovan<br />

(4), J. Duan (6), M. Albus (10), M. Alexander (1), D. Cohen<br />

(8), R. Ribble (2), K. Liang (11), N. Norton (4), W. Maier<br />

(12), G. Papadimitriou (13), D. Walsh (14), M. Jay (8), A.<br />

O'Neill (15), F. Lerer (16), D. Dikeos (13), M. Fallin (3), J.<br />

Silverman (17), M. Hansen (18), F. Dudbridge (19), P.<br />

Holmans (4)<br />

1. Stanford University, Stanford, CA 2. Virginia<br />

Commonwealth University, Richmond VA, USA 3. Johns<br />

Hopkins University, Baltimore, MD, USA 4. Cardiff<br />

University, Cardiff, UK 5. University <strong>of</strong> Western Australia,<br />

Perth 6. NorthShore University Healthcare Research Inst,<br />

Evanston,IL 7. University <strong>of</strong> Queensland, Brisbane, Australia<br />

8. Pierre et Marie Curie Faculty <strong>of</strong> Medicine, Paris 9. CNRS,<br />

Hôpital de la Pitié Salpêtrière, Paris 10. State Mental Hospital,<br />

Haar, Germany 11. Johns Hopkins University Bloomberg<br />

School <strong>of</strong> Public Health, Baltimore MD 12. University <strong>of</strong><br />

Bonn, Bonn, Germany 13. University <strong>of</strong> Athens <strong>Medical</strong><br />

School, Athens, Greece 14. The Health Research Board,<br />

Dublin, Ireland 15. Queens University, Belfast, Northern<br />

Ireland 16. Hadassah-Hebrew University <strong>Medical</strong> Center,<br />

Jerusalem, Israel 17. Mt. Sinai School <strong>of</strong> Medicine, New<br />

York, NY, USA. 18. Illumina, Inc. La Jolla, CA 19.<br />

Biostatistics Unit, Cambridge, UK<br />

*dflev@stanford.edu<br />

We carried out a genome-wide association study (GWAS) <strong>of</strong><br />

DSM-IIIR schizophrenia and schizoaffective disorder in<br />

multiply-affected families. Families ascertained by seven<br />

previous linkage studies (and not included previously in<br />

GWAS) were selected if they had constellations <strong>of</strong> genotyped<br />

individuals informative for family-based association analysis<br />

(≥1 affected cases with 1-2 parents and/or unaffected siblings;<br />

or ≥3 affected cases). Genotyping was carried with the<br />

Illumina 610-quad array. QC analyses excluded<br />

poorly-performing DNA samples and SNPs, confirmed family<br />

relationships and sex, and established ancestries. Following<br />

QC, there were 583 European-ancestry families with 2212<br />

genotyped individuals (1218 affected), and 50 families with<br />

other ancestries (primarily large pedigrees with known<br />

admixture, analyzed in separate subsets) with 253 genotyped<br />

individuals (139 affected). Single-SNP association analyses<br />

were carried out with TRANSMIT (autosomal SNPs) and<br />

UNPHASED (X chromosome SNPs), after preliminary testing<br />

to ensure comparability <strong>of</strong> the methods and correct Type I<br />

error rates. No genome-wide significant results (P < 5 x 10-8)<br />

were observed in analyses <strong>of</strong> European-ancestry or all<br />

families. Among genes containing one or more SNPs with<br />

P-values < 10-4, five encode cell adhesion molecules<br />

(CNTNAP5, AMIGO3, CADM2, CLEC4D, CLEC4E), and<br />

P-values < 10-3 were observed for clusters <strong>of</strong> SNPs in<br />

CNTNAP2 and CNTN5. Genes in the related CNTNAP and<br />

CNTN families (and the larger neurexin family) have been<br />

proposed as implicated in both autism and schizophrenia<br />

based on several lines <strong>of</strong> evidence, including cases with<br />

structural variants (for review see Burbach and van der<br />

Zwaag, Trends Neurosci, 2009;32:69-72). Larger samples and<br />

additional lines <strong>of</strong> evidence will be required to determine the<br />

significance <strong>of</strong> these findings.<br />

246 RUNS OF HOMOZYGOSITY NOT ASSOCIATED<br />

WITH INCREASED RISK OF SCHIZOPHRENIA<br />

M. Simonson* (1), M. McQueen (1), M. Keller (1)<br />

1. Institue for Behavioral Genetics; University <strong>of</strong> Colorado at<br />

Boulder<br />

*masimonson@gmail.com<br />

Previous research suggests that runs <strong>of</strong> homozygosity are a<br />

risk factor for schizophrenia (Lencz et al, 2007). We used<br />

700000 SNP markers on 2357 schizophreniacases and 2709<br />

controls to find runs <strong>of</strong> homozygosity. We assessed rare<br />

versus common runs as well as runs that did or did not overlap<br />

genes. Analyses showed no case-control differences in the<br />

percent <strong>of</strong> the genome in runs <strong>of</strong> homozygosity. Our analyses<br />

suggest that runs <strong>of</strong> homozygosity are not associated with<br />

increased risk <strong>of</strong> schizophrenia.<br />

247 FUNCTIONAL ANALYSIS OF GENETIC<br />

VARIATION AT THE DTNBP1 LOCUS.<br />

L. Carroll (1), A. Gerrish (1), L. Elliston (1), G. Kirov (1), M.<br />

Owen (1), M. O'Donovan (1), N. Williams* (1)<br />

1. Cardiff University<br />

*williamsnm@cf.ac.uk<br />

A large volume <strong>of</strong> genetic association data from multiple<br />

populations now supports the locus encoding Dysbindin<br />

(DTNBP1) as a schizophrenia susceptibility gene. The source<br />

<strong>of</strong> the genetic association at Dysbindin is equivocal, however<br />

studies to date show altered protein and mRNA levels in<br />

schizophrenic CNS. Furthermore, there is evidence to suggest<br />

that cis acting variation alters Dysbindin mRNA levels in<br />

post-mortem neural tissue and is correlated with a replicated<br />

schizophrenia “at-risk” haplotype. To identify the functional<br />

variation causing the association at DTNBP1 we have<br />

performed resequencing and high-density association mapping<br />

in large case-control and familial schizophrenia samples from<br />

Europe. Associated variants have been analysed to see<br />

whether they account for the changes in mRNA levels<br />

observed in post-mortem brain by performing allele specific<br />

expression studies in extended samples <strong>of</strong> those previously<br />

reported.


248 MITOCHONDRIAL DNA AND SCHIZOPHRENIA<br />

B. Verge (1), Y. Alonso (1), C. Miralles (1), J. Valero (1), S.<br />

Setó (1), A. Labad (1), E. Vilella (1), L. Martorell (1)<br />

1. HPUIPM, IISPV, URV<br />

One <strong>of</strong> the hypotheses about the genetic factors involved in<br />

schizophrenia points out the mitochondrial DNA (mtDNA), a<br />

16569 bp molecule which is inherited only from the mother<br />

(about 100.000 copies) and involved in maternally inherited<br />

mitochondrial disorders. If this hypothesis is true, one would<br />

expect a higher frequency <strong>of</strong> schizophrenia in relatives that<br />

share the mtDNA with a schizophrenic patient than in relatives<br />

that do not share. We have interviewed 100 schizophrenia<br />

patients according to DSM-IV criteria and 147 relatives to<br />

collect clinical data from both sides <strong>of</strong> the patient’s pedigree.<br />

Thirty one schizophrenia patients present at least one first-<br />

and/or one second-degree relative affected by schizophrenia,<br />

meanwhile the remaining 69 were sporadic cases. Apparent<br />

matrilineal inheritance was present in 22 <strong>of</strong> the 31 familial<br />

cases (71%). According to the 31 familial cases, information<br />

about mental health was achieved from 588 relatives through<br />

direct interview to one or two first degree relatives. Four<br />

groups were established to compare the presence <strong>of</strong><br />

schizophrenia in relatives depending on they share or not the<br />

mtDNA with a schizophrenia patient. Relatives that share the<br />

mtDNA with a schizophrenic patient have a higher risk <strong>of</strong><br />

developing the illness than those that do not share, OR=4.82<br />

(CI 2.04-13.11, p=0.00005). Evidence is accumulating that<br />

mitochondrial dysfunction is involved in the pathophysiology<br />

<strong>of</strong> schizophrenia and large studies are necessary to corroborate<br />

the involvement <strong>of</strong> mtDNA in the development <strong>of</strong> the illness.<br />

249 INITIAL LINKAGE ANALYSES OF SIX<br />

ENDOPHENOTYPES FOR SCHIZOPHRENIA FROM<br />

THE CONSORTIUM ON THE GENETICS OF<br />

SCHIZOPHRENIA (COGS)<br />

T. Greenwood* (1), K. Cadenhead (1), M. Calkins (2), D.<br />

Dobie (3), R. Freedman (4), M. Green (5), R. Gur (2), L.<br />

Lazzeroni (6), G. Light (1), K. Nuechterlein (5), A. Olincy (4),<br />

A. Radant (3), L. Seidman (7), L. Siever (8), J. Silverman (8),<br />

W. Stone (7), N. Swerdlow (1), D. Tsuang (3), M. Tsuang (1),<br />

B. Turetsky (2), N. Schork (9), D. Braff (1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California San<br />

Diego, La Jolla, CA 2. Department <strong>of</strong> Psychiatry, University<br />

<strong>of</strong> Pennsylvania, Philadelphia, PA<br />

3. Department <strong>of</strong> Psychiatry and Behavioral Sciences,<br />

University <strong>of</strong> Washington, and the VA Puget Sound Health<br />

Care System, Seattle, WA 4. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Colorado Health Sciences Center, Denver, CO<br />

5. Department <strong>of</strong> Psychiatry and Biobehavioral Sciences,<br />

Geffen School <strong>of</strong> Medicine at University <strong>of</strong> California Los<br />

Angeles, Los Angeles, CA 6. Department <strong>of</strong> Psychiatry and<br />

Behavioral Science, Stanford University, Stanford, CA<br />

7. Department <strong>of</strong> Psychiatry, Harvard <strong>Medical</strong> School,<br />

Massachusetts Mental Health Center Public Psychiatry<br />

Division <strong>of</strong> the Beth Israel Deaconess <strong>Medical</strong> Center and the<br />

Harvard Institute <strong>of</strong> Psychiatric Epidemiology and Genetics,<br />

Boston, MA 8. Department <strong>of</strong> Psychiatry, The Mount Sinai<br />

School <strong>of</strong> Medicine, and the James J. Peters VA <strong>Medical</strong><br />

Center, New York, NY 9. Scripps Genomic Medicine, La<br />

Jolla, CA<br />

*tgreenwood@ucsd.edu<br />

As part <strong>of</strong> the COGS, we have recently completed genotyping<br />

and preliminary analyses <strong>of</strong> a genome-wide SNP linkage scan<br />

for 211 families <strong>of</strong> schizophrenia probands comprising the<br />

first phase <strong>of</strong> our data release. Genotyping was performed<br />

using the Illumina Linkage Panel 12 containing 6,090 SNP<br />

markers across the genome. We used Merlin to conduct<br />

linkage analyses <strong>of</strong> the 6,001 SNPs that were successfully<br />

assayed, accounting for linkage disequilibrium between<br />

markers and accommodating the appropriate covariates (age<br />

and sex) for each endophenotype. We have currently<br />

completed these analyses for each <strong>of</strong> our six primary<br />

neurophysiologicaland neurocognitive endophenotypes:<br />

Prepulse inhibition (PPI), P50 suppression, the antisaccade<br />

task, the Continuous Performance Test<br />

(Degraded-Stimulusversion, DS-CPT), the Letter-Number<br />

Span (LNS), and the California VerbalLearning Task (CVLT).<br />

Analyses <strong>of</strong> endophenotypes from the Pennsylvania<br />

Computerized Neuropsychological Battery and several<br />

secondary endophenotypes are currently in progress. Thus far<br />

these analyses have collectively identified regions <strong>of</strong> linkage<br />

with LOD scores>3.0 on chromosomes 5p (PPI) and 10q<br />

(DS-CPT), <strong>of</strong> which the chromosome 10qlinkage meets<br />

genome-wide criteria for ‘significant’ linkage. We also<br />

observed ‘suggestive’ evidence for linkage (LOD>2.2) on<br />

chromosomes 1q (antisaccade), 3p (DS-CPT), and 14q(LNS).


These findings are encouraging and suggest that we will<br />

ultimately be successful in identifying genes contributing to<br />

the expression <strong>of</strong> these endophenotypes. However, we note<br />

that these analyses are preliminary, as this sample will be<br />

supplemented with our remaining 94 families currently being<br />

prepared for submission for genotyping, after which the final<br />

linkage analyses will be performed.<br />

250 SEARCHING THE CORRELATED MARKERS OF<br />

NRG1 IN POSTMORTEM BRAIN.<br />

M. Lee* (1), S. Yim (1)<br />

1. Seoul National Hospital<br />

*migyunglee37@gmail.com<br />

Neuregulin1- ErbB4 signal has a critical role in understanding<br />

<strong>of</strong> the schizophrenia pathophysiology. NRG1 expression<br />

abnormalities have been observed in patients and mice. So, we<br />

search the NRG1 expression by using the Stanley<br />

Neuropathology Consortium Integrative Database (SNCID)<br />

which is a web-based method to integrate the Stanley <strong>Medical</strong><br />

research Institute data sets. We found that cytoplasmic protein<br />

level <strong>of</strong> 53KD NRG1 intracelluar cleavage domain (ICD) is<br />

significantly increased in cytoplasmic fractions <strong>of</strong><br />

schizophrenia (n=15) compared with normal control(n=15,<br />

p≤0.001), depression(n=15, p≤0.01) and bipolar affective<br />

disorder(n=15, p≤0.01). Spearman’s test exhibited that 53 KD<br />

NRG1 level is significant correlated with glutamic acid<br />

decarboxylase( GAD)(p=0.007) at orbit<strong>of</strong>rontal cortex/III,<br />

NMDA receptor subunit zeta(p=0.009) at frontal lobe and total<br />

cathechol –o-methyltransferase (COMT) (p=0.007) at<br />

Brodmann area 46. It is well known about relation between<br />

NRG1 and glutamate system in schizophrenia, but not in<br />

NRG1 and COMT. COMT is a risk factor for psychosis and<br />

metastatic cancer. Sei et al reported the evidence <strong>of</strong> epistatic<br />

interaction between COMT gene and NRG1 gene at<br />

schizophrenia derived B lymphocytes. Although it was not<br />

elucidated about COMT gene and NRG1 gene interaction in<br />

these results, it addressed the novel possibility <strong>of</strong> epistatic<br />

interaction evidence in postmortem brain.<br />

251 QKI-7 REGULATES EXPRESSION OF<br />

INTERFERON-RELATED GENES IN HUMAN<br />

ASTROCYTES<br />

L. Jiang (1), E. Lindholm Carlstrom* (1), P. Saetre (2), E.<br />

Jazin (1)<br />

1. Uppsala University 2. Karolinska Institute<br />

*eva.lindholm@ebc.uu.se<br />

The gene the quaking homolog, KH domain RNA binding<br />

(mouse) (QKI), is a candidate gene for schizophrenia encoding<br />

an RNA-binding protein and was recently shown to be<br />

essential for myelination in oligodendrocytes. QKI is also<br />

expressed in astrocytes, but its function in these cells is not<br />

known. We studied the effect <strong>of</strong> small interference RNA<br />

(siRNA)-mediated QKI depletion on global gene expression in<br />

human astrocyte glioma cells. The most significant alteration<br />

after QKI silencing was the decreased expression <strong>of</strong> genes<br />

involved in interferon (IFN) induction (P = 6.3×10-10). These<br />

genes were down-regulated after silencing <strong>of</strong> the splice variant<br />

QKI-7, but were not affected by QKI-5 silencing.<br />

Interestingly, four <strong>of</strong> them were up-regulated after treatment<br />

with the antipsychotic haloperidol that also resulted in<br />

increased QKI-7 mRNA levels. Our results suggest a novel<br />

role for QKI-7 as a regulator <strong>of</strong> IFN production in astrocytes.


252 ALLELIC VARIATION IN THE MICRORNA<br />

GENE, HSA-MIR-943, AFFECTS THE RISK OF<br />

SCHIZOPHRENIA<br />

M. Barenboim* (1), R. Straub (1), R. Vakkalanka (1), D.<br />

Weinberger (1)<br />

1. Clinical Brain Disorders Branch, Genes, Cognition, and<br />

Psychosis Program, National Institute <strong>of</strong> Mental Health, NIH<br />

*barenboimm@mail.nih.gov<br />

MicroRNAs (miRNAs) are short (~22 nt) regulatory,<br />

non-coding RNAs. Primary miRNA transcripts form<br />

characteristic stem-loop structures. In the nucleus, pri-miRNA<br />

is first processed into pre-miRNA (~100 nt) by Drosha and<br />

after exported in the cytoplasm, into mature miRNA by Dicer.<br />

miRNA integrated into the RISC complex, binds to the<br />

3’UTR <strong>of</strong> a target mRNA and represses gene expression on<br />

the post-transcriptional level. Disease-associated SNPs are<br />

known to modulate gene expression. Hence, the study <strong>of</strong> SNPs<br />

located either in a miRNA gene or in the miRNA target site is<br />

<strong>of</strong> importance. We have identified 12 HapMap CEU SNPs<br />

located in miRNA genes. 7 SNPs were tested with the<br />

s<strong>of</strong>tware suite, Unphased, for association with schizophrenia<br />

using family-based data from the GCAP/NIMH Sibling Study.<br />

One <strong>of</strong> these SNPs (rs1077020), which was located in the<br />

stem-loop structure <strong>of</strong> hsa-miR-943, was nominally associated<br />

with schizophrenia (p = 0.03). A subsequent<br />

semi-independent case-control test <strong>of</strong> rs1077020 corroborated<br />

the association <strong>of</strong> this SNP with schizophrenia (p = 0.0005).<br />

hsa-miR-943 is located in the junction <strong>of</strong> an intron and ORF <strong>of</strong><br />

the WHSC2 gene and also in the 5’UTR <strong>of</strong> one is<strong>of</strong>orm. The<br />

test for association with schizophrenia <strong>of</strong> 13 HapMap SNPs in<br />

WHSC2 was negative. RNA secondary structure analysis<br />

shows that allele ‘G’ reduced the stability <strong>of</strong> the stem-loop<br />

structure. This could lead to a reduction in the yield <strong>of</strong> mature<br />

hsa-miR-943 and weaken the regulation and control <strong>of</strong> the<br />

target gene. The predicted list <strong>of</strong> genes targeted by<br />

hsa-miR-943 includes prominent candidate schizophrenia<br />

genes such as DISC1, BDNF, STX18, SLC1A3.<br />

253 THE SCHIZOPHRENIA SUSCEPTIBILITY GENE<br />

ZNF804A CONTAINS POLYMORPHISM<br />

INFLUENCING ITS EXPRESSION IN BRAIN<br />

M. Hill* (1), F. Buonocore (1), N. Bray (1)<br />

1. Centre for the Cellular Basis <strong>of</strong> Behaviour, Department <strong>of</strong><br />

Neuroscience, The James Black Centre, Institute <strong>of</strong><br />

Psychiatry, Kings College London, 125 Coldharbour Lane,<br />

London, SE5 9NU, UK<br />

*matthew.hill@kcl.ac.uk<br />

Genome-wide association studies have identified ZNF804A as<br />

a susceptibility locus for schizophrenia. Although ZNF804A is<br />

known to contain several non-synonymous variants, these do<br />

not appear to fully account for the observed association signal.<br />

Indeed, the most strongly associated polymorphism is located<br />

in a non-coding region <strong>of</strong> the gene, suggesting that the<br />

susceptibility is mediated, at least in part, by effects on<br />

ZNF804A expression. We screened 10 human brain regions<br />

for the presence <strong>of</strong> cis-acting effects on ZNF804A expression<br />

using highly quantitative measures <strong>of</strong> relative allelic<br />

expression. We found statistically significant departures from<br />

the expected 1:1 allelic ratio <strong>of</strong> ZNF804A expression in<br />

several individuals, indicating the presence <strong>of</strong> cis-regulatory<br />

polymorphism. Furthermore, within individuals, these<br />

cis-regulatory effects were limited to specific brain regions.<br />

Investigation <strong>of</strong> the relationship between<br />

schizophrenia-associated variants and observed distortions in<br />

allelic expression will enable assessment <strong>of</strong> the role <strong>of</strong><br />

ZNF804A expression in schizophrenia susceptibility and the<br />

brain regions that are likely to be primarily involved.


254 EVALUATION OF GENETIC ASSOCIATION<br />

BETWEEN SCHIZOPHRENIA AND RHEUMATOID<br />

ARTHRITIS<br />

J. Park* (1), H. Lee (2), H. Kim (3), S. Kang (3), S. Kim (3),<br />

A. Cho (1), J. Kim (4), J. Chung (3)<br />

1. Department <strong>of</strong> Psychiatry, Kyung Hee University East-West<br />

Neo <strong>Medical</strong> Center, Seoul, Korea 2. Department <strong>of</strong><br />

Pharmacology, College <strong>of</strong> Medicine, Kangwon National<br />

University, Chuncheon, Korea 3. Kohwang <strong>Medical</strong> Research<br />

Institute, School <strong>of</strong> Medicine, Kyung Hee University, Seoul,<br />

Korea 4. Department <strong>of</strong> Neuropsychiatry, School <strong>of</strong> Medicine,<br />

Kyung Hee University, Seoul, Korea<br />

*parkdawit@naver.com<br />

Introduction: Schizophrenia and rheumatoid arthritis (RA) are<br />

both chronic diseases with a strong genetic component. The<br />

association between schizophrenia and RA was confirmed in<br />

many studies, providing clues as to the common genetic<br />

etiology. To find common genetic factors between two<br />

diseases, a genome-wide association study was performed.<br />

Methods: 100,000 single nucleotide polymorphisms (SNPs)<br />

were evaluated by Illumina Infinium® DNA Analysis<br />

BeadChips in 24 patients with schizophrenia, 24 patients with<br />

RA, and 34 controls <strong>of</strong> Korean population.<br />

Results: Two SNPs (one from EPM2AIP1 gene on<br />

chromosome 3; the other from KLHL1 gene on chromosome<br />

13) were found to be associated with SPR and RA<br />

simultaneously. There were no significant differences in the<br />

genotype and allele frequencies <strong>of</strong> the two SNPs between the<br />

two diseases. Discussion: These results suggest the possibility<br />

that schizophrenia and RA have a common etiologic factor in<br />

the genetic level, and the genetic factor may explain the<br />

association between the two diseases.<br />

291 MICRODUPLICATIONS OF 16P11.2 ARE<br />

ASSOCIATED WITH SCHIZOPHRENIA<br />

S. McCarthy (1), G. Kirov (2), A. Addington (3), J.<br />

McClellan (4), M. Kusenda (1), O. Krastoshevsky (5), R.<br />

Kumar (6), T. Crow (7), S. Christian (6), J. Lieberman (8), F.<br />

McMahon (3), A. Malhotra (9), J. Potash (10), T. Schulze (3),<br />

E. Leibenluft (3), J. Sutcliffe (11), D. Skuse (12), M. Gill (13),<br />

N. Mendell (14), N. Craddock (2), M. Owen (2), M.<br />

O’Donovan (2), T. Shaikh (15), E. Susser (16), L. DeLisi (17),<br />

P. Sullivan (18), C. Deutsch (19), J. Rapoport (3), D. Levy (5),<br />

M. King (4)<br />

1. Cold Spring Harbor Laboratory 2. Cardiff University 3.<br />

National Institute for Mental Health 4. University <strong>of</strong><br />

Washington 5. McLean Hospital 6. University <strong>of</strong> Chicago 7.<br />

The Prince <strong>of</strong> Wales International Center for SANE Research<br />

8. Columbia University 9. The Zucker Hillside Hospital 10.<br />

Johns Hopkins School <strong>of</strong> Medicine 11. Vanderbilt University<br />

12. University College London 13. Trinity College Dublin 14.<br />

State University <strong>of</strong> New York 15. The Children's Hospital <strong>of</strong><br />

Philadelphia 16. Columbia University 17. Harvard <strong>Medical</strong><br />

School 18. University <strong>of</strong> North Carolina 19. University <strong>of</strong><br />

Massachusetts <strong>Medical</strong> School<br />

An increase in the burden <strong>of</strong> rare copy number variants<br />

(rCNVs) and the association <strong>of</strong> recurrent rCNVs at 1q21,<br />

15q11, 15q13 and 22q11 in schizophrenia implicate rCNVs in<br />

the etiology <strong>of</strong> the disorder.We present the association <strong>of</strong> rare<br />

16p11.2 microduplications with schizophrenia in a cohort <strong>of</strong><br />

1921 cases and 4062 controls (P=1.4x10-5, OR=25.4). This<br />

association was replicated using an independent sample <strong>of</strong><br />

2645 cases and 2420 controls (P=0.022, OR=8.3) from GAIN<br />

(phs000021.v2.p1). In a meta-analysis <strong>of</strong> psychiatric disorders,<br />

the 16p11.2 microduplication was consistently observed at a<br />

significantly greater frequency in schizophrenia and autism<br />

than in corresponding controls, 0.3% vs. 0.03% (P=5x10-7)<br />

and 0.46% vs. 0.02% (P=1.9x10-7) respectively. The<br />

microduplication was observed at a higher frequency in<br />

bipolar disorder (0.1% vs 0.03%, P=0.055). In contrast, the<br />

reciprocal 16p11.2 microdeletion increased the risk <strong>of</strong> autism<br />

specifically (0.78% vs. 0.02% OR=38.7, P=2.3x10-13).<br />

Analysis <strong>of</strong> quantitative patient clinical data showed that<br />

standardized head circumference based on the CDC growth<br />

charts was significantly larger in microdeletion carriers and<br />

moderately smaller in microduplication carriers (1.25 vs.<br />

-0.28, P=0.0007). Our findings add 16p11.2 to the growing list<br />

<strong>of</strong> hotspots that considerably increase the risk <strong>of</strong><br />

schizophrenia. The spectrum <strong>of</strong> diseases associated with<br />

16p11.2 is consistent with phenotypic heterogeneity at other<br />

schizophrenia risk loci and suggests that common<br />

neurobiological pathways maybe shared between phenotypes<br />

associated with 16p11.2 rearrangements. Finally the<br />

association <strong>of</strong> 16p11.2 rearrangements with head<br />

circumference point to a potential genetic basis for early brain<br />

overgrowth in autism and observations <strong>of</strong> smaller brain<br />

volume in schizophrenia.


