16.12.2012 Views

Presentation Abstracts - XIXth World Congress of Psychiatric Genetics

Presentation Abstracts - XIXth World Congress of Psychiatric Genetics

Presentation Abstracts - XIXth World Congress of Psychiatric Genetics

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

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

EDUCATIONAL SESSION ABSTRACTS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2<br />

PLENARY ABSTRACTS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10<br />

SYMPOSIA ABSTRACTS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14<br />

INDIVIDUAL ORAL PRESENTATIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41<br />

POSTER SESSION I ABSTRACTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67<br />

GENOMICS (GWAS, SEQUENCING) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68<br />

POSTER SESSION II ABSTRACTS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106<br />

ELSI, COUNSELING, AND GENETIC TESTING . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107<br />

EPIGENOMICS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109<br />

PHENOTYPES & ENDOPHENOTYPES. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112<br />

POSTER SESSION III ABSTRACTS: SPECIAL SESSION ON SUBSTANCE ABUSE GENETICS . . . . . . . . . . . . . . . . . 144<br />

POSTER SESSION IV ABSTRACTS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168<br />

STATISTICAL GENETICS, GENETIC EPIDEMIOLOGY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169<br />

FUNCTIONAL GENOMICS & MODEL ORGANISMS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182<br />

OTHER. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186<br />

PHARMACOGENOMICS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196<br />

AUTHOR INDEX. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203<br />

LEGEND<br />

E-EDUCATIONAL SESSION<br />

P-PLENARY<br />

S-SYMPOSIA<br />

OPS-ORAL PRESENTATIONS<br />

PP-POSTER PRESENTATIONS<br />

ECIP - EARLY CAREER INVESTIGATOR’S PROGRAM (ORAL AND POSTER AWARD FINALISTS)<br />

1


EDUCATIONAL<br />

SESSION<br />

ABSTRACTS<br />

2


RECRUITMENT OF SUBJECTS: LESSONS LEARNED OVER<br />

25 YEARS<br />

L. Kassem*(1), D. Kazuba(1), C. Drain(2), C. Fisher(3), E.<br />

Murphy(1)<br />

1. NIMH/NIH 2. Washington University School <strong>of</strong> Medicine 3.<br />

Indiana University School <strong>of</strong> Medicine<br />

*kasseml@mail.nih.gov<br />

This educational session will discuss subject recruitment and<br />

retention for genetic studies. The correlation between the success <strong>of</strong> a<br />

study and the strength and skill <strong>of</strong> its recruitment team has been<br />

shown to be consistently high. Members <strong>of</strong> the panel have been<br />

involved in subject recruitment for 25 years, and their collective and<br />

diverse experience demonstrates that success requires a delicate<br />

balance between simplicity and complexity, learned techniques and<br />

art. This is particularly the case where studies are focused on diverse<br />

or culturally-sensitive and unique populations, and where the advent<br />

<strong>of</strong> novel advertising and screening approaches needs to be cognizant<br />

<strong>of</strong> current legal, ethical and other sensitivities. The panel will briefly<br />

review recruitment history, variables contributing to success and<br />

approaches to be avoided.<br />

The proposed panel <strong>of</strong> speakers includes:<br />

Caroline Drain, <strong>of</strong> Washington University School <strong>of</strong> Medicine, will<br />

discuss advertisement methods and report on the best and worst<br />

approaches. In this section, “spreading the word” will be addressed<br />

specifically the panel will review the preparation <strong>of</strong> advertisement<br />

material to be used in different forms <strong>of</strong> media (e.g. fliers,<br />

newspapers, use <strong>of</strong> websites and radio advertisements). We will<br />

present examples <strong>of</strong> successful as well as unsuccessful<br />

advertisements, and we will discuss the art <strong>of</strong> tailoring<br />

advertisements for different populations. Data demonstrating the<br />

success <strong>of</strong> different forms <strong>of</strong> advertising will be presented.<br />

Carrie Fisher, <strong>of</strong> Indiana University School <strong>of</strong> Medicine, will review<br />

efficient methods for obtaining informed consent. Here the panel will<br />

examine the meaning <strong>of</strong> “informed” and “consent”. The discussion<br />

will be aimed at identifying “subject friendly” methods for obtaining<br />

informed consent. For example, is it best to obtain consent in person,<br />

by mail or over the telephone? Data comparing the return rate <strong>of</strong><br />

consent forms from the different approaches will be presented.<br />

Diane Kazuba, <strong>of</strong> the National Institutes <strong>of</strong> Mental Health/NIH, will<br />

discuss the construction <strong>of</strong> screening questionnaires and the<br />

conducting <strong>of</strong> the screening interviews - the screening interview is<br />

our subject’s first interaction with us. In this section, the screening<br />

process, starting with the construction <strong>of</strong> the interview and ending<br />

with techniques for ensuring our subjects’ comfort with the process<br />

and willingness to cooperate, will be reviewed. Data reflecting the<br />

difference between the number <strong>of</strong> subjects screened and the number<br />

<strong>of</strong> subjects actually recruited will be presented a discussion on how to<br />

decrease that difference will follow.<br />

The introduction and discussion will be lead by Layla Kassem who<br />

will also review issues, and resultant approaches, arising in the<br />

recruitment <strong>of</strong> diverse (and culturally-sensitive and unique)<br />

populations. At the end <strong>of</strong> the session, it is hoped that participants<br />

will have a better understanding <strong>of</strong> effective and efficient<br />

advertisement methods, the construction <strong>of</strong> screening tools, informed<br />

consent, and the recruitment <strong>of</strong> diverse populations.<br />

3<br />

WHAT CAN WE LEARN FROM LONGITUDINAL STUDY<br />

DESIGN? EXPERIENCES AND STUDIES OF INDIVIDUALS<br />

AT GENETIC HIGH-RISK FOR BIPOLAR DISORDER<br />

EARLY CLINICAL STAGES OF BIPOLAR DISORDER:<br />

FINDINGS FROM A LONGITUDINAL STUDY OF<br />

GENETICALLY AT-RISK OFFSPRING<br />

A. Duffy*(1), J. Horrocks(2), U. Lewitzka(3), M. Alda(1), S.<br />

Doucette(1), P. Gr<strong>of</strong>(4)<br />

1. Dalhousie University 2. Guelph University 3. University Hospital<br />

Dresden 4. University <strong>of</strong> Toronto<br />

*anne.duffy@dal.ca<br />

Introduction: There is a relative lack <strong>of</strong> information regarding the<br />

early natural history <strong>of</strong> bipolar disorder, compared to our<br />

understanding <strong>of</strong> the course <strong>of</strong> established (full-blown) illness. Given<br />

the substantial genetic contribution to bipolar disorder, longitudinal<br />

study <strong>of</strong> the <strong>of</strong>fspring <strong>of</strong> affected parents represents an important<br />

research strategy to map the trajectory into illness and study<br />

intermediate phenotypes and causal pathways, while minimizing the<br />

effect <strong>of</strong> the burden <strong>of</strong> the illness.<br />

Methodology: Eligible probands were identified from participation<br />

in neurobiological studies which confirmed a diagnosis <strong>of</strong> BDI based<br />

on SADS-L research interviews by pairs <strong>of</strong> research<br />

psychiatrists. Blind consensus diagnostic reviews confirmed DSM-<br />

IV diagnosis using research interviews and reviewing all available<br />

clinical and treatment history. Lithium response (LiR) or nonresponse<br />

(LiNR) was determined in accordance with published<br />

research protocol. Agreeable probands and any <strong>of</strong> their affected first<br />

degree relatives (BDI, II, recurrent MDD) with children in the age<br />

range <strong>of</strong> 7-25 years were approached for the study. Families were<br />

excluded if the other biological parent had a lifetime history <strong>of</strong> a<br />

major psychiatric disorder. A comparison group <strong>of</strong> families were<br />

included in which both parents had no lifetime major psychiatric<br />

disorder based on SADS-L interviews or ongoing acute or chronic<br />

major medical illness.All assenting/consenting children <strong>of</strong> eligible<br />

families and a parent were interviewed by a research psychiatrist<br />

using KSADS-PL format, blind to family affiliation and study<br />

group. Lifetime diagnoses were made using best-estimate procedures<br />

as described above.<br />

Results: This analysis includes 108 high-risk (HR) families and 55<br />

low-risk (C) families with 216 and 87 <strong>of</strong>fspring, respectively. HR<br />

<strong>of</strong>fspring had higher rates <strong>of</strong> lifetime psychopathology compared to C<br />

<strong>of</strong>fspring specifically, 69.9% vs 19.5% Axis I disorders, 39.3% vs<br />

3.4% Depressive Disorders and 14.9% vs 1.1% BD, respectively.<br />

In separate analyses, the age-corrected morbid risk <strong>of</strong> mood disorder,<br />

given a prior history <strong>of</strong> anxiety disorder was 85% compared to 40%<br />

in those without a prior history <strong>of</strong> an anxiety disorder (hazard <strong>of</strong> 2.6<br />

CI=1.59-4.25). Using Cox proportional hazards models with timevarying<br />

covariates, the risk <strong>of</strong> MDD was multiplied by a factor <strong>of</strong><br />

1.96 having experienced SUD (p=.039, 95% CI=1.03, 3.72). The risk<br />

<strong>of</strong> SUD was multiplied by a factor <strong>of</strong> 2.04 having experienced MDD<br />

(p=.052, 95% CI=0.99, 4.18). The risk <strong>of</strong> BD was multiplied by a<br />

factor <strong>of</strong> 2.66 once subjects had experienced SUD (p=.010, 95%<br />

CI=1.26, 5.61). The temporal sequence <strong>of</strong> psychopathology in those<br />

<strong>of</strong>fspring who met lifetime criteria for BD I or II disorder followed a<br />

predictable pattern (see figure 1).<br />

Conclusions: A developmental approach appears important to<br />

refining the diagnosis <strong>of</strong> BD in children and youth at genetic<br />

risk. Data from the present high-risk longitudinal study suggests<br />

evidence <strong>of</strong> clinical staging. This has implications for both<br />

intervention/prevention studies and studies <strong>of</strong> genetically sensitive<br />

intermediate pathways.<br />

H.C. PRECHTER LONGITUDINAL STUDY OF BIPOLAR


DISORDER: BASELINE DESCRIPTIVE ANALYSIS OF<br />

INDIVIDUALS WITH AND WITHOUT BIPOLAR DISORDER<br />

M. McInnis*<br />

University <strong>of</strong> Michigan<br />

*mmcinnis@umich.edu<br />

Introduction: The HC Prechter Longitudinal Study <strong>of</strong> Bipolar<br />

Disorder is an open cohort study designed to gather detailed clinical<br />

and biological data for research on the course and outcome <strong>of</strong> bipolar<br />

individuals.<br />

Methodology: Diagnoses are made with the DIGS. Current<br />

symptoms <strong>of</strong> depression and mania are assessed using the Hamilton<br />

Depression Rating Scale and the Young Mania Rating Scale.<br />

Personality parameters are estimated by the NEO-PI. Self-report<br />

instruments are used to capture histories <strong>of</strong> social and family support,<br />

life events and history <strong>of</strong> abuse as well as sleep parameters and<br />

patterns <strong>of</strong> circadian rhythms. Neuropsychological measures<br />

determine domains <strong>of</strong> functioning, including attention, memory, and<br />

executive functioning. Biological materials including DNA, salivary<br />

cortisol, and plasma for biomarkers are collected. The central<br />

hypothesis is that response and illness patterns will be predicted from<br />

the clinical, psychological, environmental, and genetic parameters.<br />

The longitudinal nature <strong>of</strong> the study will allow for both state and trait<br />

based analyses.<br />

Results: Currently 624 participants are enrolled. The average age <strong>of</strong><br />

onset for the bipolar I and II individuals is 19 years. The average age<br />

at interview for bipolar individuals is 40 years versus 32 years for<br />

controls. Recent findings show differences in cognitive functioning<br />

between individuals with and without bipolar disorder, as well as<br />

differential patterns in the depressed and hypomanic bipolar states. A<br />

lower socioeconomic burden <strong>of</strong> illness (disability) is associated with<br />

lower impact on cognitive and emotional factors.<br />

Conclusions: This research supports further pursuit <strong>of</strong> disease traits<br />

through longitudinal study designs that combine cognitive,<br />

behavioral, social, environmental, and biological factors, which is the<br />

basis <strong>of</strong> the H.C. Prechter Longitudinal Study. Building a<br />

longitudinal database with ongoing and additional studies with an<br />

open cohort that can be engaged in further research is key for the<br />

future <strong>of</strong> translational research in mood disorders.<br />

4<br />

IMPAIRED INFERIOR FRONTAL GYRUS RESPONSE TO<br />

AN EMOTIONAL INHIBITION TASK IN YOUNG FIRST-<br />

DEGREE RELATIVES OF BIPOLAR DISORDER PATIENTS<br />

COMPARED TO CONTROLS<br />

P. Mitchell*(1, 2), G. Roberts(1, 2), M. Green(1, 2), M.<br />

Breakspear(1, 2, 3), C. McCormack(1, 2), A. Frankland(1, 2), A.<br />

Wright(1, 2), C. McCue(1, 2), D. Hadzi-Pavlovic(1, 2), F. Levy(1),<br />

B. Lino(1, 2), R. Lenroot(1, ), J. Corry(1, 2)<br />

1. University <strong>of</strong> New South Wales 2. Black Dog Institute 3.<br />

Queensland Institute <strong>of</strong> Medical Research<br />

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

Introduction: Current evidence for cognitive and neuroanatomical<br />

correlates <strong>of</strong> the genetic predisposition to bipolar disorder implicate<br />

frontal cortical, and sub-cortical striatal and limbic systems involved<br />

in the cognitive control <strong>of</strong> emotion. However, there have been few<br />

functional imaging studies <strong>of</strong> individuals at high genetic risk for<br />

bipolar disorder. We examined neural activity associated with the<br />

inhibition <strong>of</strong> emotional material in young people at-risk for<br />

developing bipolar disorder, using an emotional inhibition paradigm,<br />

with the hypothesis that emotional material would impede the<br />

efficiency <strong>of</strong> cortical inhibitory networks.<br />

Methodology: Forty-eight genetically defined "high-risk”<br />

individuals, aged between 18 and 30 years, with at least one firstdegree<br />

relative with a diagnosis <strong>of</strong> bipolar disorder, and 49 healthy<br />

comparison participants performed a facial emotion go/no-go task<br />

during the acquisition <strong>of</strong> functional magnetic resonance imaging data.<br />

Results: All participants exceeded 75% accuracy. Whole brain<br />

analyses, corrected for family-wise error, revealed that at-risk<br />

participants demonstrated a highly specific and significant lack <strong>of</strong><br />

recruitment <strong>of</strong> the inferior frontal gyrus when inhibiting fearful faces,<br />

compared to healthy controls (p=0.027, FWE corrected).<br />

Conclusions: This finding suggests a dysregulated frontal inhibitory<br />

network for the inhibition <strong>of</strong> threatening emotion which may<br />

represent a neurocognitive endophenotype for bipolar disorder. This<br />

extends previous reports that the inferior frontal gyrus is under-active<br />

in patients with established bipolar disorder compared to normal<br />

controls by suggesting that it is also present in those genetically atrisk<br />

<strong>of</strong> the disorder.


CHILDHOOD DISORDERS PREDICT ADOLESCENT ONSET<br />

MOOD DISORDER IN FAMILIES WITH A BIPOLAR<br />

PROBAND<br />

J. Nurnberger *(1), M. McInnis(2), H. Wilcox(3), A. Glowinski(4),<br />

P. Mitchell(5), E. Gershon(6), W. Berrettini(7), G. Laite(1), R.<br />

Schweitzer(1), X. Cai(8), F. Azzouz(1), H. Liu(1), M. Kamali(2)<br />

1. Indiana University 2. University <strong>of</strong> Michigan 3. Johns Hopkins<br />

University 4. Washington University 5. University <strong>of</strong> New South<br />

Wales 6. University <strong>of</strong> Chicago 7. University <strong>of</strong> Pennsylvania 8.<br />

University <strong>of</strong> Iowa<br />

*jnurnber@iupui.edu<br />

Introduction: The childhood precursors <strong>of</strong> adult bipolar disorder<br />

(BP) are still a matter <strong>of</strong> controversy. Our objective was to report the<br />

lifetime prevalence and early clinical predictors <strong>of</strong> psychiatric<br />

disorders in <strong>of</strong>fspring from families <strong>of</strong> probands with DSM-IV<br />

Bipolar Disorder compared to <strong>of</strong>fspring <strong>of</strong> controls. We are carrying<br />

out a longitudinal, prospective study <strong>of</strong> subjects at risk for BP and<br />

related disorders. We report initial (cross-sectional and retrospective)<br />

diagnostic and clinical characteristics following best estimate<br />

procedures.<br />

Methodology: Assessment was performed at four university medical<br />

centers in the U.S. between June 2006 and September 2009.<br />

Participants were <strong>of</strong>fspring ages 12-21 in families with a BP proband<br />

(n=141, designated as cases) as well as similarly aged <strong>of</strong>fspring <strong>of</strong><br />

control parents (n=91). The main outcome measure was lifetime<br />

DSM IV diagnosis <strong>of</strong> major affective disorder (BPI, SABP, BPII, or<br />

Major Depression) as determined by a best estimate diagnostic<br />

process that included information from the K-SADS-BP on both<br />

child and parent and also medical records.<br />

Results: At an average age <strong>of</strong> 17, cases showed 23.4% lifetime<br />

prevalence <strong>of</strong> major affective disorder compared to 4.4% in controls<br />

(p = 0.002, adjusting for age, gender, ethnicity, and correlation<br />

between siblings). Prevalence <strong>of</strong> bipolar disorder in cases was 8.5%<br />

vs. 0% in controls (adjusted p = 0.007). No significant difference was<br />

seen in prevalence <strong>of</strong> other affective, anxiety, disruptive behavior<br />

disorders, or substance use disorders. Among case subjects<br />

manifesting major affective disorder (n=33), there was an increased<br />

risk for anxiety and externalizing disorders in comparison to cases<br />

without mood disorder. In cases but not controls, a childhood<br />

diagnosis <strong>of</strong> an anxiety disorder (Relative Risk (RR) = 2.6 (1.1 –<br />

6.3), p = 0.039) or an externalizing disorder (RR = 3.6 (1.4 – 9.0), p =<br />

0.007) was predictive <strong>of</strong> later onset <strong>of</strong> major affective disorder.<br />

Conclusions: Childhood anxiety and externalizing diagnoses predict<br />

major affective illness in adolescent <strong>of</strong>fspring in families with BP<br />

probands.<br />

5<br />

TRANSLATIONAL RESEARCH IN ANXIETY: HOW<br />

ANIMAL-BASED MODELS CAN COMPLEMENT HUMAN<br />

GENETICS<br />

EPIGENETIC MODIFICATION OF THE STRONG GENETIC<br />

PREDISPOSITION TOWARDS ANXIETY-RELATED<br />

BEHAVIOR IN PSYCHOPATHOLOGICAL HAB MICE<br />

R. Landgraf*, P. Markt, C. Avrabos, R. Naik, M. Eder, L. Czibere<br />

Max Planck Institute <strong>of</strong> Psychiatry<br />

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

Introduction: To mimic clinically relevant endophenotypes, we<br />

created extremes in the genetic predisposition to anxiety-related<br />

behavior by inbreeding CD-1 mice for either high (HAB),<br />

intermediate (NAB) or low (LAB) anxiety for >40 generations. This<br />

intra-strain, tri-directional approach provides the advantage that<br />

selection pressure is exerted on anxiety only, while maintaining a<br />

high degree <strong>of</strong> similarity in non-selected traits. Furthermore, due to<br />

their seemingly rigid predisposition, HAB/NAB/LAB mice are<br />

virtually independent <strong>of</strong> confounding variables such as uterine<br />

environment (embryo transfer), maternal care (cross-fostering), sex<br />

and age. Importantly, trait anxiety is associated with depression-like<br />

behaviors, resembling clinical comorbidity.<br />

Methodology: Chip technologies for expression pr<strong>of</strong>iling and<br />

genotyping sequencing allele-specific transcription gene reporter<br />

assays association studies.<br />

Results: As exemplarily shown in a number <strong>of</strong> candidate genes (e.g.,<br />

Avp, Tmem132d, Npsr1, Crhr1, cathepsin), polymorphisms differing<br />

between HAB and LAB do not only underlie differences in<br />

expression, but are also associated with the anxiety phenotype in a<br />

freely segregating F2 panel, suggesting a causal contribution. Despite<br />

the robust genetic predisposition, environmental stimuli were found<br />

to shift both HAB (enriched environment) and LAB (unpredictable<br />

chronic mild stress) towards NAB, highlighting an efficient<br />

phenotypic rescue. In the amygdala <strong>of</strong> HAB mice, correlates <strong>of</strong> that<br />

rescue include (i) neuronal activity, which accompanies the<br />

phenotypic shift, as well as (ii) epigenetic modifications with<br />

expression <strong>of</strong> the Crhr1 gene being reduced and its methylation status<br />

being altered upon enriched environment.<br />

Conclusions: This animal model thus <strong>of</strong>fers the opportunity to study<br />

possible interactions between genetic predisposition and epigenetic<br />

modifications in more detail, both genome-wide and at single gene<br />

level.


TMEM132D: A NEW CANDIDATE FOR ANXIETY<br />

PHENOTYPES - EVIDENCE FROM HUMAN AND MOUSE<br />

STUDIES<br />

A. Erhardt*<br />

Max Planck Institute for Psychiatry<br />

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

Introduction: Family and twin studies indicate an estimated<br />

heritability for anxiety and panic disorders up to 30-40%. In contrast<br />

to candidate gene studies, genome-wide association investigations<br />

present an unbiased approach and are helpful to reveal novel<br />

pathological pathways in complex diseases.<br />

Methodology: We performed a whole-genome association study in<br />

three different German samples with panic disorder patients and<br />

healthy controls (909 cases and 915 controls).<br />

Results: SNPs in TMEM132D were found to be associated with<br />

panic disorder, with a two SNP haplotype associated in each <strong>of</strong> three<br />

samples in the same direction, with a p-value <strong>of</strong> 1.2e-7in the<br />

combined sample. Independent SNPs in this gene were also<br />

associated with the severity <strong>of</strong> anxiety symptoms, in patients affected<br />

by panic disorder or panic attacks as well as patients suffering from<br />

unipolar depression. Risk genotypes for panic disorder were<br />

associated with higher TMEM132D mRNA expression levels in the<br />

frontal cortex. In parallel, using a mouse model <strong>of</strong> extremes in trait<br />

anxiety, we could further demonstrate that anxiety-related behavior<br />

was positively correlated with Tmem132d mRNA expression in the<br />

anterior cingulate cortex, central to the processing <strong>of</strong> anxiety/fearrelated<br />

stimuli, and that in this animal model a Tmem132d SNP is<br />

associated with anxiety-related behavior in an F2 panel. Further<br />

validation studies using next-generation sequencing revealed high<br />

number <strong>of</strong> rare variants in this gene. Additionally, behavioural and<br />

molecular phenotyping in the mouse knock-out model is ongoing to<br />

investigate, how TMEM132D is implicated in the regulation <strong>of</strong><br />

anxiety-related behaviour.<br />

Conclusions: TMEM132D is an important new candidate gene for<br />

panic disorder as well as more generally for anxiety-related behavior.<br />

6<br />

MICRORNA EXPRESSION PATTERNS IN A MOUSE<br />

MODEL OF ANXIETY<br />

T. Sipilä(1,2,3), K. Icay(1,2), J. Juhila(1,2,4), P. Ellonen(4), D.<br />

Greco(5), I. Hovatta*(1,2,3,4)<br />

1. Research Programs Unit, Molecular Neurology, Biomedicum-<br />

Helsinki, University <strong>of</strong> Helsinki 2. Department <strong>of</strong> Medical <strong>Genetics</strong>,<br />

Haartman Institute, University <strong>of</strong> Helsinki 3. Department <strong>of</strong> Mental<br />

Health and Substance Abuse Services, National Institute for Health<br />

and Welfare 4. Institute <strong>of</strong> Molecular Medicine Finland (FIMM),<br />

University <strong>of</strong> Helsinki 5. Department <strong>of</strong> Bioscience and Nutrition,<br />

Karolinska Institutet<br />

*iiris.hovatta@helsinki.fi<br />

Introduction: MicroRNAs (miRNAs) are small regulatory molecules<br />

that suppress mRNA activity. They have been isolated and<br />

characterized in all organisms and tissues, and in mammalian brain<br />

they are shown to be involved in neuronal differentiation and<br />

synaptic plasticity. Recent evidence points at the involvement <strong>of</strong><br />

miRNA expression differences in neuropsychiatric diseases. Our aim<br />

was to identify miRNAs that participate in the regulation <strong>of</strong> anxietylike<br />

behavior in a mouse model. This model consists <strong>of</strong> six inbred<br />

mouse strains that differ in their innate anxiety like behavior: A/J,<br />

DBA/2J, 129S1/SvImJ, C3H/HeJ, C57BL/6J, and FVB/NJ. We have<br />

earlier carried out gene expression pr<strong>of</strong>iling in refined brain regions<br />

<strong>of</strong> these strains and identified gene expression differences underlying<br />

anxiety-like behavior.<br />

Methodology: We have setup a miRNA-seq protocol for quantitative<br />

miRNA expression analysis from mouse brain regions. Using this<br />

approach, we carried out miRNA expression pr<strong>of</strong>iling from three<br />

brain regions playing a key role in the regulation <strong>of</strong> anxiety: frontal<br />

cortex, hippocampus and hypothalamus. We correlated the miRNA<br />

expression pr<strong>of</strong>iles with anxiety-like behavior, measured by the open<br />

field and light dark box tests, across the six strains.<br />

Results: We identified 53 miRNAs with expression levels that<br />

correlate with anxiety-like behavior. Using the miRWalk tool, which<br />

combines information from several target prediction algorithms, we<br />

predicted target genes for each differentially expressed miRNA. To<br />

assess the putative biological functions <strong>of</strong> the differentially expressed<br />

miRNAs and their predicted targets, we carried out pathway analyses<br />

with the Ingenuity Pathways Analysis tool. Several differentially<br />

expressed miRNAs were predicted to regulate target genes within the<br />

same biological pathways.<br />

Conclusions: Identification <strong>of</strong> miRNAs with expression pattern<br />

correlating with anxiety-like behavior enables formation <strong>of</strong> specific<br />

testable hypotheses regarding the gene regulatory networks<br />

underlying anxiety. These hypotheses can be tested by functional<br />

studies in mouse models or in human anxiety disorder cohorts.


NEURAL CORRELATES OF GENETICALLY-DRIVEN<br />

STRAIN DIFFERENCES IN FEAR.<br />

A. Holmes*<br />

NIAAA<br />

*holmesan@mail.nih.gov<br />

Introduction: Only a fraction <strong>of</strong> individual exposed to severe<br />

psychological trauma will meet diagnostic criteria for a clinical<br />

anxiety disorder. Risk likely results from genetic influences and gene<br />

x environment interactions that drive functional alterations in neural<br />

circuits regulating emotion. One approach to disentangle these risk<br />

factors is to identify strain, subpopulation or selectively bred rodent<br />

lines that show excessive fear after a trauma such as fear<br />

conditioning.<br />

Methodology: This presentation will discuss multi-level analysis <strong>of</strong><br />

two isogenic mouse strains (S1 and B6) exhibiting divergent fear<br />

extinction and corticoamygdala circuit function.<br />

Results: Results will show that S1 exhibited deficient extinction,<br />

overgeneralized fear to ambiguous cues and failed to effectively use<br />

safety cues to inhibit fear. Fear abnormalities in S1 were associated<br />

with autonomic disturbances and HPA-axis abnormalities, and were<br />

fully rescued by chronic treatment with the anti-anxiety medication,<br />

fluoxetine. Within the fear-circuit, S1 had enlarged dendritic arbors<br />

in basolateral amygdala neurons in S1, but normal dendritic<br />

arborization in infralimbic and prelimbic cortices. Moreover,<br />

multichannel unit recording revealed aberrant extinction-related<br />

infralimbic cortex single-unit activity and uncoupling <strong>of</strong> neuronal<br />

bursting from extinction, in S1.<br />

Conclusions: Together, these data provide novel, convergent insights<br />

into the factors underlying genetically-driven differences in fear<br />

regulation.<br />

7<br />

FROM GENE TO DRUG RESPONSE: MULTI-METHOD<br />

APPROACH TO THE PHARMACOGENOMICS OF<br />

RESPONSE TO ANTIDEPRESSANTS<br />

FROM GENE TO DRUG RESPONSE: MAKING MORE OF<br />

GENOME-WIDE PHARMACOGENETIC DATA<br />

R. Uher*(1), K. Tansey(1), M. Guipponi(2), N. Perroud(2), A.<br />

Malafosse(2), X. Hu(3), G. Lewis(4), M. O'Donovan(5), E.<br />

Domenici(6,7), M. Rietschel(8), W. Maier(9), O. Mors(10), J.<br />

Hauser(11)<br />

1. King's College London 2. University <strong>of</strong> Geneva 3. Pfizer, Groton<br />

4. 4University <strong>of</strong> Bristol 5. University <strong>of</strong> Cardiff 6. Glaxo Smith<br />

Kline 7. Roche 8. Central Institute <strong>of</strong> Mental Health 9. University <strong>of</strong><br />

Bonn 10. Aarhus University Hospital 11. Poznan University <strong>of</strong><br />

Medical Sciences<br />

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

Introduction: Genomic determinants <strong>of</strong> response to antidepressants<br />

can be translated into clinical care through personalized treatment<br />

choice and inform development <strong>of</strong> new therapeutics to target resistant<br />

cases. However, identification <strong>of</strong> genomic variants that predict<br />

individual differences in treatment response has been hindered by<br />

several obstacles including a complex longitudinal phenotype,<br />

difficulty in obtaining large high-quality clinical samples, nonspecific<br />

‘placebo’ response and limits inherent in the design <strong>of</strong><br />

clinical trials.<br />

Methodology: NEWMEDS, a collaboration <strong>of</strong> academic and<br />

industrial partners (http://www.newmeds-europe.com/), assembled a<br />

combined sample <strong>of</strong> over 2000 cases <strong>of</strong> moderate to severe<br />

depression with prospective outcome data on up to 12 weeks<br />

treatment with either a serotonin reuptake inhibiting antidepressant<br />

(SRI) or a norepinephrine reuptake-inhibiting (NRI) antidepressant.<br />

The genome-wide analyses were powered to detect a clinically<br />

significant effect for polymorphisms with a minor allele frequency <strong>of</strong><br />

10% or more. Four primary genome-wide analyses tested the<br />

hypotheses that common genetic variant exist that have a clinically<br />

significant effects on (1) response to any antidepressant, (2) response<br />

to SRI, (3) response to NRI, or (4) that differentially predict response<br />

to SRI and NRI antidepressants. These primary analyses were<br />

followed up with a variety <strong>of</strong> secondary approaches.<br />

Results: No association reached genome-wide significance (p


TRANSCRIPTOMIC AND EPIGENETIC CORRELATES OF<br />

ANTIDEPRESSANT RESPONSE<br />

F. Mamdani, J. Lopez, M. Berlim, A. Labbe, G. Turecki*<br />

McGill University<br />

*gustavo.turecki@mcgill.ca<br />

Introduction: There is significant variability in antidepressant<br />

treatment outcome, with approximately 30-40% <strong>of</strong> patients with<br />

major depressive disorder (MDD) not presenting with adequate<br />

response even following several trials. To identify potential gene<br />

expression and epigenetic biomarkers <strong>of</strong> response we investigated<br />

peripheral gene expression patterns <strong>of</strong> response to antidepressant<br />

treatment in MDD.<br />

Methodology: We used Affymetrix HG-U133 Plus2 microarrays in<br />

blood samples from untreated individuals with MDD (N = 63)<br />

ascertained at a community outpatient clinic, pre- and post eightweek<br />

treatment with citalopram and used a regression model to assess<br />

the impact <strong>of</strong> gene expression differences on antidepressant response.<br />

We carried out technical validation <strong>of</strong> significant probesets by qRT-<br />

PCR and conducted CNS follow-up <strong>of</strong> the most significant result in<br />

post-mortem brain samples from MDD and control individuals.<br />

Epigenetic investigation <strong>of</strong> differentially expressed genes was carried<br />

out.<br />

Results: A total <strong>of</strong> 32 probesests were differentially expressed<br />

according to response to citalopram treatment following FDR<br />

correction. Interferon Regulatory Factor 7 (IRF7) was the most<br />

significant differentially expressed gene and its expression was<br />

upregulated by citalopram treatment in individuals who responded to<br />

treatment. Consistent results were found in postmortem brain tissue<br />

<strong>of</strong> MDD subjects and epigenetic mechanisms seem to play a relevant<br />

role.<br />

Conclusions: These findings point to IRF7 as a gene <strong>of</strong> interest in<br />

studies investigating genomic factors associated with antidepressant<br />

response.<br />

8<br />

AN APPROACH TO EMPLOY ANIMAL STUDIES TO<br />

INFORM HUMAN PHARMACOGENETICS<br />

M. Popoli*<br />

Center <strong>of</strong> Neuropharmacology-Department <strong>of</strong> Pharmacological<br />

Sciences, University <strong>of</strong> Milano<br />

*maurizio.popoli@unimi.it<br />

Introduction: Research in rodent models may contribute to identification <strong>of</strong><br />

genomic determinants <strong>of</strong> response to drugs. This objective can be pursued by<br />

employing different rodent models, with the use <strong>of</strong> stress paradigms or<br />

transgenics reproducing genetic variants linked to neuropsychiatric disorders,<br />

and treating the animals with different drugs. The kinds <strong>of</strong> techniques that can<br />

be used range from transcriptomics and proteomics to the more recent<br />

epigenetic methods. Aim <strong>of</strong> this presentation will be to draw from different<br />

animal studies a number <strong>of</strong> converging, validated genes and pathways that<br />

may inform human pharmacogenetic studies.<br />

Methodology: Select results will be presented from a number <strong>of</strong> studies<br />

including: (1) Transcriptomics <strong>of</strong> the Flinders Sensitive Line (FSL) genetic<br />

model <strong>of</strong> depression (2) Synaptoproteomics <strong>of</strong> FSL subjected to early-life<br />

stress (gene x environment model) (3) Synaptoproteomics <strong>of</strong> the Learned<br />

Helplessness (LH) model (4) Epigenetic analysis <strong>of</strong> transgenic mice carrying<br />

the BDNF Val66Met human mutation (5) Analysis <strong>of</strong> microRNAs (miRs)<br />

involved in the action <strong>of</strong> antidepressants.The studies on FSL and LH models<br />

<strong>of</strong> depression have been carried out within the GENDEP Consortium, funded<br />

from EU in the FP6 Program. Both FSL and LH are validated and previously<br />

characterized animal models <strong>of</strong> depression. Some <strong>of</strong> these studies have been<br />

published [1-3].Epigenetic mechanisms, self-perpetuating changes in<br />

chromatin that alter gene expression, regulate CNS physiology and have a role<br />

in both neuropsychiatric pathophysiology and drug action. The BDNF<br />

Val66Met transgenic mouse is the first animal model that fully recapitulates<br />

the established phenotypic effects <strong>of</strong> a common human polymorphism<br />

expressed in brain, including reduced hippocampal size and dendritic<br />

complexity, deficit in extinction learning and NMDA receptor-dependent<br />

synaptic plasticity. The data that will be presented come from the first<br />

epigenetic analysis <strong>of</strong> BDNFMet mice and may contribute to explain some <strong>of</strong><br />

the phenotypic features <strong>of</strong> Met allele carriers.miRs have recently emerged as<br />

key regulators <strong>of</strong> complex temporal and spatial patterns <strong>of</strong> gene/protein<br />

expression changes and, thereby, neuroplasticity. The present study is the first<br />

analysis <strong>of</strong> the effects <strong>of</strong> different chronic antidepressant treatments on miRs<br />

and respective target genes.<br />

Results: A common result <strong>of</strong> the studies on FSL and LH models was the<br />

finding that energy metabolism and cellular remodeling pathways are<br />

involved in both the depressive-like phenotype and in the response to<br />

antidepressants. A number <strong>of</strong> interesting biomarkers was regulated in opposite<br />

ways by stress and antidepressants in both models. Results with the BDNF<br />

Val66Met mice have been obtained both from candidate gene studies and<br />

from a genome-wide ChIP-Seq study. Bioinformatic analysis <strong>of</strong> genes with<br />

epigenetic changes in their promoters revealed the involvement <strong>of</strong> the KEGG<br />

“Neuroactive ligand-receptor”, “MAPK signaling” and “Regulation <strong>of</strong> actin<br />

cytoskeleton” pathways. In addition, consistent changes in epigenetic tags and<br />

in expression have been found in select splice variants <strong>of</strong> BDNF transcripts.<br />

Conclusions: The results obtained in the different rodent studies summarized<br />

above allowed to select a number <strong>of</strong> genes and pathways that were involved in<br />

pathophysiology as well as in response to drug treatments. Converging results<br />

from different studies and models will be implemented to inform human<br />

pharmacogenetic research.<br />

References<br />

1. Blaveri et al. (2010) PloS ONE5(9):e12596.<br />

2. Carboni et al. (2010) Progr Neuropsychopharmacol & Biol<br />

Psychiatry16:1037-48.<br />

3. Mallei et al. (2010) Neuropharmacology2010 Dec 31. [Epub ahead <strong>of</strong><br />

print].


IMAGING INTERMEDIATE PHENOTYPES OF<br />

DEPRESSION INCLUDING RESPONSE TO THERAPY:<br />

OVERVIEW AND CONCEPTUAL LINKS TO GENETICS<br />

P. Sämann*(1), D. Höhn(1), M. Czisch(1), M. Kohli(1), S. Lucae(1),<br />

B. Inkster(2,3), P. Matthews(2,3), L. Dixson(2), T. Kam-Thong(1),<br />

B. Müller-Myhsok(1), E. Binder(1), F. Holsboer(1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry 2. GlaxoSmithKline Clinical<br />

Imaging Centre, HammersmithHospital 3. Department <strong>of</strong> Clinical<br />

Neuroscience, Imperial College<br />

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

Introduction: Imaging genomics builds associations between genetic<br />

variability and variability <strong>of</strong> neuroimaging measures. In the context<br />

<strong>of</strong> depressive disorders, imaging genomics may pursue different<br />

goals: (i) imaging studies performed on known genetic variants<br />

associated with depression phenotypes to explore their functional<br />

impact, (ii) validation <strong>of</strong> newly detected genetic variants by<br />

projection onto an intermediate phenotype, (iii) search for genetic<br />

underpinnings <strong>of</strong> depression phenotypes by the use <strong>of</strong> intermediate<br />

phenotypes. While (i) is the most commonly used approach, (ii) and<br />

(iii) are now also increasingly employed with(iii) representing a<br />

particularly promising application <strong>of</strong> the original endophenotype<br />

concept.<br />

Methodology: An example <strong>of</strong> a candidate gene approach (i) is given<br />

from recent analyses on Methyl CpG binding protein (MECP2)<br />

variants directed to brain morphology and 1H-MR-spectroscopy (1H-<br />

MRS). MRI data for analysis (i) and (iii) were acquired at the Max<br />

Planck Institute <strong>of</strong> Psychiatry, Munich, comprising 525 structural<br />

MRI scans (225 healthy adults, 300 patients with single episode or<br />

recurrent major depression), including a subgroup <strong>of</strong> 176 subjects<br />

with 1H-MRS. Concept (ii) is employed within the convergent<br />

approach framework that aims at proving evidence for true causal<br />

gene-phenotype relationship by integrating different functional<br />

assays. As an example, results from a genetic variant revealed by a<br />

whole genome case/control study are picked up. For (iii), literature<br />

based imaging based endophenotypes <strong>of</strong> response to antidepressant<br />

treatment are given. Further, a whole genome association study<br />

focussing on hippocampal structural measures is presented.<br />

Results: Ad (i): The analysis <strong>of</strong> MECP2-rs2239464 revealed<br />

genotype X group interactions on cortex volume <strong>of</strong> lower medial<br />

prefrontal cortex (mPFC) areas including the subgenual area. A<br />

similar interaction was also found with respect to mPFC glutamate/<br />

glutamine levels, pointing out an interaction between MECP2 and<br />

recurrent depression potentially related to epigenetic mechanisms. Ad<br />

(ii): A newly detected genetical variant in a neuronal transporter gene<br />

conveying risk for depression was found associated with reduced<br />

hippocampal integrity and glutamate levels in healthy risk allele<br />

carriers, and with altered hippocampal grey matter volumes in risk<br />

allele carrying patients. Ad (iii): A whole genome association study<br />

focussing on whole genome wide SNP X SNP interaction effects on<br />

hippocampal structural markers is described.<br />

Conclusions: By exemplary studies it is demonstrated that the<br />

intermediate phenotype idea is a flexible and powerful concept to<br />

study the influence <strong>of</strong> genetic variants that convey risk to develop<br />

depression or that are associated with treatment response. Imaging<br />

studies on candidate genes have been successfully employed to build<br />

new hypotheses on how these variants translate into behavioral<br />

patterns. Strict adherence to the endophenotype concept is needed<br />

when newly detected phenotype-genotype associations are validated<br />

by neuroimaging. Last, gene search studies into which imaging based<br />

phenotypes are entered are a rather new approach in depression<br />

research.<br />

9


PLENARY<br />

ABSTRACTS<br />

10


PLENARY 1: THE ROLE IN DISEASE OF SYNAPSE<br />

PROTEIN COMPLEXES AND COMPLEXITY<br />

S. Grant*<br />

The Wellcome Trust Sanger Institute<br />

*sg3@sanger.ac.uk<br />

Recent work combining synapse proteomics and human genetics<br />

shows multiprotein complexes in the postsynaptic proteome are<br />

disrupted by mutations in over 200 genes causing over 130 brain<br />

diseases (Bayes et al, 2011). A subset <strong>of</strong> these proteins assemble into<br />

multiprotein complexes known as the MAGUK Associated Signaling<br />

Complex (MASC) that is involved in over 50 diseases including<br />

schizophrenia, bipolar disease and autism. A large scale mouse<br />

genetic screen identifies sets <strong>of</strong> MASC proteins that mediate adaptive<br />

behaviours and higher cognitive functions. MASC is a signaling<br />

complex linking neurotransmitter receptors to intracellular pathways<br />

and thereby mediates behavioural responses. Comparative genomic,<br />

proteomic and behavioural genetic studies show a strong<br />

conservation <strong>of</strong> function in these complexes in mammals and provide<br />

new approaches to animal models <strong>of</strong> human disease.<br />

11<br />

PLENARY 2: ASSOCIATION METHODS FOR RARE<br />

ALLELES<br />

S. Leal*<br />

Baylor College <strong>of</strong> Medicine<br />

*sleal@bcm.edu<br />

There is currently great interest in detecting rare variant associations<br />

using next generation sequence data. A large number <strong>of</strong> association<br />

methods which aggregate variants across a region e.g. a gene have<br />

been developed specifically to analyze rare variant data. It is not clear<br />

which existing method is the most powerful and should be applied to<br />

test for associations using exome/genome sequence data. It is difficult<br />

to compare rare variant association methods because there is no<br />

standard to generate data and <strong>of</strong>ten the comparisons are biased. To<br />

fairly compare rare variant association methods it is necessary to<br />

generate data using realistic population demographic and phenotype<br />

models. The power was compared for a variety <strong>of</strong> methods to detect<br />

associations for qualitative and quantitative traits. Power was<br />

evaluated for case-control, extreme quantitative trait sampling and<br />

population based study designs. For each method, power was<br />

determined for scenarios which included 1. analysis <strong>of</strong> a. only rare<br />

variants & b. rare (


PLENARY 3: NIMH FUNDING PRIORITIES FOR GENETICS<br />

T. Insel*, T. Lehner*<br />

National Institute <strong>of</strong> Mental Health<br />

*tinsel@mail.nih.gov, *tlehner@mail.nih.gov<br />

In spite <strong>of</strong> a flattening <strong>of</strong> the NIH budget, the NIMH has continued to<br />

invest in and grow the genetics portfolio relevant to mental<br />

disorders. The conceptual and technological advances <strong>of</strong><br />

contemporary genomics have presented unprecedented opportunities<br />

to map the biological basis <strong>of</strong> risk for mental disorders. The<br />

American Recovery and Reinvestment Act <strong>of</strong> 2009 presented an<br />

unexpected opportunity for NIMH to make additional large<br />

investments in psychiatric genetics and to generate additional<br />

resources for the community as we move from genotyping to<br />

sequencing to multiplexed approaches with epigenomics,<br />

transcriptomics, proteomics, and imaging. The advent <strong>of</strong> iPS cells<br />

promises a powerful new approach for moving from genetic variation<br />

to a deeper understanding <strong>of</strong> the mechanisms by which<br />

neurodevelopmental disorders, such as schizophrenia and autism,<br />

may develop. The speakers will discuss funding trends and priorities<br />

in genetics for NIMH, including the large investments by NIMH into<br />

genomic resources for the global investigator community. The level<br />

<strong>of</strong> NIH funding in 2012 remains uncertain, but most predict cuts<br />

below the President's recommended budget. The potential impact <strong>of</strong><br />

budgetary constraints on the field going into the future will be<br />

discussed.<br />

12<br />

PLENARY 4: MODELING HUMAN GENETIC DISEASE IN<br />

THE LABORATORY<br />

N. Katsanis*<br />

Duke University Medical Center<br />

*katsanis@cellbio.duke.edu<br />

Defects <strong>of</strong> the primary cilium and its anchoring structure, the basal<br />

body, cause a number <strong>of</strong> human genetic disorders, collectively<br />

termed ciliopathies, since they are characterized by an overlapping<br />

range <strong>of</strong> phenotypes that include retinal degeneration, polydactyly,<br />

renal and hepatic fibrosis, obesity and a complex range <strong>of</strong> cognitive<br />

and neurodevelopmental defects. Recent data have also shown that<br />

some ciliopathies overlap not only phenotypically, but also<br />

genetically by contributing epistatic alleles that can modulate the<br />

phenotypic expressivity and penetrance. As such, the primary cilium<br />

and its associated signaling represent a useful model to understand<br />

the mechanism <strong>of</strong> total mutational load in a biological system.<br />

Towards that end, we have initiated systematic sequencing and<br />

functional evaluation <strong>of</strong> mutations <strong>of</strong> ciliary genes in a range <strong>of</strong><br />

ciliopathy phenotypes and, using a large allelic series, have<br />

constructed models <strong>of</strong> epistasis in oligogenic disorders that suggest<br />

an intricate interaction between rare and common alleles to modulate<br />

penetrance and expressivity. Such studies will ultimately empower<br />

the predictive nature <strong>of</strong> the genotype and inform clinical management<br />

and treatment. Importantly, coupling deep medical resequencing with<br />

systematic in vivo and in vitro functional testing <strong>of</strong>fers significantly<br />

improved resolution and increased appreciation <strong>of</strong> the complexity and<br />

architecture <strong>of</strong> genetic disease.