STATISTICS AND BIOINFORMATICS<br />

255 ESTIMATING THE ACCURACY AND<br />

CONCORDANCE OF CURRENT METHODS FOR THE<br />

DETECTION OF COPY NUMBER POLYMORPHISMS<br />

IN WHOLE GENOME ASSOCIATION STUDIES<br />

C. Vogler* (1), L. Gschwind (2), B. Röthlisberger (3), A.<br />

Huber (3), J. Sigmund (2), D. de Quervain (6), A.<br />

Papassotiropoulos (2)<br />

1. Division <strong>of</strong> Molecular Psychology, University <strong>of</strong> Basel,<br />

Basel, Switzerland. Life Science Training Facility, Biocentre,<br />

University <strong>of</strong> Basel, Basel, Switzerland 2. Division <strong>of</strong><br />

Molecular Psychology, University <strong>of</strong> Basel, Basel,<br />

Switzerland. 3. Center <strong>of</strong> Laboratory Medicine, Cantonal<br />

Hospital, Aarau, Switzerland 4. Division <strong>of</strong> Psychiatry<br />

Research, University <strong>of</strong> Zurich, Lenggstr. 31, 8029 Zurich,<br />

Switzerland<br />

*christian.vogler@unibas.ch<br />

Background: Since the discovery that Copy Number<br />

Polymorphisms (CNPs) - common Copy Number Variations -<br />

may account for a large amount <strong>of</strong> genetic variation in<br />

humans, great efforts have been put in the thorough and<br />

reliable detection <strong>of</strong> CNPs. A special hybrid genotyping array<br />

(Affymetrix SNP 6.0) has been designed to assess copy<br />

number states throughout the genome using a total <strong>of</strong> 1.8<br />

million genomic markers (~900,000 SNPs and ~900,000 Copy<br />

Number Markers). Furthermore a s<strong>of</strong>tware package (birdsuite)<br />

applying the canary algorithm has been provided by the<br />

BROAD Institute, which can be used to assign copy number<br />

states across regions <strong>of</strong> common copy number polymorphisms<br />

.Methods: In our study we individually genotyped 771 healthy<br />

Swiss subjects using the Affymetrix SNP 6.0 Genechip. We<br />

aimed at conducting a CNP analysis applying the canary<br />

algorithm and to subsequently assess the accuracy <strong>of</strong> this<br />

approach using Real-Time-quantitative PCR (RT qPCR) and<br />

array Comparative Genomic Hybridzation (aCGH) as a<br />

validation tool for CNPs. Results and Conclusion: Processing<br />

<strong>of</strong> Affymetrix SNP 6.0 GeneChips worked well and the bird<br />

suite s<strong>of</strong>tware package could be successfully applied. CNP<br />

analysis has been carried out using the Broad-CNP-Map<br />

comprising 1316 CNPs. Applying the recommended quality<br />

controls given by the BROAD institute, in a remaining sample<br />

size <strong>of</strong> 724 individuals a total number <strong>of</strong> 11943 CNPs > 90kb<br />

were detected. Evaluation <strong>of</strong> results with RT qPCR and aCGH<br />

is currently under progress.<br />

256 COMPARISON BETWEEN THREE DIFFERENT<br />

APPROACHES FOR GENOME-WIDE<br />

IDENTIFICATION AND ANALYSIS OF COPY<br />

NUMBER VARIATIONS<br />

L. Gschwind* (1), C. Vogler (1), I. Filges (2), J. Sigmund (1),<br />

D. de Quervain (3), A. Papassotiropoulos (1)<br />

1. Division <strong>of</strong> Molecular Psychology, University <strong>of</strong> Basel,<br />

Basel, Switzerland Life Science Training Facility, Biocentre,<br />

University <strong>of</strong> Basel, Basel, Switzerland<br />

2. Division <strong>of</strong> <strong>Medical</strong> Genetics, Department <strong>of</strong> Biomedicine,<br />

University Children’s Hospital, Basel, Switzerland<br />

3. Division <strong>of</strong> Psychiatry Research, University <strong>of</strong> Zurich,<br />

Zurich, Switzerland<br />

*leo.gschwind@stud.unibas.ch<br />

Background: Very recently Copy Number Variations (CNVs)<br />

have been shown to affect different phenotypic traits such as<br />

susceptibility to disease. Currently, methods for in silico<br />

analysis to identify CNVs using hybrid genotyping array<br />

intensity data are being developed. So far an extensive<br />

evaluation <strong>of</strong> the robustness <strong>of</strong> these algorithms is still<br />

lacking. Methods: We used Affymetrix SNP 6.0 GeneChip<br />

data from 724 healthy Swiss individuals and investigated three<br />

different CNV detecting algorithms (PennCNV, Birdseye and<br />

CNAM). Subsequently to analysis, results <strong>of</strong> all three CNV<br />

detection tools were compared to determine concordance. As<br />

an independent quality control, we also used the Affymetrix<br />

SNP 6.0 GeneChip to process DNA from three clinical<br />

samples with verified large-scale structural variations. We<br />

examined the viability <strong>of</strong> the three different algorithms to<br />

detect the known genomic aberrations. Results and<br />

Conclusion: Affymetrix SNP 6.0 GeneChips were processed<br />

and algorithms could be successfully applied. Preliminary<br />

results indicate that all three algorithms reliably detect sizable<br />

structural variants. Analysis <strong>of</strong> concordance between the three<br />

methods is currently under progress.


257 MACHINE-LEARNING APPROACHES FOR<br />

GENOME-WIDE ASSOCIATION STUDIES OF MOOD<br />

DISORDERS*FAYAZ SEIFUDDIN, MEHDI<br />

PIROOZNIA, JENNIFER JUDY, PAMELA MAHON,<br />

JAMES POTASH, PETER ZANDI<br />

F. Seifuddin* (1), M. Pirooznia (1), J. Judy (1), P. Mahon (1),<br />

J. Potash (1), P. Zandi (1)<br />

1. The Johns Hopkins University School <strong>of</strong> Medicine, Mood<br />

Disorders Program<br />

*fseifud1@jhmi.edu<br />

Mood disorders are highly heritable forms <strong>of</strong> major mental<br />

illnesses. A major breakthrough in genetic association <strong>of</strong><br />

mood disorders was anticipated with the advent <strong>of</strong> genome<br />

wide association studies (GWAS). However, to date few<br />

major significant findings have been identified, which<br />

suggests the complex genetic etiology <strong>of</strong> mood disorders,<br />

requiring alternative approaches to analyzing GWAS data.<br />

Machine learning (ML) and data mining methods have been<br />

successfully employed for both supervised and unsupervised<br />

learning to mine and identify biomarkers in gene expression<br />

studies. In this study, we compared four ML methods, namely,<br />

decision tree, Bayesian networks, support vector machine, and<br />

neural networks, in the analysis <strong>of</strong> GWAS data on bipolar<br />

disorder and major depression. Bipolar datasets were obtained<br />

from the Genetic Association Information Network (GAIN)<br />

and Welcome Trust Case Control Consortium (WTCCC)<br />

bipolar samples. Major depression datasets were obtained<br />

from GAIN depression and Genetics <strong>of</strong> Recurrent Early-onset<br />

Depression (GENRED) samples. We chose one dataset for<br />

each disorder as the training dataset and built SNP classifier<br />

models to distinguish cases and controls. The classifiers were<br />

then applied to the second test datasets and their accuracy for<br />

classification was compared. Different feature selection<br />

methods were explored in order to address the dimensionality<br />

problem and improve the performance <strong>of</strong> the classifiers.<br />

Results across the four methods will be presented for each<br />

disorder. ML methods <strong>of</strong>fer a novel approach to analyze<br />

GWAS data, but their performance characteristics need to be<br />

evaluated to better understand how they may be used to<br />

unravel the complex genetic etiology <strong>of</strong> common mood<br />

disorders.<br />

258 BAYESIAN APPROACHES TO ANALYSING TRIO<br />

DATA<br />

E. Heron* (1), R. Anney (1), L. Gallagher (1), M. Gill (1)<br />

1. Neuropsychiatric Genetics Research Group, Dept. <strong>of</strong><br />

Psychiatry, Trinity College Dublin, Ireland.<br />

*eaheron@tcd.ie<br />

For early onset psychiatric diseases such as ADHD and<br />

Autism, trio data, consisting <strong>of</strong> two parents and an affected<br />

child, can sometimes be collected for GWAS type analyses.<br />

These statistical analyses aim to detect an association between<br />

a disease locus (or a locus in LD with the disease locus) and<br />

the particular disease phenotype under study. This trio data<br />

differs from the usual case control type data used in the<br />

majority <strong>of</strong> GWAS studies, as data from an explicit control<br />

group is not collected. GWAS analyses for trio data are<br />

usually carried out using the transmission disequilibrium test<br />

(TDT). The TDT examines transmissions and<br />

non-transmissions <strong>of</strong> alleles from heterozygous parents to<br />

probands, thus creating pseudo-controls for comparisons. A<br />

Bayesian approach to examine associations for trio data is<br />

presented. This approach uses Bayes factors to compare<br />

models (or hypotheses), ultimately resulting in posterior odds<br />

for a model <strong>of</strong> association for each individual SNP (Equation<br />

1, Y is data). p(H1|Y)/p(H0|Y) = [p(Y|H1)/p(Y|H0)] x<br />

[p(H1)/p(H0)](1)Posterior Odds <strong>of</strong> H1 = Bayes factor x Prior<br />

Odds <strong>of</strong> H1Unlike the p-value approach to testing for<br />

association, the Bayes factor does not require a multiple<br />

testing correction. Instead, meaningful prior information in the<br />

form <strong>of</strong> prior odds <strong>of</strong> association can be incorporated. This<br />

prior information depends on the number <strong>of</strong> SNPs expected to<br />

be disease variants (or in LD with disease variants), not on the<br />

number <strong>of</strong> SNPs genotyped or on the number <strong>of</strong> tests carried<br />

out. We explore the Bayes factor approach using both<br />

simulated and real trio datasets.


259 PHENOMIC ANALYSIS OF PSYCHIATRIC<br />

SYMPTOMS AND RATING SCALES IN PARKINSON’S<br />

DISEASE<br />

R. Schork* (1), M. Houser (2), C. Bloss (1), N. Schork (3)<br />

1. Scripps Genomic Medicine; Elon University 2. Division <strong>of</strong><br />

Neurology, Scripps Clinic 3. Department <strong>of</strong> Molecular and<br />

Experimental Medicine, The Scripps Research Institute<br />

*rschork@elon.edu<br />

Non-motor symptoms in Parkinson’s disease (PD) are<br />

common, but poorly understood. One issue is a paucity <strong>of</strong><br />

research in this area, and another is evidence that traditional<br />

analysis <strong>of</strong> total scores on standard psychiatric rating scales<br />

may be inadequate for assessment <strong>of</strong> these symptoms (e.g.,<br />

anxiety) in PD. As such, we conducted association analysis <strong>of</strong><br />

psychiatric symptoms in PD, comparing findings from<br />

analysis <strong>of</strong> total rating scale scores versus item-level pr<strong>of</strong>iles.<br />

Four scales administered to 23 PD patients were evaluated,<br />

including the State-Trait Anxiety Inventory, Geriatric<br />

Depression Scale, Beck Depression Inventory, and Marin<br />

Apathy Scale. Independent variables (IVs) hypothesized to be<br />

associated with psychiatric symptomatology were identified<br />

from the literature, and two sets <strong>of</strong> analyses were performed:<br />

(1) linear regression <strong>of</strong> total scores for each scale on each IV;<br />

and (2) multivariate distance matrix regression (MDMR) <strong>of</strong><br />

item-level pr<strong>of</strong>iles on each IV. Fourteen IVs were identified,<br />

including demographic and disease-related variables,<br />

cognitive indices, daytime sleepiness, and comorbid disease.<br />

Results revealed differences in the IVs found to be associated<br />

with total scores versus item-level pr<strong>of</strong>iles on each <strong>of</strong> the four<br />

scales. Across all <strong>of</strong> the scales, IVs with the most discrepant<br />

association statistics between the two methods included<br />

daytime sleepiness, Mini-Mental Status, and perceived<br />

memory problems. Furthermore, follow-up analyses suggest<br />

that certain items on each scale may be more salient than<br />

others for psychiatric assessment in PD populations. Findings<br />

suggest that item-level “phenomic” analysis <strong>of</strong> psychiatric<br />

rating scales may provide useful additional information in the<br />

assessment <strong>of</strong> psychiatric symptomatology in PD.<br />

260 BLOCK-WISE GENOME-WIDE ASSOCIATION<br />

ANALYSIS IDENTIFIES A LIMITED NUMBER OF<br />

REPLICABLE CANDIDATE REGIONS LINKED WITH<br />

SCHIZOPHRENIA<br />

T. Gladwin (1), E. Derks* (1), W. Cahn (1), R. Kahn (1), R.<br />

Oph<strong>of</strong>f (1)<br />

1. Department <strong>of</strong> Psychiatry, Rudolf Magnus Institute <strong>of</strong><br />

Neuroscience, University <strong>Medical</strong> Centre Utrecht, Utrecht,<br />

The Netherlands<br />

*e.m.derks@umcutrecht.nl<br />

Schizophrenia is a complex disorder with high heritability.<br />

Genome Wide Association (GWA) studies have been hindered<br />

by low statistical power to detect risk alleles with small effect<br />

sizes. We will introduce a new approach to the analysis <strong>of</strong><br />

GWA data. Genotypic data were available for 728 cases and<br />

653 controls from the Netherlands; GAIN data <strong>of</strong> 1,172 cases<br />

and 1,378 were used for replication. The aim is to identify<br />

regions with a significantly increased number <strong>of</strong> nominally<br />

significant tests. To this end, 22 autosomal chromosomes were<br />

split into 1390 2 MB regions. While single SNP and haplotype<br />

analyses did not result in statistically significant findings, we<br />

identified 13 regions with an increase in nominally significant<br />

SNPs. Five <strong>of</strong> these regions could be replicated (p


261 POWER DIFFERENCES FOR FAMILY-BASED<br />

EPISTASIS DETECTION METHODS<br />

S. Bergen* (1), A. Fanous (2), K. Kendler (3), B. Maher (4)<br />

1. Department <strong>of</strong> Human and Molecular Genetics, Virginia<br />

Commonwealth University 2. Washington VA <strong>Medical</strong><br />

Center, Georgetown University <strong>Medical</strong> Center, Virginia<br />

Commonwealth University 3. Virginia Institute for Psychiatric<br />

and Behavrioral Genetics, Department <strong>of</strong> Psychiatry, Virginia<br />

Commonwealth University 4. Departments <strong>of</strong> Psychiatry and<br />

Human and Molecular Genetics, Virginia Commonwealth<br />

University<br />

*sbergen@gmail.com<br />

The impact <strong>of</strong> genetic variation on common diseases is<br />

complex, involving many loci and their interactions. In<br />

population-based association samples, modeling gene-gene<br />

interaction, or epistasis, the conditional impact <strong>of</strong> one genetic<br />

polymorphism on another, is a relatively straightforward<br />

exercise. In these samples, genotypes at multiple loci, with<br />

their interaction term(s), can be included in a regression model<br />

as predictors. Detection <strong>of</strong> gene-gene interactions in<br />

family-based association samples is more complicated, but<br />

several methods have been proposed. In these approaches<br />

pseudocontrols are created based on the alleles or genotypes<br />

that were not transmitted to an affected <strong>of</strong>fspring from<br />

genotyped parents. Two main approaches to generating<br />

pseudocontrols involve using nontransmitted two-locus<br />

genotypes (Cordell approach) and nontransmitted alleles<br />

(two-locus haplotype relative risk approach; HRR). In the<br />

former, 15 pseudocontrols would be generated, while the latter<br />

yields only three. These cases and pseudocontrols are then<br />

analyzed using logistic regression conditional on the parental<br />

genotypes. The core distinction between these approaches is<br />

the inclusion <strong>of</strong> transmitted alleles in the creation <strong>of</strong><br />

pseudocontrol genotypes in the Cordell approach. We have<br />

explored the power for both <strong>of</strong> these methods under a variety<br />

<strong>of</strong> models <strong>of</strong> epistasis in 1000 simulated samples <strong>of</strong> 500<br />

parent-child trios. Under “classic” epistasis models, the HRR<br />

approach yields greater power, but under most other models<br />

(recessive, additive, heterogeneity, and crossover) the Cordell<br />

approach demonstrates increased power for detecting epistasis.<br />

These results suggest different approaches to family based<br />

epistasis testing are indicated depending on the expected<br />

model <strong>of</strong> epistasis in effect.<br />

262 PSYCHIATRIC GWAS CONSORTIUM -<br />

CHALLENGES IN WORLD WIDE GENETIC<br />

ASSOCIATION STUDIES<br />

S. Ripke* (1), M. Daly (1), B. Neale (1)<br />

1. CHGR<br />

* ripke@chgr.mgh.harvard.edu<br />

Here we present a quality control pipeline for genome-wide<br />

association SNP data. This pipeline was developed at MGH in<br />

Boston, as a direct outgrowth <strong>of</strong> the Psychiatric GWAS<br />

Consortium Analytic Committee. The aims <strong>of</strong> this pipeline are<br />

to execute the quality control <strong>of</strong> GWAS-SNP data, impute<br />

variation based on a reference sample and combine the data in<br />

a single analysis. The quality control steps include the<br />

capacity to identify relatives and overlapping samples in and<br />

across studies, as well as produce principal components<br />

representing population variation to control for stratification in<br />

downstream-analysis and ancestry outlier exclusion. Through<br />

the use <strong>of</strong> parallelization and massive computer clusters such<br />

as the GCC (Netherlands), preliminary results can be<br />

generated in as little as one week, even for study sizes <strong>of</strong> over<br />

40,000 individuals. This poster is meant as an addition to the<br />

Plenary Session 4 – Psychiatric GWAS Consortium on<br />

Sunday, November 8th, and should educate the field about the<br />

challenges from handling multiple datasets from all over the<br />

world, as well as provide insight into emerging solutions to<br />

these difficulties.


263 EVIDENCE FOR EPISTASIS IN GENOME-WIDE<br />

ASSOCIATION STUDIES OF BIPOLAR DISORDER<br />

J. Judy* (1), P. Belmonte (1), F. Seifuddin (1), M. Pirooznia<br />

(1), Bipolar Genome Study (BiGS), J. Potash (1), P. Zandi (1)<br />

1. Johns Hopkins School <strong>of</strong> Medicine, Johns Hopkins School<br />

<strong>of</strong> Public Health<br />

* jtoolan1@jhmi.edu<br />

Bipolar Disorder (BD) is highly heritable, suggesting an<br />

underlying genetic etiology. However, efforts to identify<br />

causal genetic variants in BD have been markedly<br />

unsuccessful. Epistasis, whereby a genetic variant at one locus<br />

influences the phenotypic effect <strong>of</strong> a different locus, may<br />

contribute to this lack <strong>of</strong> discovery. Data from large scale<br />

genome-wide association studies (GWAS) affords researchers<br />

the opportunity to explore this phenomenon. We examined the<br />

potential role <strong>of</strong> epistasis in BD by analyzing data from two<br />

GWAS from the BiGS Consortium: an initial sample <strong>of</strong> 1001<br />

cases and 1034 controls from the GAIN data, and a follow-up<br />

sample <strong>of</strong> 1199 cases and 403 controls from the TGEN data.<br />

We used PLINK to search for statistically significant pairwise<br />

interactions, followed by querying functional annotation<br />

databases to find evidence for biological interactions. We<br />

followed two complementary strategies. We first employed<br />

the “clumping” routine to identify the 513 most significantly<br />

associated loci that tagged the remaining SNPs, and tested for<br />

interactions between them. We then examined the pairwise<br />

interactions between SNPs in ANK3, which has been<br />

implicated as genome-wide significant in BD, and SNPs<br />

across the genome. The most significant interaction consistent<br />

between the two samples was between rs4948254 in ANK3<br />

and 3 SNPs (rs2206135, rs2235460, and rs4811247) in<br />

CTNNBL1 (p-values between 10e-03 to 10e-05).<br />

Interestingly, CTNNBL1 is a homologue to beta-catenin, a<br />

key component <strong>of</strong> the Wnt signaling pathway, and is involved<br />

in apoptosis. We plan to test for replication <strong>of</strong> this interaction<br />

using the WTCCC data.<br />

264 GENOME WIDE STRATEGIES TO CORRECT<br />

FOR MULTIPLE TESTING IN THE STUDY OF<br />

GENETIC INTERACTIONS<br />

G. Guffanti* (1), H. Stern (1), J. Turner (1), S. Potkin (1), F.<br />

Macciardi (1), N. Schork (2)<br />

1. University <strong>of</strong> California, Irvine 2. Scripps Health and The<br />

Scripps Research Institute<br />

*gguffant@uci.edu<br />

Success in identifying susceptibility genes for complex traits<br />

is strongly affected by the ability to describe the effects <strong>of</strong><br />

gene-environment (GxE) interactions. We present a<br />

comprehensive genome-wide strategy to test GxE interactions<br />

through an integrated methodological framework. This<br />

methodology performs iterative resampling procedures for<br />

interaction terms to correct for multiple comparisons. Our<br />

methodology tests separately tests GxE interaction effects<br />

from the main effects using the traditional regression-based<br />

general linear models (GLMs). We isolate the proportion <strong>of</strong><br />

variance explained by the interaction term factoring out main<br />

effects from the GLM that tests the hypothesis <strong>of</strong> no<br />

interaction. Then we apply non-parametric inferential<br />

data-driven procedures to the variance explained by the<br />

interaction and contrast the results with<br />

null-quantile-transformed distribution. The rejection regions<br />

for the test statistics are defined using stepwise multiple<br />

testing procedures to derive the appropriate adjustment <strong>of</strong> the<br />

p-values. We evaluated the accuracy <strong>of</strong> the multiple testing<br />

corrections in both simulation and real dataset studies<br />

assessing effect size, power and sample size. We validated our<br />

novel measure to describe GxE interactions’ effects on disease<br />

risk by contrasting the E+,BB events that identify the high risk<br />

pr<strong>of</strong>ile subjects to the rest <strong>of</strong> the individuals involved in the<br />

study, the negative risk pr<strong>of</strong>ile subjects. This is the first<br />

statistical approach that recasts the relationship between the<br />

four main components <strong>of</strong> statistical hypothesis testing (i.e.,<br />

significance level α, power, effect size, sample size) so that it<br />

can account for the specific characteristics <strong>of</strong> a complex<br />

phenomenon like GxE interaction.