PLENARY 5: GENETIC DISSECTION OF COMPLEX<br />

TRAITS<br />

A. Chakravarti*<br />

Johns Hopkins University<br />

*aravinda@jhmi.edu<br />

Introduction: The genetic bases for complex diseases and<br />

quantitative traits have remained an open problem in genetics for<br />

close to a century.<br />

Although much progress have been made in our understanding, from<br />

theoretical models and in model systems, the natural variation that<br />

contributes to these traits remain elusive. I will discuss our efforts at<br />

understanding two medically relevant human neuronal disorders,<br />

Hirschsprung disease and autism, and how progress in these disorders<br />

have taught us the relevant lessons as to why traits and diseases are<br />

complex.<br />

13<br />

PLENARY 7: GENETIC INFLUENCES ON IMPULSIVITY<br />

D. Goldman*<br />

National Institute <strong>of</strong> Health<br />

*davidgoldman@mail.nih.gov<br />

Impulsivity, describing action without foresight, is a precursor and<br />

mediating factor in several psychiatric diseases including addictions<br />

and personality disorders. Impulsivity is a modifier <strong>of</strong> other<br />

psychiatric diseases, for example contributing to suicidality.<br />

Functional alleles mediate the heritability <strong>of</strong> impulsivity and<br />

disorders with which it is associated. Alleles mediating risk for<br />

psychiatric disease are both common and uncommon, and <strong>of</strong> different<br />

effect sizes, however their actions appear to be stronger on<br />

intermediate phenotypes, including heritable endophenotypes. For<br />

example, a functional variant <strong>of</strong> Neuropeptide Y exerts strong effects<br />

on molecules, and intermediate effects on brain responses, but<br />

weaker effects on anxiety (Zhou et al, Nature, 2008). Via genome<br />

wide association, CHRNA5 Asp398Asn was identified as a risk factor<br />

in nicotine addiction. Via a study <strong>of</strong> functional connectivity, we<br />

found that the risk allele, Asn398, exerts a much larger effect (11% <strong>of</strong><br />

the variance) on the connectivity <strong>of</strong> multiple brain circuits (Hong et<br />

al, PNAS, 2010), potentially clarifying how Asn398 could modify<br />

vulnerability. The circuits weakened by Asn398 include a dorsal<br />

anterior cingulate/ventral striatal circuit that predicts nicotine craving.<br />

To find rare and uncommon alleles contributing to impulsivity we<br />

used next generation sequencing in severely impulsive individuals<br />

from a founder population, Finland. Echoing the discovery <strong>of</strong> Han<br />

Brunner <strong>of</strong> an MAOA stop codon that led to impulsivity in one Dutch<br />

family, we identified a stop codon, Q20*in the serotonin HTR2B<br />

receptor. *20 led to variable nonsense-mediated decay <strong>of</strong> the HTR2B<br />

mRNA and complete blockade <strong>of</strong> receptor expression. *20 was<br />

necessary for severe impulsivity displayed by some individuals in our<br />

sample <strong>of</strong> violent <strong>of</strong>fenders, but not alone sufficient. Other important<br />

factors were male sex and inebriation with alcohol. *20 was<br />

associated with disorders marked by impulsivity and co-segregated in<br />

families. It did not lead to global cognitive deficits. *20 is apparently<br />

population-specific but common in Finns, with an allele frequency<br />

>1%, echoing findings made for various medical genetic diseases,<br />

where genetic heterogeneity is dramatically reduced in this<br />

population. Mice knocked out for htr2b are impulsive and novelty<br />

seeking. Genomic approaches, tied to deep phenotyping and study <strong>of</strong><br />

appropriate human populations and animal models, are identifying<br />

rare and common alleles that underly heritability <strong>of</strong> traits such as<br />

impulsivity, and <strong>of</strong>fer an avenue to the identification <strong>of</strong> genes whose<br />

rare and uncommon alleles modulate behavior.


SYMPOSIA<br />

ABSTRACTS<br />

14


SYMPOSIUM 1: NEW INSIGHTS FROM CNV STUDIES IN<br />

SCHIZOPHRENIA<br />

S1.1 DE NOVO CNVS IN SCHIZOPHRENIA ARE<br />

ENRICHED FOR PATHOGENIC EVENTS AND DIFFER<br />

FROM INHERITED CNVS<br />

G. Kirov*(1), M. O'Donovan(1), D. Ivanov(1), M. Ikeda(1), E.<br />

Rees(2), D. Ruderfer(3,4,5), J. Moran(5), K. Chambert(5), P.<br />

Sklar(5), S. Purcell(5), D. Grozeva(1), P. Olason(6), Y. Böttcher(6),<br />

H. Stefansson(6), K. Stefansson(6), M. Owen(1)<br />

1. Cardiff University 2. Fujita Health University 3. Massachusetts<br />

General Hospital and Harvard Medical School 4. Massachusetts<br />

General Hospital 5. Broad Institute 6. deCODE genetics<br />

*kirov@cardiff.ac.uk<br />

Introduction: We hypothesized that CNVs occurring de novo as new<br />

mutations in individuals affected with schizophrenia are particularly<br />

likely to be relevant to the pathogenesis <strong>of</strong> this disorder.<br />

Methodology: Samples were genotyped with Affymetrix 6.0 arrays<br />

and filtered with the z-score method.<br />

Results: Using 638 informative families, we found 34 rare de novo<br />

CNVs in 662 schizophrenia probands, a rate <strong>of</strong> 5.1%. This is over 2fold<br />

higher compared with a rate <strong>of</strong> 2.2% in 2623 Icelandic<br />

individuals without a history <strong>of</strong> neurodevelopmental disorders.<br />

Several <strong>of</strong> the de novo CNVs found in schizophrenia are in loci<br />

already implicated in disease pathogenesis: 3q29, 15q11.2, 15q13.3<br />

and 16p11.2. This supports the conclusion that they are enriched for<br />

pathogenic loci. The median size <strong>of</strong> the de novo CNVs was 320.8kb.<br />

This makes them much larger than the 1300 rare CNVs found in 605<br />

unaffected controls from the Bulgarian population, genotyped and<br />

called using the same methods (median size = 69.4kb, P=1.6x10 -7 ).<br />

However the de novo CNVs found in schizophrenia were very similar<br />

in size to de novo CNVs identified in unaffected controls from the<br />

Icelandic population.<br />

Conclusions: We postulate that de novo CNVs are more pathogenic<br />

in general, as they have yet to subjected to natural selection. This<br />

makes them different in size and other properties to CNVs found in<br />

the general population.<br />

15<br />

S1.2 DE NOVO COPY NUMBER VARIANTS CONFER RISK<br />

FOR EARLY ONSET BIPOLAR DISORDER<br />

AND SCHIZOPHRENIA<br />

D. Malhotra*(1), S. Mccarthy(2), K. burdick(3), A. malhotra(4), E.<br />

leibenluft(5), J. potash(6), F. mcmahon(7), T. schulze(8), S.<br />

cichon(9), M. reitschel(10), J. kelsoe(1), E. gershon(12), D. levy(13),<br />

A. corvin(14), M. karayiorgou(15), J. sebat(1)<br />

1. Department <strong>of</strong> Psychiatry,University <strong>of</strong> California, San Diego 2.<br />

Stanley Center for Cognitive Genomics, Cold Spring Harbor<br />

Laboratory 3. Zucker Hillside hospital, NorthShore Long Island<br />

Jewish health system 4. Zucker Hillside hospital, NorthShore Long<br />

Island Jewish health system 5. Section on Bipolar Spectrum<br />

Disorders, Mood and Anxiety Disorders Program, NIMH Building<br />

15K -MSC 2670 6. The Johns Hopkins Hospital, John Hopkins<br />

University, 600 North Wolfe Street 7. Genetic Basis <strong>of</strong> Mood and<br />

Anxiety Disorders, National Institute <strong>of</strong> Mental Health, NIH,<br />

Convent Drive MSC 3719 8. Department <strong>of</strong> Psychiatry and<br />

Psychotherapy, University <strong>of</strong> Gottingen 9. Department <strong>of</strong> Genomics,<br />

Life and Brain Center, University <strong>of</strong> Bonn 10. Central Institute <strong>of</strong><br />

Mental Health, Square J5, 68159 11. Department <strong>of</strong><br />

Psychiatry,University <strong>of</strong> California, San Diego 12. Department <strong>of</strong><br />

Psychiatry and Behavioral Neuroscience, University <strong>of</strong> Chicago 13.<br />

McLean Hospital 14. Neuropsychiatric <strong>Genetics</strong> Research Group,<br />

Institute <strong>of</strong> Molecular Medicine and Department <strong>of</strong> Psychiatry,<br />

Trinity College 15. Department <strong>of</strong> Psychiatry, Columbia University<br />

16. Department <strong>of</strong> Psychiatry,University <strong>of</strong> California, San Diego<br />

*dmalhotra@ucsd.edu<br />

Introduction: Family-based studies <strong>of</strong> copy number variation (CNV)<br />

have demonstrated that rare spontaneous mutations play a role in<br />

neuropsychiatric disorders including autism (ASD) and schizophrenia<br />

(SZ). However, the relevance <strong>of</strong> such mutations to bipolar disorder<br />

(BD) has not been established. We tested the hypothesis that de novo<br />

CNVs are enriched in BD subjects with early age-at-onset (AAO≤18)<br />

and we sought to confirm the strong association <strong>of</strong> de novo CNVs in<br />

SZ.<br />

Methodology: Using a microarray consisting <strong>of</strong> 2.1 million probes,<br />

we screened for de novo CNVs ≥10 kb in size in blood derived DNAs<br />

from 788 subject-mother-father trios. Diagnoses <strong>of</strong> subjects included<br />

BD (N=185), SZ (N=177) and healthy controls (N=426). CNVs were<br />

subsequently validated by a custom tiling-resolution array<br />

comparative genomic hybridization (CGH) platform.<br />

Results: A total <strong>of</strong> 23 de novo CNVs were detected. Mutations were<br />

detected in 0.9% <strong>of</strong> controls and at significantly increased rates in BD<br />

(4.3%, P=0.009) and SZ (4.5%, P=0.007). In BD, the observed effect<br />

was mainly in probands with AAO≤18 (5.6%, P=0.006) and not in<br />

late onset BD (2.6%, P=0.22), as hypothesized, while in SZ, we did<br />

not observe a similar trend. De novo CNVs had a median size <strong>of</strong> 112<br />

kb and contained a median <strong>of</strong> 2 genes.<br />

Conclusions: Our findings provide evidence that de novo structural<br />

mutations are associated with BD, particularly in cases with an early<br />

disease onset. Genes identified in this study may help to elucidate the<br />

neurobiological basis <strong>of</strong> mood disorders and other psychiatric<br />

disorders.


S1.3 DE NOVO CNV ANALYSIS IMPLICATES SPECIFIC<br />

GENETIC ABNORMALITIES IN THE PATHOGENESIS OF<br />

SCHIZOPHRENIA<br />

M. Owen*(1), A. Pocklington(1), P. Holmans(1), D. Ivanov(1), S.<br />

Grant(2), P. Sklar(3,4,5), S. Purcell(3,4,5), D. Ruderfer(3,4,5), J.<br />

Moran(5), K. Chambert(5), L. van de Lagemaat(2), À. Bayés(2), E.<br />

Fernandez(2), N. Komiyama(2), G. Kirov(1), M. O'Donovan(1)<br />

1. Cardiff University 2. Wellcome Trust Sanger Institute 3.<br />

Massachusetts General Hospital and Harvard Medical School 4.<br />

Center for Human <strong>Genetics</strong> Research 5. Broad Institute<br />

*owenmj@cf.ac.uk<br />

Introduction: Rare, recurrent copy number variants (CNVs) are<br />

known to substantially increase susceptibility to schizophrenia. We<br />

postulated that de novo CNVs are particularly likely to be relevant to<br />

pathogenesis.<br />

Methodology: We used Affymetrix 6.0 arrays to genotype 662<br />

patients suffering with schziophrenia and all their parents. Data were<br />

filtered for the presence <strong>of</strong> LCRs, frequency >1%, >15kb size, and<br />

with the median z-score method. Potential de novo CNVs were<br />

validated with Agilent custom arrays.<br />

Results: We found that de novo CNVsoccurred in 5.1% <strong>of</strong> 662<br />

schizophrenia probands and were enriched for known and novel<br />

pathogenic loci. The occurrence <strong>of</strong> multiple de novo CNVs<br />

implicated EHMT1 as well as DLG2, whose orthog is directly<br />

regulated in Drosophila by the orthlog <strong>of</strong> EHMT1, and other<br />

members <strong>of</strong> the membrane-associated guanylate kinase (MAGUK)<br />

family <strong>of</strong> proteins. Genes within the de novo CNVs were<br />

significantly enriched for membership <strong>of</strong> the postsynaptic density<br />

(PSD p=1.72 x 10 -5 ) largely as a consequence <strong>of</strong> enrichment for<br />

membership <strong>of</strong> N-Methyl-D-Aspartate receptor (NMDAR<br />

p=1.30x10 -5 ) and neuronal activity-regulated cytoskeleton-associated<br />

protein complexes (ARC p=1.07x10 -6 ).<br />

Conclusions: This work provides strong evidence that specific<br />

synaptic complexes implicated in cognitive function and neuronal<br />

plasticity are highly enriched for rare mutations that contribute to<br />

schizophrenia pathogenesis.<br />

16<br />

S1.4 CNVS BETWEEN SCHIZOPHRENIA AND<br />

NEURODEVELOPMENTAL DISORDERS<br />

D. Rujescu*(1), H. Stefanson(2), A. Ingason(1), S. Steinberg(2), S.<br />

Cichon(3), R. Oph<strong>of</strong>f(4), E. Sigurdsson(5), S. Tosato(6), A.<br />

Palotie(7), M. Franziska Degenhardt(3), O. Andreassen(8), T.<br />

Werge(9), M. Rietschel(10), D. Collier(11), D. St Clair(12), K.<br />

Stefansson(1)<br />

1. Division <strong>of</strong> Molecular and Clinical Neurobiology, Department <strong>of</strong><br />

Psychiatry, Ludwig-Maximilians-University, Nußbaumstrasse 7,<br />

80336 Munich, Germany 2. deCODE genetics, Sturlugata 8, IS-101,<br />

Reykjavik, Iceland 3. Institute <strong>of</strong> Human <strong>Genetics</strong> Department <strong>of</strong><br />

Genomics Life & Brain Center University <strong>of</strong> Bonn Sigmund-Freud-<br />

Strasse 25 D-53105 Bonn, Germany 4. Rudolf Magnus Institute <strong>of</strong><br />

Neuroscience and Department <strong>of</strong> Medical <strong>Genetics</strong>, University<br />

Medical Center, 3584 CG Utrecht, The Netherlands 5. Department <strong>of</strong><br />

Psychiatry, National University Hospital, Hringbraut, IS-101<br />

Reykjavik, Iceland 6. Section <strong>of</strong> Psychiatry and Clinical Psychology,<br />

University <strong>of</strong> Verona, Verona, 37134 Verona, Italy 7. Wellcome<br />

Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton,<br />

Cambridge, CB10 1SA, UK 8. Division <strong>of</strong> Mental Health and<br />

Addiction, Oslo University Hospital & Institute <strong>of</strong> Clinical Medicine,<br />

University <strong>of</strong> Oslo, Kirkeveien 166, PO Box 4956 Nydalen, 0424<br />

Oslo, Norway 9. Institute <strong>of</strong> Biological Psychiatry, Mental Health<br />

Centre Sct. Hans Copenhagen University Hospital, DK-4000<br />

Roskilde, Denmark 10. Department <strong>of</strong> Genetic Epidemiology in<br />

Psychiatry, Central Institute <strong>of</strong> Mental Health, University <strong>of</strong><br />

Heidelberg, J5, D-68159 Mannheim, Germany 11. Social, Genetic<br />

and Developmental Psychiatry Centre, Institute <strong>of</strong> Psychiatry, King's<br />

College, London SE5 8AF, UK 12. Department <strong>of</strong> Mental Health,<br />

University <strong>of</strong> Aberdeen, Royal Cornhill Hospital, Aberdeen AB25<br />

2ZD, UK<br />

*dan.rujescu@med.uni-muenchen.de<br />

Introduction: A major challenge in medicine is to understand<br />

genetic, molecular and cellular mechanisms underlying mental<br />

disorders including schizophrenia, which involves complex genetic<br />

and environmental determinants. The last few years brought up a<br />

series <strong>of</strong> studies in schizophrenia which substantially advanced the<br />

knowledge on the genetic causes and had a major impact on the field.<br />

Especially genome-wide studies on Copy Number Variants (CNVs)<br />

raised highest interests. These techniques have shown that a much<br />

higher number <strong>of</strong> CNVs exists in humans than previously recognized.<br />

Methodology: Interestingly, some <strong>of</strong> the discovered deletions in<br />

schizophrenia have been shown to associate with a number <strong>of</strong><br />

unexpected phenotypes, including minor dysmorphic features,<br />

abnormal EEG, significant expressive language deficits, and a<br />

spectrum <strong>of</strong> neuropsychiatric impairments that include epilepsy,<br />

ADHD, autism spectrum disorder, and cognitive impairment varying<br />

from moderate to mild learning disability. Thus, CNV carriers need<br />

to be carefully characterised regarding clinical signs and symptoms,<br />

as well as cognitive and behavioural features across diagnostic<br />

boundaries.<br />

Results: In a genome-wide search for CNVs associating with<br />

schizophrenia performed by the SGENE consortium, a population<br />

based sample was used to identify de novo CNVs. Beside these<br />

finding at 1q21.1, 15q11.2 and 15q13.3, further CNV studies were<br />

performed. e.g. CNVs were reported in the NRXN1 gene (2p16.3),<br />

furthermore, 15q11-q13 duplications were detected in a<br />

region overlapping the Prader-Willi/Angelman syndrome or at<br />

16p13.1, which has been implicated in childhood-onset<br />

developmental disorders.<br />

Conclusions: All these studies as well as new results will be<br />

presented and critically discussed.


SYMPOSIUM 2: GENES TO BIOLOGY IN<br />

UNDERSTANDING OF NICOTINE DEPENDENCE<br />

S2.1 SMOKING AND GENETIC RISK VARIATION ACROSS<br />

POPULATIONS OF EUROPEAN, ASIAN, AND AFRICAN<br />

ANCESTRY - A META-ANALYSIS OF CHROMOSOME<br />

15Q25<br />

L. Chen*, L. Bierut, C. Meta-Analysis Group<br />

Department <strong>of</strong> Psychiatry, Washington University School <strong>of</strong><br />

Medicine<br />

*chenli@psychiatry.wustl.edu<br />

Introduction: Recent meta-analyses <strong>of</strong> genetic data from subjects <strong>of</strong><br />

European ancestry show strong evidence for association between<br />

smoking quantity and multiple genetic variants on chromosome<br />

15q25. This study extends the examination <strong>of</strong> association between<br />

distinct loci in the CHRNA5-CHRNA3-CHRNB4 region and smoking<br />

quantity to populations <strong>of</strong> Asian and African ancestry to refine<br />

specific reported associations.<br />

Methodology: The analyses <strong>of</strong> a dichotomized cigarettes smoked per<br />

day (CPD) phenotype in 27 datasets (subjects <strong>of</strong> European, Asian,<br />

and African American ancestry N=14,786, 6,889, 10,912<br />

respectively, for a total <strong>of</strong> 32,587 subjects who smoked) were metaanalyzed.<br />

To refine the reported loci associated with heavy smoking,<br />

we examined all available variants in each population.<br />

Results: We demonstrate the association between smoking quantity<br />

and genetic loci in the chromosome 15q25 region across all three<br />

populations and narrow the region <strong>of</strong> association. The most strongly<br />

associated variant seen consistently across the three populations is<br />

rs16969968 (OR=1.33, 95%C.I.=1.25-1.42, p=1.1x10 -17 ). Another<br />

locus displayed a consistent signal in both European and Asian<br />

ancestry, but not in African American ancestry.<br />

Conclusions: Multiple distinct genetic variants on chromosome<br />

15q25 are associated with smoking quantity across diverse<br />

populations. Further, our results narrow the regions <strong>of</strong> association.<br />

The observed consistent associations <strong>of</strong> rs16969968 with heavy<br />

smoking across different populations, combined with its functional<br />

significance, suggest that rs16969968 is most likely a causative<br />

variant in this region. Additional distinct genetic variants may also<br />

be associated with heavy smoking across populations. Using the<br />

cross-population study paradigm provides valuable insights to inform<br />

future biological experiments.<br />

17<br />

S2.2 NICOTINIC RECEPTORS IN THE HABENULO-<br />

INTERPEDUNCULAR TRACT REGULATE NICOTINE<br />

INTAKE<br />

P. Kenny*<br />

Scripps Florida<br />

*pjkenny@scripps.edu<br />

Introduction: Tobacco smoking results in more than 5 million<br />

deaths each year and accounts for almost 90% <strong>of</strong> all deaths from lung<br />

cancer. Nicotine is the major reinforcing component in tobacco<br />

smoke responsible for addiction. Nicotine acts in the brain through<br />

neuronal nicotinic acetylcholine receptors (nAChRs). Twelve<br />

neuronal nAChR subunits have been identified: nine αsubunits (α2–<br />

α10) and three β subunits (β2–β4). The predominant nAChR subtypes<br />

in brain are those containing α4 and β2 subunits. A major advance in<br />

our understanding <strong>of</strong> smoking behaviour is the finding that variation<br />

in the α5/α3/β4 nAChR subunit gene cluster located in chromosome<br />

region 15q25 significantly increases risk <strong>of</strong> tobacco addiction. In<br />

particular, polymorphisms in the α5 subunit gene (CHRNA5), which<br />

result in decreased function <strong>of</strong> the subunit, increase vulnerability to<br />

tobacco addiction. In addition, polymorphisms in the α3 subunit also<br />

greatly increase vulnerability to tobacco dependence. Here, we<br />

investigated the mechanisms through which α5-containing (denoted<br />

as α5*) nAChRs and a3* nAChRs influence the reinforcing<br />

properties <strong>of</strong> nicotine. As α5* and a3* nAChRs are densely expressed<br />

in the medial habenula (MHb) and/or interpeduncular nucleus (IPN),<br />

we also assessed the role for these nAChRs in the habenulointerpeduncular<br />

tract in regulating nicotine intake.<br />

Methodology: The reinforcing properties <strong>of</strong> nicotine were measured<br />

in rats and mice by means <strong>of</strong> the intravenous nicotine selfadministration<br />

behavior. Nicotine reinforcement was assessed in<br />

wildtype (WT) and α5 nAChR subunit knockout (α5 KO) mice. The<br />

expression <strong>of</strong> α5 nAChR subunits was “rescued” in discrete brain<br />

regions <strong>of</strong> the α5 KO mice through lenti-virus-mediated reexpression.<br />

Conversely, α5 or α3 naChR subunits were knocked<br />

down in discrete brain regions in rats by means <strong>of</strong> lentivirus-mediated<br />

delivery <strong>of</strong> short-hairpin interfering RNAs against these subunits.<br />

Finally, the effects <strong>of</strong> nicotine on brain reward systems were<br />

measured using alterations in intracranial self-stimulation (ICSS)<br />

thresholds in rats.<br />

Results: The α5 KO mice self-administered far greater quantities <strong>of</strong><br />

nicotine than their WT counterparts, and this effect was most<br />

evidence when high unit doses <strong>of</strong> nicotine were available. Virusmediated<br />

re-expression <strong>of</strong> α5 subunits in the habenulointerpeduncular<br />

tract <strong>of</strong> KO mice, achieved through virus infusions<br />

into MHb, normalized the increased intake detected in KO mice.<br />

Conversely, virus-mediated knockdown <strong>of</strong> α5 subunits in the<br />

habenulo-interpeduncular tract resulted in greater nicotine intake in<br />

rats. Knockdown <strong>of</strong> α5 subunits in MHb <strong>of</strong> rats did not alter the<br />

rewarding effects <strong>of</strong> nicotine, measured as nicotine-lowered ICSS<br />

thresholds, but abolished the inhibitory effects <strong>of</strong> higher nicotine<br />

doses on brain reward systems, reflected in nicotine-elevated ICSS<br />

thresholds. Finally, knockdown <strong>of</strong> α3 subunits in MHb or IPN <strong>of</strong> rats<br />

also increased their responding for nicotine.<br />

Conclusions: Taken together, these findings demonstrate that α5*<br />

and a3* nAChRs in the habenulo-interpeduncular tract negatively<br />

regulate nicotine self-administration behavior in rats and mice. This<br />

action is related to the fact that these nAChRs regulate the inhibitory<br />

effects <strong>of</strong> higher nicotine doses on brain reward circuitries. Genetic<br />

variation that results in diminished function <strong>of</strong> nAChRs containing α5<br />

and/or a3 subunits may therefore increase vulnerability to tobacco<br />

addiction by reducing the aversive effects <strong>of</strong> nicotine, permitting<br />

greater quantities <strong>of</strong> tobacco to be consumed.


S2.3 IMPORTANCE OF α4β2α5 AND α6β2β3 * ACHRS IN<br />

POTENTIAL DRUGS FOR NICOTINE DEPENDENCE<br />

J. Lindstrom*, A. Kuryatov<br />

University <strong>of</strong> Pennsylvania<br />

*jslkk@mail.med.upenn.edu<br />

Introduction: Previous studies showed that α5 398N is a risk variant<br />

for nicotine dependence, lung cancer and certain cognitive defects<br />

and that (α4β2)2α5 AChRs with the risk sequence exhibited reduced<br />

function. α5, α3, and β4 genes are located contiguously in the<br />

genome. (α3β4)2α5 and (α3β2)2α5 are prominent postsynaptic AChR<br />

subtypes in autonomic ganglia and enteric neurons, but are rare in<br />

brain. (α4β2)2α5 AChRs are a significant presynaptic subtype among<br />

brain α4β2 * AChRs. Knock out <strong>of</strong> α5 in the medial habenula<br />

increases nicotine self-administration. Previous studies showed that<br />

(α6β2)(α4β2)β3 presynaptic AChRs on dopaminergic neurons are the<br />

most sensitive subtype to activation by nicotine. Knock out <strong>of</strong> either<br />

α4, α6 or β2 subunits in the ventral tegmental area prevents selfadministration<br />

<strong>of</strong> nicotine. Mixtures <strong>of</strong> free α6, α4, β2 and β3<br />

subunits do not result in expression <strong>of</strong> homogenous populations <strong>of</strong><br />

functional AChRs <strong>of</strong> the desired complex subtypes.<br />

Methodology: Human α3 and α4 AChR subtypes containing α5<br />

subunits with either D or N398 were assayed for function<br />

electrophysiologically after expression in Xenopusoocytes. Human<br />

AChR subtypes containing α5 subunits with D398 expressed in<br />

permanently transfected HEK cell lines were assayed for function<br />

using a fluorescent indicator <strong>of</strong> membrane potential. Functional<br />

human α6β2β3 * AChR subtypes <strong>of</strong> defined subunit composition and<br />

order were expressed in Xenopusoocytes using concatameric<br />

constructs with (AGS)n linkers between the C-terminal amino acid <strong>of</strong><br />

one subunit and the N-terminal amino acid <strong>of</strong> the next.<br />

Results: The α5 N398 risk variant reduced Ca ++ permeability and<br />

increased desensitization in (α4β2)2α5 but not (α3β4)2α5 or<br />

(α3β2)2α5 AChRs, compared to the α5 D398 variant. Sensitivity to<br />

activation was not altered. The high sensitivity to activation and<br />

desensitization <strong>of</strong> (α4β2)2α5 AChRs results in a narrow concentration<br />

range in which activation and desensitization curves overlap. This<br />

region centers at 0.2 µM nicotine, a concentration sustained in<br />

smokers. This concentration desensitizes 60% <strong>of</strong> these AChRs and<br />

permits smoldering activation <strong>of</strong> the remainder. The low sensitivity to<br />

activation and desensitization <strong>of</strong> (α3β4)2α5 by nicotine results in<br />

negligible activation or desensitization by 0.2 µM nicotine. α6β2β3 *<br />

AChRs can be expressed as concatamers using 1, 2, 3, or 4 linkers.<br />

Linking the accessory β3 subunit to α6 or α4 is critical to insure<br />

assembly. Concatameric pentamers are sensitive to activation by<br />

nicotine and by putative α4β2 AChR-specific agonists such as<br />

varenicline and sazetidine.<br />

Conclusions: The reduced function <strong>of</strong> (α4β2)2α5 AChRs with the<br />

risk variant α5 N398 and the increased nicotine consumption<br />

observed when α5 is knocked out suggests that it would be valuable<br />

to develop positive allosteric modulator drugs which could increase<br />

the function <strong>of</strong> AChRs with α5 subunits. The use <strong>of</strong> cell lines<br />

expressing (α4β2)2α5 and other subtypes provides methods for<br />

selecting such drugs. The sensitivity <strong>of</strong> (α6β2)(α4β2)β3 AChRs to<br />

activation by nicotine and smoking cessation drugs such as<br />

varenicline, as well as the observation that knock out <strong>of</strong> α4, α6, or β2<br />

subunits prevents nicotine self-administration, suggests that<br />

antagonists or negative allosteric modulators specific for α6β2β3 *<br />

AChRs would be useful for smoking cessation. The use <strong>of</strong><br />

concatameric α6β2β3 * AChRs provides a method for selecting such<br />

drugs.<br />

18<br />

S2.4 REGULATORY POLYMORPHISMS IN NICOTINE<br />

DEPENDENCE RISK GENES<br />

R. Smith*, W. Sadee<br />

The Ohio State University<br />

*smith.4051@osu.edu<br />

Introduction: Genetic variants within nicotinic receptors have been<br />

shown to modulate an individual’s risk for nicotine dependence.<br />

Among the most reproducible risk factor is a non-synonymous<br />

polymorphism (rs16969968) within the alpha5 nicotinic receptor<br />

subunit gene, CHRNA5. Genetic factors also contribute to CHRNA5<br />

mRNA expression and risk for substance abuse, but the specific<br />

variants responsible for changes in mRNA expression are unclear.<br />

Methodology: To determine the presence <strong>of</strong> cis-acting functional<br />

genetic variants contributing to CHRNA5 mRNA expression, we<br />

measured allelic expression <strong>of</strong> CHRNA5 within post-mortem human<br />

prefrontal cortex brain tissue. Using allelic expression as a<br />

phenotype, we scanned the CHRNA5 gene locus for polymorphisms<br />

responsible for imbalanced allelic expression. Polymorphisms<br />

determined to be responsible for allelic expression were genotyped in<br />

our full brain cohort and total CHRNA5 expression was compared<br />

across genotype using quantitative PCR. We then tested these<br />

expression-related variants for clinical association with nicotine<br />

dependence.<br />

Results: We uncovered a functional genetic locus 13.5kb upstream <strong>of</strong><br />

CHRNA5 acting as an enhancer <strong>of</strong> CHRNA5 mRNA<br />

expression. Minor allele carriers for enhancer variants displayed >2fold<br />

differences in allelic expression, while individuals homozygous<br />

for the enhancer minor alleles expressed >4-fold more cortical<br />

CHRNA5 mRNA. Haplotype structure analysis predicts that the<br />

enhancer SNPs rarely occur on the same allele as the previously<br />

implicated non-synonymous variant. Taking into account this<br />

relationship, enhancer region SNPs confer significant risk only when<br />

you control for the presence <strong>of</strong> the non-synonymous variant in a joint<br />

SNP analysis.<br />

Conclusions: Polymorphisms within a transcriptional enhancer<br />

located 13.5kb upstream <strong>of</strong> CHRNA5 modulate CHRNA5 mRNA<br />

transcription. These enhancer variants confer risk for nicotine<br />

dependence in a context-dependent manner. Understanding the<br />

biological functions <strong>of</strong> these clinically relevant genetic variants<br />

revealed a SNP-SNP interaction increasing risk for nicotine<br />

dependence that is otherwise obscured by haplotype structure when<br />

using traditional clinical association methods. These findings aid in<br />

the construction <strong>of</strong> complex biological or genetic models <strong>of</strong> nicotine<br />

dependence that mimic human factors. The relevance <strong>of</strong> the enhancer<br />

CHRNA5 polymorphism in other brain regions and peripheral tissues<br />

(e.g., the lung) needs further study. Moreover, identification <strong>of</strong><br />

functional CHRNA5 variants facilitates the analysis <strong>of</strong> gene-gene<br />

interactions with respect to nicotine and other drug addictions.


S2.5 A META-ANALYSIS OF GENE-ENVIRONMENT<br />

INTERACTIONS OF HEAVY SMOKING WITH THE<br />

CHRNA5-A3-B4 GENE CLUSTER<br />

S. Hartz*(1), S. Short(2), P. Kraft(3), L. Bierut(1)<br />

1. Washington University 2. Brown University 3. Harvard University<br />

*hartzs@wustl.edu<br />

Introduction: Recent studies have shown an association between<br />

cigarettes per day (CPD) and a nonsynonymous SNP in CHRNA5,<br />

rs16969968. There is conflicting evidence regarding whether age <strong>of</strong><br />

onset <strong>of</strong> regular smoking, a strong predictor <strong>of</strong> nicotine dependence,<br />

modifies this association.<br />

Methodology: We conducted a large meta-analysis to determine<br />

whether the association between rs16969968 and CPD is modified by<br />

age <strong>of</strong> onset <strong>of</strong> regular smoking. Other variables related to both CPD<br />

and age <strong>of</strong> onset, including decade <strong>of</strong> birth, educational attainment,<br />

and gender, were also included in analysis.<br />

Results: The association between rs16969968 and CPD is higher<br />

among early-onset smokers (age <strong>of</strong> onset <strong>of</strong> regular use ≤ 16)<br />

compared to later-onset smokers.<br />

Conclusions: The increased association between CPD and<br />

rs16969968 that we found among early-onset smokers supports the<br />

epidemiological observation <strong>of</strong> an increased rate <strong>of</strong> nicotine<br />

dependence among early-onset smokers. This gives further credence<br />

to public health interventions targeting adolescent smoking.<br />

19<br />

SYMPOSIUM 3: EXPLORING THE MECHANISMS<br />

UNDERLYING GENOME-WIDE SIGNIFICANT<br />

ASSOCIATIONS WITH PSYCHOSIS<br />

S3.1<br />

GENOME-WIDE SIGNIFICANT RISK VARIANTS FOR<br />

PSYCHOSIS AND NEURAL INTERMEDIATE PHENOTYPES<br />

A. Meyer-Lindenberg*<br />

Central Institute <strong>of</strong> Mental Health<br />

*a.meyer-lindenberg@zi-mannheim.de<br />

Introduction: While GWAS will probably not provide all answers<br />

about the genetics <strong>of</strong> schizophrenia, any common variant that does<br />

survive the extreme amount <strong>of</strong> statistical thresholding that this<br />

method requires certainly merits study using intermediate imaging<br />

phenotypes. Of those variants, the one with the strongest support is<br />

zinc finger protein 804A (ZNF804A) encoding a zinc-finger protein<br />

<strong>of</strong> unknown, but possibly regulatory function. Like many candidate<br />

gene variants, ZNF804A is pleiotropic on the level <strong>of</strong> psychiatric<br />

diagnoses, also being associated with bipolar disorder.<br />

Methodology: We use an imaging genetics approach in a sample <strong>of</strong><br />

n=110 Germans <strong>of</strong> German descent genotyped for ZNF804A risk<br />

SNPs and studied using functional neuroimaging,<br />

Results: In functional neuroimaging with an n-back working memory<br />

probe, healthy carriers <strong>of</strong> risk genotypes exhibit no changes in<br />

regional activity. However, they did exhibit pronounced gene dosagedependent<br />

alterations in functional connectivity, which was decreased<br />

from DLPFC across hemispheres and increased with hippocampus,<br />

similar to findings in patients. These connectivity abnormalities were<br />

specific to working memory. Subsequent workshowed an inability to<br />

downregulate key parts <strong>of</strong> the mentalizing system in conjunction with<br />

impaired connectivity <strong>of</strong> this system to DLPFC, suggesting possible<br />

downstream functional activation effects <strong>of</strong> impaired prefrontal<br />

connectivity that mirror findings in patients. Interestingly,<br />

abnormally increased coupling <strong>of</strong> amygdala was also observed, a<br />

phenotype unlikely to be related to heritable risk for schizophrenia<br />

and therefore possibly related to risk for bipolar disorder, where<br />

similar findings in patients have been described.<br />

Conclusions: Neural intermediate phenotypes related to risk for<br />

schizophrenia and bipolar disorder through Z NF804A genetic risk<br />

variants can be defined and map onto systems previously implicated<br />

in patients.


S3.2 COGNITIVE & IMAGING CORRELATES OF<br />

GENOME-WIDE SIGNIFICANT RISK VARIANTS FOR<br />

SCHIZOPHRENIA<br />

G. Donohoe, E. Rose, D. Morris, A. Hargreaves, M. Gill, A. Corvin<br />

Neuropsychiatric genetics research group, Department <strong>of</strong> psychiatry,<br />

Trinity College Institute <strong>of</strong> Neuroscience and Institute <strong>of</strong> Molecular<br />

Medicine, Trinity College Dublin<br />

Introduction: Genome wide association studies have resulted in the<br />

identification <strong>of</strong> a number <strong>of</strong> novel genetic loci for schizophrenia and<br />

related disorders. Understanding the functional impact <strong>of</strong> these<br />

variants on brain structure and function is crucial to understand their<br />

role in disease pathology.<br />

Methodology: We pr<strong>of</strong>ile the neuropsychological,<br />

electrophysiological, and neuro-imaging characteristics <strong>of</strong> a number<br />

<strong>of</strong> these variants, including ZNF804A.<br />

Results: Specifically, we will use this data to suggest that while the<br />

risk associated with some genetics variants is being mediated by an<br />

influence on these intermediate phenotypes, other risk variants<br />

delineate illness subtypes in which cognitive deficits are a less<br />

prominent feature.<br />

Conclusions: The implications <strong>of</strong> these distinctions for the<br />

intermediate phenotypes approach will be discussed.<br />

20<br />

S3.3 FUNCTIONAL ANALYSIS OF THE ZNF804A AND TCF4<br />

SCHIZOPHRENIA SUSCEPTIBILITY GENES<br />

D. Blake, C. Tinsley*<br />

Cardiff University<br />

*tinsleycl@cardiff.ac.uk<br />

Introduction: Recent genome-wide association (GWA) studies have<br />

led to the discovery <strong>of</strong> several new risk alleles for schizophrenia. For<br />

example, intragenic SNPs in the ZNF804A (2q32.1) and TCF4<br />

(18q21.1) genes are associated with a moderate increase in<br />

schizophrenia risk. Whilst TCF4 is a relatively well-characterised<br />

transcription factor little is known about the biological function <strong>of</strong><br />

ZNF804A. The presence <strong>of</strong> a C2H2 class <strong>of</strong> zinc finger at the Nterminus<br />

<strong>of</strong> the protein has led to the speculation that ZNF804A may<br />

be involved in transcription. The aim <strong>of</strong> this study was to determine<br />

the function <strong>of</strong> ZNF804A and to identify other genes whose<br />

expression may be regulated by ZNF804A and TCF4.<br />

Methodology: Exon array, interaction trap, chromatin<br />

immunoprecipitation, mass spectrometry.<br />

Results: Complementary strategies have been used to define the<br />

interactomes <strong>of</strong> ZNF804A and TCF4. We have generated a network<br />

<strong>of</strong> protein:protein interactions around the ZNF804A and TCF4 nodes<br />

that implicate both proteins as regulators <strong>of</strong> gene expression. Several<br />

<strong>of</strong> these interactors include genes that are either transcriptional<br />

activators or co-activators and epigenetic modulators <strong>of</strong> gene<br />

expression. We have also created cell lines that inducibly overexpress<br />

ZNF804A and TCF4 or where each <strong>of</strong> the two genes has<br />

been knocked-down. These experiments have shown that bidirectional<br />

changes in the levels <strong>of</strong> ZNF804A are associated with<br />

altered gene expression defining an initial set <strong>of</strong> target genes in<br />

human cells. Finally, we have independently validated a number <strong>of</strong><br />

physiological targets for TCF4 using luciferase assays and chromatin<br />

immunoprecipitation. Similar experiments with ZNF804A are<br />

ongoing.<br />

Conclusions: Our data show that ZNF804A and TCF4 regulate the<br />

expression <strong>of</strong> a subset <strong>of</strong> genes in human cells. The relevance <strong>of</strong><br />

these targets to the biology <strong>of</strong> schizophrenia and allied<br />

neuropsychiatric disorders is currently being determined.


S3.4 MOLECULAR AND CELLULAR MECHANISMS<br />

UNDERLYING GENOME-WIDE SIGNIFICANT<br />

ASSOCIATION BETWEEN ZNF804A AND PSYCHOSIS<br />

M. Hill*, F. Buonocore, A. Jeffries, R. Dobson, J. Price, N. Bray<br />

Institute <strong>of</strong> Psychiatry, King's College London<br />

*matthew.hill@kcl.ac.uk<br />

Introduction: The first genome-wide significant finding for the<br />

broad phenotype <strong>of</strong> psychosis (encompassing both schizophrenia and<br />

bipolar disorder) was with SNP rs1344706 within the ZNF804A gene.<br />

Re-sequencing and detailed association analyses have not uncovered<br />

a variant at this locus that is more strongly associated. However, the<br />

molecular and cellular mechanisms by which it impacts on risk for<br />

psychosis are unknown. To address the fundamental mechanisms<br />

underlying ZNF804A association with psychosis, we 1) performed<br />

electrophoretic mobility shift assays (EMSA) to assess potential<br />

allelic differences in the capacity <strong>of</strong> rs1344706 to bind nuclear<br />

proteins, 2) measured allelic expression <strong>of</strong> ZNF804A in foetal and<br />

adult human brain and 3) manipulated the expression <strong>of</strong> ZNF804A in<br />

human neural cells to determine its impact on the cellular<br />

transcriptome.<br />

Methodology: DNA-protein interactions were investigated by EMSA<br />

using extracts from three human neural cell lines and subsequent<br />

incubation with allele specific probes. Relative allelic expression <strong>of</strong><br />

ZNF804A was determined in 12 foetal brain samples and across<br />

multiple brain regions in 13 adult subjects. For 3 brain regions<br />

additional subjects were also investigated. Statistically significant<br />

departure from the expected 1:1 allelic ratio <strong>of</strong> gene expression was<br />

assessed by Mann-Whitney tests. RNA interference using two<br />

separate small interfering RNAs (siRNA) was used to manipulate<br />

ZNF804A expression in a human neural progenitor cell line. The<br />

effects <strong>of</strong> ZNF804A knockdown on the cellular transcriptome were<br />

compared to a control siRNA by gene expression microarray (n=4 for<br />

each <strong>of</strong> the 3 conditions). Gene Ontology analysis was performed on<br />

all nominally significant differentially expressed transcripts that were<br />

shared by the two ZNF804A siRNA. High confidence single<br />

transcript differences were determined by Significance Analysis <strong>of</strong><br />

Microarrays (SAM) at a false discovery rate <strong>of</strong>


S4.2 INTEGRATING RNA-SEQ AND GENOME-WIDE<br />

ASSOCIATION TO IDENTIFY RISK GENES FOR<br />

SCHIZOPHRENIA<br />

X. Chen*(1), Y. Zhang(2), J. Chen(1), J. Xu(2), F. Xu(2), Z.<br />

Peng(2), T. Guennel(3), M. Reimers(3), S. Bacanu(1), J. Sun(4), Z.<br />

Zhao(4), K. Kendler(1)<br />

1. Virginia Commonwealth University 2. Beijing Genomic Institute<br />

3. Virginia Commonwealth University 4. Vanderbilt University<br />

*xchen@vcu.edu<br />

Introduction: Genome-wide association studies have identified<br />

several promising candidate genes for schizophrenia, many more risk<br />

genes remain to be discovered. Due to the small effects <strong>of</strong> these<br />

candidate genes, GWA alone is not effective to identify them. In the<br />

last few years, the progress <strong>of</strong> DNA sequencing technologies has<br />

made it practical to sequence the entire transcriptome for a reasonable<br />

number <strong>of</strong> subjects. Both GWA and RNA-seq are systematic<br />

approaches to study the entire genome. Integration <strong>of</strong> RNA-seq data<br />

with GWA may increase the power <strong>of</strong> genetic association studies and<br />

improve selection <strong>of</strong> top candidates.<br />

Methodology: We conducted mRNA sequencing for 82 post-mortem<br />

brain samples (Brodmann area 24) from Stanley Medical Research<br />

Institute, and performed differential expression analyses between<br />

patients <strong>of</strong> schizophrenia and bipolar disorder and healthy controls.<br />