265 MODELING THE COMBINED EFFECTS OF SNPS<br />

USING BINOMIAL LOGISTIC REGRESSION:<br />

SELECTION CRITERIA AND THE USE OF<br />

PRINCIPAL COMPONENTS ANALYSIS<br />

T. Gladwin (1), W. Cahn (1), R. Kahn (1), R. Oph<strong>of</strong>f (1), E.<br />

Derks* (1)<br />

1. Department <strong>of</strong> Psychiatry, Rudolf Magnus Institute <strong>of</strong><br />

Neuroscience, University <strong>Medical</strong> Centre Utrecht, Utrecht,<br />

The Netherlands<br />

*e.m.derks@umcutrecht.nl<br />

Genome Wide Association (GWA) studies suggest that many<br />

loci with small effect sizes play a role in schizophrenia. We<br />

present the results <strong>of</strong> a modeling approach that combines these<br />

effects. Genome wide data were available for 728 cases and<br />

653 controls. Principal component analysis was performed on<br />

1618 SNP-variabless which were associated (P


267 SZGR: A COMPREHENSIVE SCHIZOPHRENIA<br />

GENE RESOURCE<br />

P. Jia* (1), J. Sun (1), A. Guo (2), Z. Zhao (1)<br />

1. Vanderbilt University 2. Virginia Institute for Psychiatric<br />

and Behavioral Genetics, Virginia Commonwealth University<br />

*peilin.jia@vanderbilt.edu<br />

Introduction: We have seen during the past two decades an<br />

exponential growth <strong>of</strong> vast amounts <strong>of</strong> biological data in<br />

schizophrenia genetics, including those generated by<br />

traditional positional cloning approach, individual<br />

gene/marker association studies and emerging genome-wide<br />

association studies, more than 32 genome-wide linkage scans<br />

and several meta-analyses, and a large number <strong>of</strong> microarray<br />

experiments. Besides these genetic datasets, abundant<br />

biological information for the schizophrenia candidate genes<br />

can be extracted from public databases such as Gene Ontology<br />

annotations, protein-protein interaction (PPI) networks, and<br />

regulatory and cellular pathways. At present, there is a strong<br />

trend towards integrating the data from various genetic studies<br />

and their related biological information in the cellular systems<br />

so that promising candidate genes can be prioritized for follow<br />

up bioinformatics analysis and experimental verification.<br />

Methods: We designed and implemented Schizophrenia Gene<br />

Resource (SZGR), a comprehensive database with<br />

user-friendly web interface. SZGR was implemented as a<br />

relational database using the open source MySQL database<br />

system and is freely accessible through a web interface<br />

developed in JSP technology. Results: SZGR deposits genetic<br />

data from all available sources including association studies,<br />

linkage scans, gene expression, literature, Gene Ontology<br />

(GO) annotations, gene networks, cellular and regulatory<br />

pathways, and microRNAs and their target sites. Moreover,<br />

SZGR provides online tools for data browse and search, data<br />

integration, custom gene ranking, and graphical presentation.<br />

Conclusions: To our understanding, SZGR is the most<br />

comprehensive resource for schizophrenia, or such kind in<br />

psychiatric genetics. This system can be easily applied to other<br />

complex diseases, especially other psychiatric disorders. The<br />

SZGR database is available at<br />

http://bioinfo.mc.vanderbilt.edu/SZGR/.


SUBSTANCE ABUSE<br />

268 ASSOCIATION BETWEEN THE NMDA<br />

RECEPTOR SUBUNIT 2B GENE (GRIN2B) AND<br />

NICOTINE DEPENDENCE IN A SAMPLE OF<br />

JAPANESE WORKERS<br />

S. Sakata* (1), T. Shinkai (1), H. Chen (2), K. Utsunomiya<br />

(1), Y. Fukunaka (1), K. Yamada (1), O. Ohmori (1), J.<br />

Nakamura (1)<br />

1. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu, Japan<br />

2. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu,<br />

Japan; Department <strong>of</strong> Public Health, Kaohsiung <strong>Medical</strong><br />

University, Kaohsiung, Taiwan<br />

*bossnokobun@yahoo.co.jp<br />

Preclinical research findings in laboratory animals indicate<br />

that the glutamatergic system may be involved in nicotine<br />

dependence. In animals, N-methyl-D-aspartate (NMDA)<br />

receptor antagonists decreased nicotine self-administration or<br />

reinstatement <strong>of</strong> nicotine-seeking behavior. NMDA receptor<br />

dysfunction may be involved in the pathophysiology <strong>of</strong><br />

smoking behavior. The human gene cording for the 2B subunit<br />

<strong>of</strong> the NMDA receptor (GRIN2B) is thus considered a<br />

candidate gene for nicotine dependence. To examine the<br />

hypothesis, we examined the association between smoking<br />

behavior and three GRIN2B polymorphisms, G-200T<br />

(5'UTR), C2664T (exon 13) and T5072G (3'UTR), using a<br />

sample <strong>of</strong> Japanese healthy workers (N = 673). This study<br />

was approved by the Ethics Committee <strong>of</strong> the University <strong>of</strong><br />

Occupational and Environmental Health, and informed<br />

consent was obtained from all subjects prior to participation to<br />

the study. Among smokers, the -200G allele, which has been<br />

associated with lower gene activity, was associated with<br />

having smoking cessation histories (for more than 1 month)<br />

with non-significant trend (P=0.09). Our results suggest that<br />

GRIN2B G-200T polymorphism may play a role in smoking<br />

cessation in our sample. Details <strong>of</strong> association analyses for<br />

other markers as well as marker haplotypes will be presented.<br />

269 POSSIBLE ASSOCIATION BETWEEN THE<br />

ALA72SER POLYMORPHISM OF THE<br />

CATECHOL-O-METHYL TRANSFERASE GENE<br />

(COMT) AND NICOTINE DEPENDENCE IN A<br />

SAMPLE OF JAPANESE WORKERS<br />

K. Utsunomiya* (1), T. Shinkai (2), S. Sakata (3), H. Chen<br />

(4), Y. Fukunaka (5), K. Yamada (6), O. Ohmori (7), J.<br />

Nakamura (8)<br />

1. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health<br />

*k-utsu@med.uoeh-u.ac.jp<br />

Catechol-O-methyl transferase gene (COMT) is one <strong>of</strong> the<br />

enzymes involved in the degradation and inactivation <strong>of</strong><br />

catecholamine transmitters including dopamine, which is<br />

present in dopaminergic brain regions. Nicotine stimulates<br />

dopamine release and activates dopaminergic reward neurons.<br />

The dopaminergic reward system may be a key mechanism in<br />

nicotine dependence. Recently, new functional single<br />

nucleotide polymorphism (SNP) Ala72Ser in the COMT gene,<br />

which accounts for the enzyme activity, was found. The<br />

purpose <strong>of</strong> our study is to examine the association between the<br />

COMT Ala72Ser polymorphism and nicotine dependence in<br />

its each stage (e.g., smoking initiation, dependence<br />

development, and smoking cessation). Smoking status was<br />

confirmed by in-person structured interviews. We have<br />

recruited the present sample from one <strong>of</strong> the Japanese major<br />

manufacturing company, with advantage <strong>of</strong> relatively<br />

homogenous background (total N = 673). Among them, we<br />

defined 435 smoking initiators (SI) and 234 non-initiators<br />

(who reported never having smoked a single cigarette: NI).<br />

Among SI, 417 were high nicotine dependent smokers (who<br />

reported having smoked > 100 cigarettes or total smoking<br />

duration > 6 months in their lifetime: HND), and 16 low<br />

nicotine dependent smokers (LND). Although there was no<br />

significant difference in both genotype and allele frequencies<br />

between SI and NI, we found a significant difference in both<br />

genotype (chi-square= 13.56, d.f. = 2, p= 0.019) and allele<br />

frequencies (chi-square = 5.089, d.f. = 1, p = 0.038, OR =<br />

0.34, 95%CI = 0.14-0.81) <strong>of</strong> the COMT Ala72Ser between<br />

HND and LND. These data suggest that the COMT Ala72Ser<br />

polymorphism is possibly associated with the degree <strong>of</strong><br />

nicotine dependence in Japanese workers.


270 THE ASSOCIATION OF NEURONAL NICOTINE<br />

ACETYLCHOLINE RECEPTOR ALPHA4 AND BETA2<br />

GENE POLYMORPHISMS WITH NICOTINE<br />

DEPENDENCE IN JAPANESE MALE WORKERS<br />

H. Chen* (1), T. Shinkai (2), S. Sakata (2), K. Utsunomiya<br />

(3), Y. Fukunaka (2), K. Yamada (2), O. Ohmori (4), J.<br />

Nakamura (2)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Occupational and<br />

Environmental Health, Kitakyushu, Japan; Department <strong>of</strong><br />

Public Health, Kaohsiung <strong>Medical</strong> University, Kaohsiung,<br />

Taiwan 2. Department <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Occupational and Environmental Health, Kitakyushu, Japan<br />

3. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Occupational and<br />

Environmental Health, Kitakyushu, Japan; Health<br />

Management Department, Shimonoseki Shipyard &<br />

Machinery Works, Mitsubishi Heavy Industries, Ltd.,<br />

Shimonoseki, Japan 4. Department <strong>of</strong> Psychiatry, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu,<br />

Japan; Wakato Hospital, Kitakyushu, Japan<br />

*stbchen@med.uoeh-u.ac.jp<br />

Polymorphisms in the genes coding the nicotinic acetylcholine<br />

receptors have been suggested to play a role in the<br />

determination <strong>of</strong> smoking-related phenotypes because<br />

smoking activates the brain reward system through these<br />

receptors. To examine this hypothesis, we conducted a<br />

genetic association study in a well-defined sample <strong>of</strong> Japanese<br />

workers (N =673). We analyzed 5 SNPs in the CHRNA4<br />

gene and 3 SNPs in the CHRNB2. We assessed nicotine<br />

dependence using the Fagerström Test <strong>of</strong> Nicotine<br />

Dependence (FTND) and the Tobacco Dependence Screener<br />

(TDS). Among 284 current male smokers, 168 were<br />

dependent smokers (DS) (FTND score 4 or more) and 116<br />

were non-dependent smokers (non-DS). We found a<br />

significant difference <strong>of</strong> the CHRNA4 (rs1044397) genotype<br />

distribution between DS and non-DS (chi-square= 7.84, df =<br />

2, p-value = 0.02). As for the CHRNB2 gene polymorphism<br />

(rs4845652), we found non-significant differences in both<br />

genotype (chi-square =2.99, df=1, p-value=0.08) and allele<br />

frequencies (chi-square=3.78, df=1, p-value=0.05) between<br />

DS and non-DS groups. Our data suggests that CHRNA4 and<br />

CHRNB2 gene may correlate to nicotine dependence in our<br />

sample. The gene-gene interaction data will be presented.<br />

271 ASSOCIATION OF POLYMORPHISM IN THE<br />

HUMAN MU-OPIOID RECEPTOR OPRM1 GENE<br />

WITH NICOTINE DEPENDENCE IN JAPANESE<br />

WORKERS<br />

T. Shinkai* (1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Occupational and<br />

Environmental Health, Kitakyushu, Japan<br />

*shinkai@med.uoeh-u.ac.jp<br />

The mu-opioid receptor has been implicated in the<br />

pathogenesis <strong>of</strong> substance dependence including nicotine<br />

dependence. Studies examining the association <strong>of</strong> the<br />

mu-opioid receptor gene (OPRM1) with substance<br />

dependence have focused on the Asn40Asp (A118G) single<br />

nucleotide polymorphism (SNP). In the present study, we<br />

tested association between the functional OPRM1 A118G<br />

polymorphism (rs1799971) and nicotine dependence in its<br />

each stage (e.g., smoking initiation, dependence development,<br />

and smoking cessation) in a well-defined sample <strong>of</strong> Japanese<br />

workers. We have recruited the present sample from one <strong>of</strong> the<br />

Japanese major manufacturing company (total N = 673), with<br />

advantage <strong>of</strong> relatively homogeneous background. Smoking<br />

status was confirmed by in-person structured interviews.<br />

Among our sample, we identified 233 never smokers (who<br />

reported never having smoked a single cigarette in their<br />

lifetime) and 167 heavy smokers (who smoke >20 cigarettes<br />

daily). We found a non-significant difference in allele<br />

frequency (?2 = 3.54, df = 1, p = 0.06) between never smokers<br />

and heavy smokers. Our data suggests that OPRM1 gene may<br />

confer the risk for nicotine dependence in our sample.


272 ASSOCIATION BETWEEN THE BDNF VAL66MET<br />

POLYMORPHISM AND NICOTINE DEPENDENCE IN<br />

A SAMPLE OF JAPANESE WORKER<br />

K. Yamada* (1), T. Shinkai (1), K. Utsunomiya (2), H. Chen<br />

(3), S. Sakata (1), Y. Fukunaka (1), O. Ohmori (4), J.<br />

Nakamura (1)<br />

1. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu, Japan<br />

2. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu,<br />

Japan; Health Management Department, Shimonoseki<br />

Shipyard & Machinery Works, Mitsubishi Heavy Industries,<br />

Ltd., Shimonoseki, Japan 3. Department <strong>of</strong> Psychiatry, School<br />

<strong>of</strong> Medicine, University <strong>of</strong> Occupational and Environmental<br />

Health, Kitakyushu, Japan; Department <strong>of</strong> Public Health,<br />

Kaohsiung <strong>Medical</strong> University, Kaohsiung , Taiwan<br />

4. Department <strong>of</strong> Psychiatry, School <strong>of</strong> Medicine, University<br />

<strong>of</strong> Occupational and Environmental Health, Kitakyushu,<br />

Japan; Wakato Hospital, Wakamatsu-ku, Kitakyushu, Japan<br />

*hokuto_no_ken_1225@yahoo.co.jp<br />

Brain-derived neurotrophic factor (BDNF) is a nerve growth<br />

factor that plays an important role in the development and<br />

maintenance <strong>of</strong> adult neurons and is important regulator <strong>of</strong><br />

synaptic plasticity in human brain. It has been reported that<br />

BDNF and dopamine (DA) systems interact within a number<br />

<strong>of</strong> neurobiological processes (Guillin et al., 2001). BDNF's<br />

influence on DA responsiveness might be an important factor<br />

in the etiopathology <strong>of</strong> substance abuse including nicotine<br />

dependence, which is implicated in DA reward system<br />

(Guillin et al., 2001, 2007). Interestingly, preclinical data<br />

show that acute administration decreases, whereas chronic<br />

nicotine increases, BDNF mRNA levels in the hippocampus<br />

(Kenny et al., 2000). Further, genome-wide linkage scans<br />

indicate that the region <strong>of</strong> chromosome 11p13, where the<br />

BDNF gene is located, likely harbors susceptibility genes for<br />

nicotine dependence (Li et al., 2003). Moreover, Beuten et al.<br />

(2005) provide evidence <strong>of</strong> an association between allelic<br />

variants <strong>of</strong> BDNF and nicotine dependence in male Caucasian<br />

smokers (Beuten et al., 2005). More recent reports also<br />

showed an association between smoking behavior and the<br />

BDNF Val66Met polymorphism (Lang et al., 2007). Taken<br />

together, these findings suggest that BDNF may play an<br />

important role in the pathophysiology <strong>of</strong> nicotine dependence.<br />

We have recruited the present sample from one <strong>of</strong> the<br />

Japanese major manufacturing companies (total N = 673),<br />

with advantage <strong>of</strong> relatively homogeneous background.<br />

Among our sample, we identified 233 never smokers (who<br />

reported never having smoked a single cigarette in their<br />

lifetime) and 167 heavy smokers (who smoke ?20 cigarettes<br />

daily). We could not find an association between smoking and<br />

the BDNF Val66Met polymorphism in our sample. Our data<br />

suggests that BDNF gene may not confer the risk for nicotine<br />

dependence in Japanese workers.<br />

273 PRINCIPAL COMPONENT AND BAYESIAN<br />

NETWORK ANALYSES OF GENOME WIDE<br />

ASSOCIATION RESULTS FROM SUBSTANCE<br />

DEPENDENCE AND SMOKING CESSATION<br />

SUCCESS.<br />

T. Drgon* (1), C. Johnson (1), J. Rose (2), G. Uhl (1)<br />

1. NIDA, NIH 2. Duke University<br />

*tdrgon@intra.nida.nih.gov<br />

Complex traits with polygenic genetic architecture provide<br />

challenges for genome wide association. When expected effect<br />

sizes for individual polymorphisms are modest, extremely<br />

large sample sizes may be required to achieve “genome wide<br />

significance” using eg 2 tests with corrections for multiple<br />

comparisons. Principal Component Analysis (PCA) reduces<br />

multidimensional datasets to simpler covariance or distance<br />

matrices and is a priori agnostic to structure in the data. PCA<br />

components readily distinguish individuals <strong>of</strong> different<br />

racial/ethnic backgrounds, as we have demonstrated in<br />

comparison <strong>of</strong> 500k GWA data from f European American,<br />

African American and Asian samples. However, other<br />

principal components clearly distinguish samples <strong>of</strong> substance<br />

dependent individuals from controls, and successful vs.<br />

unsuccessful quitters in smoking cessation trials. We have<br />

developed permutation and Monte Carlo tests that allow us to<br />

assign p values for the significance <strong>of</strong> these differences.<br />

Bayesian networks (BN) provide probabilistic graphical<br />

models that help visualize a set <strong>of</strong> variables and their<br />

conditional independencies. Bayesian networks can thus help<br />

us to classify individuals based on genotypes and other<br />

relevant phenotypic variables. We have constructed Bayesian<br />

network models for predicting success in smoking. Each <strong>of</strong><br />

these models provides support for the idea that additive<br />

genetic influences can describe substantial proportions <strong>of</strong> the<br />

genetic contributions to these phenotypes. Each <strong>of</strong> these<br />

models is internally consistent and provides a basis for<br />

development <strong>of</strong> clinically relevant probabilistic decision<br />

making.


274 TYROSINE HYDROXYLASE GENE AND<br />

METHAMPHETAMINE USE DISORDERS<br />

H. Ujike* (1), Y. Okahisa (1), M. Takaki (1), M. Kodama (1),<br />

T. Inada (2), N. Uchimura (2), M. Yamada (2), M. Iyo (2), I.<br />

Sora (2), N. Iwata (10), N. Ozaki (2)<br />

1. Department <strong>of</strong> Neuropsychiatry, Okayama University<br />

Graduate School <strong>of</strong> Medicine, Dentistry and Pharmaceutical<br />

Sciences 2. JGIDA<br />

* hujike@cc.okayama-u.ac.jp<br />

Tyrosine hydroxylase (TH) is a rate-limiting enzyme <strong>of</strong><br />

dopamine synthesis and may be involved in several<br />

dopamine-related neuropsychiatric disorders including<br />

substance dependence and schizophrenia. Previous reports<br />

showed significant genetic association between the TH gene<br />

and nicotine dependence and alcoholism. We examined two<br />

polymorphisms <strong>of</strong> the TH gene, HUMTH01-VNTR and<br />

Val81Met (rs6356), in 221 patients with methamphetamine<br />

dependence and/or psychosis and 279 age- and sex-matched<br />

healthy controls. There was no significant difference in allelic<br />

and genotypic distribution <strong>of</strong> HUMTH01-VNTR and<br />

Val81Met between methamphetamine dependence/psychosis<br />

and control. Several clinical phenotypes <strong>of</strong> methamphetamine<br />

dependence/psychosis, such as age at first abuse, duration <strong>of</strong><br />

latency from the first abuse to onset <strong>of</strong> psychosis, prognosis <strong>of</strong><br />

psychosis after therapy, and complication <strong>of</strong> spontaneous<br />

relapse <strong>of</strong> psychotic state were examined, however, there was<br />

no association between any polymorphism and the clinical<br />

phenotype, either. HUMTH01-VNTR is a tetra-nucleotide<br />

repeat located in the intron 1, which motif indicates binding to<br />

transcription factors. K1 allele <strong>of</strong> HUMTH01-VNTR was<br />

shown to be associated with heart rate and HVA concentration<br />

in CSF and serum. Val81Met polymorphism changes TH<br />

structure. Accordingly, the two polymorphisms examined in<br />

the present study may be physiologically functional.<br />

Therefore, the present findings may indicate that individual<br />

variation <strong>of</strong> TH activity may not be involved in susceptibility<br />

to methamphetamine use disorders and those clinical severity<br />

or prognosis <strong>of</strong> psychosis.<br />

275 LINKAGE SCAN OF NICOTINE DEPENDENCE IN<br />

THE UCSF FAMILY ALCOHOLISM STUDY<br />

I. Gizer* (1), C. Ehlers (2), C. Vietan (3), K. Seaton-Smith<br />

(4), H. Feiler (5), J. Lee (6), S. Segall (7), D. Gilder (1), K.<br />

Wilhelmsen (1)<br />

1. University <strong>of</strong> North Carolina 2. Scripps Research Institute<br />

3. California Pacific <strong>Medical</strong> Center 4. Arizona College <strong>of</strong><br />

Osteopathic Medicine 5. University <strong>of</strong> California at San<br />

Francisco 6. University <strong>of</strong> Illinois at Urbana-Champaign<br />

7. University <strong>of</strong> North Carolina at Chapel Hill<br />

* igizer@unc.edu<br />

Ample data suggest nicotine dependence represents a heritable<br />

condition, and several research groups have performed linkage<br />

analysis to identify genomic regions associated with this<br />

disorder. In the present study, a genome-wide linkage scan for<br />

nicotine dependence was conducted in a community sample<br />

that included 950 probands and 1204 first-degree relatives<br />

recruited as part <strong>of</strong> the UCSF Family Alcoholism Study. The<br />

Semi-Structured Assessment for the Genetics <strong>of</strong> Alcoholism<br />

(SSAGA) was used to derive DSM-IV nicotine dependence<br />

diagnoses, and linkage analysis was conducted using variance<br />

components analysis. The strongest linkage signal was<br />

observed on chromosome 2q31.1 at 184 centimorgans nearest<br />

to marker D2S2188 (LOD score = 3.52, nominal p-value =<br />

.00005). Additionally, suggestive linkage peaks were<br />

identified on chromosomes 2q13, 4p15.33-31, 11q25, and<br />

12p11.23-21. Notably, analyses <strong>of</strong> co-occurring alcohol<br />

dependence suggested that the 2q31.1 locus is not linked to<br />

alcohol dependence in the present population, and thus may<br />

influence nicotine dependence specifically. In addition,<br />

linkage analysis <strong>of</strong> the 14 individual nicotine dependence<br />

symptoms assessed by the SSAGA to the 2q31.1 locus showed<br />

that a broad range <strong>of</strong> nicotine dependence symptoms provided<br />

modest contributions to the observed linkage signal suggesting<br />

that this locus confers a general risk towards nicotine<br />

dependence rather than influencing a specific facet <strong>of</strong> the<br />

disorder. In sum, these data provide compelling evidence that<br />

chromosome 2q31.1 harbors a susceptibility locus for nicotine<br />

dependence.