We also identified SNPs influencing gene expression (eQTL<br />

analyses) in this and other datasets. These data were integrated with<br />

GWA data from the Molecular <strong>Genetics</strong> <strong>of</strong> Schizophrenia sample to<br />

identify promising candidates for further studies.<br />

Results: In the RNA-seq expression analyses, we found that<br />

substantially more genes (FDR q ≤ 0.05, N = 608) are differentially<br />

expressed between schizophrenic subjects and healthy controls than<br />

that observed in microarray studies using the same subjects. We also<br />

found that the differential expression <strong>of</strong> many genes in schizophrenia<br />

patients relative to controls is correlated with the expression changes<br />

<strong>of</strong> bipolar disorder patients, suggesting a shared liability between the<br />

two disorders. In eQTL analyses <strong>of</strong> the SPATA7-PTPN21-EML5<br />

locus, one <strong>of</strong> the top ranked candidate genes from gene-based GWA<br />

analyses <strong>of</strong> the MGS dataset, SNPs influencing the expression <strong>of</strong><br />

these genes are found to be associated with schizophrenia in a metaanalysis<br />

<strong>of</strong> several independent samples, supporting the idea that<br />

integration <strong>of</strong> expression data with GWA can improve identification<br />

<strong>of</strong> candidate genes with small effects.<br />

Conclusions: Expression pr<strong>of</strong>iling by RNA-seq is a powerful<br />

approach to identify differentially expressed genes between diseased<br />

and control subjects. Our expression data provided empirical<br />

evidence that schizophrenia and bipolar disorder may share broad<br />

liability. Integration <strong>of</strong> eQTL and GWA analyses can improve the<br />

power to detect candidate genes with modest effect sizes.<br />

22<br />

S4.3 BRAIN EQTLS AND FUNCTION-BASED GWAS OF<br />

BIPOLAR DISORDER IDENTIFIED NOVEL DISEASE RISK<br />

GENES<br />

N. Cox*(1), E. Gamazon(1), J. Badner(2), L. Cheng(2), F. Pibiri(2),<br />

C. Zhang(2), C. Chen(2), K. Grennan(2), E. Gershon(2), D.<br />

Nicolae(1), B. Consortium(1,2), C. Liu(2)<br />

1. Section <strong>of</strong> Genetic Medicine University <strong>of</strong> Chicago 2. Dept. <strong>of</strong><br />

Psychiatry and Behavioral Medicine University <strong>of</strong> Chicago<br />

*ncox@bsd.uchicago.edu<br />

Introduction: Genome-wide association studies (GWAS) with<br />

psychiatric diseases have yielded limited findings, including a few<br />

genome-wide significant associated genes, one polygenic model, and<br />

a few rare copy number variations. Functional annotation <strong>of</strong> SNPs<br />

may be able to assist the re-evaluation <strong>of</strong> the genome-wide<br />

association results to recover some lost heritability. One major<br />

function <strong>of</strong> SNPs is to affect gene expression.<br />

Methodology: We used the genome-wide association study to map<br />

quantitative trait loci <strong>of</strong> gene expression in human brain. Extensive<br />

statistical analyses take into account covariates (such as brain pH)<br />

and batch effects in the microarray data. Functional SNPs are then<br />

used to perform function-based GWAS analysis to identify novel<br />

disease susceptibility genes <strong>of</strong> bipolar disorder.<br />

Results: We have identified thousands <strong>of</strong> SNPs that are associated<br />

with gene expression levels, including different splicing is<strong>of</strong>orms in<br />

two brain regions. We are applying these data into the re-evaluation<br />

<strong>of</strong> existing genome-wide association studies <strong>of</strong> bipolar disorder and<br />

schizophrenia, and have identified novel genes that are associated<br />

with disease but have been missed in previous gene hunting. Three<br />

genes showed genome-wide significant associations (p < 1E-6) in one<br />

sample set and were replicated at nominal significance level in a<br />

second dataset.<br />

Conclusions: Biological understanding <strong>of</strong> genetic variants could<br />

lead to new understanding <strong>of</strong> GWAS data, and to the discovery <strong>of</strong><br />

novel disease risk genes.


S4.4 WHOLE GENOME SEQUENCING OF INDIVIDUALS<br />

FROM THE PORTUGUESE ISLAND COHORT (PIC)<br />

A. Fanous(1,2,3,4), Z. Zhao(5), J. Sun(5), R. Amdur(1,2), O.<br />

Evgrafov(4), A. Clark(4), H. Medeiros(4), M. Pato(4), C. Pato(4),<br />

J. Knowles*(4)<br />

1. 1Washington VA Medical Center 2. Georgetown University<br />

School <strong>of</strong> Medicine 3. Virginia Commonwealth University School <strong>of</strong><br />

Medicine 4. Keck School <strong>of</strong> Medicine <strong>of</strong> the University <strong>of</strong> Southern<br />

California 5. Departments <strong>of</strong> Psychiatry, Biomedical Informatics, and<br />

Cancer Biology, Vanderbilt University Medical Center<br />

*knowles@med.usc.edu<br />

Introduction: We have piloted using whole genome sequencing<br />

(PE100@32X) with Illumina DNA sequencers for one individual in<br />

each <strong>of</strong> 23 multiplex PIC pedigrees segregating schizophrenia (n=17)<br />

or bipolar disorder (n=5).<br />

Methodology: These data were mapped with BWA, or PerM and<br />

variants were called with SAMtools and GATK, and then annotated<br />

with either SVA or ANNOVAR.<br />

Results: Each individual has approximately 3.5 million SNPs, <strong>of</strong><br />

which ~100-120 are nonsense and ~10,000 are missense. Each<br />

annotated sequence variation was then weighted by the familyspecific<br />

evidence <strong>of</strong> linkage derived from an autosomal dominant,<br />

affecteds only, analyses.<br />

Conclusions: We will present the results <strong>of</strong> these analyses and the<br />

plan for our future work.<br />

23<br />

S4.5 WHOLE METHYLOME STUDIES OF PSYCHIATRIC<br />

CONDITIONS<br />

E. van den Oord*, G. Rudolf, S. Nerella, J. Bukszár, A. Khachane,<br />

J. McClay, K. Åberg<br />

VCU<br />

*ejvandenoord@vcu.edu<br />

Introduction: Several challenges exist when performing DNA<br />

methylation studies <strong>of</strong> psychiatric conditions. First, many <strong>of</strong> the<br />

disease processes involve the brain. However, as the procurement <strong>of</strong><br />

brain tissue is not possible in (living) patients, we <strong>of</strong>ten have to<br />

perform such studies in DNA from peripheral whole blood or<br />

Epstein-Barr virus (EBV) transformed cell lines. Second, CpG sites<br />

may not show inter-individual methylation variation in the tissue that<br />

is being studied. These “non-variable” sites need to be eliminated<br />

prior to analyses to avoid false discoveries and improve statistical<br />

power to detect biologically relevant markers. A third challenge<br />

involves the interpretation <strong>of</strong> significant associations between<br />

methylation markers and case-control status. For example, an<br />

association may be the results <strong>of</strong> the methylation marker affecting<br />

disease susceptibility or could be “spurious” as a result <strong>of</strong>, for<br />

instance, medication use. Finally, methylation studies have<br />

historically been restricted to the study <strong>of</strong> CpG sites in a limited<br />

number <strong>of</strong> candidate genes. However, to find methylation markers it<br />

may be needed to screen all CpG sites across the entire methylome<br />

for association with disease status.<br />

Methodology: We focus on next-generation sequencing as a costeffective<br />

approach for whole methylome studies. To process the<br />

massive amount <strong>of</strong> data generated in such studies and address the<br />

above mentioned concerns, we present a data analysis pipeline we<br />

developed consisting <strong>of</strong> the following steps 1) alignment & basic QC,<br />

2) estimation <strong>of</strong> methylation levels, 3) selection <strong>of</strong> methylation<br />

variable sites, 4) annotation, and 5) association analyses. Our data<br />

analysis pipeline is illustrated using a sample <strong>of</strong> 1,500 subjects + 75<br />

technical replicates. To study the use <strong>of</strong> whole blood and EBV DNA<br />

in methylation studies <strong>of</strong> psychiatric conditions, we compared 1)<br />

whole methylome pr<strong>of</strong>iles in cortex, hippocampus, and whole blood<br />

from 10 inbred mice and 2) whole blood and EBV DNA from 10<br />

human subjects.<br />

Results: Consistent with the literature, our estimator suggests that<br />

over 80% <strong>of</strong> all CpG are methylated. However, only a proportion <strong>of</strong><br />

these sites show substantial inter-individual methylation variation in<br />

whole blood and should be used in subsequent association studies.<br />

We find evidence that methylation variable sites share bioinformatics<br />

features and can be correlated across larger genomic regions.<br />

Methylation levels can be measured reliably in whole blood and<br />

methylation pr<strong>of</strong>iles in whole blood shows overlap with methylation<br />

pr<strong>of</strong>iles in brain. Caution seems required when interpreting results<br />

from methylation studies using EBV DNA.<br />

Conclusions: Whole methylome studies <strong>of</strong> psychiatric conditions are<br />

economically and technically feasible and <strong>of</strong>fer great potential to<br />

supplement tradition sequence based genetic studies.


S4.6 PATHWAY AND NETWORK ANALYSIS OF GWAS<br />

AND RNA-SEQ FOR SCHIZOPHRENIA<br />

Z. Zhao*(1), P. Jia(1), L. Wang(1), J. Sun(1), A. Fanous(2,3), J.<br />

Xu(4), I. Consortium(5), K. Kendler(2), X. Chen(2)<br />

1. Vanderbilt University 2. Virginia Commonwealth University 3.<br />

Washington VA Medical Center 4. SUNY at Buffalo School <strong>of</strong><br />

Medicine 5. International Schizophrenia Consortium<br />

*zhongming.zhao@vanderbilt.edu<br />

Introduction: Genome-wide association studies (GWAS) have<br />

become a popular approach for searching for common genetic<br />

variants which increase susceptibility to complex diseases or traits.<br />

At the transcriptome level, RNA sequencing (RNA-Seq) is rapidly<br />

emerging as a powerful tool for identifying differentially expressed<br />

genes in diseases. Recently, pathway or network-based analysis <strong>of</strong><br />

genomic datasets has emerged as an alternative but potentially<br />

powerful approach to searching for disease causal genes, assuming a<br />

complex disease might have resulted from a number <strong>of</strong> genes which<br />

disrupt one or more pathways or protein complexes.<br />

Methodology: We applied several pathway enrichment methods to<br />

schizophrenia GWAS datasets including Gene Set Enrichment<br />

Analysis (GSEA), hypergeometric test, and a generalized additive<br />

model (GAM). At the network level, we developed a dense module<br />

searching (DMS) method to identify candidate subnetworks or genes<br />

for complex diseases by integrating the association signal from<br />

GWAS datasets into the human protein-protein interaction (PPI)<br />

network. Finally, we performed a preliminary pathway analysis <strong>of</strong><br />

schizophrenia RNA-Seq data.<br />

Results: We found several pathways that were top ranked and likely<br />

associated with schizophrenia by these methods. These pathways are<br />

related to metabolism <strong>of</strong> glutamate, the process <strong>of</strong> apoptosis,<br />

inflammation, and immune system. Our pathway analysis <strong>of</strong> RNA-<br />

Seq data generally supported these findings, including the<br />

identification <strong>of</strong> the cell adhesion molecules (CAMs) pathway. Our<br />

DMS analysis <strong>of</strong> schizophrenia GWAS datasets successfully<br />

identified 164 network module genes, 12 <strong>of</strong> which were significant<br />

by meta-analysis after multiple testing correction.<br />

Conclusions: Our analysis suggested pathway and network<br />

approaches are promising in post-GWAS era and may complement<br />

the original analysis <strong>of</strong> genomics datasets such as GWAS and RNA-<br />

Seq. However, biases and challenges in these methods exist and<br />

improvement <strong>of</strong> methods is expected.<br />

24<br />

SYMPOSIUM 5: APPLICATION OF EXOME SEQUENCING<br />

TECHNOLOGIES TO AUTISM<br />

S5.1 THE GENETIC ARCHITECTURE OF AUTISM<br />

SPECTRUM- AND RELATED- NEURODEVELOPMENTAL<br />

DISORDERS REVEALED THROUGH HIGH-RESOLUTION<br />

GENOME ANALYSIS<br />

S. Scherer*<br />

Hospital for Sick Children and University <strong>of</strong> Toronto<br />

*stephen.scherer@sickkids.ca<br />

We will provide an update <strong>of</strong> our Canadian efforts aimed at<br />

cataloguing all highly-penetrant copy number variation (CNV) and<br />

DNA sequence-level variants in autism spectrum and related<br />

neurodevelopmental disorders. We are using the highest-resolution<br />

genomic technologies to (i) examine a new Ontario-wide ASD<br />

consecutive case cohort for the impact <strong>of</strong> CNVs in autism and related<br />

neurodevelopmental disorders for diagnostic validity assessment and<br />

discovery and (ii) through next generation sequencing (NGS) define<br />

the allelic 'mutation' architecture in ASD creating a resource for<br />

translational research and validated diagnostics.<br />

We will present our latest data from the first few hundred sequenced<br />

exomes/genomes describing both characteristic and new findings. We<br />

will discuss our NGS sequencing on individuals carrying potentially<br />

pathogenic large CNVs under the hypothesis that there may be<br />

additional sequence changes or other contributing loci. Further<br />

characterization <strong>of</strong> the variants identified is achieved by assessment<br />

<strong>of</strong> segregation in families and by determining frequencies in<br />

independent case and control populations, as well as through<br />

functional studies.<br />

We speculate that our approach will not only yield novel genes and<br />

variants influencing ASD susceptibility, but will also shed light on<br />

the broader issues <strong>of</strong> allelic variant architecture, variable penetrance<br />

and expressivity, and phenotypic heterogeneity in complex<br />

disorders. Some novel ‘pathogenic’ variants have been identified and<br />

will this data will be presented at the meeting. We will also discuss<br />

our experiences within a social health care system in translating this<br />

information back to the participating families.


S5.2 EXOME SEQUENCING OF AUTISM AND<br />

SCHIZOPHRENIA IN THE UK10K PROJECT<br />

J. Barrett, A. Palotie*<br />

The Wellcome Trust Sanger Institute<br />

*ap8@sanger.ac.uk<br />

Introduction: The UK10K project aims at sequencing 10 000<br />

individuals to provide a catalogue <strong>of</strong> low frequency variants and to<br />

study their association to disease traits. The project will generate<br />

low-coverage (6X) whole genome sequences for 4000 individuals<br />

from two deeply phenotyped UK population based cohorts, and deep<br />

whole exome sequences for 6000 individuals with extreme clinical<br />

phenotypes related to obesity, neurodevelopmental disorders and 8<br />

rare severe diseases.<br />

Methodology: We will describe the neurodevelopmental arm <strong>of</strong> the<br />

project, which will sequence 3000 cases with autism or schizophrenia<br />

in total. Approximately 800 non-syndromic autism spectrum disorder<br />

cases have been selected from three UK and two Finnish centres.<br />

These cases have been weighted in favor <strong>of</strong> a number <strong>of</strong> “high value”<br />

criteria, including multiply affected families, and depth <strong>of</strong> additional<br />

phenotype data<br />

Results: Among the first cases being sequenced are a number <strong>of</strong><br />

large Finnish families with multiple affected individuals. We plan to<br />

use genome-wide SNP data generated in these families to combine<br />

exome sequence data with gene flow within families (i.e. withinfamily<br />

linkage) to prioritize potential family specific high-penetrance<br />

autism variants<br />

Conclusions: A progress report <strong>of</strong> this startegy will be presented.<br />

25<br />

S5.3 WHOLE-EXOME SEQUENCING IN SIMPLEX AUTISM<br />

QUARTETS<br />

S. Sanders, M. Murtha, A. Gupta, J. Murdoch, M. Roubeson,<br />

M. State*<br />

Yale University School <strong>of</strong> Medicine<br />

*matthew.state@yale.edu<br />

Introduction: Over the last several years, the importance <strong>of</strong> rare<br />

mutation for the genetic etiology <strong>of</strong> autism spectrum disorders (ASD)<br />

has been repeatedly demonstrated. Recent studies comparing affected<br />

to unaffected siblings within simplex families ascertained as part <strong>of</strong><br />

the Simons Simplex Collection (SSC) (Sanders et al 2011) has<br />

confirmed and extended prior reports <strong>of</strong> the significant contribution<br />

<strong>of</strong> large, de novo copy number variations (CNVs). These findings<br />

have pointed to the marked effect sizes carried by de novo variants,<br />

with odd ratios approaching 6 for the overall class <strong>of</strong> mutation, as<br />

well as to the particular importance <strong>of</strong> specific regions <strong>of</strong> recurrence,<br />

with population frequencies in affected individuals in some intervals<br />

reaching 1%. Overall, from our empiric CNV data in more than 1000<br />

SSC families, we have estimated the presence <strong>of</strong> 250 de novo genic<br />

CNV regions carrying risk in the human genome, the vast majority <strong>of</strong><br />

which have not yet been identified. Whole- exome sequencing within<br />

this same SSC cohort provides a highly efficient means to pursue<br />

these findings in greater depth, <strong>of</strong>fering to further elaborate the<br />

genomic architecture <strong>of</strong> de novo variations in discordant sibling pairs<br />

and to identify, via multiple recurrences within a given gene, specific<br />

mutations contributing to ASD.<br />

Methodology: We have undertaken whole-exome sequencing <strong>of</strong> 268<br />

individuals from within 70 families ascertained by the Simons<br />

Simplex Collection, a comprehensively phenotyped simplex cohort,<br />

where the majority <strong>of</strong> pedigrees include a proband, an unaffected<br />

mother and father and an unaffected sibling. The sequenced cohort<br />

included 12 parent-child trios and 58 “quartets”, with the latter<br />

providing a direct comparison <strong>of</strong> de novo mutation rates in matched<br />

discordant sibling pairs. Within these 58 families, 11 carried<br />

previously identified ASD-risk CNVs. Within the additional 12 trios,<br />

4 carried established ASD risk CNVs, allowing for an assessment <strong>of</strong><br />

the relationship <strong>of</strong> de novo structural and sequence variation within<br />

individuals and across the cohort. DNA selection was carried out<br />

using either the Nimblegen custom whole-exome (35Mbp target) or<br />

SeqCap EZExome v2 array (45Mbp target) and samples were<br />

sequenced on either an Illumina GAIIx or Illumina HiSeq 2000<br />

instrument<br />

Results: We will first provide an overview <strong>of</strong> the alignment, variant<br />

calling and quality control procedures for our whole exome data. We<br />

will then present data on the overall burden <strong>of</strong> rare de novo<br />

mutations, comparing affected individuals to their unaffected<br />

siblings. We will address the question <strong>of</strong> recurrence <strong>of</strong> de novo<br />

variants and their statistical significance in assessing association<br />

Conclusions: Whole exome sequencing in simplex quartets with<br />

ASD provides an important opportunity for gene discovery,<br />

reaffirmes the contirbution <strong>of</strong> de novo variation to ASD, and provides<br />

insight into the frequency, distribution, and character <strong>of</strong> de novo<br />

sequence mutation in both affected and unaffected individuals.


S5.4 FULL EXOME SEQUENCING OF AUTISM CASES,<br />

FAMILIES, AND CONTROLS<br />

ECIP<br />

B. Neale*(1,2), J. Buxbaum(3), B. Devlin(4), G. Schellenberg(5), J.<br />

Sutcliffe(6), R. Gibbs(7), M. Daly(1,2)<br />

1. Analytic and Translational <strong>Genetics</strong> Unit, Massachusetts General<br />

Hospital 2. Program in Medical and Population <strong>Genetics</strong>, Broad<br />

Institute <strong>of</strong> MIT and Harvard 3. Mount Sinai School <strong>of</strong> Medicine 4.<br />

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

Pathology and Laboratory Medicine, University <strong>of</strong> Pennsylvania 6.<br />

Department <strong>of</strong> Molecular Physiology and Biophysics, Vanderbilt<br />

University 7. Baylor College <strong>of</strong> Medicine<br />

*bmneale@gmail.com<br />

Introduction: We conducted whole-exome sequencing on a case<br />

control cohort <strong>of</strong> approximately 1,000 cases and 1,000 controls for<br />

autism.<br />

Methodology: For half <strong>of</strong> these samples, data were generated at<br />

Baylor College <strong>of</strong> Medicine, using Nimblegen capture and ABI Solid<br />

Sequencing and the other half were generated at the Broad Institute<br />

<strong>of</strong> MIT and Harvard using Agilent capture and Illumina GAII and<br />

HiSeq technologies. We will describe the alignment, variant calling<br />

and general quality control analyses <strong>of</strong> both <strong>of</strong> these datasets<br />

including a description <strong>of</strong> the relative merits <strong>of</strong> both capture and<br />

sequencing technologies. As part <strong>of</strong> our study design, we have<br />

sequenced a handful <strong>of</strong> samples at both centers, enabling a more<br />

comprehensive comparison <strong>of</strong> genotype calls. We will also<br />

demonstrate that these two datasets are largely comparable in terms<br />

<strong>of</strong> more common variation, indicating that even though the entire<br />

data generation pipeline is radically different between these two sites,<br />

the assessment <strong>of</strong> common variation shows striking consistency.<br />

Results: We have conducted a range <strong>of</strong> association tests on these two<br />

datasets including variant-specific and gene-based approaches. For<br />

variant-specific analyses, we test variation that is too rare to have<br />

been effectively captured using genome-wide association (i.e.<br />

frequency < 5%), with a particular focus on obviously functional<br />

variation. For gene-based analysis, we have calculated burden <strong>of</strong><br />

nonsense and missense mutations for autism as well as applied Calpha<br />

approach. We demonstrate that the genetics for autism must be<br />

complex from the case-control analysis.<br />

Conclusions: In addition to the case-control sequencing, we have<br />

also generated data on 100 trios, which enables the assessment <strong>of</strong> de<br />

novo mutations for risk to autism. We identified de novo mutations at<br />

approximately the rate <strong>of</strong> 1 per trio and that these mutations follow a<br />

Poisson distribution across the families analyzed. We have assessed<br />

the extent to which the genes that carry de novo mutations are<br />

excessively connected via protein-protein interactions.<br />

26<br />

SYMPOSIUM 6: BIOLOGICALLY INFORMED ANALYSIS<br />

OF GWAS DATA: FROM BIOLOGY TO GENES AND BACK<br />

S6.1 INFERENCE OF FUNCTIONAL GENE NETWORKS IN<br />

THE SYNAPSE<br />

L. Cornelisse*(1), E. Tsivtsivadze(2), T. Dijkstra(2,3), T. Heskes(2),<br />

M. Verhage(1)<br />

1. Functional Genomics, Center for Neurogenomics and Cognitive<br />

Research (CNCR), Dept. <strong>of</strong> Clinical <strong>Genetics</strong>, VUmc, Neuroscience<br />

Campus Amsterdam (NCA), VU University (VUA) and VU<br />

University medical center (VUmc) 2. Machine Learning Group,<br />

Intelligent Systems, Institute for Computing and Information<br />

Sciences, Radboud University 3. Signal Processing Systems, Dept <strong>of</strong><br />

Electrical Engineering, Technical University Eindhoven<br />

*niels.cornelisse@cncr.vu.nl<br />

Brain disease is one <strong>of</strong> the most pressing health problems in today's<br />

western society. For most brain disorders heritability estimates are<br />

known to be high, (e.g. 90% for autism, 80% for schizophrenia).<br />

However, the genetic variants identified so far with genome-wide<br />

association studies (GWAS) explain only a small percentage <strong>of</strong> the<br />

trait variance. One explanation <strong>of</strong> this 'missing heritability' is that<br />

epistatic interactions between multiple genes have a major<br />

contribution to the disease risk. These interactions will be undetected<br />

due to necessary multiple testing corrections when 500,000+ genetic<br />

variants are tested in parallel in a GWA approach. An alternative way<br />

to model multiple small genetic effects and consider dependency<br />

between different genes is to evaluate the combined, additive effects<br />

<strong>of</strong> variations in multiple genes by means <strong>of</strong> pathway or functional<br />

gene group analysis. Such a systems level approach relies heavily on<br />

sound characterization <strong>of</strong> gene network architecture and the<br />

functional interactions in the network. We focus on genes expressed<br />

in synapses, which are small structures in the brain <strong>of</strong> about 1 µm 3 in<br />

size with a crucial role in basic brain functions such as information<br />

processing, learning and memory. About 10% <strong>of</strong> all genes is<br />

expressed in the synapse <strong>of</strong> which several have been implicated in<br />

neurological disorders over the past few years. We use machinelearning<br />

techniques to infer functional gene networks in the<br />

presynaptic terminal, which is responsible for the precisely timed<br />

release <strong>of</strong> neurotransmitter during neuronal activity. High-content<br />

immunoprecipitation mass spectrometry (IP-MS) interaction data<br />

generated in our institute and functional data from genetic<br />

perturbation studies in Hippocampal autaptic neurons (a reduced<br />

model system for assessing synaptic transmission) are used to cluster<br />

presynaptic genes in functional groups and pathways. We show that<br />

presynaptic genes do not cluster exclusively to one pathway but can<br />

be involved in multiple steps in the release process, which argues for<br />

weighted functional grouping <strong>of</strong> genes in GWAS <strong>of</strong> complex traits.