276 ASSOCIATION ANALYSIS BETWEEN<br />

POLYMORPHISMS IN MONOAMINE OXIDASE A<br />

(MAOA) AND DOPAMINE RECETOR D2 (DRD2)<br />

GENES AND COCAINE DEPENDENCE<br />

A. Negrão* (1), A. Pereira (2), R. Laranjeira (3), J. Krieger<br />

(2), H. Vallada (1)<br />

1. Program <strong>of</strong> Genetics and Pharmacogenetics, Department<br />

and Institute <strong>of</strong> Psychiatry, University <strong>of</strong> São Paulo <strong>Medical</strong><br />

School, São Paulo, Brazil 2. Laboratory <strong>of</strong> Genetic and<br />

Molecular Cardiology/LIM-13, Heart Institute (InCor),<br />

University <strong>of</strong> São Paulo <strong>Medical</strong> School, São Paulo, Brazil 3.<br />

Alcohol and Drug Research Unit, Psychiatry Department,<br />

Federal University <strong>of</strong> São Paulo, São Paulo, Brazil<br />

*abnegrao@usp.br<br />

Dopaminergic brain systems have a major role in drug reward,<br />

thus making genes involved in these circuits candidates for<br />

susceptibility to substance use disorders. An association<br />

between cocaine dependence and a dopamine transporter gene<br />

in a sample <strong>of</strong> cocaine users in Brazil has previously been<br />

published. In the present study we investigate other candidate<br />

genes involved in dopaminergic neurotransmission in that<br />

same sample: monoamine oxidase A (MAOA) and dopamine<br />

D2 receptor (DRD2). MAOA is an enzyme involved in the<br />

oxidative metabolism <strong>of</strong> dopamine. Antisocial behavior<br />

associated with alcohol consumption has been related to a<br />

variation in the MAOA gene. In addition, epidemiological<br />

evidence demonstrated a high association between antisocial<br />

behavior and substance dependence. Therefore, MAOA gene<br />

polymorphism could be linked to substance dependence but no<br />

study has documented its association to cocaine dependence to<br />

date. Polymorphisms at the DRD2 have been reported in many<br />

alcohol-dependent studies. 681 patients who met ICD10<br />

diagnosis <strong>of</strong> cocaine dependence and 756 controls were<br />

genotyped for three SNPs in the MAOA gene (rs 6323, rs<br />

2235185, rs 1137070), one SNP (rs1801028) and one<br />

insertion/deletion polymorphism (rs 1799732) in the DRD2<br />

gene. We observed no significant differences in allele,<br />

genotype, or haplotype frequencies between cases and controls<br />

for any <strong>of</strong> the tested markers. Our study suggests that there is<br />

no association between variants in the MAOA and DRD2<br />

genes and cocaine dependence.<br />

277 GENETIC EPIDEMIOLOGY OF OPIOID<br />

DEPENDENCE IN BULGARIA<br />

A. Todorov* (1), M. Lynskey (1), M. Nikolov (2), O.<br />

Beltcheva (2), R. Kaneva (2), I. Kremensky (2), D.<br />

Krasteva (3), E. Nesheva (3), E. Yankova (4)<br />

1. Washington Univ. Saint Louis 2. Molecular Medicine<br />

Center, S<strong>of</strong>ia, Bulgaria 3. Bulgarian Addictions Institute<br />

4. Initiative for Health Foundation<br />

*todorov@wustl.edu<br />

This ongoing study <strong>of</strong> heroin dependence in Bulgaria is<br />

collecting semi-structured psychiatric assessments and blood<br />

samples from 2,500 injecting drug users 18 years and older<br />

who have used heroin regularly for more than one year. We<br />

present the study methodology and preliminary findings from<br />

the first ~ 1,500 participants. Briefly, 62% <strong>of</strong> participants<br />

agreed to an HIV test with 2.5% <strong>of</strong> samples testing positive<br />

(24 <strong>of</strong> 959) – higher than previous reports for Bulgaria.<br />

Preliminary genotyping is complete for ~1,100 cases and<br />

~1,100 matched controls; for SNPs in OPRM1, DRD2, CRH,<br />

COMT, BDNF, SLC6A4, TPH2 and CLOCK. Controls were<br />

drawn from the population-representative repository at the<br />

National Genetics Laboratory. In OPRM1, we find no<br />

evidence for association with either A118G (rs1799971) or<br />

with rs2075572 (both p > 0.50); but a possible association<br />

with rs932245 in intron 1 (p < 0.01). In other genes, possible<br />

associations include rs3176921, upstream <strong>of</strong> CRH and in<br />

linkage disequilibrium with a block spanning the CRH coding<br />

region (p < 0.01); and with the Val66Met polymorphisms in<br />

BDNF (p = 0.03).Funded by DA018823.


278 ASSOCIATION ANALYSIS BETWEEN<br />

POLYMORPHISMS IN THE<br />

BUTYRYLCHOLINESTERASE (BCHE) GENE AND<br />

COCAINE DEPENDENCE<br />

A. Negrão* (1), A. Pereira (2), R. Laranjeira (3), J. Krieger<br />

(2), H. Vallada (1)<br />

1. Program <strong>of</strong> Genetics and Pharmacogenetics, Department<br />

and Institute <strong>of</strong> Psychiatry, University <strong>of</strong> São Paulo <strong>Medical</strong><br />

School, São Paulo, Brazil 2. Laboratory <strong>of</strong> Genetic and<br />

Molecular Cardiology/LIM-13, Heart Institute (InCor),<br />

University <strong>of</strong> São Paulo <strong>Medical</strong> School, São Paulo, Brazil<br />

3. Alcohol and Drug Research Unit, Psychiatry Department,<br />

Federal University <strong>of</strong> São Paulo, São Paulo, Brazil<br />

* abnegrao@usp.br<br />

Genetic markers in the dopaminergic brain systems are<br />

associated with drug dependence but other biological<br />

pathways can contribute to the susceptibility to this disorder.<br />

Butyrylcholinesterase (BChE) is codified by the BChE gene<br />

and it hydrolyzes cocaine to metabolites with little biologic<br />

activity. Point mutations in the BChE gene have been<br />

described in the general population; some <strong>of</strong> them being<br />

“silent” mutations. BChE has also been tested as a novel<br />

therapeutic agent for cocaine dependence. Brimijoin et al,<br />

2008 demonstrated that a quadruple mutant hydrolase derived<br />

from human butyrylcholinesterase suppressed cocaine toxicity<br />

and abolished drug-primed reinstatement in rats.<br />

Polymorphisms in the BChE gene can lead to different<br />

enzyme pr<strong>of</strong>iles generating individuals with<br />

increased/decreased susceptibility to engage in addictive<br />

behaviors due to varying concentrations <strong>of</strong> cocaine reaching<br />

the reward system in the brain. In this study we hypothesize<br />

that genetic variations in the BChE gene can be risk factos for<br />

cocaine dependence. 681 patients who met ICD10 diagnosis <strong>of</strong><br />

cocaine dependence and 756 controls were genotyped for<br />

three SNPs in the BChE gene (rs1803274, rs4263329,<br />

rs4680662). All markers were in HWE. Genotype differed<br />

significantly for rs4263329 between cases (f(GG)=0,6%) and<br />

controls (f(GG)=1,4%), (p=0.03; ?2=4,48). Genotypes from<br />

the other two markers and allele frequencies did not differ<br />

between cases and controls. No haplotypic effect was<br />

detected. We suggest that the GG genotype in rs 4263329 can<br />

be a protective factor for cocaine dependence. Additional<br />

studies are required to elucidate the role <strong>of</strong> BChE in the<br />

pathophysiology <strong>of</strong> cocaine dependence.<br />

279 CHRM2 GENOMIC REGIONS DIFFERENTIALLY<br />

ASSOCIATE WITH INTERNALIZING AND<br />

EXTERNALIZING COMORBIDITIES IN ALCOHOL<br />

DEPENDENT INDIVIDUALS<br />

D. Kertes* (1), D. Dick (2), C. Prescott (3), K. Kendler (2), B.<br />

Riley (2)<br />

1. University <strong>of</strong> Florida 2. Virginia Commonwealth University<br />

3. University <strong>of</strong> Southern California<br />

*dkertes@gmail.com<br />

Background: Genetic epidemiology studies in psychiatry have<br />

demonstrated that genetic factors contribute to two broad sets<br />

<strong>of</strong> behaviors: externalizing disorders and internalizing<br />

disorders. The muscarinic acetylcholine receptor M2<br />

(CHRM2) is one <strong>of</strong> several genes commonly studied for its<br />

role in alcohol dependence which also show associations with<br />

both internalizing and externalizing comorbidities in this<br />

population. This research aims to identify whether CHRM2<br />

loci differentially associate with comorbid internalizing and<br />

externalizing phenotypes that commonly occur in alcohol<br />

dependent individuals. Method: Lifetime history <strong>of</strong> major<br />

depression (MD), conduct disorder (CD) and antisocial<br />

personality disorder (ASPD) were ascertained via structured<br />

clinical interviews in 552 alcohol dependent individuals. MD,<br />

CD and ASPD symptom counts and diagnoses were associated<br />

with allele frequencies in 20 CHRM2 markers via<br />

regression-based analyses. Results: Results revealed<br />

significant associations for five SNPs with CD symptoms, CD<br />

diagnosis, and ASPD symptoms (.05>p>.001). Three SNPs<br />

showed significant associations with MD symptoms or<br />

diagnosis (.05>p>.01). Examination <strong>of</strong> LD structure revealed<br />

that MD and CD/ASPD phenotypes were associated with<br />

markers in different genomic regions that were consistent with<br />

previous reports <strong>of</strong> association with externalizing and<br />

internalizing phenotypes. Conclusions: Both MD and<br />

CD/ASPD phenotypes in alcohol dependent individuals are<br />

associated with SNP variants in CHRM2 and are distinctly<br />

associated with different genomic regions <strong>of</strong> CHRM2. These<br />

results suggest variants in different genomic regions in<br />

CHRM2 predict liability to different comorbid conditions in<br />

alcohol dependent individuals.


280 INTRONIC SNP FROM DRD2 GENE AND DRD2<br />

HAPLOTYPES, ASSOCIATED WITH ALCOHOLISM<br />

IN RUSSIANS<br />

M. Monakhov* (1), T. Lezheiko (2), S. Borozdina (2), O.<br />

Evdokimova (2), V. Golimbet (2)<br />

1. Engelhardt Institute <strong>of</strong> Molecular Biology, Russian<br />

Academy <strong>of</strong> Sciences, Moscow, Russia 2. Mental Health<br />

Research Center, Russian Academy <strong>of</strong> <strong>Medical</strong> Sciences,<br />

Moscow, Russia<br />

* misha.monahov@gmail.com<br />

Introduction: Dopamine receptor type 2 gene has long been<br />

studied as a candidate for addictive behaviors, including<br />

alcoholism. We present results <strong>of</strong> an association analysis <strong>of</strong><br />

alcoholism sample from Russian population. Five SNPs from<br />

DRD2 as well asVal/Met COMT polymorphism were<br />

investigated. Methods: Case sample consisted <strong>of</strong> 208 alcoholic<br />

patients, diagnosed according to criteria <strong>of</strong> ICD-10.Data on<br />

the amount <strong>of</strong> daily alcohol consumption, typical duration <strong>of</strong><br />

hard drinking periods and remission periods, parental age at<br />

birth, tobacco smoking, life stress events were collected.<br />

Control sample included 366 healthy volunteers with no<br />

history <strong>of</strong> mental disorders. DRD2 polymorphisms rs1800498,<br />

rs1801028, rs6275, rs6277, rs1800497 and COMTVal158Met<br />

were analyzed. Genotyping was performed with 5’-nuclease<br />

assay. Results and discussion: We have found an association<br />

between alcoholism affection status and intronic SNP<br />

rs1800498from DRD2 gene (chisq=5.53 df = 1 p=0.019) as<br />

well as with haplotypes containingthis polymorphism (chisq=<br />

17.03 df = 6 p=0.009). rs1800498 was also associated with<br />

duration <strong>of</strong>hard drinking periods (F(2, 173)=3.37, p=0.037).<br />

Statistically significant result was found fora two-SNP<br />

haplotype (rs1801028-rs1800497) and alcoholism in female<br />

subsample (chisq= 11.7 df = 3 p=0.0085).These data, obtained<br />

by the study <strong>of</strong> novel sample, add up to the pool <strong>of</strong> findings <strong>of</strong><br />

DRD2gene role in alcohol dependence. The same SNP is<br />

associated with schizophreniaand altruistic behavior according<br />

to our previous research that may point to its functional<br />

effect.<br />

281 NETWORK ANALYSIS OF CANDIDATE GENES<br />

OF ALCOHOL CONSUMPTION<br />

A. Lourdusamy* (1), G. Schumann (1)<br />

1. Institute <strong>of</strong> Psychiatry, King's College London<br />

* anbarasu.lourdusamy@kcl.ac.uk<br />

Microarray analyses <strong>of</strong> brain gene expression in several rodent<br />

models for excessive alcohol consumption have identified<br />

candidate genes that underlie the genetic predisposition to<br />

drink alcohol. Although the mechanism <strong>of</strong> these candidate<br />

genes is being explored in human, little is known about the<br />

molecular relationship between them. Here, we set out to<br />

systematically analyze the complex effects <strong>of</strong> interrelated<br />

genes and provide a framework for revealing their<br />

relationships in association with alcohol consumption. The<br />

alcohol network is composed <strong>of</strong> 29 human homologs <strong>of</strong><br />

proteins known to be involved in excessive alcohol<br />

consumption in mice, and 183 additional human proteins that<br />

interact with these human homolgos. Overall, the network<br />

consists <strong>of</strong> 416 binary interactions among 212 unique proteins.<br />

We observed that human protein-protein interaction networks<br />

associated with alcohol consumption have a power-law<br />

connectivity distribution as many other biological networks<br />

have. These proteins are strongly interconnected and in turn<br />

form 10 sub-networks which comprise over 50% <strong>of</strong> candidate<br />

genes. Among candidate target genes associated with alcohol<br />

consumption, the genes including, PTPN11, GNB1, PSMB7,<br />

B2M, THRA, and GRIA1 have high connectivity (hubs). The<br />

hub nodes and sub-networks are consistent with the prior<br />

knowledge about alcohol consumption. In particular, PTPN11<br />

appears to play a very important role in the alcohol network<br />

through frequent interactions with key proteins in cellular<br />

signaling. This network-based approach represents an<br />

alternative method for analyzing the complex effects <strong>of</strong><br />

alcohol candidate genes.


282 DOPAMINE D4 RECEPTOR GENE: POSSIBLE<br />

INFLUENCE ON ATTENTION DEFICIT<br />

HYPERACTIVITY DISORDER IN BRAZILIAN<br />

POPULATION<br />

G.C. Akutagava-Martins (1), G. Ferraz (1), J.P. Genro (1), G.<br />

Polanczyk (2), C. Zeni (3), M. Schmitz (3), LAP Rohde (3),<br />

MH Hutz (1), T. Roman* (1)<br />

1. Department <strong>of</strong> Genetics, Federal University <strong>of</strong> Rio Grande<br />

do Sul, Brazil 2.Department <strong>of</strong> Psychology and Neuroscience,<br />

Duke University, USA 3. Department <strong>of</strong> Psychiatry and Legal<br />

Medicine, Federal University <strong>of</strong> Rio Grande do Sul, Brazil.<br />

*troman29@yahoo.com.br<br />

The dopamine D4 receptor gene (DRD4) is one <strong>of</strong> the most<br />

investigated genes in Attention deficit/hyperactivity disorder<br />

(ADHD) and has already been accepted as a susceptibility<br />

gene. The aim <strong>of</strong> present study was to search for a possible<br />

association between ADHD and the 120 bp tandem<br />

duplication <strong>of</strong> promoter region, the SNPs -616C>G (rs747302)<br />

and -521C>T (rs1800955) and the 48 bp VNTR <strong>of</strong> DRD4 gene<br />

in a Brazilian sample composed by 488 ADHD children and<br />

adolescents, diagnosed according to DSM-IV criteria, and<br />

their biological parents. The 120 bp tandem duplication and<br />

the 48 bp VNTR were genotyped by PCR, while the SNPs -<br />

616C>G and -521C>T were genotyped by PCR-RFLP. The<br />

possibility <strong>of</strong> linkage disequilibrium among the studied<br />

polymorphisms was tested using MLocus s<strong>of</strong>tware. The<br />

hypothesis <strong>of</strong> association was verified using FBAT s<strong>of</strong>tware.<br />

There was no evidence <strong>of</strong> association between each<br />

polymorphism and ADHD for the whole sample (P values<br />

ranging from 0.200 to 0.974). However, when the analysis was<br />

restricted to families where the proband presented the<br />

combined type <strong>of</strong> the disorder, the VNTR 2R allele was<br />

preferentially not transmited (P=0.033). Haplotype analyses<br />

did not show any association (P values ranging from 0.063 to<br />

0.977). The supposed protective effect <strong>of</strong> 2R allele observed<br />

herein is intriguing. This result must be interpreted cautiously<br />

and investigated through other approaches, as dimensional<br />

analyses. It is also possible that the findings, divergent from<br />

the literature, are due to DRD4 structural complexity, which<br />

should be understood to better characterize its association with<br />

ADHD.


AUTHOR INDEX


Aarts, Esther 2<br />

Abeles, Michael 242<br />

Åberg, Elin O9.1<br />

Aberg, Karolina O2.6<br />

Abou Jamra, Rami O12.2, 149<br />

Abraham, Richard O17.1<br />

Acheson, Ruth 6<br />

Acierno, Ron 110<br />

Adati, Naoki O1.4<br />

Ades, AE 93<br />

Adkins, Daniel E. O2.6, 23<br />

Adolfsson, Rolf O7.3, 52, 122, 136<br />

Affonseca-Bressan, 170<br />

Rodrigo<br />

Agan, Noelle 106<br />

Agartz, Ingrid 53, 64<br />

Aggen, Steven S16.7<br />

Agrawal, Arpana S16.2, S16.3,<br />

S16.4, S16.6,<br />

S16.7<br />

Ågren, Hans O10.1, O10.5<br />

Ahmankhaniha,<br />

Hamidreza<br />

97<br />

Ahn, Yong-Min 47<br />

Aitchison, Katherine J S1.1, O1.3, O2.5,<br />

O7.4, O11.1,<br />

O15.2, 143, 164,<br />

190<br />

Akelai, A. O12.4<br />

Akiskal, Hagop S14.2, O7.6<br />

Akiyama, Tsuyoshi 177<br />

Akula, Nirmala ECI 19, S14.4,<br />

O2.2, O5.5, 127,<br />

163<br />

Alaerts, Maaike 52, 122<br />

Alatrouny, Mohamed 243<br />

Alblas, Margrieta 119<br />

Albrecht, E 114<br />

Albu, Crenguta 34<br />

Albu, Dinu 34<br />

Albus, M 245<br />

Al-Chalabi, Ammar O17.1<br />

Alda, Martin O9.4, 162<br />

Aleksic, Branko 176, 209, 220,<br />

221, 222<br />

Alexander, Michael O9.2, 245<br />

Alho, Hannu 189<br />

Ali, Jeffrey 137<br />

Aliev, Fazil S16.7, O16.2, 117<br />

Alizadeh, Behrooz 81<br />

Alkelai, A. 233<br />

Allardyce, Judith O11.2<br />

Allgulander, Christer 30<br />

Alliey, Ney 180<br />

Allin, Matthew 236<br />

Almasy, Laura S12.4, S12.5,<br />

S16.6, S16.7, ECI<br />

13, ECI 18, 76<br />

Alonso, Yolanda 248<br />

Alphs, Larry 199<br />

Als, Thomas D. O1.1<br />

Alth<strong>of</strong>f, Robert S8.1<br />

Amankwa, Susan 236<br />

Amann-Zalcenstein,<br />

D.<br />

O12.4<br />

Ambler, Antony O9.7<br />

Amdur, Richard ECI 3, 179, 223<br />

Amin, Farooq 115<br />

Amir, Nurmiati 57<br />

Amstadter, Ananda 110<br />

An, Seon-Sook O3.5<br />

Anand, Sonia O10.2<br />

Ancoli-Israel, Sonia 56<br />

Andersen, Johan O8.1, 39<br />

Andersson, Gerhard O3.1<br />

Andreassen, Ole 53, 64<br />

Andresen, Michael S10.2<br />

Andriuskeviciute, 109<br />

Irena<br />

Angelo, Gary W. 71<br />

Anjorin, Adebayo ECI 17, O2.1, 59,<br />

156<br />

Anney, Richard P4.6, 16, 21, 36,<br />

48, 258<br />

Anton, Eva S. 25<br />

Arcos-Burgos,<br />

Mauricio<br />

O11.6<br />

Ardau, Raffaella 162<br />

Arias Vasquez,<br />

Alejandro<br />

O8.3<br />

Arima, Kunimasa 66<br />

Arinami, Tadao 215<br />

Aristotle, Aristotle 191<br />

Arnold, Paul S10.1, S10.4,<br />

S10.5<br />

Arranz, Maria 83<br />

Aryee, Martin S2.2<br />

Aschauer, Harald N. 152<br />

Asherson, Philip 3, 9, 11, 24, 59<br />

Athanasiu, Lavinia 53<br />

Athie, Maria Carolina 75<br />

Aukes, Maartje 81<br />

Aulchenko, Yuri 147<br />

Autism Genome 36<br />

Project, The<br />

Austin, Jehannine P2.2, 43, 107<br />

Ayub, Muhammad ECI 15, 173<br />

Azzam, Amin S10.6<br />

Babatz, Timothy D. ECI 12<br />

Bachner-Melman,<br />

Rachel<br />

91<br />

Backlund, Lena O10.1, O10.5<br />

Badner, Judith S2.3, S2.5, 98,<br />

157, 180<br />

Bailey, Julia 65<br />

Bailey, Mark 45<br />

Baji, Ildikó 50<br />

Baker, Tim ECI 7<br />

Bakken, Trygve 64<br />

Bakker, Steven C. 227, 234<br />

Balachandar,<br />

218<br />

Vellingiri<br />

Balazic, Joze 49<br />

Ballinger, Dennis O16.4<br />

Banaschewski, Tobias 11<br />

Bannasch, Danika 22<br />

Bannen, Ryan 135<br />

Bansal, Vikas S13.5<br />

Barenboim, Maxim 252<br />

Barichello, Tatiana 75<br />

Barnes, Michael O1.3, O7.4, O11.1<br />

Barr, Cathy O14.3, 50, 95,<br />

171, 202<br />

Barrett, Thomas 157<br />

Barry, Edwina 4, 6, 10<br />

Barth, Alexander 120<br />

Bartling, Jürgen 174<br />

Baruch, Kuti 126<br />

Basmanav, F. Buket O8.2, O9.2<br />

Bass, Nick J ECI 17, O2.1,<br />

O17.1<br />

Batra, Anil 197<br />

Bayes, Monica 7, 8, 9<br />

Beardsley, Patrick M. O2.6<br />

Becker, Jessica 174<br />

Beckmann, J.S. O12.4<br />

Beevers, Christopher<br />

G.<br />

58<br />

Behm, Frederique 183<br />

Beitchman, Joseph 172<br />

Bell, Maureen 208<br />

Bellgrove, Mark 12, 84<br />

Belmonte, Pamela S14.1, S14.3, 154,<br />

157, 263<br />

Belmonte-de-Abreu,<br />

Paulo S.<br />

14<br />

Beltcheva, Olga 277<br />

Benák, István 50<br />

Ben-Asher, E. O12.4<br />

Benson, Gary 135<br />

Bergen, Andrew S13.2, S13.4,<br />

S13.5, O16.4<br />

Bergen, Sarah ECI 3, 223, 261<br />

Bergstrom, Jan O3.1<br />

Berlo, Jacques 140<br />

Bernard, Lerer 206<br />

Berner, Michael 197<br />

Berrettini, Wade S13.1, S13.2,<br />

S13.5, 138, 157<br />

Berry, Erin M. S15.5<br />

Bertolucci, Paulo 73<br />

Bertram, David S6.3<br />

Bertuzzi, Guilherme P. 14<br />

Berwaerts, Joris 199<br />

Besnyo, Márta 50<br />

Bespalova, Irina N. 71<br />

Bettecken, Thomas S1.4<br />

Beveridge, Natalie S7.2<br />

Bevilacqua, Laura O1.7<br />

Bevova, Marianna S4.1, S4.5<br />

Biechele, Travis S15.5<br />

Bielsa, Anna 8<br />

Bierut, Laura S16.1, S16.2,<br />

S16.3, S16.4,<br />

S16.5, S16.6,<br />

S16.7, ECI 7, 138<br />

Bigdeli, Tim ECI 3<br />

Bilder, Robert 242<br />

Bilkei-Gorzo, Andras O12.2, 120<br />

Binder, Elisabeth B. S1.4, O2.5, O3.2,<br />

O15.1, 30<br />

Binder, Elke O2.5, O3.2<br />

Bipolar Genome Study S9.4, S14.2,<br />

(BiGS)<br />

S14.3, O5.5, O7.5,<br />

O7.6, O11.4, ECI<br />

14, ECI 19, 127,<br />

166, 263, 283<br />

Birn, F. O6.2<br />

Bishop, Jeffrey S11.3<br />

Bishop, John S13.5<br />

Bitter, István 234<br />

Bizarro, Lisianne 24<br />

Björk, Karl 92<br />

Black, Donald W. 115


Blackwood, Douglas O1.1, O5.1, 208,<br />

216, 237<br />

Blangero, John S12.3, S12.4,<br />

S12.5, ECI 13, 76,<br />

240<br />

Bleotu, Coralia 149<br />

Bliziotes, Michael S11.4<br />

Bloch, Paul J. 41<br />

Bloss, Cinnamon S13.1, S13.2,<br />

S13.5, 64, 157,<br />

169, 259<br />

Blum, Kenneth 182<br />

Bochdanovits, Zoltan S4.1, S4.4, S4.5<br />

Bockholt, H.Jeremy O6.2<br />

Bogue, Molly 23<br />

Boks, Marco O11.2<br />

Bollen, Joseph 140<br />

Bonaguro, Russell J. S3.5<br />

Bonde, Jens Peter 39<br />

Bondy, Brigitta S1.4<br />

Bonsall, Robert 79<br />

Boomsma, Dorret S4.5, S8.1, 114,<br />

147<br />

Boretti-Hummer,<br />

Andrea<br />

7, 9<br />

Borgå Johansen, 18<br />

Espen<br />

Børglum, Anders D. O1.1, O8.1, 27,<br />

39, 212, 214<br />

Borozdina, Svetlana 280<br />

Bosch, Rosa 7, 8, 9<br />

Bousman, Chad S6.2, 213<br />

Boyd, Simeon 22<br />

Bozdagi, Ozlem O13.1<br />

Bradley, Bekh 79<br />

Bradshaw, John 6<br />

Bradwejn, Jacques 178<br />

Braff, David 207, 249<br />

Brain, Ursula 43<br />

Bramon, Elvira 83, 236<br />

Brandt, John O2.4<br />

Braverman, Eric 182<br />

Bray, Nick 72, 253<br />

Brayne, Carol O17.1<br />

Breen, Gerome S7.1, O1.3, O7.4,<br />

O11.1, O11.5,<br />

O17.4, 143, 164,<br />

170<br />

Bremner, Rod 95<br />

Brentani, Helena ECI 8<br />

Breslau, Naomi S16.5, ECI 7<br />

Breuer, Rene S14.3, S14.4,<br />

O5.5, O8.2, O9.2,<br />

119, 160<br />

Briones Velasco,<br />

Magdalena<br />

ECI 5<br />

Brizendine, Edward ECI 14<br />

Brockschmidt, Felix 174<br />

Brodkin, Edward S. 41<br />

Brookes, Keeley 4, 10<br />

Brotman, Melissa 163<br />

Brouwer, Rachel M. 85<br />

Brown, Sarah 208<br />

Bruce, Heather 135<br />

Bruder, Jennifer 174<br />

Brueckl, Tanja O15.1, 100, 101<br />

Bruining, Hilgo O13.3, O13.4<br />

Brunner, Han O8.3<br />

Bu, Youngmin 70, 134<br />

Buccola, Nancy G. 115<br />

Buch, Helle 39<br />

Bucholz, Kathleen S16.1, S16.3<br />

Buitelaar, Jan O4.2, 2, 3, 7, 9, 11<br />

Bukszar, Jozsef O2.6, 42<br />

Bundo, Miki O1.4, 94<br />

Bunge, Silvia A. ECI 4<br />

Bunney, William E. O7.3<br />

Buonocore, Federica 72, 253<br />

Burdick, Katherine S9.2, O8.6, 167<br />

Burmeister, Margit S6.4, 128, 129<br />

Burton, Cynthia 242<br />

Busatto, Geraldo 104, 113<br />

Butler, Amy W S1.1, O1.3, O7.4,<br />

O11.1, 143<br />

Buttenschøn, Henriette O1.1, O 8.1, 39<br />

N.<br />

Buttigieg, Neil O4.4, 19<br />

Buxbaum, Joseph O12.5, O13.1<br />

Buzas,, Beata 167<br />

Byerley, William F. 115, 157<br />

Byrnes, Andrea 25<br />

Caballero, Alejandro 108<br />

Cadenhead, Kristin 207, 249<br />

Cadigan, Jennifer 283<br />

Caesar, Sian O10.3<br />

Cagliani, R. O12.4<br />

Cahn, Wiepke O11.2, 227, 260<br />

265<br />

Cai, Xueya ECI 14<br />

Cairns, Murray S7.2<br />

Calarge, Chadi S11.4<br />

Calati, Raffaella 140<br />

Calhoun, Vince O6.2<br />

Caligiuri, Michael S6.3<br />

Calkins, Monica 249<br />

Camarena, Beatriz 108<br />

Cannon, Ronald O5.2<br />

Capogna, Michael O5.4<br />

Cappi, Carolina ECI 8<br />

Cardenas-Godinez,<br />

Marcela<br />

ECI 5<br />

Cardone, Laura 208<br />

Carless, Melanie S12.4, S12.5, ECI<br />

13, 76<br />

Carmine Belin, 142<br />

Andrea<br />

Carr, Kenneth O1.6<br />

Carroll, Adam S7.2<br />

Carroll, Liam O12.1, 247<br />

Casas, Miguel 7, 8, 9<br />

Caspi, Avshalom O9.7<br />

Cate-Carter, Tasha 171<br />

Cath, Danielle S10.1, S10.2,<br />

S10.3, S10.4,<br />

S10.7<br />

Cavallini, Maria S10.4<br />

Cristina<br />

Celestrin, Kevin O1.6<br />

Cervilla, Jorge 28<br />

Ceulemans, Shana 52, 122<br />

Chambers,<br />

84<br />

Christopher<br />

Chana, Gursharan S6.2, 213<br />

Chandler, Sharon S6.2, 213<br />

Chang, Byung-Eun 70<br />

Chang, Chien Ching 87, 235<br />

Chapagain, Ram Hari 240<br />

Charlesworth, Jac C S12.4<br />

Chavira, Denise S10.6<br />

Chen, Chao S2.3, S2.5, O8.4,<br />

98<br />

Chen, David ECI 19<br />

Chen, Guo-Lin 68<br />

Chen, Haiming 128, 129<br />

Chen, Hsin-I S6.4, 268, 269,<br />

272<br />

Chen, Jen-Yeu O8.4<br />

Chen, LiShiun S16.4, S16.5, ECI<br />

7<br />

Chen, Peining O2.4<br />

Chen, Suephy 67<br />

Chen, Xiangning O3.5, 42, 179<br />

Cheng, Lijun S2.3, S2.5, 98<br />

Cheng, Min-Chih O8.4<br />

Chiesa, Alberto 185<br />

Chillotti, Caterina 162<br />

Cho, Ah Rang 254<br />

Choi, Jung-Eun 151, 195, 196, 200<br />

Choi, Kyung Sook 46<br />

Chorlian, David ECI 18<br />

Chow, Tze Jen ECI 1, 217, 226<br />

Chowdari, Kodavali 243<br />

Christensen, Jane<br />

Hvarregaard<br />

212, 214<br />

Christensen, Kenneth O1.1<br />

V.<br />

Christian, Susan L. ECI 12<br />

Chung, Hedy 199<br />

Chung, Joo Ho 254<br />

Chung, Myeon-Woo 133<br />

Cichon, Sven S14.3, S14.4,<br />

O5.5, O8.2, O9.2,<br />

O11.6, 119, 148,<br />

149, 160, 197<br />

Cloninger, C. Robert S16.3, 115<br />

Cochrane, Lynne 125<br />

COGA S16.1, S16.3,<br />

S16.6, S16.7, ECI<br />

18, 90<br />

COGEND S16.1, S16.3,<br />

S16.7<br />

Coghill, David 4<br />

Cohen, Andrew 62<br />

Cohen, D 245<br />

Cohen, Nadine 199<br />

Cohen-Woods, Sarah S1.1, O1.3, O7.4,<br />

O11.1, 143, 152,<br />

285<br />

Cojocaru, Mariana 34<br />

Cole, James 285<br />

Coleman, Michael 80<br />

Coleman, Vegas ECI 14<br />

Collier, David S5.2, S7.1, S15.1,<br />

O11.5, O15.2, 83,<br />

153, 236<br />

Colombo, Elisa 188<br />

Colombo, Roberto 188<br />

Condie, Alison O5.2<br />

Congiu, Donatella 162<br />

Consortium on<br />

Lithium Genetics<br />

(ConLiGen)<br />

O9.4<br />

Constante, Miguel 83<br />

Contini, Verônica 14<br />

Contreras, Javier 141<br />

Cook, Ed S10.5, 61


Cools, Roshan 2<br />

Cooper, Jon 72<br />

Corbett, Blythe A. ECI 4<br />

Cordeiro, Quirino ECI 8<br />

Corina, Benjet ECI 5<br />

Cormand, Bru O9.6, 3, 5, 7, 8, 9<br />

Cormican, Paul 125<br />

Correia-Pinto, Julien 75<br />

Cortes, M Jose O6.4<br />

Corvin, Aiden S3.1, S9.1, O12.1,<br />

O15.3, 48, 125<br />

Corydon, Thomas J. 214<br />

Coryell, William S11.4, 157<br />

Cosentino, C. 233<br />

Cox, Nancy S10.3<br />

Cozzi, P. O12.4, 233<br />

Craddock, Nicholas S14.5, O1.3, O5.3,<br />

O7.4, O10.3,<br />

O12.1, O12.5,<br />

102, 143, 160,<br />

210, 285<br />

Craig, David S2.3, 157<br />

Craig, Ian W. S1.1, O1.3, O2.5,<br />

O7.4, O11.1,<br />

O15.2, 143, 152,<br />

164, 190<br />

Craig, Thomas 211<br />

Crane, Jacquelyn S10.3<br />

Crepel, An O13.2<br />

Crespo, José M. 40<br />

Crowley, James O12.3, 23, 25, 229<br />

Cubells, Joseph P4.7, 79<br />

Cuccaro, Michael O3.3, O17.2<br />

Cummins, Tarrant 84<br />

Curran, Joanne S12.4, S12.5, ECI<br />

13, 76<br />

Curtis, David O2.1, ECI 17<br />

Czerski, Piotr 190<br />

Czobor, Pál 234<br />

Dackor, Jennifer 25<br />

Dagdan, Elif 168<br />

Dahl, Hans Atli 27<br />

Dáibhis, Aoife 6<br />

Dal Fiume, C. O12.4<br />

Dale, Anders 64<br />

Daly, Mark 262<br />

Dapretto, Mirella 88<br />

Daróczi, Gabriella 50<br />

DasBanerjee, Tania S8.2, S8.3, 18<br />

Davey Smith, G 32, 93, 114<br />

David, Anthony O15.2<br />

Davies, Charlotte O4.4, 19<br />

Davies, Gail 74<br />

Davis, Jeremy S6.3<br />

Dazzan, Paola O15.2, 211<br />

de Geus, Eco S4.5, S8.1, 147<br />

de Haan, Marcel S10.7<br />

de Jong, Pieter 26<br />

de Jonge, Maretha O13.4<br />

de Kloet, E.R. 60<br />

de La Bastide, Melissa 208<br />

de la Marche, Wouter O13.2, 31, 33<br />

de la Portilla, Sonia O6.4<br />

de Leeuw, Charles N. S3.5<br />

de Luca, Vincenzo 191, 244<br />

de Quervain,<br />

Dominique<br />

255, 256<br />

de Rijk, Peter 136<br />

de Ronchi, Diana 44, 140, 185, 186,<br />

188<br />

de Saloma, Andiara 170<br />

de Sonneville, Leo O13.4<br />

de Zutter, Sonia 52<br />

Deary, Ian O5.2, 74, 208<br />

Dechairo, Bryan 15, 17<br />

Deckert, Juergen 30<br />

Dedman, Alexandra O2.1, 156<br />

Degenhardt, Franziska 119, 160<br />

Del Zompo, Maria 162<br />

Deleuran, Thomas O1.1<br />

Del-Favero, Jurgen 52, 122, 136, 237<br />

DeLisi, Lynn E. 135<br />

Deloukas, Panos O17.1<br />

Delucchi, Kevin S10.2, S10.7<br />

Demirkan, Ayse 147<br />

Demontis, Ditte O8.1, 212<br />

Dempster, Emma O9.5, O14.3,<br />

O14.5, 202, 236<br />

Deng, Hong-Wen 204<br />

Deng, Xiaoyan 184<br />

Dennis, J. Michael 115<br />

Denys, Damiaan S10.2, S10.4,<br />

O11.6<br />

DeRijk, R.H. 60<br />

Derks, Eske O11.2, 260, 265<br />

DeRosse, Pamela S9.2, O8.6, 167<br />

Derringer, Jaime S16.2, S16.4,<br />

S16.6, ECI 10<br />

Desrivieres, Sylvane O14.1<br />

Detera-Wadleigh,<br />

Sevilla<br />

S14.4, O2.2<br />

DeVane, Lindsay S11.2<br />

Devlin, Angela 43<br />

Devlin, Bernie 118, 243<br />

Devriendt, Koen O13.2<br />

Di Forti, Marta O15.2, 105<br />

Diaconu, Carmen C. 149<br />

Diaz Lacava, Amalia 120<br />

Dick, Danielle S16.1, S16.3,<br />

S16.4, S16.6,<br />

S16.7, O14.1,<br />

O16.2, O16.3,<br />

117, 279<br />

Dickinson, Dwight O6.3<br />

Dike, Craig S6.3<br />

Dikeos, D 245<br />

Diniz, Breno 73<br />

Diniz, Mateus 170<br />

Ditzen, Claudia S6.1<br />

Djurovic, Srdjan 53, 64<br />

Doan, Bridget S10.5<br />

Dobie, Dorcas 249<br />

Dobyns, William B. ECI 12<br />

Docherty, Sophia O9.5, 77<br />

Doheny, Kimberly S16.1, S16.3,<br />

S16.4<br />

Dombovári, Edit 50<br />

Domschke, Katharina S1.4, 30<br />

Donohoe, Gary S3.1, S9.1<br />

Doody, Gillian 211<br />

Dorst, Marieke 31<br />

Doud, Mary Kathryn S15.5<br />

Doudney, Kit 184<br />

Downing,<br />

AnnCatherine<br />

O2.4<br />

Downs, B. William 182<br />

Drago, Antonio 44, 186<br />

Dreolini, Lisa S3.5<br />

Drews, Eva 120<br />

Drgon, Tomas 183, 273<br />

Druid, Henrik 230<br />

Duan, Jubao 115, 245<br />

Ducci, Francesca O16.1, ECI 2, 105<br />

Dudbridge, F 245<br />

Duggirala,<br />

Ravindranath<br />

S12.4, S12.5, ECI<br />

13, 76<br />

Duncan, Laramie 103<br />

Dutt, Anirban 83, 236<br />

Dwyer, Sarah O12.1, 210<br />

Dyer, Thomas D. S12.4, S12.5, ECI<br />

13, 76<br />

Ebstein, Richard 11, 91<br />

Edenberg, Howard S16.1, S16.2,<br />

S16.3, S16.4,<br />

S16.6, S16.7, ECI<br />

14, ECI 18, 90,<br />

138, 157<br />

Edman, Gunnar O10.1, O10.5<br />

Egan, Michael O6.3<br />

Eggermann, Thomas 86<br />

Ehlers, Cindy 275<br />

Ehringer, Marissa 69<br />

Eissa, Ahmed 243<br />

Ekwall, Karl 92<br />

Elassy, Mai 243<br />

Elbert, Adrienne 171<br />

El-Boraie, Hala 243<br />

Eley, Thalia C. 144<br />

Elfving, Betina 214<br />

Elkin, Amanda O1.3, 190<br />

Ellingrod, Vicki S11.1, S11.4<br />

Elliott, Paul ECI 2<br />

Elliston, Lynne 247<br />

Elston, Robert C. 148<br />

ENGAGE Handedness 114<br />

Consortium, The<br />

Engert, James O10.2<br />

Erhardt, Angelika O3.2, 30<br />

Eriksson, Elias 30<br />

Erpe, Mariano ECI 14<br />

Escaramís, Geòrgia 40<br />

Eskin, Eleazar 157<br />

Esko, T. 114<br />

Esposito-Smythers,<br />

Christianne<br />

62<br />

Estivill, Xavier 8, 40<br />

Eubank, James O14.3<br />

Evans, DM 114<br />

Evans, Emma O4.4, 19<br />

Evans, Kathy 208<br />

Evdokimova, Oksana 280<br />

Everall, Ian S6.2, S7.3, 213<br />

Evgrafov, Oleg 239<br />

Fagerness, Jes S10.3, S10.4,<br />

S10.5, 61<br />

Fallin, MD 245<br />

Fallon, James 63, 238<br />

Fann, Cathy S-J 87, 205, 235<br />

Fanous, Ayman 42, 179, 223, 261


Faraone, Stephen S6.3, S8.1, S8.2,<br />

S8.3, 3, 9, 11, 15,<br />

17, 18, 118<br />

Farmer, Anne E. S1.1, O1.3, O2.5,<br />

O5.3, O7.4,<br />

O11.1, O11.5,<br />

102, 143, 164,<br />

190, 285<br />

Fasmer, Ole Bernt S14.2, 9<br />

Fathi, Warda 243<br />

Favis, Reyna 199<br />

Fearon, Paul 211<br />

Feier, Gustavo 75<br />

Feiler, Heidi 275<br />

Feinberg, Andrew P3.1, S2.2<br />

Feng, Guoping O3.3, O6.6<br />

Feng, GuoYin O3.3, 38<br />

Feng, Ningping O6.3<br />

Feng, Yu O14.3, 50, 95, 171<br />