S6.2 GENE-SET ENRICHMENT AND NETWORK-BASED<br />

MULTI-LOCUS ANALYSES OF GENOME-WIDE DATA<br />

P. Lee, D. Ruderfer, C. O'Dushlaine, S. Purcell*<br />

Massachusetts General Hospital<br />

*shaun@atgu.mgh.harvard.edu<br />

Introduction: Under models <strong>of</strong> multifactorial inheritance a<br />

considerable number <strong>of</strong> variants individually explain very little <strong>of</strong> the<br />

total genetic variation in the population and therefore are hard to<br />

detect. In this case, it will <strong>of</strong>ten be necessary to approach genomewide<br />

datasets (whether <strong>of</strong> common single nucleotide polymorphisms<br />

(SNPs), rare copy number variants (CNVs) from genome-wide<br />

association studies (GWAS), or rare variants from whole-exome<br />

sequencing studies) from a multilocus perspective, considering the<br />

patterns <strong>of</strong> association to disease in the context <strong>of</strong> their genomic and<br />

functional locations.<br />

Methodology: We present two approaches that layer prior biological<br />

information, in the form <strong>of</strong> either gene-sets or networks, onto the<br />

otherwise functionally agnostic statistical information arising from<br />

standard genotype-phenotype correlational analyses. We give an<br />

overview <strong>of</strong> both methods and discuss alternatives.<br />

Results: The first approach is designed to test for specific sets <strong>of</strong><br />

genes that are enriched in associated intervals in the context <strong>of</strong><br />

GWAS and CNV studies, and implemented in the INRICH s<strong>of</strong>tware<br />

package. We review applications <strong>of</strong> this approach across a range <strong>of</strong><br />

different (psychiatric and non-psychiatric) GWAS and discuss the<br />

implications for likely genetic models and the adequacy <strong>of</strong> existing<br />

“pathway” data for neuropsychiatric research. We also focus on the<br />

utility <strong>of</strong> this approach to perform in silico fine-mapping in regions<br />

where multiple genes contain variants in strong linkage<br />

disequilibrium and are therefore supported by equally strong<br />

association evidence The second approach utilizes biological network<br />

information, for example, from databases such as the STRING<br />

protein-protein interaction database, to guide gene-based rare variant<br />

analyses <strong>of</strong> whole-exome sequence datasets. Gene-based association<br />

tests for individual genes “borrow strength” from the statistics for<br />

other genes that are neighboring in biological network space. This<br />

approach is currently being implemented in the PLINK/Seq s<strong>of</strong>tware<br />

package.<br />

Conclusions: As well as potentially adding biological insight on top<br />

<strong>of</strong> genetic findings, consideration <strong>of</strong> biological information in<br />

multilocus genetic analysis can be used to a) prioritize sub-threshold<br />

associations from GWAS for follow-up, b) identify genes <strong>of</strong> small<br />

effect, particularly apt for the analysis <strong>of</strong> very rare variants and c)<br />

effectively perform in silico mapping <strong>of</strong> associated GWAS regions or<br />

genes under large CNVs. We provide free, robust s<strong>of</strong>tware packages<br />

that can be used towards these aims.<br />

27<br />

S6.3 PROTEIN COMPLEXES AND THE FUNCTIONAL<br />

BASIS OF PSYCHIATRIC DISORDERS<br />

A. Pocklington*<br />

Cardiff University<br />

*pocklingtonaj@cardiff.ac.uk<br />

Introduction: To identify cellular processes disrupted in psychiatric<br />

disorders and understand the impact <strong>of</strong> disease-associated mutations<br />

will require appropriate models <strong>of</strong> functional organisation at the<br />

molecular level. A common mechanism <strong>of</strong> organisation found in all<br />

cell-types is the physical localisation <strong>of</strong> molecules to form specialised<br />

functional components.<br />

Methodology: Proteomic characterisation <strong>of</strong> organelles and protein<br />

complexes can be used to generate a high-level model <strong>of</strong> the cellular<br />

machinery suitable for gene-set/pathway based analyses. In addition<br />

to providing a very clear link to cell-biological function, such models<br />

can drastically reduce the burden <strong>of</strong> multiple testing, a major problem<br />

associated with the fine-grained functional ontologies used to date.<br />

Results: This presentation will concentrate on proteomic studies <strong>of</strong><br />

the synapse, being the major component <strong>of</strong> the cellular machinery<br />

unique to the nervous system.<br />

Conclusions: Current models <strong>of</strong> synapse molecular organisation will<br />

be described and their relevance to psychiatric genetics discussed.


S6.4 THE USE OF PROTEIN-PROTEIN INTERACTIONS TO<br />

INTERPRET GENETIC ASSOCIATION TO DISEASE<br />

ECIP<br />

E. Rossin*(1,2,3), K. Lage(4), S. Ripke(1,2), B. Neale(1,2), K.<br />

Samocha(1,2,3), P. GWAS Consortium(1,2,3), S.<br />

Raychaudhuri(2,3,5), R. Xavier(1,2,3), C. Cotsapas(2,6), M.<br />

Daly(1,2,3)<br />

1. Massachusetts General Hospital 2. The Broad Institute 3. Harvard<br />

Medical School 4. Center for Biological Sequence Analysis,<br />

Department <strong>of</strong> Systems Biology, Technical University <strong>of</strong> Denmark 5.<br />

Brigham and Women's Hospital 6. Yale School <strong>of</strong> Medicine<br />

*rossin@broadinstitute.org<br />

Introduction: Genome-wide association studies (GWAS) have<br />

defined hundreds <strong>of</strong> genomic regions containing variation<br />

predisposing to complex disease. Inferring disease biology from these<br />

observations, however, hinges on our ability to discover the<br />

molecular processes being perturbed by these risk variants. It has<br />

previously been observed that different genes harboring causal<br />

mutations for the same Mendelian disease <strong>of</strong>ten physically interact.<br />

In a pursuit to evaluate the degree to which this is true <strong>of</strong> genes<br />

within strongly associated loci in complex disease as well as genes<br />

identified through more recent exome sequencing as potentially<br />

baring risk mutations, we have developed a method called DAPPLE<br />

(Disease Association Protein-Protein Link Evaluator).<br />

Methodology: Using a collection <strong>of</strong> 170,000 published proteinprotein<br />

interactions, DAPPLE looks for direct and indirect<br />

connections between proteins from associated loci to build a diseasespecific<br />

network that includes not only the seeded proteins but also<br />

proteins elsewhere in the genome. Because protein-protein interaction<br />

data can be biased, we then evaluate the subsequent network and its<br />

individual members for connectivity beyond chance expectation<br />

using within-degree node-label permutation, an approach that<br />

controls for the varying degree to which proteins are represented in<br />

the database. We then look for enrichment in various external<br />

properties in the network, such as association to phenotype or<br />

particular genes <strong>of</strong> interest. We looked for protein-protein<br />

interactions within the top hits in a recent schizophrenia metaanalysis<br />

(9394 cases, 12462 controls) as well as de novo mutations<br />

(missense, nonsense or splice-site) from an on-going autism exome<br />

sequencing study (100 trios).<br />

Results: In schizophrenia, we find that the emergent network from<br />

the top associations is enriched for the predicted targets <strong>of</strong> mir-137, a<br />

micro-RNA in one <strong>of</strong> top regions, and that those targets are also<br />

enriched for association to Schizophrenia. In Autism, we find that the<br />

network built from genes harboring single de novo<br />

missense/nonsense/splice-site mutations show direct and indirect<br />

connections far in excess <strong>of</strong> chance.<br />

Conclusions: We have developed a method to systematically look<br />

for protein-protein interactions between proteins from genomic loci<br />

<strong>of</strong> interest and evaluate subsequent networks for statistical<br />

enrichment in connectivity. We have shown that when applied to<br />

schizophrenia and autism, such a method can be useful to identify<br />

potentially important biochemical connections.<br />

28<br />

S6.5 EXPLORING GENOME-WIDE ASSOCIATION DATA IN<br />

BIOLOGICAL CONTEXT THROUGH PROTEIN-PROTEIN<br />

INTERACTIONS<br />

P. Sham*, J. Kwan, M. Li<br />

The Universiy <strong>of</strong> Hong Kong<br />

*pcsham@hku.hk<br />

Introduction: In recent years, gene-based tests have gained<br />

popularity in genome-wide association studies (GWAS), because the<br />

gene is generally regarded as the basic functional unit <strong>of</strong> the genome.<br />

However, genes code for proteins which <strong>of</strong>ten interact physically<br />

with each other to form more complex functional structures. A<br />

natural generalization <strong>of</strong> gene-based analysis is therefore the<br />

consideration <strong>of</strong> gene sets that code for interacting proteins.<br />

Methodology: We have developed an association test for the simple<br />

scenario, <strong>of</strong> protein-protein interaction (PPI) pairs. This test considers<br />

all PPI pairs from public databases for evidence <strong>of</strong> association<br />

between sequence variants in both genes, through the combination <strong>of</strong><br />

gene-based statistics. The test assigns differential weights to the two<br />

genes in a PPI pair according to their numbers <strong>of</strong> PPI partners, and<br />

filters out significant pairs where all the evidence for association<br />

comes primarily from only one <strong>of</strong> the two PPI partners.<br />

Results: The new test was applied to a published GWAS on Crohn’s<br />

disease, and detected a number <strong>of</strong> PPI gene pairs significantly<br />

associated with disease, even after Bonferroni adjustment for the total<br />

number <strong>of</strong> PPI pairs. Among these significant pairs are those where<br />

one or both partners are not significant according to gene-based tests.<br />

Conclusions: We conclude that association analysis <strong>of</strong> PPI pairs is a<br />

useful complement to SNP-based and gene-based analyses, and may<br />

provide additional statistical power for detecting genes associated<br />

with complex diseases. This may be particularly useful for<br />

psychiatric disorders because <strong>of</strong> the small effect sizes <strong>of</strong> the SNPs<br />

that so far have been detected.


SYMPOSIUM 7: FROM GENES TO BIOLOGY:<br />

DEVELOPMENTAL PSYCHOPATHOLOGY<br />

GENETIC EPIDEMIOLOGY OF ADHD AND DEPRESSION<br />

ACROSS THE LIFESPAN<br />

D. Boomsma(1), C. Middeldorp(1), C. Dolan(1), M. Nivard(1), K.<br />

Kan(1), M. Groen-Blokhuis(1), J. van Beek(1), L. Geels(1), L.<br />

Ligthart(1), J. Hudziak(1), R. Alth<strong>of</strong>f(1), T. van Beijsterveldt(1), J.<br />

Vink(1), M. Bartels(1), G. Willemsen(1), E. de Geus(1)<br />

1. Netherlands Twin Register, Dept <strong>of</strong> Biological Psychology, VU<br />

University 2. University <strong>of</strong> Vermont<br />

Introduction: Large genome-wide association (GWA) studies <strong>of</strong><br />

complex psychiatric phenotypes need to consider whether effects <strong>of</strong><br />

genetic variants should be evaluated separately in men and women,<br />

or as a function <strong>of</strong> age, as the effect <strong>of</strong> a particular variant may be<br />

different across sex and age. Expression <strong>of</strong> genes can differ across<br />

sex or age (genotype x sex and genotype x age interactions) but<br />

increasing the number <strong>of</strong> tests amplifies the penalty for multiple<br />

testing. We test for quantitative and qualitative sex and age<br />

differences in genetic architecture for multiple indices <strong>of</strong> ADHD and<br />

depression phenotypes using longitudinal data from same-sex and<br />

opposite-sex twin pairs, age 3 through 80 years.<br />

Methodology: The Netherlands Twin Register (NTR) has<br />

collected longitudinal data in children from age 3 years up till age<br />

20+ on multiple indices <strong>of</strong> Attention Problems and ADHD and<br />

anxious depression by parental, teacher and self-report (N ><br />

16.000 pairs). In adults, multiple inventories assessing these<br />

phenotypes by self-report were collected between 1991 and 2011 (up<br />

to 9 surveys; N >7500 adult twins). Longitudinal genetic analyses <strong>of</strong><br />

these data were performed with structural equation modeling.<br />

Genomewide SNP typing is available in subgroups <strong>of</strong> participants.<br />

Results: Longitudinal twin data on ADHD and depression show<br />

heritabilities to be only mildly affected by rater or sex, but<br />

significantly by age. Genetic correlations along the lifespan will be<br />

presented, addressing whether the same genes are operating at all<br />

ages.<br />

29<br />

S7.2 TWINS, TISSUE AND TIME: A COMPARISON OF<br />

GENOMIC STRUCTURE ACROSS TWINS, DNA AND<br />

LONGITUDINAL SAMPLES<br />

P. Scheet*(1), E. Ehli(2,3), X. Xiao(1), A. Abdellaoui(3), R.<br />

Alth<strong>of</strong>f(4), J. Hottenga(4), G. Willemsen(4), K. Nelson(2), P.<br />

Huizenga(2), Y. Hu(2), M. Bartels(4), M. Groen-Blokhuis(4), E. de<br />

Geus(4), J. Hudziak(5), G. Davies(2,3), D. Boomsma(4)<br />

1. Department <strong>of</strong> Epidemiology, University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center 2. Avera Institute for Human Behavioral <strong>Genetics</strong>,<br />

Avera McKennan Hospital and University Health Center 3.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> South Dakota, Sanford<br />

School <strong>of</strong> Medicine, Division <strong>of</strong> Basic Biomedical Sciences 4.<br />

Department <strong>of</strong> Biological Psychology, VU University 5. Department<br />

<strong>of</strong> Psychiatry, University <strong>of</strong> Vermont, College <strong>of</strong> Medicine<br />

*pscheet@alum.wustl.edu<br />

Introduction: With the desire to assess genetic variation with<br />

conveniently-obtained biological sources in large scale collaborative<br />

projects, one question is whether inference <strong>of</strong> copy number (CN) is<br />

overly sensitive to the source <strong>of</strong> material for DNA analysis (e.g.<br />

blood, buccal) and another is whether CN is stable over time. Here,<br />

we address these questions by studying 1,471 DNA samples from<br />

710 individuals in the Netherlands Twin Register, including both<br />

twins and non-twins, with the availability <strong>of</strong> longitudinal samples and<br />

samples from different tissues, to obtain a technical evaluation and<br />

confirmation <strong>of</strong> copy number inference from the Affymetrix 6.0<br />

DNA microarray technology.<br />

Methodology: We analyzed DNA from longitudinal blood and<br />

buccal sources from a large sample (n = 710 subjects), including 43<br />

MZ (23 female) and 75 DZ (26 both female, 23 both male), their<br />

parents and siblings. The participants are registered with the<br />

Netherlands Twin Register and DNA samples have been collected in<br />

multiple projects, both from blood and buccal material. In addition,<br />

we examine effects <strong>of</strong> plating individuals on the estimation <strong>of</strong><br />

CNVs. Finally, we aim to evaluate whether blood is an acceptable<br />

DNA source in studies that include twins. To assess the consistency<br />

<strong>of</strong> CN inference across DNA source material, blood and buccal<br />

derived samples from the same individuals were compared (withinindividual,<br />

between sources) and from different members <strong>of</strong> the same<br />

MZ pair. To assess the consistency over time, samples <strong>of</strong> the same<br />

individual and tissue type, obtained at different time points, were<br />

compared. Multiple algorithms were applied to obtain CNV<br />

calls. We focus on those from Birdsuite v.2 (Korn et al).<br />

Results: No greater discordance was found for CN and genotype<br />

inference between samples from the same individual (or MZ cotwin)<br />

that derived from buccal than from blood. However, there was a<br />

small but statistically-significant decrease in across-tissue<br />

concordance. We also found no evidence <strong>of</strong> a temporal effect on CN<br />

variation from 389 individuals sampled at 2 time points ranging from<br />

1 to 12 years apart. Copy number estimates between co-twins from<br />

43 MZ pairs were highly consistent for both deletions (R 2 ≈ 90%) and<br />

duplications (R 2 ≈ 86%), as was genotype concordance (R 2 > 99%).<br />

Conclusions: In light <strong>of</strong> our data and results, we feel it is prudent,<br />

and indeed cost-effective, to conduct a microarray-based GWA study<br />

<strong>of</strong> genotypes and copy number using buccal-derived DNA for efforts<br />

to map genetic variants predisposing to disease and behaviors. This<br />

allowance will lower the barrier for participation <strong>of</strong> many groups <strong>of</strong><br />

individuals, including children. Finally, our study confirms that<br />

collections <strong>of</strong> blood derived DNA from large twin registries <strong>of</strong>fer a<br />

valuable resource for mapping and characterizing complex disease,<br />

given their unique contribution to partitioning environmental and<br />

genetic effects and the stability <strong>of</strong> genomic features across samples<br />

and time.


S7.5 FURTHER UNDERSTANDING THE HETEROGENEITY<br />

OF MDD<br />

B. Penninx*<br />

VU University Medical Center, department <strong>of</strong> Psychiatry<br />

*b.penninx@vumc.nl<br />

Introduction: MDD is a highly heterogeneous disorder, which limits<br />

the search for genetic predictors.<br />

Methodology: Using GWA from the NESDA study, we examined<br />

whether evidence can be found for differential genetic predictors for<br />

chronic versus non-chronic depression using 2-year longitudinal data.<br />

Results: Results will be presented from GWA and candidate gene<br />

analyses on genetic predictors <strong>of</strong> the course <strong>of</strong> depression. For this<br />

purpose we will use the 2-year longitudinal data from more than 1000<br />

MDD cases from the NESDA study in which course trajectories were<br />

assessed.<br />

Conclusions: Acknowledging the heterogeneity <strong>of</strong> MDD, it is<br />

essential to further examine the differential genetic predictors <strong>of</strong><br />

MDD subtypes.<br />

30<br />

SYMPOSIUM 8: ELUCIDATING THE FUNCTIONAL<br />

RELEVANCE OF THE ANK3 AND CACNA1C BIPOLAR<br />

DISORDER RISK GENES<br />

S8.1 ANKYRIN G AND L-TYPE CALCIUM CHANNEL:<br />

POTENTIAL ROLES IN PSYCHIATRIC DISEASE<br />

S. Zhu(1), V. Bennett(2), C. Ross*(1)<br />

1. Johns Hopkins 2. Duke<br />

*caross@jhu.edu<br />

Introduction: Ankyrin G is one <strong>of</strong> the strongest genetic risk factors<br />

identified by GWAS for bipolar disorder. In brain, it mainly localizes<br />

at the axon initiation segment (AIS) and Node <strong>of</strong> Ranvier. Cav1.2,<br />

the alpha subunit <strong>of</strong> L-type voltage gated calcium channel, is a risk<br />

factor by GWAS for bipolar disorder and schizophrenia. We are<br />

exploring the effects <strong>of</strong> loss <strong>of</strong> Ankyrin G function, and since<br />

Ankyrin G can bind to several other ion channels, we postulated that<br />

it could also interact with the calcium channel.<br />

Methodology: We are using cellular and molecular techniques both<br />

in vitro and in vivo<br />

Results: Immunoprecipitation from SH-SY5Y cells transfected with<br />

Ankyrin G and Calcium channel constructs indicates an interaction<br />

mediated by the beta subunit. Double label immuno-fluorescence<br />

studies in primary cultured neurons and mouse brain sections suggest<br />

partial colocalization. We are probing the cellular significance <strong>of</strong> loss<br />

<strong>of</strong> function <strong>of</strong> Ankyrin G and interactions between Ankyrin G and<br />

Ca v1.2, by knockdown <strong>of</strong> Ankyrin G in neuronal cell lines and<br />

primary neurons. Bipolar disorder and schizophrenia are believed to<br />

involve dysregulation <strong>of</strong> brain development. Ankyrin G is known to<br />

regulate the innervation <strong>of</strong> inhibitory interneurons <strong>of</strong> the AIS <strong>of</strong><br />

cerebellar Purkinje cells. We are examining the effect <strong>of</strong> knockdown<br />

<strong>of</strong> Ankyrin G on interneuron innervations <strong>of</strong> pyramidal cells in the<br />

developing cerebral cortex. We are also characterizing Ankyrin G<br />

loss <strong>of</strong> function in a novel conditional mouse model, and using in<br />

utero elctroporation.<br />

Conclusions: These studies suggest a role for Ankyrin G and its<br />

protein interactions in cellular pathogenic pathways and cortical<br />

development, which may provide possible therapeutic targets for<br />

psychiatric disorders.


S8.2 ROLE OF ANKYRIN 3 IN REGULATING BIPOLAR-<br />

RELATED BEHAVIORS<br />

M. Leussis(1,2,3), E. Berry-Scott(3), H. Jhuang(4), M. Saito(2), K.<br />

Ilsley(2), T. Poggio(5), P. Sklar(6), T. Serre(4), T. Petryshen*(1,2,3)<br />

1. Dept <strong>of</strong> Psychiatry, Massachusetts General Hospital and Harvard<br />

Medical School 2. <strong>Psychiatric</strong> and Neurodevelopmental <strong>Genetics</strong><br />

Unit, Center for Human Genetic Research, Massachusetts General<br />

Hospital 3. Stanley Center for <strong>Psychiatric</strong> Research, Broad Institute<br />

4. Cognitive, Linguistic & Psychological Sciences Dept & Institute<br />

for Brain Sciences, Brown University 5. McGovern Institute for<br />

Brain Sciences, MIT 6. Mount Sinai School <strong>of</strong> Medicine<br />

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

Introduction: The ankyrin 3 gene (ANK3) has been conclusively<br />

identified as a risk gene for bipolar disorder (BD) in numerous<br />

patient GWAS and targeted association studies. ANK3 encodes the<br />

ankyrin G scaffolding protein that functions in the development and<br />

maintenance <strong>of</strong> the axon initial segment (AIS) <strong>of</strong> neurons that elicits<br />

action potentials, as well as maintenance <strong>of</strong> neuronal polarity and<br />

synaptic function. The mechanism by which ANK3 contributes to<br />

BP risk is unknown. As a first step towards elucidating this<br />

mechanism, we are investigating the behavioral and neurobiological<br />

impact <strong>of</strong> dysregulated neural expression <strong>of</strong> mouse Ank3 using RNA<br />

interference and knockout genetic models.<br />

Methodology: Lentiviral-mediated RNA interference was used to<br />

reduce Ank3 expression in hippocampus or nucleus accumbens <strong>of</strong><br />

adult male C57BL/6J mice. Lentivirus expressing a GFP marker and<br />

one <strong>of</strong> two short hairpin RNA (shRNA) sequences targeting Ank3, or<br />

a scrambled control sequence, was injected bilaterally (≥10 9 infection<br />

units/ml) into hippocampus or nucleus accumbens (N = 10-12 mice<br />

per group). Mice were assessed for behaviors modeling bipolar<br />

disorder symptoms, including novelty- and psychostimulant-induced<br />

hyperlocomotion, behavioral disinhibition, and hedonic and<br />

anhedonic behaviors, as well as intermediate phenotypes (prepulse<br />

inhibition), circadian activity, learning (fear conditioning,<br />

habituation), and sensory and motor performance. Reversal <strong>of</strong> the<br />

behavioral phenotype was examined following chronic treatment<br />

with lithium (85 mg/kg i.p.) or lamotrigine (2.5 mg/kg i.p.). To<br />

validate findings from RNA interference, knockout mice with Ank3<br />

haploinsufficiency in brain were assessed for a subset <strong>of</strong><br />

behaviors. Expression <strong>of</strong> ankyrin G and protein interactors was<br />

measured by immunohistochemistry <strong>of</strong> fixed brain to evaluate the<br />

impact <strong>of</strong> Ank3 reduction on neural functions.<br />

Results: Mice with hippocampal Ank3 silencing displayed consistent<br />

changes in several behaviors for both shRNA sequences (P < 0.05 vs.<br />

control shRNA), while other behaviors were unchanged, indicating<br />

the phenotype is not due to global impairment. Lithium treatment<br />

partially reversed some behavioral changes, whereas lamotrigine had<br />

a non-specific effect on the Ank3 and control shRNA groups. By<br />

immunohistochemistry, both Ank3 shRNA sequences reduced<br />

ankyrin G expression at the AIS by 40-50%, suggesting impaired<br />

action potential firing may underlie the behavioral changes, although<br />

other Ank3 functions could also be perturbed. Ank3 silencing in<br />

nucleus accumbens resulted in less robust behavioral changes,<br />

potentially due to insufficient virus distribution in this region.<br />

Conclusions: Viral-mediated RNA interference in mouse brain is a<br />

practical method for studying the neural function <strong>of</strong> psychiatric risk<br />

genes. The behavioral changes induced by Ank3 hippocampal<br />

reduction and their reversal by lithium treatment in mice suggest that<br />

the mechanism by which ANK3 confers BD risk is through a loss <strong>of</strong><br />

function. These results and ongoing work defining the neural<br />

aberrations underlying the behavioral changes are expected to help<br />

elucidate the disease relevance <strong>of</strong> ANK3.<br />

31<br />

S8.3 MOOD DISORDER SUSCEPTIBILITY GENE CACNA1C<br />

MODIFIES MOOD-RELATED BEHAVIORS IN MICE AND<br />

INTERACTS WITH SEX TO INFLUENCE BEHAVIOR IN<br />

MICE AND DIAGNOSIS IN HUMANS<br />

T. Gould*(1), D. Dao(1), P. Mahon(2), X. Cai(1), C. Kovacsics(1),<br />

D. Levinson(3), M. Arad(1), J. Shi(3), P. Zandi(2), J. Knowles(4), M.<br />

Weissman(5), W. Coryell(6), W. Scheftner(7), W. Lawson(8), S.<br />

Thompson(1), J. Potash(2)<br />

1. University <strong>of</strong> Maryland School <strong>of</strong> Medicine 2. Johns Hopkins<br />

University 3. Stanford University 4. University <strong>of</strong> Southern<br />

California 5. Columbia University 6. University <strong>of</strong> Iowa 7. Rush<br />

University 8. Howard University<br />

*gouldlab@me.com<br />

Introduction: Recent genome-wide association studies have<br />

associated polymorphisms in the gene CACNA1C, which codes for<br />

CaV1.2, with a bipolar disorder and depression diagnosis.<br />

Methodology: The behaviors <strong>of</strong> wild-type and Cacna1c<br />

heterozygous mice <strong>of</strong> both sexes were evaluated in a number <strong>of</strong> tests.<br />

Based upon sex differences in our mouse data, we assessed a<br />

geneXsex interaction for diagnosis <strong>of</strong> mood disorders in human<br />

subjects. Data from the NIMHBP and the GenRED Consortiums<br />

were examined using a combined dataset that included 2021 mood<br />

disorder cases (1223 females) and 1840 controls (837 females).<br />

Results: In both male and female mice, Cacna1c haploinsufficiency<br />

was associated with lower exploratory behavior, decreased response<br />

to amphetamine, and antidepressant-like behavior in the forced swim<br />

and tail suspension tests. Female, but not male, heterozygous mice<br />

displayed decreased risk-taking behavior or increased anxiety in<br />

multiple tests, greater attenuation <strong>of</strong> amphetamine-induced<br />

hyperlocomotion, decreased development <strong>of</strong> learned helplessness,<br />

and a decreased acoustic startle response, indicating a sex-specific<br />

role <strong>of</strong> Cacna1c. In humans, sex-specific genetic association was seen<br />

for two intronic single nucleotide polymorphisms, rs2370419 and<br />

rs2470411, in CACNA1C, with effects in female subjects (odds ratio<br />

= 1.64, 1.32) but not in male subjects (odds ratio = .82, .86). The<br />

interactions by sex were significant after correction for testing 190<br />

single nucleotide polymorphisms (p(corrected) = .03, .04) and were<br />

consistent across two large datasets.<br />

Conclusions: Our preclinical results support a role for CACNA1C in<br />

mood disorder pathophysiology, and the combination <strong>of</strong> human<br />

genetic and preclinical data support an interaction between sex and<br />

genotype.


S8.4 IMPACT OF THE PSYCHIATRIC SUSCEPTIBILITY<br />

GENE CACNA1C IN HUMAN IMAGING GENETIC STUDIES<br />

ON NEURAL ACTIVITY IN HEALTHY SUBJECTS - AN<br />

OVERVIEW<br />

S. Witt*(1), J. Strohmaier(1), S. Erk(2), A. Krug(3), M. Thimm(3),<br />

C. Kuehner(4), T. Schulze(5), T. Kircher(1), H. Walter(2), M.<br />

Wessa(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 2. Neurology,<br />

Neurosurgery and Psychiatry, Division <strong>of</strong> Mind and Brain Research,<br />

Charité 3. Department <strong>of</strong> Psychiatry and Psychotherapy, Philipps-<br />

University 4. RG Longitudinal and Intervention Research, Dep <strong>of</strong><br />

Psychiatry and Psychotherapy Institute <strong>of</strong> Mental Health, University<br />

<strong>of</strong> Heidelberg 5. Clinic for Psychiatry and Psychotherapy, Georg-<br />

August-University 6. Department <strong>of</strong> Cognitive and Clinical<br />

Neuroscience, Institute <strong>of</strong> Mental Health, University <strong>of</strong> Heidelberg<br />

*stephanie.witt@zi-mannheim.de<br />

Introduction: Recent genome-wide association studies report strong<br />

evidence for an association between the single nucleotide variant<br />

rs1006737 in CACNA1C and bipolar disorder, schizophrenia, and<br />

major depression. The impact <strong>of</strong> this risk variant has been tested<br />

since in endophenotype studies using an imaging genetics approach<br />

in healthy individuals – a strategy which has repeatedly proven to be<br />

successful as it is not hampered by confounding variables that are<br />

typically present in patients.<br />

Methodology: This presentation gives an overview on imaging<br />

genetic studies which analyzed association <strong>of</strong> rs1006737 with<br />

endophenotypes from functional MRI studies carried out in the<br />

Department <strong>of</strong> Genetic Epidemiology together with different clinical<br />

collaboration partners with sample sizes ranging between 63 and 110<br />

participants. Studies vary with regard to the functional MRI<br />

paradigms which were applied in the specific contexts.<br />

Results: Brain functions associated with the genetic variant include<br />

neural activity related to episodic memory, attention networks, verbal<br />

fluency and reward circuits. Differential activation with regard to<br />

rs1006737 is found in different regions <strong>of</strong> the brain including<br />

hippocampus, amygdala, frontal and temporal gyrus and precuneus.<br />

Moreover, we will give an outlook on association <strong>of</strong> rs1006737 with<br />

behavioral and personality measurements.<br />

Conclusions: The results <strong>of</strong> our studies are put into context with<br />

other recent imaging genetic studies on rs1006737 from the literature.<br />

32<br />

S8.5 HUMAN FUNCTIONAL STUDIES OF ANK3 AND<br />

CACNA1C FROM THE GREEK LOGOS COHORT<br />

P. Bitsios*(1), P. Roussos(1,2), S. Giakoumaki(1), M. Pasparakis(1)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Sciences, University <strong>of</strong><br />

Cretel 2. Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong> Medicine<br />

*pbitsios@med.uoc.gr<br />

Introduction: The rs1006737 CACNA1C and rs10994336 ANK3<br />

genetic variants have been recently identified as the most consistent,<br />

genome-wide significant risk factors for bipolar disorder, while the<br />

CACNA1C variant has also been associated with schizophrenia and<br />

major depression. Genetic association studies in confounder-free,<br />

healthy subjects using multiple disease endophenotypes is a useful<br />

strategy that may elucidate the functional role <strong>of</strong> GWAS-supported<br />

risk polymorphisms and, in particular, the mechanism by which they<br />

increase risk for mental illness.<br />

Methodology: We examined the phenotypic consequences <strong>of</strong> these<br />

risk alleles in a large homogeneous cohort <strong>of</strong> healthy young males<br />

(n=543 mean age 22.1±3) recruited from the first wave <strong>of</strong> the<br />

LOGOS project (Learning On <strong>Genetics</strong> Of Schizophrenia) in<br />

Heraklion, Crete. Subjects were tested for sensorimotor gating as<br />

assessed by prepulse inhibition (PPI), working and verbal memory,<br />

executive function, startle reactivity and temperament and personality<br />

traits. A subsample <strong>of</strong> 220 subjects underwent testing for affective<br />

startle modulation using the IAPS (International Affective Picture<br />

System).<br />

Results: UNPHASED analysis revealed that the CACNA1C risk<br />

allele (rs1006737_A) was associated with lower extraversion and<br />

higher harm avoidance, trait anxiety and paranoid ideation consistent<br />

with a non-specific proneness to anxiety and paranoia, while the<br />

ANK3 risk allele (rs10994336_T) was associated with lower novelty<br />

seeking and behavioral response to reward, consistent with proneness<br />

to anhedonia. Both risk alleles were associated with high startle<br />

reactivity. CACNA1C risk allele carriers also presented with<br />

enhanced startle reactivity in the aversive-pictures condition. The risk<br />

genotypes did not affect sensorimotor gating, working memory or<br />

executive function in this cohort <strong>of</strong> healthy males.<br />

Conclusions: The personality configuration and affective startle<br />

reactivity data suggest that these GWAS-supported risk<br />

polymorphisms are associated with emotional dysregulation,<br />

penetrant in otherwise asymptomatic and apparently cognitively<br />

intact individuals. At least as far as the CACNA1c variant is<br />

concerned this is in keeping with recent reports suggesting a role <strong>of</strong><br />

this variant in abnormal threat signal processing within the<br />

hippocampus and/or amygdala which may lie in the causal pathway<br />

that links genetic risk to affective illness, or indeed, schizophrenia.<br />

The relative risk and the environmental/epistatic conditions required<br />

for the conversion <strong>of</strong> risk allele carriers to mild symptomatic stages<br />

and/or to overt illness need to be assessed in longitudinal designs.<br />

This strategy may facilitate early, effective and cost-efficient<br />

genotype-dependent therapeutic interventions.


SYMPOSIUM 9: ADVANCES IN UNDERSTANDING<br />

DYSBINDIN-1 FUNCTIONS RELEVANT TO<br />

SCHIZOPHRENIA<br />

S9.1 REGULATION OF SYNAPSE COMPOSITION BY THE<br />

DYSBINDIN ASSOCIATED COMPLEXES, BLOC-1 AND AP-3<br />

V. Faundez*<br />

Emory University<br />

*vfaunde@emory.edu<br />

Introduction: Dysbindin assembles into the biogenesis <strong>of</strong> lysosome<br />

related organelles complex 1 (BLOC-1), which interacts with the<br />

clathrin adaptor complex 3 (AP-3). Both complexes participate in a<br />

common endosome trafficking route. Deficiencies in AP-3 and<br />

BLOC-1 subunits affect synapse composition suggesting roles for the<br />

two complexes in protein sorting to or from synapses.<br />

Methodology: We tested this hypothesis by analyzing the targeting<br />

<strong>of</strong> an AP-3 and BLOC-1-dependent membrane protein cargo targeted<br />

to in pre and postsynaptic compartments, PI4KIIa.<br />

Results: PI4KIIaco-purifies with BLOC-1 and AP-3 in neuronal<br />

cells, an association sensitive to the BLOC-1 genetic dosage. These<br />

biochemical interactions were corroborated genetically as since a<br />

decreased PI4KIIacontent in the dentate gyrus neuropil <strong>of</strong> dysbindinnull<br />

sandy mice was phenocopied in BLOC-1 subunit deficiencies,<br />

pallid and muted, and the AP-3-null mocha allele. Reduction <strong>of</strong><br />

PI4KIIain the dentate reflects a failure to traffic from the cell body.<br />

PI4KIIawas targeted to processes in wild type primary cultured<br />

cortical neurons and PC12 cells, but failed to reach neurites in cells<br />

lacking either AP-3 or BLOC-1 complexes. Similarly, disruption <strong>of</strong><br />

an AP-3 sorting motif in PI4KIIaimpaired its sorting into processes <strong>of</strong><br />

PC12 and primary cultured cortical neuronal cells.<br />

Conclusions: Our findings indicate a novel vesicle transport<br />

mechanism requiring dysbindin-associated complexes for cargo<br />

targeting from neuronal cell bodies to neurites and nerve terminals,<br />

and suggest that mistargeting <strong>of</strong> specific vesicular cargos may<br />

underlie, in part, the molecular pathogenesis <strong>of</strong> schizophrenia<br />

33<br />

S9.2 EPISTATIC INTERACTIONS BETWEEN DTNBP-1 AND<br />

COMT: EFFECTS ON WORKING MEMORY IN MICE AND<br />

PREFRONTAL PHYSIOLOGICAL ACTIVITY IN HUMANS<br />

D. Weinberger*, F. Papeleo, G. Carr, J. Chen, M. Burdick, R.<br />

Straub, J. Callicott<br />

NIMH/NIH<br />

*weinberd@mail.nih.gov<br />

Introduction: <strong>Psychiatric</strong> genes are involved in networks and<br />

pathways that subserve risk relevant cell biology, and the risk valence<br />

<strong>of</strong> a particular gene likely depends on the integrity <strong>of</strong> other related<br />

proteins. DTNBP1 has been linked with DA synaptic activity via D2<br />

receptor trafficking after ligand induced internalization, which has<br />

been demonstrated in cell culture (Iizuki et al 2008) and in mice<br />

(Yuanyuan et al 2009). We tested the cognitive effects <strong>of</strong> the<br />

DTNBP1-DA interaction in mice and the cortical physiologic effects<br />

in humans with fMRI by genetically varying dysbindin expression<br />

and COMT enzyme activity.<br />

Methodology: Based on the D2 trafficking effects and the well<br />

characterized inverted U shaped DA response relationship in<br />

prefrontal cortex, it was predicted that decreased dysbindin<br />

expression and increased synaptic dopamine would translate into<br />

abnormal D2 signaling which would be disadvantageous in both the<br />

mouse and human paradigms. Single (COMT or DTNBP1) and dual<br />

(COMT plus DTNBP1) C57 background knockout mice were created<br />

(total N= 71 animals) and tested in the forced choice alternating T<br />

maze working memory task. It was predicted that individual gene<br />

alterations would enhance working memory, but combined defects<br />

would diminish it. For the human fMRI experiment, normal subjects<br />

(N=115) performed the N back working memory task. Similarly, it<br />

was predicted that there would be nonlinear, epistatic effects <strong>of</strong><br />

COMT and DTNBP1 on physiological efficiency during working<br />

memory task in normal subjects, associated with exaggerated D2<br />

signaling. The effect <strong>of</strong> a DTNBP1 haplotype associated with<br />

decreased expression <strong>of</strong> dysbindin in human prefrontal cortex (Bray<br />

et al 2005) was tested on the background <strong>of</strong> COMT val/met<br />

genotypes.<br />

Results: Compared with wild type animals, each single KO mouse<br />

required fewer days to achieve criteria, but the combined gene KO,<br />

either combined heterozygotes or combined homozygotes, showed a<br />

dramatic failure <strong>of</strong> learning the task (p


S9.3 DYSBINDIN-1 MODULATES BRAIN STRUCTURE,<br />

GLUTAMATERGIC FUNCTION AND MEMORY IN MICE<br />

J. Jentsch*(1,2)<br />

1. Department <strong>of</strong> Psychology, UCLA 2. Department <strong>of</strong> Psychiatry &<br />

Bio-behavioral Sciences, UCLA 3.<br />

*jentsch@psych.ucla.edu<br />

Introduction: Though dysbindin-1 has been repeatedly associated<br />

with schizophrenia, much more needs to be learned about the specific<br />

role the gene product plays in the pathogenesis <strong>of</strong> the disorder.<br />

Fortunately, many <strong>of</strong> the biological roles and functional properties <strong>of</strong><br />

dysbindin-1 are conserved between mice and humans, permitting a<br />

direct study <strong>of</strong> the cellular, network and systems abnormalities that<br />

mediate mnemonic and cognitive deficits caused by functional<br />

variation in the gene. Here, we take use <strong>of</strong> a genetic model <strong>of</strong><br />

dysbindin-1 deficiency in mice to explore the mechanisms by which<br />

this gene affects memory-related systems, including the prefrontal<br />

cortex and hippocampus.<br />

Methodology: Mice carrying a spontaneous, null mutation <strong>of</strong> the<br />

gene encoding dysbindin-1, on the C57Bl6 background, were used in<br />

the studies. A combination <strong>of</strong> in vitro electrophysiological recordings<br />

from prefrontal cortical and hippocampal neurons, in vivo<br />

microdialysis studies, in vivo micro-magnetic resonance imaging<br />

scans, ex vivo gene expression analyses and behavioral assessments<br />

<strong>of</strong> memory function (delayed non-match-to-position, object/social<br />

recognition, contextual fear conditioning) were used.<br />

Results: Genetic dysbindin deficiency (including haploinsufficiency)<br />

is associated with alterations in glutamatergic signaling. In vitro,<br />

spontaneous and evoked excitatory post-synaptic currents are reduced<br />

in prefrontal cortex and hippocampus a variety <strong>of</strong> evidence supports<br />

the notion that this is due to compromised pre-synaptic release <strong>of</strong><br />

glutamate during sustained activity conditions. Additionally,<br />

adaptations in post-synaptic gluamate receptor function (specifically,<br />

NMDA/glutamate receptor hypoactivity) has been identified in both<br />

regions, and this effect is at least in part transcriptional in nature. In<br />

vivo, evoked gluamate release is - as expected - diminished. Finally,<br />

functional activity within hippocampus and dopaminergic networks<br />

has been identified with manganese-enhanced microMRI.<br />

Collectively, these glutamatergic abnormalities within hippocampus<br />

and prefrontal cortex combine to underlie complex memory systems<br />

abnormalities, including difficulties with working memory, object<br />

recognition and contextual fear learning.<br />

Conclusions: A range <strong>of</strong> pre- and post-synaptic neuroadaptations<br />

affecting glutamatergic transmission appear to be caused by genetic<br />

deficiency in dysbindin-1 function. By identifying the proximal<br />

changes in neurotransmission that are caused by reduced dysbindin-1<br />

function and that link to the memory systems complications<br />

stemming from neurochemical changes, rationale therapeutic<br />

strategies can be developed.<br />

34<br />

S9.4 DYSBINDIN-1 MUTANT MICE IMPLICATE REDUCED<br />

PARVALBUMIN-CELL MEDIATED INHIBITION AS A<br />

FINAL COMMON PATHWAY IN THE<br />

PATHOPHYSIOLOGY OF SCHIZOPHRENIA<br />

K. Talbot*, G. Carlson, M. Gandal, R. Gur, S. Arnold, S. Siegel<br />

University <strong>of</strong> Pennsylvania<br />

*talbotk2@mail.med.upenn.edu<br />

Introduction: At least twenty studies have reported an association<br />

between schizophrenia and variation in the dystrobrevin binding<br />

protein 1 gene (DTNBP1). An increasing number <strong>of</strong> studies report<br />

that several <strong>of</strong> these DTNBP1 risk variants are associated with<br />

severity <strong>of</strong> the disorder's positive and negative symptoms, as well as<br />

its cognitive deficits. While the relationship <strong>of</strong> risk variants in<br />