Fennell, Alanna 173<br />

Fernandez, Cathy O13.3<br />

Fernandez, Guillen O8.3<br />

Ferraro, Thomas N. 41<br />

Ferraz Alves, Tania 113<br />

Ferrier, Nicol O5.3, O10.3<br />

Filges, Isabel 256<br />

Filiou, Michaela S6.1<br />

Fisher, Helen O15.2, 211<br />

Fisher, Sherri S16.3<br />

Flint, Jonathan O3.5<br />

Flint, Tracey J O1.1, O8.1<br />

Flodman, Pamela O5.1<br />

Flores, Deborah 141<br />

Foldager, Leslie O1.1<br />

Forero, Diego 52, 122<br />

Forlenza, Orestes 73<br />

Foroud, Tatiana S16.6, ECI 14,<br />

ECI 18, 90, 157<br />

Forsell, Yvonne O1.2, O7.3, O9.1<br />

Forstner, Andreas J. O8.2, O9.2<br />

Fox, Helen 74<br />

Fox, Louis S16.3, S16.5<br />

Fox, Peter T S12.4<br />

Frangou, Sophia 153<br />

Frank, Christopher L. S15.5<br />

Frank, Josef 197<br />

Franke, Barbara O4.2, O8.3, O9.6,<br />

2,3,5,7, 9, 11<br />

Franke, Thomas O1.6<br />

Franti, Lindsay 89<br />

Fraser, Christine O10.3<br />

Freedman, Robert 115, 249<br />

Freeman, Natalie 202<br />

Freimer, Nelson S10.3, ECI 2<br />

Freitag, Christine O4.1<br />

Freudenberg, Jan O9.2<br />

Frisen, Louise O10.1, O10.5<br />

Fryland, Tue 214<br />

Fryns, Jean-Pierre O13.2<br />

Fu, Cynthia 285<br />

Fu, Dong-Jing 199<br />

Fuchs, Karoline 152<br />

Fujii, Takashi 66<br />

Fukuhara, Chiaki 67<br />

Fukunaka, Yuko 268, 269, 272<br />

Fumagalli, M. O12.4<br />

Furney, Simon O17.4<br />

Gaafar, Hanan 243<br />

Gádoros, Julia 50<br />

Gage, Fred P1.1<br />

Galea, Sandro 110<br />

Gallagher, Louise O15.3, 258<br />

Galter, Dagmar 142<br />

Ganda, Clarissa 57<br />

Gao, Cheng-Ge 204<br />

Gardiner, Erin S7.2<br />

Gargus, Jay O5.1<br />

Garrido, Helena S10.6<br />

Garriock, Holly S1.2<br />

Gastaminza, Xavier 8<br />

Gates, Amy 125<br />

Gattaz, Wagner 73, 75, 198<br />

Gaudi, Simona 238<br />

Gaysina, Dasha 285<br />

Ge, Dongliang O11.3, 254<br />

Ge, Xuecai S15.5<br />

Gejman, Pablo V. 115, 245<br />

Gelernter, Joel 110<br />

GENEVA S10.3, S16.1,<br />

S16.2, S16.3,<br />

S16.6, S16.7<br />

Genro, Julia 13<br />

George, Charles J. 202<br />

Georgieva, Lyudmila O12.1, O12.5<br />

Gerrish, Amy O17.1, 247<br />

Gershon, Elliot S2.3, S2.5, S5.3,<br />

127, 157, 180<br />

Geschwind, Daniel 94<br />

Geyer, Mark S6.3, 146, 207<br />

Ghadiri, Mohammad 97, 158<br />

Giacometti, Emanuela S3.6<br />

Gibbs, Anne 80<br />

Gibbs, Richard O12.3<br />

Giegling, Ina O12.1, 186<br />

Gilbert, Donald L. ECI 4<br />

Gilder, David 275<br />

Gilks, Will 125<br />

Gill, Michael S3.1, S9.1, O12.1,<br />

O17.1, 4, 6, 10,<br />

11, 12, 16, 21, 48,<br />

125, 143, 258<br />

Gillespie, Charles 79<br />

Giltay, E.J. 60<br />

Girardi, Paolo 153<br />

Gizer, Ian 275<br />

Gladwin, Thomas 260, 265<br />

Glahn, David S12.4, S12.5, ECI<br />

13, 76<br />

Glaser, B 32, 93<br />

Glatt, Stephen S6.2, 82, 213<br />

Glessner, J 32<br />

Goate, Alison S16.3, S16.5,<br />

O17.1, ECI 7<br />

Goenjian, Armen 65<br />

Goes, Fernando 150<br />

Gogos, Joseph A. S3.4<br />

Golding, J 32<br />

Goldman, David O1.7, O16.1, 167<br />

Goldman, Lynn R O16.1<br />

Goldstein, David O11.3, 254<br />

Golimbet, Vera 280<br />

Gomez, Lissette O14.3, 171<br />

Gómez-Barros, Nuria 8<br />

Gomez-Sanchez,<br />

Ariadna<br />

ECI 5<br />

Gonzalez, Laura 108<br />

Goodman, Marianne O1.7<br />

Goossens, Dirk 122, 136, 237<br />

Gordon-Smith,<br />

Katherine<br />

O10.3<br />

Göring, Harald S12.4, S12.5, ECI<br />

13, 76<br />

Gow, Alan 74<br />

Graae, Lisette 142<br />

Grant, SFA 32<br />

Gratacòs, Mònica 40<br />

Gray, Joanna 285<br />

Green, Elaine O5.3, O10.3<br />

Green, Michael 249<br />

Greenwood, Tiffany S9.4, S14.2, O7.6,<br />

157, 166, 180,<br />

207, 249, 283<br />

Gregersen, Noomi 27<br />

Grevet, Eugênio H. 14<br />

Grigoroiu-Serbanescu,<br />

Maria<br />

148, 149<br />

Groß-Lesch, Silke 8<br />

Grove, Tyler S11.1<br />

Grozeva, Detelina O5.3, O10.3, 19<br />

Grucza, Richard S16.2, S16.3,<br />

S16.4, S16.5,<br />

S16.6, ECI 7<br />

Gruenewald, Ellen 51<br />

Gschwind, Leo 255, 256<br />

Guerrini, Irene ECI 17<br />

Guffanti, Guia O6.2, 63, 132,<br />

238, 264<br />

Guidry, Jami 242<br />

Guijarro-Domingo,<br />

Silvina<br />

8<br />

Guilherme, Luiza ECI 8<br />

Guillozet-Bongaarts,<br />

Angela<br />

232<br />

Guindallini, Camilla 170<br />

Guitiérrez-Zotes,<br />

Alfonso<br />

40<br />

Gunasinghe, Cerisse 285<br />

Guo, An-Yuan O3.5, 266, 267<br />

Guo, Ting-Wei 204<br />

Gupta, Ajay O2.2<br />

Gur, Raquel 76, 249<br />

Gurling, Hugh O2.1, ECI 17, 59,<br />

156<br />

Gutiérrez, Blanca 28<br />

Gwilliam, Rhian O17.1<br />

Haas, Magali 199<br />

Haavik, Jan S14.2, O9.6, 3, 5,<br />

7, 8, 9<br />

Hack, Laura O16.2<br />

Haggarty, Stephen S15.5<br />

Hagoort, Peter O8.3<br />

Haines, Jonathan O17.2<br />

Hakonaron, Hakon S13.1, S13.2, 32<br />

Hall, Mei Hua S12.3, 80, 240<br />

Hall, Stephanie 15, 17<br />

Halmoy, Anne S14.2, 7<br />

Hamdan, Adnan 206<br />

Hamilton, Steven S1.2, ECI 6<br />

Hamshere, Marian O10.3, O12.1,<br />

O17.1, 160


Han, Baoguang O2.4<br />

Han, Yan 204<br />

Hanna, Gregory S10.4, S10.5, 61<br />

Hansen, Aase 39<br />

Hansen, M. O1.1, 245<br />

Hansen, Thomas 212<br />

Haraksingh, Rajini 130<br />

Hardin, Jill O16.4<br />

Hare, Elizabeth 141<br />

Harold, Denise O17.1<br />

Haroutunian, Vahram O12.5<br />

Harris, Adam S13.5<br />

Harris, Sarah 74, 208<br />

Hartikainen, Anna-<br />

Liisa<br />

ECI 2<br />

Hartmann, Annette O12.1, 186<br />

Hartz, Sarah 138<br />

Hashimoto, Eri 94<br />

Hashimoto, Hitoshi O6.5<br />

Hashimoto, Ryota 66, 215<br />

Hatsukami, Dorothy S16.5, ECI 7<br />

Hattori, Kotaro 66<br />

Hauser, Joanna S1.1, O2.5, 190<br />

Hawi, Ziarah 4, 10, 12, 21<br />

Hawkin, William O5.2<br />

He, Guang O6.1, 38<br />

He, Lin O6.1, O7.1, 38<br />

Head, Jennifer 283<br />

Heath, Simon O1.3, O7.4, O11.1<br />

Heck, J. Denis O5.1<br />

Hedemand, Anne O8.1, 27, 212<br />

Heine, Monika 9<br />

Heinz, Andreas 197<br />

Henigsberg, Neven S1.1, O2.5, 190<br />

Hennah, William S15.2, 189, 208<br />

Hennings, Johannes S1.4<br />

M.<br />

Her, Charles-Henry 156<br />

Heresco-Levy, Uriel 91<br />

Heriani, H 57<br />

Herms, Stefan 119, 149<br />

Hernández, Sandra 108<br />

Heron, Eleisa 258<br />

Heron, Jon O4.3<br />

Hervás, Amaya 8<br />

Hesselbrock, Victor S16.1, S16.2,<br />

S16.3, S16.6,<br />

S16.7<br />

Hessl, David 22<br />

Hester, Robert 84<br />

Hettema, John O3.5<br />

Heutink, Peter S4.1, S4.5<br />

Heyrman, L 237<br />

Higuchi, Teruhiko 66<br />

Higuchi, Ttsuro 121<br />

Hill, Francesca 72<br />

Hill, Matthew 21, 48, 168, 253<br />

Hinds, David O16.4<br />

Hinrichs, Anthony S16.3, S16.5<br />

Hipolito, Maria O11.4, 157<br />

Hippman, Catriona 107<br />

Hirata, Yuko 172<br />

Hoda, Farzana 285<br />

Hodge, Susan ECI 6<br />

Hodges, Angela O17.4<br />

Hodgkinson, Colin A O1.7, O16.1, 167<br />

H<strong>of</strong>, Patrick O13.1<br />

H<strong>of</strong>fmann, Per 174<br />

H<strong>of</strong>man, Albert O7.2<br />

H<strong>of</strong>t, Nicole 69<br />

Hohmann, John 232<br />

Hokyo, Akira 82<br />

Hollander, Jonathan S7.4<br />

Hollingworth, Paul O17.1<br />

Holmans, Peter O5.3, O10.3,<br />

O12.1, O12.5,<br />

O17.1, 245<br />

Holsboer, Florian S1.4, O3.2, O15.1,<br />

100, 101<br />

Holst, Martin O12.2<br />

Holt, Robert A. S3.5<br />

Honda, Hiroyuku 215<br />

Honer, William 107<br />

Hoogendijk, Witte S4.1, S4.5<br />

Hoogman, Martine 2<br />

Hoop, Jinger P2.3<br />

Horan, Michael 74<br />

Hori, Hiroaki 66<br />

Horstmann, Sonja S1.4<br />

Hosang, Georgina 102<br />

Hottenga, Jouke-Jan 114, 147<br />

Hou, Wei-Kun 204<br />

Hougaard, David<br />

Michael<br />

O8.1<br />

Hounie, Ana S10.4, S10.5, ECI<br />

8, 61<br />

Houser, Melissa 259<br />

Hovatta, Iiris O3.4<br />

Howells, William S16.1, S16.3<br />

Howrigan, Daniel 118<br />

Hsu, Pei-Chun 87, 205, 235<br />

Hu, Xiaolan 15<br />

Hua Hall, Mei 83<br />

Huang, Ke O6.1<br />

Huber, Andreas 255<br />

Hudziak, James S8.1<br />

Huezo-Diaz, Patricia<br />

Hukic, Diana<br />

190<br />

Hulse, Gary 192<br />

Hulsh<strong>of</strong>f Pol, Hilleke<br />

E.<br />

85, 227<br />

Hultman, Christina 224, 225<br />

Hunt, Stephen 59<br />

Hunter, Jason R. 71<br />

Hunter, Mary-Jill 107<br />

Hussain, Hamid 139<br />

Hutchison, Kent 69<br />

Hutz, Mara 13<br />

Hwang, Rudi S11.5<br />

Hwu, Hai-Gwo 87, 205, 228, 235<br />

Hyde, Thomas 232<br />

Iacono, William G. ECI 10<br />

Iaenisch, Rudolf S3.6<br />

Ibrahim, Ibtihal 243<br />

Ibrahim, Nahed 243<br />

Ichiba, Mio 121, 123<br />

Ickovicz, Abel 171<br />

Iijima, Yoshimi 66<br />

Iizuka, Yukihiko O6.3<br />

Ikeda, Masashi 215<br />

Ikram, MA 114<br />

Ilott, Nicholas 24<br />

Im, Heh-In S7.4<br />

Inada, Toshiya 176, 220, 222, 274<br />

Ingason, Andres S5.2<br />

Inoue, Ken 29<br />

International Group<br />

for The Study <strong>of</strong><br />

Lithium Treated<br />

Patients, IGSLI<br />

O9.4<br />

Interheart<br />

Investigators<br />

O10.2<br />

Ioannidis, John 30<br />

Irizarry, Rafael S2.2<br />

Irmansyah, I 57<br />

Irons, Daniel E. ECI 10<br />

Ishigooka, Jun O1.4<br />

Ishihara, Ryoko 231<br />

Ising, Marcus S1.4, O3.2, O15.1,<br />

100, 101<br />

Israel, Salomon 91<br />

Ito, Yoshihito 176, 209, 220,<br />

221, 222, 231<br />

Iuvone, Michael 67<br />

Ivanov, Dobril O5.3<br />

Iwamoto, Kazuya S2.1, O1.4, 94<br />

Iwata, Nakao 215, 220, 221,<br />

222, 274<br />

Iyegbe, Conrad 105<br />

Iyo, Masaomi 274<br />

Izzo, Giselle 73<br />

Jablensky, Assen S3.1<br />

Jack, Lisa O16.4<br />

Jackson, Sarah 106<br />

Jacob, Christian P. O4.1, O9.6, 5, 7,<br />

8, 9<br />

Jacobsen, Kaya S14.2, 7<br />

Jancic, Dunya S14.3<br />

Janiri, Luigi 188<br />

Jansen, Andreas 86<br />

Janssens, Cecile 147<br />

Järvelin, Marjo-Riitta ECI 2<br />

Jay, M 245<br />

Jayathilake, Karu O6.5<br />

Jazin, Elena 251<br />

Jenkins, Gregory S1.2<br />

Jerez, Alvaro 141<br />

Jerman, Borut S1.1, O2.5<br />

Jia, Peiin O2.6, 42, 266, 267<br />

Jiang, Lin 251<br />

Jin, Choon-Jo 151<br />

Joensen, S<strong>of</strong>us 27<br />

Johansson, Stefan S14.2, 3, 7, 8, 9<br />

Johnson, Catherine 183, 273<br />

Johnson, Eric S16.3, S16.4,<br />

S16.5, ECI 7<br />

Johnson, Katherine 6<br />

Johnson, Matthew P. S12.4<br />

Johnson, Paul 45<br />

Jonathan, Sebat S5.4<br />

Jones, Allan 232<br />

Jones, Ian O5.3, O7.4,<br />

O10.3, 102<br />

Jones, Lisa O5.3, O7.4,<br />

O10.3, 102, 285<br />

Jones, Peter 211<br />

Joo, Eun-Jeong 46, 47<br />

Joyce, Peter 184<br />

Joyner, Alexander 64


Judy, Jennifer S14.1, 154, 257,<br />

263<br />

Jugurnauth, Sarah S7.1<br />

Jurkeniene, Lina 109<br />

Kaakinen, Marika ECI 2<br />

Kaczvinszk, Emília 50<br />

Kærgaard, Anette 39<br />

Kærlev, Linda 39<br />

Kahn, Rene O11.2, 81, 85,<br />

227, 234, 260, 265<br />

Kaiya, Hisanobu ECI 11, 29<br />

Kakiuchi, Chihiro 37, 177<br />

Kaladjieva, Luba S3.1<br />

Kalita, J 78<br />

Kallunki, Pekka O1.1<br />

Kalsi, Gursharan O16.2<br />

Kamail, Masoud 89, 161<br />

Kamar, Sher Bahadur 240<br />

Kan, Cees 2, 7, 9<br />

Kananen, Laura O3.4<br />

Kanazawa, Tetsufumi 82, 193<br />

Kand, Tae Sun 133<br />

Kandaswamy, ECI 17, 156<br />

Radhika<br />

Kandil, Karim 243<br />

Kane, John S9.3<br />

Kaneva, Radka 277<br />

Kang, Seung-Gul 195, 196, 200<br />

Kang, Sung Wook 254<br />

Kang, Xin 239<br />

Kano, Yukiko 35, 37<br />

Kanyas, K. O12.4<br />

Kaplan, Allan S 20<br />

Kapoor, Manav 111<br />

Kapornai, Krisztina 50, 202<br />

Kaprio, Jakko O14.1<br />

Kapur, Suman 111, 112<br />

Karayan, Ida 65<br />

Karlsson, Robert 142<br />

Kas, Martien O13.3, O13.4<br />

Kasai, Kiyoto 37<br />

Kasper, Siegfried 140<br />

Katerberg, Hilga S10.2<br />

Kato, Maiko 121<br />

Kato, Nobumasa 35<br />

Kato, Tadafumi S2.1, O1.4, 94<br />

Kaufman, Liana ECI 15<br />

Kawamura, Yoshiya 29, 35, 37, 177<br />

Kawashige, Seiya 82, 193<br />

Kaye, Walter S13.2, S13.3,<br />

S13.5<br />

Ke, Xiayi 74<br />

Keavney, Bernard O10.2<br />

Keefe, Richard S. E. 229<br />

Keers, Robert 102, 190<br />

Keller, Matthew 103, 246<br />

Kelly, Brian 68<br />

Kelsoe, John P4.5, S9.4, S14.2,<br />

O7.3, O7.5, O7.6,<br />

56, 146, 157, 166,<br />

169, 180, 201,<br />

242, 283<br />

Kember, Rachel 294<br />

Kemner, Chantal 81<br />

Kendler, Kenneth S16.7, O3.5,<br />

O16.2, O16.3, ECI<br />

3, 42, 115, 179,<br />

223, 245, 261, 279<br />

Kennedy, James S10.5, S11.5, 20,<br />

30, 50, 61, 172,<br />

178, 191, 202<br />

Kennedy, Martin 184<br />

Kennemer, Michael O16.4<br />

Kenny, Elaine 125<br />

Kenny, Paul S7.4<br />

Kent, Lindsey 4, 10<br />

Kerem, Zekeria 139<br />

Kerner, Mallory 182<br />

Kerr, Elizabeth 171<br />

Kertes, Darlene A. 279<br />

Khemka, Deepak 137<br />

Kiefer, Falk 197<br />

Kiemeney, Lambertus 7<br />

Kikuyama, Hiroki 82, 193<br />

Kilpatrick, Dean 110<br />

Kim, Hak Jae 254<br />

Kim, Hyun Sook 46<br />

Kim, Jeong-Hyun 70, 134<br />

Kim, Jong Woo 254<br />

Kim, Leen 195, 196<br />

Kim, Se Hyun 47<br />

Kim, Su Kang 254<br />

Kim, Yong Sik 47<br />

King, Mary-Claire S5.1<br />

King, Nicole 172<br />

Kinnell, Hazel 51<br />

Kinon, Bruce O2.4<br />

Kinoshita, Yoko 215<br />

Kircher, Thilo 86<br />

Kirley, Aiveen 4, 10<br />

Kirov, George S5.2, O5.3, O10.3,<br />

O12.1, O12.5, 19,<br />

247<br />

Kirschbaum, Clemens O7.2<br />

Kiss, Eniko 50, 202<br />

Klei, Lambertus 243<br />

Kleinman, Joel 232<br />

Klimecki, Walt 56<br />

Kloiber, Stefan S1.4, O3.2, 100<br />

Klok, M.D. 60<br />

Knappskog, Per 7, 8, 9<br />

Knight, Helen O5.2<br />

Knowles, James S10.4, 239<br />

Ko, Seong-Gyu 70, 134<br />

Kochunov, Peter S12.4<br />

Kockum, Ingrid 142<br />

Kodama, Masafumi 274<br />

Koehler, Angela N. S15.5<br />

Koenen, Karestan 110<br />

Koenigsberg, Harold O1.7<br />

Koga, Minori 215<br />

Koh, Jun 82, 193<br />

Kohli, Martin A. S1.4<br />

Koide, Takayoshi 220<br />

Koishi, Shinko 35<br />

Kojima, Toshio O1.4<br />

Kolachana, Bhaskar O6.3<br />

Koller, Daniel O10.4, ECI 14,<br />

157<br />

Kolstad, Henrik 39<br />

Komel, Radovan 49<br />

Konishi, Yoshiaki ECI 11<br />

Konneker, Thomas 25, 229<br />

Kooij, J.J. Sandra 7, 9<br />

Korbel, Jan O. 130<br />

Korszun, Ania O1.3, O7.4, 102,<br />

143<br />

Koszycki, Diana 178<br />

Kothencné, Viola<br />

Osváth<br />

50<br />

Kovacic, Zrnka 190<br />

Kovacs, Maria 50, 202<br />

Kovas, Yulia 77<br />

Kozel, Dejan 190<br />

Kracht, Sören 86<br />

Kraft, Jeffrey S1.2<br />

Kramer, John S16.1, S16.2,<br />

S16.3, S16.4,<br />

S16.6, S16.7<br />

Krasteva, Dorita 277<br />

Krastoshevsky, Olga 80<br />

Krause, Verena 80<br />

Kreiker, Susanne 7, 8<br />

Kremen, William S6.2, 213<br />

Kremensky, Ivo 277<br />

Krieger, Jose Eduardo 276, 278<br />

Kripke, Daniel O7.5, 56, 201<br />

Kristensen, Ann Suhl 30, 39<br />

Kronholm, Erkki O1.5<br />

Krueger, Robert S16.1, S16.2,<br />

S16.3, S16.4,<br />

S16.6, S16.7, ECI<br />

10<br />

Krug, Axel 86<br />

Kruggel, Frithj<strong>of</strong> 63, 132<br />

Kruse, Torben 27<br />

Kuan, Leonard 232<br />

Kukita, Camila 73<br />

Kumar, Ravinesh A. ECI 12<br />

Kumari, Meena O7.2<br />

Kuntsi, Jonna 11<br />

Kunugi, Hiroshi 66<br />

Kuo, Po-Hsiu O3.5, O16.2<br />

Kuperman, Samuel S16.1, S16.2,<br />

S16.3, S16.6,<br />

S16.7<br />

Kurian, Sunil S6.3<br />

Kusenda, Mary O13.5<br />

Kushima, Itaru 174, 209, 220,<br />

221, 222<br />

Kusumawardhani,<br />

Agung<br />

57<br />

Kusumi, Ichiro O1.4<br />

Kyra, Kanyas 206<br />

Labad, Antonio 248<br />

Lachman, Herb O5.4<br />

Laenerts, A-S 237<br />

Lahiri, Debomoy S6.3<br />

Lahti, Jari 189<br />

Lai, I-Ching O8.4<br />

Laird, Nan 118<br />

Lakatos, Anita 63, 132<br />

Lambert, Christophe O17.3<br />

Lancet, D. O12.4<br />

Landaas, Elisabeth 7<br />

Landen, Mikael O10.1, O10.5<br />

Landgraf, Rainer S6.1<br />

Langenecker, Scott 89, 161<br />

Langley, Kate O4.3, O4.4, 1, 19<br />

Laranjeira, Ronaldo 276, 278<br />

Larsen, Eric 190


Lasseter, VK 245<br />

Lathrop, Mark O1.3, O7.4,<br />

O11.1, 164<br />

Laurent, C. 245<br />

Laurie, Cathy S16.1<br />

Lavebratt, Catharina O1.2, O7.3, O9.1,<br />

92<br />

Law, Matthew 203<br />

Lawlor, Brian O17.1<br />

Lawrence, Jacob O2.1, ECI 17, 59,<br />

156<br />

Lawson, William O11.4, 157<br />

Lazzeroni, Laura 249<br />

Lea, Vella 242<br />

Lecrubier, Yves 140<br />

Lee, Hee Jae 254<br />

Lee, Heon-Jeong 151, 195, 196, 200<br />

Lee, James 275<br />

Lee, Kyu Young 46, 47<br />

Lee, Migyung 250<br />

Lee, Richard 99<br />

Lee, Sang Kil 67<br />

Lee, Sun-Young<br />

Lee, Yohan<br />

151<br />

Leff, Julian 211<br />

Leibenluft, Ellen 163, 165<br />

Lekman, Magnus 142<br />

Lemenager, Tagrid 197<br />

Lenaerts, An-S<strong>of</strong>ie 122<br />

Lencz, Todd S9.2, S9.3, O8.6<br />

Le-Niculescu, Helen S6.3, O10.4<br />

Lennartsson, Andreas 92<br />

Lepard, Tiffany 106<br />

Leppä, Virpi O1.5<br />

Lerer, Bernard O12.4, 233, 238,<br />

245<br />

Lesch, Klaus-Peter O4.1, O9.6, 3, 5,<br />

7, 8, 9<br />

Levin, Raz 91<br />

Levinson, Douglas F. 115, 245<br />

Levitan, Robert D 20<br />

Levy, Deborah 80<br />

Lewis, Cathryn M S1.1, O1.3, O2.5,<br />

O7.4, O11.1, 143,<br />

153, 164, 170<br />

Lezheiko, Tatyana 280<br />

Li, Chao O6.3<br />

Li, Dawei ECI 16<br />

Li, Qingqin 199<br />

Li, Rick 115<br />

Li, Tao O6.1<br />

Li, Wenbiao 79<br />

Liang, KY 245<br />

Liao, Ding-Lieh O8.4<br />

Liao, Isaac ECI 4<br />

Libiger, Ondrej S13.5<br />

Lichtenstein, Paul 224, 225<br />

Lieb, Roselind O15.1, 100, 101<br />

Lieberman, Jeffrey A. S11.5, O2.6,<br />

O12.3, 191, 229<br />

Liedel, Stephanie O9.6, 5<br />

Lienard, Patricia 140<br />

Liewald, David 74<br />

Light, Gregory 207, 249<br />

Lin, Michelle O4.1<br />

Lin, Peng 138<br />

Lindefors, Nils O3.1<br />

Lindgren, CM 114<br />

Lindholm Carlstrom,<br />

Eva<br />

251<br />

Linotte, Sylvie 140<br />

Linse Peterson, Greta O17.3<br />

Lionel, Anath 158<br />

Liou, Ying-Jay O8.4<br />

Lit, Lisa ECI 4, 22<br />

Liu, Chih-Min 87, 205, 228, 235<br />

Liu, Chunyu S2.3, S2.5, S5.3,<br />

127, 157, 180<br />

Liu, Qing Rong 183<br />

Liu, Tze-Tze 205<br />

Liu, Wenlei O2.4<br />

Liu, Xiao-Gang 204<br />

Liu, Xiaoxi 35, 37, 177<br />

Liu, Xinmin 163<br />

Liu, Youfang 224, 225<br />

Liu, Yue 204<br />

Liu, Yu-Li 87, 205, 228, 235<br />

Livingston, Gill O17.1<br />

Lloyd, K.C. Kent 26<br />

Lochner, Christine S10.2, S10.4<br />

Logan, Richard O14.3<br />

Loh, Han Chern ECI 1, 217, 226<br />

Loh<strong>of</strong>f, Falk W. 41, 157<br />

Lohr, James S6.3, 213<br />

Lönnqvist, Jouko O3.4, 189<br />

Lonsdorf, Tina O3.1<br />

Loo, Sandra 15, 17<br />

Lorentzen, Steinar 53<br />

Loukola, Anu O1.5<br />

Lourdusamy,<br />

Anbarasu<br />

O14.1, 281<br />

Lovestone, Simon S15.1, O17.1,<br />

O17.4<br />

Lovett, Maureen 171<br />

Low, Nancy C. O9.3<br />

Lu, Q. Richard S15.4<br />

Lu, She-Min 204<br />

Lucae, Susanne S1.4, 100<br />

Luciano, Michelle 74<br />

Ludwig, Kerstin 174<br />

Luis, Rohde 13<br />

Lunnon, Katie O17.4<br />

Luo, Wei 98<br />

Lupoli, Sara O12.4, 63, 233<br />

Luzi, Sonija O15.2, 105<br />

Lydall, Gregory J. ECI 17, 59<br />

Lynskey, Michael S16.2, S16.3,<br />

S16.4, 277<br />

Lysaker, Paul S6.3<br />

Ma, Jie 204<br />

Maccarrone,<br />

Giuseppina<br />

S6.1<br />

Macciardi, Fabio O6.2, O12.4, 233,<br />

63, 132, 238, 264<br />

Macintosh, Andrew S3.2<br />

Macintyre, Donald O5.2<br />

Maclean, Alan O5.2<br />

Maddox, Connie 67<br />

Madison, Jon S15.5<br />

Magi, R. 114<br />

Magistretti, Pierre S13.2, S13.5<br />

Maher, Brion O3.5, ECI 3, 223,<br />

261<br />

Mahon, Pamela 257<br />

Maia, Jessica O11.3, 254<br />

Maier, Wolfgang S1.1, S14.4, O2.5,<br />

O17.1, 143, 190,<br />

245<br />

Makarov, Vladimir 165<br />

Malaspina, Dolores 239<br />

Malhotra, Anil S9.2, S9.3, O8.6,<br />

ECI 9, 165<br />

Malhotra, Dheeraj 165<br />

Malki, Karim O2.5<br />

Mallet, J. 245<br />

Malloy, Mary O5.2, 216, 237<br />

Malone, Molly 171<br />

Manchia, Mirko O9.4, 162<br />

Mandelli, Laura 185, 188<br />

Mangino, M. 114<br />

Manji, Husseini 199<br />

Mann, Karl 197<br />

Manor, Iris 11<br />

Mansour, Hader 243<br />

Mantripragada, Kiran O4.4, 19<br />

Manz, Niklas ECI 18, 90<br />

Mao, Yingwei S15.5<br />

Marco, Di Nicola 188<br />

Margolis, Russell L. 135<br />

Mari, Jair 170<br />

Markov, Valentin 86<br />

Marks, Michael 59<br />

Maron, Eduard 30<br />

Marques, Andrea ECI 8<br />

Marshall, Nicolette 236<br />

Martin, Andrew O4.4, 19<br />

Martin, Nick S12.1, 114<br />

Martinez Levy,<br />

Gabriela Ariadna<br />

ECI 5<br />

Martinotti, Giovanni 188<br />

Martorell, Lourdes 40, 248<br />

Marusic, Andrej 190<br />

Massat, Isabelle 140<br />

Mathé, Aleksander 92<br />

Mathews, Carol S10.1, S10.2,<br />

S10.3, S10.4,<br />

S10.5, S10.6,<br />

S10.7, 61<br />

Mathur, Aditi 203<br />

Mattheisen, Manuel 119, 160<br />

Mattingley, Jason 84<br />

Mattingsdal, Morten 53<br />

Mavroconstanti,<br />

Thegna<br />

7<br />

May, Todd 213<br />

Mayer, László 50<br />

Mazza, Marianna 188<br />

McCarthy, Mark ECI 2, 114<br />

McCarthy, Michael 201<br />

McCarthy, Shane 165, 291<br />

McClay, Joseph L. O2.6<br />

McClellan, Jon S5.1<br />

McClure, Jennifer O16.4<br />

McCombie, W.<br />

Richard<br />

208<br />

McCracken, Jim 17<br />

McDonald, Colm 236<br />

McEvoy, Joseph O11.3, 254<br />

McGeary, John E. 58, 62<br />

McGhee, Kevin O5.2, 74<br />

McGrath, Patrick S1.2


McGuffin, Peter S1.1, O1.3. O5.3,<br />

O7.4, O10.3,<br />

O11.1, O11.5,<br />

O15.2, 102, 143,<br />

152, 164, 190, 211<br />

McHugh, Patrick 184<br />

McInnis, Melvin S6.4, 89, 128, 129,<br />

157, 161<br />

McKeigue, Paul 170<br />

McKeon, Patrick 168<br />

McKinney, Rebecca O7.5, 166, 201,<br />

283<br />

McLeod, Howard 23<br />

McMahon, Francis S14.3, S14.4,<br />

O2.2, 5.5, O11.6,<br />

ECI 19, 30, 127,<br />

157, 163, 165<br />

McNealy, Kristin 88<br />

McNeill, Geraldine 74<br />

McQueen, Matthew 118, 246<br />

McQuillin, Andrew O2.1, ECI 17, 59,<br />

156<br />

McRae, Allan O5.2, 208<br />

Meaburn, Emma O14.5<br />

Medland, SE 114, 116<br />

Megson, Ian 203<br />

Meier, Sandra 160<br />

Meiser, Bettina P2.1<br />

Melas, Philippe 92<br />

Melle, Ingrid 53, 64<br />

Meltzer, Herbert O6.5, 191<br />

Menchón, José<br />

Manuel<br />

40<br />

Mendez, Enrique ECI 5<br />

Mendlewicz, Julien 140, 190<br />

Mendoza, Rickardo 141<br />

Menezes, Paulo 104, 113<br />

Menke, Andreas S1.4<br />

Mercer, Kristie 79<br />

Merikangas, Alison O15.3, O15.4<br />

Metspalu, Andres 30<br />

Meye, Frank O13.3<br />

Meyer, Jerrold O4.1, 68<br />

Meyer, Jobst O4.1<br />

Michelon, Leandro 104, 113<br />

Mick, Eric 3, 9, 15, 17, 155<br />

Middeldorp, Christel 147<br />

Middleton, Frank S8.1, S8.2, S8.3,<br />

18<br />

Miguel, Euripedes ECI 8<br />

Mikhailov, Anna 173<br />

Mikkelsen, Sigurd 39<br />

Milanesi, L. 233<br />

Mill, Jonathan O9.5, O9.7,<br />

O14.1, O14.5, 24<br />

Millar, Kirsty 208<br />

Miller, Del S11.1<br />

Miller, Gregory 68<br />

Minassian, Arpi 146, 242<br />

Minato, Takanobu 29<br />

Mir, Asif ECI 15, 173<br />

Mira, Korner 206<br />

Miralles, Carmen 248<br />

Miranda, Ana 11<br />

Miranda, Eduardo 54, 55<br />

Mishra, U.K. 78<br />

Missaglia, Sara 188<br />

Mitchell, Philip P2.1<br />

Miyagawa, Taku 29<br />

Miyamoto, Jill 69<br />

Moebus, Susanne O8.2, O9.2, 119<br />

Moens, Lotte 52, 122, 136<br />

Moessner, Rainald O3.3<br />

M<strong>of</strong>fitt, Terrie O9.7<br />

Moghimi, Narges 158<br />

Molina, Esther 28<br />

Moline, Jessica S11.1<br />

Moller, Hans-Jurgen O12.1, 186<br />

Monakhov, Mikhail 91, 280<br />

Monnier, Philippe O14.3<br />

Moon, Randall S15.5<br />

Moreno, Ricardo 198<br />

Morgan, Craig O15.2, 211<br />

Morgan, Kevin O17.1, 211<br />

Morgan, Margaret O12.3<br />

Morgan, Marsha 59<br />

Morken, Gunnar 53<br />

Morris, Brian 45<br />

Morris, Derek S3.1, S9.1, O12.1,<br />

48, 125<br />

Morris, Noella Marie 192<br />

Mors, Ole S1.1, O1.1, O2.5,<br />

O8.1, 27, 30, 39,<br />

143, 190, 212, 214<br />

Mortensen, Preben Bo O8.1<br />

Moses, Eric K. S12.4, ECI 13<br />

Moskvina, Valentina O10.3, O12.1,<br />

O12.5<br />

Mostafavi-<br />

Abdolmaleky, Hamid<br />

O14.4<br />

Motala, Farhana 16<br />

Mouri, Akihiro 215<br />

Mowry, Bryan J. 115, 245<br />

Mueller, Daniel 191<br />

Mueller-Myhsok,<br />

Bertram<br />

S1.4<br />

Muglia, Pierandrea S1.1, O1.3, O7.4,<br />

O11.1, 164<br />

Mühleisen, Thomas S14.4, O8.2, O9.2,<br />

86, 119, 160<br />

Muhonen, Leea 189<br />

Muir, Walter O1.1, O5.2, 208,<br />

216, 237<br />

Mulas, Fernando 11<br />

Müller, Daniel J. S11.5<br />

Müller-Myhsok,<br />

Bertram<br />

S4.2, O3.2, 174<br />

Mulligan, Aisling 6, 16<br />

Muniz, Renan ECI 8<br />

Munsie, Leanne O2.4<br />

Murphy, Dennis S10.4, O11.6<br />

Murray, Robin O15.2, 54, 55, 83,<br />

104, 105, 211, 236<br />

Murray, Sarah S. S13.5<br />

MvGough, Jim 17<br />

Na, Han Sung 133<br />

Nabeshima, Toshitaka 215<br />

Nagai, Taku 209, 231<br />

Nakamura, Jun 215, 268, 269, 272<br />

Nakamura, Masayuki 121, 123, 124<br />

Nakamura, Yukako 176, 209, 220,<br />

221, 222<br />

Nakano, Yoko O1.4, 94<br />

Nalls, Michael O5.5<br />

Nandam, Sanjay 84<br />

Nasrun, Martina 57<br />

Nathan, Pradeep 84<br />

Navon, Ruth 126<br />

Neagu, Ana I. 149<br />

Neale, Benjamin P4.4, S10.3,<br />

S10.4, 15, 17, 114,<br />

116, 262<br />

Neale, MC 116<br />

Need, Anna O11.3, 254<br />

Negrão, André<br />

Brooking<br />

276, 278<br />

Negri, Gloria 188<br />

Nelson, Elliot 138<br />

Nelson, Stanley 15, 17<br />

Nertney, D. 245<br />

Nesheva, Eleonora 277<br />

Nestadt, Gerald S10.4, 245<br />

Neuh<strong>of</strong>f, Nina 174<br />

Neuman, Rosalind S16.1, S16.3<br />

Neuroimaging<br />

Initiative, Alzheimer’s<br />

Diesease<br />

63<br />

New, Antonia O1.7<br />

Ng, Mandy Y S1.1, O1.3, O7.4,<br />

O11.1, 143<br />

Nguyen, Trang O4.1, O9.6, 5<br />

Ni, Xingqun 191<br />

Nicodemus, Kristin 219<br />

Nicolini, Humberto S10.4, 141<br />

Niculescu, Alexander S6.3, O10.4<br />

Nielsen, Anders Lade 214<br />

Nieratschker, Vanessa O8.2, O9.2, 86,<br />

119<br />

Nievergelt, Caroline O7.5, 56, 157<br />

Nigg, Joel S8.4<br />

Nikamo, Pernilla O10.1, O10.5<br />

Nikolov, Ivan O10.3, O12.1<br />

Nikolov, Momchil 277<br />

Nimgaonkar,<br />

76, 118, 243<br />

Vishwajit<br />

Nisenbaum, Laura O2.4<br />

Nishida, Hisami 37<br />

Nishida, Nao 29<br />

Njau, Reuben O2.4<br />

Nnadi, Charles 167<br />

Noble, Ernest 65, 181<br />

Nocon, Agnes 101<br />

Noda, Yukihiro 209, 231<br />

Noens, Ilse O13.2, 31<br />

Noethen, Markus M. O5.5, 148, 149<br />

Nogueira, Mariana 8<br />

Nohesara, Shabnam 97<br />

Nolen, Willem 147<br />

Nonkens, Lourens O13.3<br />

Noor, Abdul ECI 15, 158, 173<br />

Nordent<strong>of</strong>t, Merete O1.1, O8.1<br />

Nørgaard-Pedersen,<br />

Bent<br />

O8.1<br />

Nørmølle<br />

Buttenschøn, Henriette<br />

27


Norrback, Karl- 52, 122, 136<br />

Fredrik<br />

Norton, Nadine O12.1, 245<br />

Nothen, Markus S14.3, S14.4,<br />

O8.2, O9.2,<br />

O17.1, 119, 160,<br />

174, 197<br />

Novak, Melinda 68<br />

Nowrouzi, Behdin 172<br />

Nuechterlein, Keith 249<br />

Nugent, Nicole 110<br />

Nurnberger Jr., John S6.3, S16.1,<br />

S16.2, S16.3,<br />

S16.6, S16.7,<br />

O10.4, O11.4, ECI<br />

14, 30, 138, 157<br />

Nwulia, Evaristus O11.4, 157<br />

Nyegaard, Mette O1.1, O8.1, 27,<br />

212, 214<br />

Nymberg, Charlotte O14.1<br />

O’Donovan, Michael 160<br />

Oades, Robert D 11<br />

Oberlander, Tim 43<br />

O'Donoghue, Therese S9.1<br />

O'Donovan, Lucy 45<br />

O'Donovan, Michael S3.1, S5.2, S9.1,<br />

O4.3, O4.4, O5.3,<br />

O10.3, O12.1,<br />

O12.5, O17.1, 19,<br />

210, 215, 245, 247<br />

O'Dushlaine, Colm 131, 159<br />

Oedegaard, Ketil<br />

Joachim<br />

S9.4, S14.2<br />

Oh, S 245<br />

Ohadi, Mina 158<br />

Ohashi, Mitsuki 231<br />

Ohman, Arne O3.1<br />

Ohmori, Osamu 268, 269, 272<br />

Ohnishi, Tetsuo O2.3<br />

Ohno, Kinji 209<br />

Ojha, Saroj Prasad S12.3, 240<br />

Ojopi, Elida 73, 75, 198<br />

Okahisa, Yuko 274<br />

Okamoto, Nagahisa 66<br />

Okazaki, Yuji ECI 11, 29<br />

Okochi, Tomo 215<br />

Oldham, Michael<br />

Olga, Likhodi<br />

94<br />

Olincy, Ann 249<br />

Ollier, William 74<br />

Olsen, Inger Marie L. O1.1<br />

Olsson, Eric 230<br />

Olvera, Rene O9.8, ECI 13, 76<br />

Omori, Mayu 66<br />

Omri, Teltsh 206<br />

O'Neill, A 245<br />

O'Neill, Francis ECI 3, 179, 223<br />

O'Neill, Raymond R. 26<br />

Ono, Yutaka 176<br />

Ontiveros, Alfonso 141<br />

Oostra, Ben 147<br />

Oph<strong>of</strong>f, Roel O11.2, 81, 227,<br />

260, 265<br />

Oppelaar, Hugo O13.3<br />

Orro, Alessandro O12.4, 63, 233<br />

Osby, Urban O10.1, O10.5, 230<br />

Oscar-Berman, 182<br />

Marlene<br />

Osimo, Emanuele O12.4, 238<br />

Osnat, Karni 206<br />

Ospina, Luz O13.1<br />

Otowa, Takeshi ECI 11, 29, 30, 35,<br />

37, 177<br />

Otte, David-Marian O12.2<br />

O'Tuathaigh, Colm S3.3<br />

Owen, Michael S3.1, S5.2, S9.1,<br />

O1.3, O4.3, O4.4,<br />

O5.3, O7.4,<br />

O10.3, O11.1,<br />

O12.1, O12.5,<br />

O17.1, 1, 19, 102,<br />

143, 210, 215,<br />

245, 247<br />

Ozaki, Norio 176, 209, 215,<br />

220, 221, 222,<br />

231, 274<br />

Ozkaragoz, Tulin 181<br />

Ozomaro, Uzoezi O3.3<br />

Ozturk, Mine 117<br />

Paddock, Silvia 142<br />

Pae, Chi-Un 185<br />

Palácios, Selma ECI 8<br />

Palejev, Dean 130<br />

Palmason, Haukur O4.1<br />

Panariello, Fabio 244<br />

Panganiban, Corrie 157<br />

Papadimitriou, G 245<br />

Papageorgis, Panos O14.4<br />

Papapetrou, Danae O13.1<br />

Papassotiropoulos,<br />

Andreas<br />

255, 256<br />

Pare, Guillaume O10.2<br />

Paredes, Ursula 170<br />

Pariante, Carmine O15.2<br />

Park, Hyung-Seok 151<br />

Park, HyunJoo 133<br />

Park, Jeong-Su 70, 134<br />

Park, Jin Kyung 254<br />

Park, Joanne 4, 10<br />

Park, Sohee S12.3<br />

Park, Sunju 70, 134<br />

Park, Young-Min 195<br />

Parker, Gordon P2.1<br />

Parla, Jennifer 208<br />

Partonen, Timo O1.2, O1.5, O7.3<br />

Paschall, Justin 157<br />

Passmore, Peter O17.1<br />

Pásztor, Péter I. 234<br />

Patel, Darshana 175<br />

Patel, Sagar S6.3, O10.4<br />

Patterson, Diana O16.2<br />

Paul, Torsten 148, 160<br />

Pauls, David S10.1, S10.3,<br />

S10.4, S10.5,<br />

O4.2, 61<br />

Paunio, Tiina S12.2, O1.5, O7.3<br />

Payo-Cano, Jose L O2.5<br />

Payton, Antony 74<br />

Pedersen, Carsten<br />

Bøcker<br />

O8.1<br />

Pedrosa, Erika O5.4<br />

Pedroso, Inti O11.5<br />

Peeters, Hilde O13.2<br />

Peirce, Timothy O12.1<br />

Peltonen, Leena O3.4, ECI 2, 114,<br />

189<br />

Pendleton, Neil 74<br />

Penninx, Brenda S4.1, S4.5, 147<br />

Pereira, Alexandre<br />

Costa<br />

276, 278<br />

Pereira, Ana-Caterina ECI 17<br />

Perez, Jorge 190<br />

Perez-Rodriguez,<br />

Mercedes<br />

O1.7<br />

Pericak-Vance,<br />

Margaret<br />

O3.3, 17.2<br />

Perkins, Diana O. 229<br />

Perlis, Roy S1.5<br />

Perroud, Nader S1.1<br />

Perry, William 146<br />

Peters, Eric S1.2<br />

Petryshen, Tracey S15.5<br />

Pfister, Hildegard O15.1, 100, 101<br />

PGC Schizophrenia<br />

GWAS Collaboration,<br />

P4.3<br />

The<br />

Phenome Group, The<br />

Bipolar Disorder<br />

S14.1<br />

Philibert, Rob 30<br />

Piccardi, Paola 162<br />

Picchioni, Marco 83<br />

Pickard, Ben O5.2, 216, 237<br />

Pickles, Andrew 74<br />

Pidsley, Ruth O9.5<br />

Pirkola, Sami O1.5, O3.4<br />

Pirlo, Katrina O1.3, O7.4,<br />

O11.1, 143<br />

Pirooznia, Mehidi S14.1, 154, 257,<br />

263<br />

Pitch, Ashley 171<br />

Pitts, Steve O16.4<br />

Pjetri, Eneda O13.3<br />

Placentino, Anna S1.1, O2.5, 190<br />

Plomin, Robert O14.5, 77<br />

Pluzhnikov, Anna S10.3<br />

Poelmans, Geert O4.2<br />

Pogue-Geile, Michael 76<br />

Polanczyk, Guilherme 13<br />

Polgár, Patrícia 234<br />

Porjesz, Bernice S16.1, S16.6,<br />

S16.7, ECI 18, 90<br />

Porkka-Heiskanen,<br />

Tarja<br />

O1.5<br />

Porteous, David S15.2, O5.2, 51,<br />

74, 208<br />

Potash, James S2.2, S14.1,<br />

S14.3, 99, 150,<br />

154, 157, 165,<br />

257, 263<br />

Potkin, Steve O6.2, 63, 132,<br />

191, 238, 264<br />

Pouta, Anneli ECI 2<br />

Powell, John 54, 55, 83, 105<br />

Prado, Carolina 198<br />

Prata, Diana 236<br />

Pratt, Amanda 23<br />

Pratt, Judy 45<br />

Pregelj, Peter 49<br />

Preisig, Martin 143<br />

Prescott, Carol O16.2, O16.3, 279


Price Foundation<br />

Collaborative Group,<br />

The<br />

S13.1, S13.4,<br />

S13.5<br />

Priebe, Lutz O9.2, 119<br />

Prince, Martin 170<br />

Priotsi, P S15.1<br />

Propping, Peter O12.2<br />

Prossin, Alan 89, 161<br />

Psychiatric GWAS P4.2, P4.3, P4.4,<br />

Consortium, The P4.5<br />

Pugh, Elizabeth S16.1, S16.3,<br />

S16.4<br />

Pulver, AE 245<br />

Purcell, Shaun S5.5, S10.3,<br />

S10.4, O2.1,<br />

O12.3, ECI 17,<br />

118, 131, 159<br />

Qi, Hongshi 92<br />

Qian, Yudong 127<br />

Qiu, Chuan 204<br />

Quackenbush, Corey 23, 229<br />

Quesenberry, Charles S1.3<br />

Quevedo, João 75<br />

Quinn, Emma S9.1, O12.1, 48<br />

Racz, Ildiko 120<br />

Radant, Allen 249<br />

Ramakers, Geert O13.3<br />

Ramakrishnan, Kamna 208<br />

Ramos-Quiroga, Josep<br />

Antoni<br />

3, 7, 8, 9<br />

Rangarajan, Sumathy O10.2<br />

Rangaswamy,<br />

ECI 18, 90<br />

Madhavi<br />

Rasmussen, Jerod 132<br />

Raventos, Henriette 141<br />

Rebolini, Danea 208<br />

Redman, Margot S2.3<br />

Reif, Andreas O9.6, 3, 5, 7, 8, 9,<br />

30<br />

Reinalda, Megan S1.2<br />

Remington, Gary 191<br />

Remschmidt, Helmut 174<br />

Renner, Tobias O4.1<br />

Ressler, Kerry 79<br />

Réthelyi, János M. 234<br />

Reus, Victor S1.3<br />

Rex, Katharine 56<br />

Ribases, Marta 7, 8, 9<br />

Ribble, R 245<br />

Ribeiro, Sidarta 75<br />

Rice, John S16.1, S16.3,<br />

S16.5, ECI 7, 115,<br />

138, 143, 157<br />

Richmond, Todd 135<br />

Richter, Margaret S10.5<br />

Richter, Anne 197<br />

Richter, Peggy M. S11.5<br />

Rietschel, J. O2.5<br />

Rietschel, Marcella S1.1, S14.3,<br />

S14.4, O5.5, O8.2,<br />

O9.2, O11.6, 30,<br />

86, 119, 143, 148,<br />

160, 190, 197<br />

Rijpkema, Mark O8.3<br />

Rijsdijk, Fruhling 11, 83, 144<br />

Riley, Brien O16.2, ECI 3, 42,<br />

179, 223, 245, 279<br />

Rimol, Lars 64<br />

Ripatti, Samuli O3.4, 114<br />

Ripke, Stephan P4.1, S1.4, O3.2,<br />

262<br />

Ritchie, Terry 65<br />

Ritter, Benjamin P. 71<br />

Rivera, Margarita 28, 143<br />

Rizzu, Patrizia S8.1<br />

Roberts, Rachel O4.4, 19<br />

Robertson, Ian 6<br />

Robinson, Delbert S9.3<br />

Robinson, M. ECI 17<br />

Rochberg, Nanette 138<br />

Roche, Siobhan 168<br />

Roddey, Cooper 64<br />

Roeske, Darina O3.2<br />

Roeyers, Herbert 11<br />

Rogdaki, Maria 92<br />

Roig, Barbara O6.4<br />

Roman, Tatiana 13, 292<br />

Romanos, Marcel O4.1, O9.6, 5<br />

Romer Ek, Inger O10.1, O10.5<br />

Rommelse, Nanda 11<br />

Rose, Emma S9.1<br />

Rose, Jed 183, 273<br />

Rose, Richard O14.1<br />

Rosenfeld, Jill O6.6<br />

Rosengren, Annika O10.2<br />

Ross, Jessica S10.6<br />

Rothenberger, Aribert 11<br />

Röthlisberger, Benno 255<br />

Rouleau, Guy S10.3<br />

Roy, Alec O16.1<br />

Royall, Josh 232<br />

Ruberto, Gaia 153<br />

Rubinsztein, David O17.1<br />

Ruck, Christian O3.1<br />

Rucker, James 164<br />

Rückert, IM 114<br />

Ruderfer, Douglas 131, 159<br />

Rugulies, Reinar 39<br />

Ruiz, Aida 54, 55<br />

Ruiz-Linares, Andres S10.3<br />

Rujescu, Dan S3.1, S5.2, S9.1,<br />

O12.1, 186<br />

Rupprecht, Rainer S1.4<br />

Rush, A. John S1.4<br />

Russell, Elen O10.3<br />

Saarikoski, Sirkku 189<br />

Sabatti, Chiara S10.3<br />

Sabunciyan, Sarven S2.2<br />

Saccone, Nancy S16.1, S16.3,<br />

S16.5, ECI 7<br />

Saccone, Scott S16.3, 138<br />

Saetre, Peter 251<br />

Sagvolden, Terje 18<br />

Sahoo, Trilochan O6.6<br />

Saito, Toshikazu 94<br />

Sakai, Yoshie 177<br />

Sakata, Shinichi 268, 269, 272<br />

Sakurai, Takeshi O12.5, O13.1<br />

Salah, Hala 243<br />

Salgado, Carlos A. I 14<br />

Salisbury, Dean 80<br />

Salomon, Daniel S6.3<br />

Salvi, Erika O12.4, 63, 233<br />

Samaan, Zainab O10.2<br />

Sampaio, Aline ECI 8<br />

Sánchez-Mora,<br />

Cristina<br />

8, 9<br />

Sanders, Alan R. 115, 245<br />

Sano, Akira 121, 123, 124<br />

Santangelo, Susan L. S12.3, 240<br />

Santarelli, Danielle S7.2<br />

Sarah, Murray 157<br />

Sasaki, Tsukasa ECI 11, 29, 30, 35,<br />

37, 177<br />

Saunders, Erika 89<br />

Saus, Ester 40<br />

Savla, Gauri 242<br />

Saykin, Andrew 63<br />

Scazufca, Marcia 104, 113<br />

Schaefer, Catherine S1.3<br />

Schäfer, Helmut O9.6, 5<br />

Schafer, Martin 186<br />

Schalkwyk, Leonard C O2.5, O14.5<br />

Schalling, Martin O1.2, O3.1, O7.3,<br />

O10.1, O10.5, 230<br />

Scharf, Jeremiah S10.1, S10.3<br />

Scheftner, William 157<br />

Schilling, Karl O12.2<br />

Schilling, Paul 207<br />

Schizophrenia<br />

Genomics Ireland<br />

Consortium, The<br />

O8.5<br />

Schlegel, Kristina 156<br />

Schloegelh<strong>of</strong>er,<br />

Monika<br />

152<br />

Schmouth, Jean-<br />

Francois<br />

S3.5, 290<br />

Schnack, Hugo G. 85<br />

Sch<strong>of</strong>ield, Peter P2.1<br />

Scholte, Evert 31<br />

Schork, Andrew 169<br />

Schork, Nicholas S6.3, S13.1,<br />

S13.2, S13.5,<br />

O10.4, 64, 88,<br />

157, 169, 249,<br />

259, 264<br />

Schork, Ryan 259<br />

Schosser, Alexandra O1.3, O11.1, 143,<br />

152<br />

Schreiber, Stefan 119<br />

Schuckit, Marc S16.6, S16.7<br />

Schulze, Katja 83, 236<br />

Schulze, Thomas S14.3, S14.4,<br />

O2.2, O5.5,<br />

O11.6, 30, 148,<br />

157, 160, 165, 190<br />

Schumacher, Johannes 30<br />

Schumann, Gunter P1.2, O14.1, ECI<br />

2, 281<br />

Schwab, Sibylle<br />

Gabriele<br />

57, 192, 245<br />

Scolnick, Ed ECI 17<br />

Scott, Adrian Phillip 192<br />

Scott, Ashley A. S13.5, 88<br />

Scott, Sarah 4<br />

Seaton-Smith,<br />

Kimberly<br />

275<br />

Sebat, Jonathan P3.2, O13.5<br />

Segall, Judith O6.2


Segall, Samantha 275<br />

Segurado, Ricardo 12<br />

Seidman, Larry 249<br />

Seifuddin, Fayaz S14.1, 154, 257,<br />

263<br />

Seitz, Christiane O4.1<br />

Sens-Espel, Roser 140<br />

Sergeant, Joseph 11<br />

Serretti, Alessandro 44, 140, 185, 186,<br />

187<br />

Service, Susan S10.3<br />

Setó, Sonia 248<br />

Severin, Emilia 34<br />

Severino, Giovanni 162<br />

Severinsen, Jacob 212<br />

SGENE Consortium,<br />

The<br />

S15.1<br />

Sha, Li 216<br />

Shaffer, Lisa O6.6<br />

Shah, Abhishek O5.4<br />

Shah, N Jon 86<br />

Shaheen, S-M S10.5<br />

Shaikh, Madiha 83, 236<br />

Shaikh, Sajid 202<br />

Sham, Pak 54, 55<br />

Sharad, Shashwat 112<br />

Sharma, Rajiv S2.4<br />

Sharp, Frank R. ECI 4<br />

Sharp, Sally 59, 156<br />

Shaw, Duncan 203<br />

Shaw, Marian S13.5<br />

Shekhtman, Tatyana 56, 201<br />

Shen, Elaine 232<br />

Shen, Lei O2.4<br />

Shen, Ling S1.3<br />

Shen, Pei-Hong O16.1<br />

Shi, Jianxin 115, 245<br />

Shi, Yongyong O6.1, O7.1, 38<br />

Shianna, Kevin O11.3, 254<br />

Shimada, Takafumi 29, 35, 37, 177<br />

Shimo, Hirochika 121, 123, 124<br />

Shin, Hee Jung 133<br />

Shinkai, Takahiro 268, 269, 271, 272<br />

Shiokawa, Nari 123, 124<br />

Shirazi, Elham 97<br />

Shrestha, Payas 240<br />

Shtir, Corina 15, 17<br />

Shyn, Stanley S1.2<br />

Sibony, David 191<br />

Sidhu, Shrada 111<br />

Siever, Larry O1.7, 249<br />

Sigmund, Jessica<br />

Charlotte<br />

255, 256<br />

Silberberg, Gilad 126<br />

Silverman, Jeremy M. 71, 115, 245, 249<br />

Simmons, Andrew O17.4<br />

Simon, Mariella O5.1<br />

Simonson, Matthew 246<br />

Simpson, Elizabeth M. S3.5<br />

Simpson, George 239<br />

Sims, Rebecca O17.1<br />

Singh, Karun K. S15.5<br />

Singhapakdi, Kanya 67<br />

Singleton, Andrew O5.5<br />

Sinke, Richard 81<br />

Sinkus, Algimantas 109<br />

Sinkute, Gintare 109<br />

Sironi, M. O12.4<br />

Sitnikova, Tatiana S12.3, 240<br />

Sitskoorn, Margriet 81<br />

Sjöholm, Louise O1.2, O7.3, O9.1<br />

Sklar, Pamela O2.1, O12.3, ECI<br />

17, 118, 131, 159,<br />

224, 225<br />

Skup, Martha 163<br />

Slaats-Willemse,<br />

Dorine<br />

2<br />

Slager, Susan S1.2<br />

Slifer, Michael O17.2<br />

Sliwa, Karen O10.2<br />

Smalley, Susan 15, 17<br />

Smith, Erin 157, 169<br />

Smith, Moyra O5.1<br />

Smith, Rebecca O14.5, 190<br />

Smith, Stevens ECI 7<br />

Smolka, Michael 197<br />

Smoller, Jordan P4.7, S14.6, 118<br />

Snyder, Michael 130<br />

Soares, Dinesh O5.2, 208<br />

Soda, Takahiro S15.5<br />

Sonuga-Barke,<br />

Edmund<br />

11<br />

Soon, Siew Choo ECI 1<br />

Sora, Ichiro 274<br />

Sørensen, Karina<br />

Meden<br />

O8.1<br />

Soria, Virginia 40<br />

Soronen, Pia O1.5, O7.3<br />

Souery, Daniel S1.1, O2.5, 140,<br />

190<br />

Spanagel, Rainer 197<br />

Specht, Michael O3.2<br />

Spector, TD 114<br />

Spirito, Anthony 62<br />

Sprock, Joyce 207<br />

Squassina, Alessio 162<br />

Strik Lievers, L. O12.4<br />

St. Clair, David S15.3, O5.2, O5.3,<br />

O6.1, O10.3, 237<br />

Stanford, Clare 59<br />

Starr, John O5.2, 74, 208<br />

State, Matthew S10.3<br />

Steele, CJM S14.4, ECI 19<br />

Steer, C. 32<br />

Stein, Dan S10.2<br />

Steinhausen, Hans-<br />

Christoph<br />

11<br />

Stergiakouli,<br />

Evangelia<br />

1<br />

Stern, Hal 264<br />

Stewart, S. Evelyn S10.1, S10.2,<br />

S10.4, S10.5<br />

Steyaert, Jean O13.2, 31<br />

Stice, Eric 182<br />

Stiedl, Oliver O13.3<br />

Stitzel, Jerry 69<br />

Stöcker, Tony 86<br />

Stojanovic, Mirjana 152<br />

Stokowski, Renee O16.4<br />

Stolerman, Ian 24<br />

Stone, William 249<br />

Straarup, Steen E. O1.1<br />

Straub, Richard O6.3, 219, 252<br />

Strauss, John 28<br />

Strengman, Eric 81, 234<br />

Strohmaier, Jana S14.4, 160<br />

Stroup, T. Scott 229<br />

Sturgess, Jessica E. S11.5<br />

Subedi, Janardan S12.3, 240<br />

Sudi, Jyotsna ECI 12<br />

Sudic, Diana 230<br />

Sugawara, Hiroko O1.4<br />

Sugaya, Nagisa 29<br />

Sugiyama, Toshiro 37<br />

Suhl Kristensen, Ann 27<br />

Sullivan, Patrick P4.2, S4.1, S4.3,<br />

O2.6, O12.3, 23,<br />

25, 147, 224, 225,<br />

229<br />

Sun, Cuie O3.5<br />

Sun, Jingchun 42, 266, 267<br />

Sung, In-Kyung 151<br />

Sur, Mriganka S3.6<br />

Surakka, Ida O3.4<br />

Suvisaari, Jaana O3.4<br />

Svenningsson, Per 92<br />

Swaab, Hanna O13.3, O13.4<br />

Swan, Gary O16.4<br />

Swanson, Beryl 232<br />

Swerdlow, Neal 207, 249<br />

Syed, Sharifah O4.4, 19<br />

Székely, Judit 50<br />

Szeszko, Philip O8.6<br />

Taillefer, Stephanie S10.5<br />

Takaki, Manabu 274<br />

Takeda, Masatoshi 215<br />

Talati, Ardesheer ECI 6<br />

Talib, Leda 73<br />

Tamang, Swarneem 240<br />

Tamás, Zsuzsa 50<br />

Tamashiro, Kellie 99<br />

Tang, Pek Yee ECI 1, 217, 226<br />

Tang, Yilang 79<br />

Tanii, Hisashi ECI 11, 29<br />

Tassa, Carlos S15.5<br />

Tatarelli, Roberto 153<br />

Tatro, Erick S7.3, 213<br />

Tatsumi, Masahiko 66<br />

Tavian, Daniela 188<br />

Taylor, Grantley 80<br />

Taylor, Stephan S11.1<br />

Tee, Shiau Foon ECI 1, 217, 226<br />

Tenesa, Albert 74<br />

Tenore, Christopher O5.4<br />

Teraishi, Toshiya 66<br />

Terwisscha van<br />

Scheltinga, Afke F.<br />

227<br />

Tesli, Martin 145<br />

Tewari, Deepshikha 78<br />

Tewhey, Ryan S. S13.5<br />

Thapar, Anita O4.3, O4.4, 1, 19,<br />

59<br />

Thiagalingam, Sam O14.4<br />

Thiselton, Dawn 179<br />

Thode, Kirstin O9.8<br />

Thomsen, Jane 39


Thomson, Pippa 51, 208<br />

Thrall, Jenny O11.3, 254<br />

Thrush, Carol 106<br />

Tiemeier, Henning O7.2<br />

Tieran, V. 233<br />

Tikhomorov, Anna S10.3<br />

Tischfield, Jay S10.3, S16.1,<br />

S16.2, S16.3,<br />

S16.4, S16.7<br />

Tiwari, Arun K. S11.5<br />

Tobar, Salwa 243<br />

Tobin, Claire 6<br />

Tochigi, Mamoru 29<br />

Todd, Richard 15, 17<br />

Todorov, Alexandre 15, 17, 277<br />

Tokunaga, Katsushi 29, 35<br />

Tomita, Yasuyuki 215<br />

Tomiyasu, Akiyuki 121, 123, 124<br />

Tooney, Paul S7.2<br />

Topol, Eric J. S13.5, 64<br />

Torkamani, Ali S13.5<br />

Torri, Federica O12.4, 132, 233,<br />

Toulopoulou,<br />

Timothea<br />

Tourette GWAS<br />

Consortium<br />

238<br />

83<br />

S8.3<br />

Toye, Caroline 10<br />

Traskman-Bendz, Lil O10.1, O10.5<br />

Treutlein, Jens 86, 197<br />

Troakes, Claire 72<br />

Tropea, Daniela S3.6<br />

Tsai, Li-Huei S15.5<br />

Tsai, Shih-Feng 205<br />

Tsuang, Debby 249<br />

Tsuang, Ming S6.3, 213, 249<br />

Tsutsumi, Atsushi 82, 193<br />

Turck, Chris S6.1, O12.2<br />

Turetsky, Bruce 249<br />

Turner, Jessica O6.2, 63, 132,<br />

238, 264<br />

Turner, Norris 199<br />

Twamley, Elizabeth 146, 242<br />

Twyman, Roy 199<br />

Uchimura, Naohiko 274<br />

Uchiyama, Hir<strong>of</strong>umi 66<br />

Ueda, Junko O1.4, 94<br />

Uenishi, Hiroyuki 82<br />

Ueno, Shu-ichi 121, 123, 124<br />

Uher, Rudolf S1.1, O1.3, O2.5,<br />

O7.4, O11.1, 102,<br />

143, 190<br />

Uhl, George 183, 273<br />

Uhr, Manfred S1.4, O3.2, O15.1<br />

Ujike, Hiroshi 274<br />

Ukai, Wataru 94<br />

Umekage, Tadashi 29, 37, 177<br />

Unschuld, Paul G. O3.2<br />

Urban, Alexander<br />

Eckehart<br />

130<br />

Urretavizcaya, Mikel 40<br />

Utge, Siddheshwar O1.5<br />

Utsunomiya, Kensuke 268, 269, 272<br />

Vacic, Vladimir O13.5<br />

Vakkalanka, Krishna O6.3, 252<br />

Valero, Joaquín O6.4, 40, 248<br />

Vallada, Homero ECI 8, 104, 113,<br />

276, 278<br />

Vallender, Eric 68<br />

Valvassori, Samira 75<br />

van Baal, G. Caroline<br />

M.<br />

85<br />

van Beck, Margaret O5.2<br />

van Beijesterveldt,<br />

CEM<br />

S8.1<br />

van Berckelaer-Onnes,<br />

Ina<br />

31<br />

van den Oord, Edwin<br />

JCG.<br />

O2.6, O3.5, ECI 3,<br />

23, 42, 223, 229<br />

van der Does, A.J.W. 60<br />

van Duijn, Cornelia O7.2, 114, 147<br />

van Dyck, Richard 147<br />

van Engeland, Herman O13.3, O13.4<br />

van Haren, Neeltje<br />

E.M.<br />

227<br />

van Lith, Hein O13.3<br />

van Rooij, F. 114<br />

van 't Slot, Ruben 81<br />

VandeBerg, John S12.3, 240<br />

Vasquez, Alejandro 11<br />

Vassos, Evangelos 153<br />

Vawter, Marquis S6.5, O7.3, 238<br />

Vederman, Aaron 89, 161<br />

Veijola, Juha ECI 2<br />

Velders, Fleur O7.2<br />

Veltman, Joris O8.3<br />

Verge, Begoña 248<br />

Verhulst, Frank O7.2<br />

Vetró, Ágnes 50, 202<br />

Vetuz, Glen 10<br />

Victor, Marcelo M. 14<br />

Videtic, Alja 49<br />

Vietan, Cassandra 275<br />

Vilella, Elisabet O6.4, 40, 248<br />

Villa, Catalina O5.4<br />

Vincent, John ECI 15, 158, 173<br />

Vine, Anna ECI 17<br />

Visscher, Peter O5.2, 74, 208<br />

Vladimirov, Vladimir<br />

I.<br />

O2.6, 179<br />

Vogler, Christian 255, 256<br />

von Plessen, Kerstin 8<br />

Vorstman, Jacob O13.4<br />

Vukcevic, Damjan O10.3<br />

Waddington, John S3.3<br />

Wade, Rachel 43<br />

Wagner, Joseph 84<br />

Wagner, Michael S10.4, O3.3<br />

Waite, Roger 182<br />

Walitza, Susanne O4.1, O11.6<br />

Wallace, Douglas O5.1<br />

Walsh, Dermot O16.2, ECI 3, 179,<br />

223, 245<br />

Walshe, Muriel 83, 236<br />

Walss-Bass, Consuelo O9.8<br />

Walters, James S3.1, S9.1<br />

Walther, Donna 183<br />

Wand, Gary 99<br />

Wang, Dai 199<br />

Wang, Jen S16.1, S16.5<br />

Wang, Kai S13.1, S13.2, 32<br />

Wang, Ti O6.1<br />

Wang, Wei 204<br />

Wang, Xiaobin O13.1<br />

Wang, Ying-Chieh O8.4<br />

Warburton, Peter E. 135<br />

Warnke, Andreas O9.6, 5, 174<br />

Watchorn, Amy 6<br />

Watson, Annie 243<br />

Watson, James D. 208<br />

Webb, Bradley O3.5, ECI 3, 42,<br />

223<br />

Webh<strong>of</strong>er, Christian S6.1<br />

Webster, Maree S2.2<br />

Wegener, Gregers 92, 214<br />

Wei, Jun 203<br />

Weinberger, Daniel O6.3, 219, 252<br />

Weiner, Michael 63<br />

Weinstein, Shauna O1.7<br />

Weissflog, Lena O9.6, 5<br />

Weissman, Myrna ECI 6<br />

Weissman, Sherman 130<br />

Wellcome Trust Case<br />

Control Consortium,<br />

WTCCC<br />

O5.3, O10.3<br />

Weller, Andrew E. 41<br />

Wells, Tony T. 58<br />

Wen, Chun-Chiang 87, 205, 228, 235<br />

Wendland, Jens S14.4, O11.6<br />

Werge, Thomas O8.1, 212<br />

Wessel, Jennifer O16.4<br />

Whalley, Lawrence 74<br />

Wheelan, Sarah 150<br />

White, Angela O5.2<br />

Wickramaratne, Priya ECI 6<br />

Widyawati, Ika 57<br />

Wiedemann, Klaus 197<br />

Wienker, Thomas F. 120<br />

Wigg, Karen O14.3, 171<br />

Wilde, Alex P2.4<br />

Wildenauer, Dieter<br />

Bernd<br />

57, 192, 245<br />

Wilhelm, Kay P2.1<br />

Wilhelmsen, Kirk 225, 275<br />

Willcutt, Erik 103<br />

Willemsen, Gonneke S4.5, 147<br />

Williams, Ben O9.7<br />

Williams, Hywel O12.1, 1, 210, 215<br />

Williams, Ian 83, 236<br />

Williams, Julie O17.1<br />

Williams, Nigel O4.4, O12.1, 19,<br />

245, 247<br />

Williams-Blangero,<br />

Sarah<br />

S12.3, 240<br />

Williamson, Douglas O9.8<br />

Wilson, Nathan S3.6<br />

Winchester, Catherine 45<br />

Winkler, Anderson S12.4<br />

Witasp, Anna 92<br />

Witt, Stephanie 119, 197<br />

Wittchen, Hans-Ulrich 100<br />

Wodarz, Norbert 197<br />

Woldbye, David 27, 30<br />

Wong, Chloe O9.5, O9.7<br />

Wood, Alexis 11


Wood, Joel 243<br />

Wormley, B 245<br />

Woudstra, Saskia S4.1<br />

Wray, Naomi S16.2, 147<br />

Wright, Fred A. 25<br />

Wrobel, Bozena 239<br />

Wu, Joseph O7.3<br />

Wu, Xiaodong 199<br />

Xia, Xuhua 178<br />

Xie, Changchun O10.2<br />

Xu, Xiaohui 170<br />

Xuei, Xiaoling ECI 14<br />

Yamada, Kenji 268, 269, 272<br />

Yamada, Kiy<strong>of</strong>umi 231<br />

Yamada, Mitsuhiko 274<br />

Yamada, Shinnosuke 209, 231<br />

Yamamoto, Maki 58<br />

Yamamoto, Noriko 66<br />

Yamanouchi, Yoshio 215<br />

Yan, Han 204<br />

Yan, Jia S16.7<br />

Yang, Ueng-Cheng 87, 205, 235<br />

Yang, Wei-Chih 205<br />

Yang, Xiaoju 99<br />

Yankova, Elena 277<br />

Yaqubi, Sahab O14.4<br />

Yassin, Amal 243<br />

Yasui-Furukori, Norio 176<br />

Yehyawi, Nabeel S6.3<br />

Yilmaz, Öznur O12.2<br />

Yilmaz, Zeynep 20<br />

Yim, Seon-Jin 250<br />

Yoav, Kohn 206<br />

Yokoyama, Jennifer S1.2<br />

Yolken, Robert S2.2<br />

Yoneda, Hiroshi 82, 193<br />

Yoon, Seungtai O13.5<br />

Yoshikawa, Takeo O2.3<br />

Yoshimi, Akira 209, 231<br />

Yoshimura, Reiji 215<br />

Young, Allan O5.3, O10.3<br />

Young, James O12.5<br />

Younkin, Steven O17.1<br />

Yu, Dongmei S10.3, 240<br />

Yu, Lan 135<br />

Yuan, Nicole P O16.1<br />

Yuan, Qiaoping O1.7, O16.1<br />

Yusuf, Salim O10.2<br />

Zaharieva, Irina O4.4, 19<br />

Zai, Clement C. S11.5, 191, 244<br />

Zaitlen, Noah 157<br />

Zammit, Stanley O12.1<br />

Zandi, Peter S14.1, S14.3,<br />

O11.4, 154, 157,<br />

257, 263<br />

Zavos, Helena M.S. 144<br />

Zeng, Hongkui 232<br />

Zeng, Zhen O6.1<br />

Zeni, Cristian 13<br />

Zerres, Klaus 86<br />

Zhang, Dandan S2.3, S2.5, S5.3,<br />

98, 127, 180<br />

Zhang, Haitao S13.1<br />

Zhang, Jian-Ping S9.3<br />

Zhang, Michelle O10.2<br />

Zhang, Peng 157<br />

Zhang, Rui 204<br />

Zhang, Yaoyang S6.1<br />

Zhang, Ying 130<br />

Zhang, Zhao O6.1<br />

Zhang-James, Yanli S8.1, S8.3<br />

Zhao, Hongyu ECI 16<br />

Zhao, Qian O6.1, O7.1<br />

Zhao, XinZhi O6.1<br />

Zhao, Zhongming O2.6, O3.5, ECI 3,<br />

42, 223, 266, 267<br />

Zhong, Nan-Nan 204<br />

Zhou, Kaixin 11<br />

Zhou, Qiang O13.1<br />

Zhou, Zhifeng O1.7<br />

Ziaolhagh, Ali 158<br />

Zimmer, Andreas O12.2, 120<br />

Zimmermann, Petra O15.1, 100, 101<br />

Zimmermann, Ulrich<br />

S.<br />

197<br />

Zitman, Frans 60, 147<br />

Zlojutro, Mark S12.5, 76<br />

Zoë, Prichard 178<br />

Zohar, Joseph 140<br />

Zollner, Sebastian 157<br />

Zubaid, Mohammed O10.2<br />

Zuchner, Stephan O3.3<br />

Zupanc, Tomaz 49<br />

Zwiers, Marcel 2

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!