DTNBP1 and the expression <strong>of</strong> the encoded protein, dysbindin-1,<br />

remains unclear, reductions in that protein appear to be a common<br />

feature <strong>of</strong> synaptic tissue in at least three brain areas in<br />

schizophrenia: auditory cortices, hippocampal formation, and<br />

dorsolateral prefrontal cortex. Studies on sandy mice homozygous for<br />

a deletion mutation in Dtnbp1 (dys -/-) resulting in loss <strong>of</strong> dysbindin-<br />

1 have shown that this protein directly or indirectly regulates<br />

glutamate, dopamine, and GABAergic transmitter systems. All <strong>of</strong><br />

those systems regulate cerebrocortical gamma-band oscillations,<br />

which are generated by interactions among parvalbumin (PV) cell<br />

networks and pyramidal cells. Abnormalities in gamma oscillations<br />

are common features in schizophrenia and are associated (like<br />

DTNBP1 risk variants) with the diagnostic symptoms and cognitive<br />

deficits <strong>of</strong> the disorder. Since disruption <strong>of</strong> gamma oscillations may<br />

be a final common pathway in the pathophysiology <strong>of</strong> schizophrenia,<br />

we tested the effects <strong>of</strong> dysbindin-1 loss on auditory-induced gamma<br />

activity, the PV cells and inhibitory drive critical to that activity, and<br />

endophenotypes <strong>of</strong> schizophrenia associated with gamma activity.<br />

Methodology: The auditory cortex and hippocampus <strong>of</strong> 12 male dys<br />

-/- and 12 male wild-type littermates on a C57BL6/J background<br />

were studied. PV cell populations in both areas were characterized<br />

immunohistochemically. Auditory evoked response potentials were<br />

recorded to study acoustic responses, sensory gating, and prepulse<br />

inhibition (PPI) and later used in time-frequency analyses to evaluate<br />

auditory evoked gamma oscillations recorded from the hippocampus.<br />

Voltage-sensitive dye (VSD) imaging <strong>of</strong> hippocampal slice<br />

preparations was used to study inhibitory drive in CA1 following<br />

Schaffer collateral stimulation.<br />

Results: While showing normal acoustic brain stem responses, dys -<br />

/- mice displayed auditory endophenotypes <strong>of</strong> schizophrenia, namely<br />

reduced sensory gating and PPI, both <strong>of</strong> which were correlated with<br />

reduced power and phase-locking <strong>of</strong> evoked, early gamma activity.<br />

The basis for these abnormalities was suggested by reduced PV<br />

immunoreactivity in PV cells <strong>of</strong> the auditory cortex and hippocampus<br />

in dys -/- mice. The potential impact <strong>of</strong> this abnormality in fastspiking<br />

inhibitory neurons on gamma activity was shown by<br />

markedly reduced inhibitory CA1 responses <strong>of</strong> dys -/- mice to<br />

Schaffer collateral activation with no apparent effect on evoked<br />

excitation.<br />

Conclusions: These findings collectively indicate that dysbindin-<br />

1 promotes generation <strong>of</strong> gamma oscillations by PV-pyramidal cell<br />

networks and that disruption <strong>of</strong> that role promotes auditory<br />

endophenotypes <strong>of</strong> schizophrenia potentially promoting both its<br />

clinical symptoms and cognitive deficits. The same cell networks<br />

may be a common target <strong>of</strong> other genetic risk factors for this<br />

disorder.


SYMPOSIUM 10: RECENT ADVANCES IN THE GENETICS<br />

OF POST-TRAUMATIC STRESS DISORDERS (PTSD):<br />

GOING BEYOND GENETIC ASSOCIATIONS<br />

S10.1 FKBP5, EARLY TRAUMA AND POST TRAUMATIC<br />

STRESS DISORDER<br />

E. Binder*(1,2)<br />

1. Max-Planck Institute <strong>of</strong> Psychiatry 2. Emory University School <strong>of</strong><br />

Medicine, Dept. <strong>of</strong> Psychiatry and Behavioral Sciences<br />

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

Introduction: Early trauma has been shown to increase the risk for a<br />

number <strong>of</strong> psychiatric disorders, including post traumatic stress<br />

disorder (PTSD). This environmental effect can be moderated by<br />

genetic polymorphisms in a number <strong>of</strong> genes, including the gene<br />

encoding FKBP5, a co-chaperone <strong>of</strong> the glucocorticoid receptor<br />

(GR). This effect is most likely mediated by differences in the<br />

response <strong>of</strong> the stress response system in carriers <strong>of</strong> the opposite<br />

alleles <strong>of</strong> these polymorphisms. However, the molecular mechanism<br />

<strong>of</strong> this gene x environment interaction has not yet been investigated.<br />

Methodology: We assessed the effects <strong>of</strong> early trauma and FKBP5<br />

genotype on GR-mediated FKBP5 transcription, endocrine measures<br />

and DNA methylation in peripheral blood in two different cohorts.<br />

This was followed by reporter gene assays to investigate allele- and<br />

DNA methylation-specific effects in specific FKBP5 enhancer<br />

regions containing GR response elements (GREs).<br />

Results: We observed that early trauma disrupts the positive<br />

feedback between GR-activation and FKBP5 transcription in an<br />

allele-specific manner, which is paralleled by allele-specific effects<br />

<strong>of</strong> early trauma on GR-sensitivity. DNA-methylation <strong>of</strong> specific CGs<br />

close to functional intronic GREs was also significantly associated<br />

with exposure to early trauma. In two independent cohorts (N = 68<br />

and N = 93) we observed an FKBP5 genotype x early trauma<br />

interaction on FKBP5 DNA methylation. In addition, these specific<br />

methylation patterns predicted ex-vivo GR sensitivity as well as the<br />

cortisol response to a psychological stressor (Trier Social Stress<br />

Test). The functionality <strong>of</strong> these methylation changes was confirmed<br />

in reported gene assays.<br />

Conclusions: The presented data suggest that the interaction <strong>of</strong><br />

specific functional FKBP5 polymorphisms and early trauma on adult<br />

PTSD symptoms could be mediated by allele-specific effects <strong>of</strong> early<br />

trauma on DNA methylation in critical enhancer regions <strong>of</strong> this gene.<br />

35<br />

S10.2 CORTICOTROPHIN-RELEASING HORMONE TYPE 1<br />

RECEPTOR GENE (CRHR1) VARIANTS PREDICT POST-<br />

TRAUMATIC STRESS DISORDER ONSET AND COURSE IN<br />

PEDIATRIC INJURY PATIENTS<br />

K. Koenen*<br />

Columbia Mailman School <strong>of</strong> Public Health<br />

*kck5@columbia.edu<br />

Introduction: Posttraumatic stress disorder (PTSD) is a common and<br />

disabling anxiety disorder that may occur in the aftermath <strong>of</strong><br />

exposure to potentially traumatic life events. PTSD is moderately<br />

heritable, but few specific molecular variants accounting for this<br />

heritability have been identified. Genes regulating the hypothalamicpituitary-adrenal<br />

(HPA) axis, such as corticotrophin-releasing<br />

hormone type 1 receptor gene (CRHR1), have been implicated in<br />

traumatic-stress related phenotypes but have yet to be studied in<br />

relation to PTSD.<br />

Methodology: The present study sought to examine the relation<br />

between 9 single nucleotide polymorphisms (SNPs) in the CRHR1<br />

gene and posttraumatic stress symptoms in a prospective study <strong>of</strong><br />

pediatric injury patients (n=103) who were first assessed in the acute<br />

aftermath <strong>of</strong> their injury at the hospital.<br />

Results: Results indicated that multiple SNPs were associated with<br />

acute symptoms at a univariate level, and after correction for multiple<br />

testing, rs12944712 was significantly related to acute PTSD<br />

symptoms. Longitudinal latent growth curve analyses suggest that<br />

rs12944712 is also related to both acute symptom level and trajectory<br />

<strong>of</strong> symptoms over time.<br />

Conclusions: The present study adds support for the role <strong>of</strong> CRHR1<br />

in the stress response following potentially traumatic event exposure<br />

in youth.


S10.3 EVIDENCE FOR ASSOCIATION OF THE<br />

PACAP/PAC1 RECEPTOR PATHWAY WITH POST-<br />

TRAUMATIC STRESS SYMPTOMS<br />

K. Ressler*(1,2), K. Mercer(1,2), B. Bradley(1,3), T. Jovanovic(1),<br />

E. Binder(1,4), A. Myers(5), V. May(6)<br />

1. Emory University 2. HHMI 3. VA Medical Center 4. Max Planck<br />

Institute <strong>of</strong> Psychiatry 5. University <strong>of</strong> Miami 6. University <strong>of</strong><br />

Vermont<br />

*kressle@emory.edu<br />

Introduction: Post traumatic stress disorder is a prevalent and<br />

debilitating psychiatric disorder. Although a number <strong>of</strong> studies have<br />

examined genes within stress response pathways, none have yet<br />

examined the role <strong>of</strong> the PACAP/PAC1 receptor pathway with<br />

Posttraumatic Stress symptoms. Pituitary adenylate cyclaseactivating<br />

polypeptide (PACAP) is known to broadly regulate the<br />

cellular stress response. In contrast, it is unclear if the PACAP-PAC1<br />

receptor pathway has a role in human psychological stress responses,<br />

such as post-traumatic stress disorder (PTSD).<br />

Methodology: We use a combination <strong>of</strong> convergent genomic<br />

approaches, gene association studies in an all-traumatized population,<br />

and convergent biological studies examining the association <strong>of</strong><br />

PACAP levels and PAC1 genotypes with PTSD symptoms and fearrelated<br />

physiology. We then examine effects <strong>of</strong> PAC1 genotypes on<br />

PAC1 gene expression levels, and effects <strong>of</strong> methylation at the PAC1<br />

locus on PTSD symptoms. Finally, we examine differential<br />

expression <strong>of</strong> the PAC1 / PACAP genes within the rodent brain<br />

following fear conditioning and as a function <strong>of</strong> estrogen.<br />

Results: Here we find, in heavily traumatized African American<br />

subjects, a sex-specific association <strong>of</strong> PACAP blood levels with fear<br />

physiology, PTSD diagnosis and symptoms in females. We examined<br />

44 single nucleotide polymorphisms (SNPs) spanning the PACAP<br />

(encoded by ADCYAP1) and PAC1 (encoded by ADCYAP1R1)<br />

genes, demonstrating a sex-specific association with PTSD<br />

(N>1200). A single SNP in a putative estrogen response element<br />

(ERE) within ADCYAP1R1, rs2267735, predicts PTSD diagnosis and<br />

symptoms in females only. This SNP also associates with fear<br />

discrimination and with ADCYAP1R1 messenger RNA expression in<br />

human brain. Methylation <strong>of</strong> ADCYAP1R1 in peripheral blood is also<br />

associated with PTSD. Complementing these human data,<br />

ADCYAP1R1<br />

Conclusions: These data suggest that perturbations in the PACAP-<br />

PAC1 pathway are involved in abnormal stress responses underlying<br />

PTSD. These sex-specific effects may occur via estrogen regulation<br />

<strong>of</strong> ADCYAP1R1 gene expression. PACAP levels and ADCYAP1R1<br />

SNPs may serve as useful biomarkers to further our mechanistic<br />

understanding <strong>of</strong> PTSD.<br />

36<br />

S10.4 A GENETIC VARIANT BDNF POLYMORPHISM<br />

ALTERS EXTINCTION LEARNING IN BOTH MOUSE AND<br />

HUMAN<br />

F. Soliman, S. Pattwell, C. Glatt, B. Casey, F. Lee*<br />

Weill Cornell Medical College<br />

*fslee@med.cornell.edu<br />

Introduction: Highly conserved neural circuitry between rodents and<br />

humans has allowed for in-depth characterization <strong>of</strong> behavioral and<br />

molecular processes associated with emotional learning and memory.<br />

In this study we focused on identifying biologically valid fear-related<br />

phenotypes across species.<br />

Methodology: We focussed on a common single-nucleotide<br />

polymorphism (SNP) in the brain-derived neurotrophic factor<br />

(BDNF) gene that leads to a valine (Val) to methionine (Met)<br />

substitution at codon 66 (Val66Met) in both mouse and human<br />

behavioral studies as well human functional imaging studies.<br />

Results: In an inbred genetic knock-in mouse strain that expresses<br />

the variant BDNF allele to recapitulate the specific phenotypic<br />

properties <strong>of</strong> the human polymorphism in vivo, we found the BDNF<br />

Val66Met genotype was associated with treatment-resistant forms <strong>of</strong><br />

anxiety-like behavior. An additional objective <strong>of</strong> this study was to<br />

test if the Val66Met genotype could affect extinction learning in our<br />

mouse model and whether such findings could be generalized to<br />

human populations. Both mice and human carrying the Met allele<br />

were impaired in extinguishing a conditioned fear response, which<br />

was paralleled by atypical frontoamygdala activity in humans.<br />

Conclusions: Thus, this variant BDNF allele may play a role in<br />

anxiety disorders, such as post-traumatic stress disorder, showing<br />

impaired learning <strong>of</strong> cues that signal safety versus threat and in the<br />

efficacy <strong>of</strong> treatments that rely on extinction mechanisms, such as<br />

exposure therapy.


SYMPOSIUM 11: GENOMICS OF SUICIDAL BEHAVIOR<br />

AND INTERMEDIATE PHENOTYPES<br />

S11.1 AN INTERNATIONAL GENOME-WIDE STUDY OF<br />

SUICIDAL BEHAVIOR AND INTERMEDIATE<br />

PHENOTYPES IN MOOD DISORDERS<br />

J. Mann*(1), D. Rujescu(2), G. Turecki(3), H. Galfalvy(1), C.<br />

Hodgkinson(4), D. Goldman(4), M. Oquendo(1), Y. Huang(1), A.<br />

Burke(1), D. Currier(1)<br />

1. Columbia University 2. Ludwig Maximilians University 3. McGill<br />

University 4. NIAAA<br />

*jjm@columbia.edu<br />

Introduction: The diathesis for suicidal behavior is partly heritable<br />

with estimates from twin studies apporaching 50% for suicide.<br />

Adoption studies also support a genetic component for suicide.What<br />

is not known are the responsible genes. Most studies have been<br />

guidied by the known neurobiolgy and ahve concentrated on genes<br />

associated with the serotonergic system. This NIMH funded study<br />

sought to find genes using a sample <strong>of</strong> almost 2800 cases and<br />

controls. The phenotype was suicide or nonfatal suicide attempts.<br />

Methodology: About 2800 cases and controls, all Caucasians,<br />

including suicides, nonfatal suicides, most with a major depressive<br />

disorder, and controls <strong>of</strong> two types: one psychiatric ocntrol group<br />

with a major depressive disorder and one group <strong>of</strong> healthy volunteers<br />

found to be free from axis I psychiatric diagnosis. Diagnoses were<br />

done by a SCID I structured clinical interview. Genotyping was done<br />

using the Infinium HumanHap550 Genotyping BeadChip that<br />

contains >555,000 SNP loci with high-density tag SNP content .<br />

Results: The sample is made up <strong>of</strong> apporximately 590 suicides and<br />

158 sudden death controls, over 2000 nonfatal suicide attempters<br />

with a major depressive disorder (MDD) and controls that include<br />

healthy volunteers and 579 psychiatric controls (MDD with no life<br />

time suicide attempt). The basic analysis compares 596 suicides and<br />

427 nonfatal attempters to 1740 nonattempters. Next we will check<br />

for the effects <strong>of</strong> major depression by comparing 596 suicide and 423<br />

nonfatal attempts with major depression to 579 depressed controls.<br />

Finally we will explore differences between suicides and nonfatal<br />

attempters.<br />

Conclusions: This study will generate new findings from the largest<br />

such GWAS reported todate on suicidal behavior in major<br />

depression.<br />

37<br />

S11.2 GENOME-WIDE ASSOCIATION STUDIES OF<br />

INTERMEDIATE PHENOTYPES OF SUICIDAL BEHAVIOR:<br />

FOCUS ON AGGRESSION AND IMPULSIVITY<br />

D. Rujescu*<br />

University <strong>of</strong> Munich<br />

*dan.rujescu@med.uni-muenchen.de<br />

Introduction: Every year over 1 million people commit and over 10<br />

million people attempt suicide. This is one suicide every 40 seconds<br />

and one suicide attempt every 3 seconds worldwide. Suicide accounts<br />

for almost 2% <strong>of</strong> the world's death and it has emerged as one <strong>of</strong> the<br />

leading causes <strong>of</strong> death among individuals aged 15-34 years in most<br />

<strong>of</strong> the countries. The risk <strong>of</strong> suicide-related behavior is supposed to<br />

be determined by a complex interplay <strong>of</strong> sociocultural factors,<br />

psychiatric history, personality traits, and genetic vulnerability. This<br />

view is supported by adoption and family studies indicating that<br />

suicidal acts have a genetic contribution <strong>of</strong> ca. 55% that is<br />

independent <strong>of</strong> the heritability <strong>of</strong> Axis I and II psychopathology.<br />

Beside all other risk factors, especially personality seems to have a<br />

high impact on suicidal behaviour.<br />

Methodology: Aggression has been associated with suicidal<br />

behaviour alt least since the early findings by Asberg et al. (1976).<br />

Since then a high amount <strong>of</strong> studies could replicate this link.<br />

Results: For that reasons we have initiated a large scale case control<br />

genetic association study and investigated especially the role <strong>of</strong><br />

personality traits like aggression, anger or impulsivity as intermediate<br />

phenotypes <strong>of</strong> suicidal behaviour.<br />

Conclusions: We performed genome-wide association studies on<br />

these traits in over 4000 patients and controls and will discuss these<br />

results in relation to suicidal behaviour.


S11.3 META-ANALYSIS OF GENOME-WIDE<br />

ASSOCIATION STUDIES OF SUICIDE ATTEMPT IN MOOD<br />

DISORDER<br />

R. Perlis*<br />

Massachusetts General Hospital<br />

*rperlis@partners.org<br />

Introduction: Risk for suicide attempt appears to be in part heritable.<br />

Other neuropsychiatric studies suggest that large sample sizes may be<br />

required to detect association with common variation, and that such<br />

studies are facilitated by meta-analysis.<br />

Methodology: Fixed and random-effects meta-analysis was used to<br />

investigate genome-wide association data from multiple mood<br />

disorder cohorts. In addition, aggregate risk scores and pathwaybased<br />

tests were used to investigate association across cohorts.<br />

Results: No single locus reached a threshold for genomewide<br />

significant. However, polygenic and pathway-based tests suggest<br />

some consistency in liability across cohorts.<br />

Conclusions: Individual loci <strong>of</strong> large effect do not appear to<br />

contribute to inherited liability for suicide attempt among mood<br />

disorder patients. However, tests which aggregate risk across loci<br />

suggest the utility <strong>of</strong> alternative methodologies for follow-up <strong>of</strong><br />

GWAS results.<br />

38<br />

S11.4 GENOME-WIDE EPIGENETIC REGULATION BY<br />

EARLY-LIFE TRAUMA AND SUICIDE<br />

B. Labonte, M. Suderman, G. Maussion, M. Szyf, M. Meaney,<br />

G. Turecki*<br />

McGill University<br />

*gustavo.turecki@mcgill.ca<br />

Introduction: Our genome adapts to the environment pressures in<br />

part through epigenetic mechanisms. Specific DNA methylation<br />

changes occur in animals as a response to variation in early<br />

environment, and recent data suggest that similar processes take place<br />

in humans who have histories <strong>of</strong> early life adversity. Here we report a<br />

genome-wide study <strong>of</strong> promoter methylation in individuals who died<br />

by suicide and were victims <strong>of</strong> severe abuse during childhood.<br />

Methodology: Promoter DNA methylation levels were pr<strong>of</strong>iled using<br />

meDIP followed by microarray hybridization in hippocampal tissue<br />

from 41 French-Canadian men (25 with a history <strong>of</strong> severe childhood<br />

abuse and 16 controls). Methylation pr<strong>of</strong>iles were compared to<br />

corresponding genome-wide gene expression pr<strong>of</strong>iles obtained by<br />

mRNA arrays. Methylation differences between groups were<br />

validated on neuronal and non-neuronal DNA fractions isolated by<br />

fluorescence-assisted cell sorting (FACS).<br />

Results: We identified 362 differentially methylated promoters in<br />

individuals with a history <strong>of</strong> abuse compared to controls. Among<br />

these promoters, 248 indicated hypermethylation and 114<br />

hypomethylation. Validation and site–specific quantification <strong>of</strong> DNA<br />

methylation in the most significantly differentially methylated<br />

gene promoters indicated that methylation differences were mainly<br />

driven by the neuronal cellular fraction. Artificially<br />

methylated constructs mimicking the methylation state in samples<br />

from abused suicide completers showed decreased promoter<br />

transcriptional activity associated with decreased hippocampal<br />

expression <strong>of</strong> the most significantly differentially methylated gene.<br />

Conclusions: Our results suggest that childhood adversity triggers<br />

epigenetic alterations in the promoters <strong>of</strong> several genes in<br />

hippocampal neurons increasing risk for suicide.


SYMPOSIUM 12: PHARMACOGENETICS IN PSYCHIATRIC<br />

PRACTICE: "YES WE CAN"<br />

S12.1 ANXIOUS DEPRESSION AND PHARMACOGENETICS<br />

OF ANTIDEPRESSANT DRUGS<br />

E. Binder*(1,2)<br />

1. Emory University School <strong>of</strong> Medicine, Dept. <strong>of</strong> Psychiatry and<br />

Behavioral Sciences 2. Max-Planck Institute <strong>of</strong> Psychiatry<br />

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

Introduction: A number <strong>of</strong> studies have shown that suffering from<br />

anxious depression is one <strong>of</strong> the most consistent predictors for a<br />

negative treatment outcome. This might be an indication for the fact<br />

that anxious depression could be biologically and genetically distinct<br />

subtype <strong>of</strong> major depression, resulting in different genetic predictors<br />

for antidepressant treatment response.<br />

Methodology: Genome-wide and candidate gene-base association<br />

data from two large pharmacogenetics cohorts, the "Munich<br />

Antidepressant Signature"-MARS project and the "Sequenced<br />

Treatment Alternatives to Relieve Depression" - STAR*D study will<br />

be presented. The analyses will focus on gene x anxious depression<br />

interactions on antidepressant treatment response.<br />

Results: In both studies, anxious depression is a strong negative<br />

predictor <strong>of</strong> treatment outcome. Genetic polymorphisms in genes<br />

within the stress hormone system, CRHR1 and CRHBP interact with<br />

anxious depression to predict treatment response. In both genes, the<br />

pharmacogenetic associations are only observed in patients with<br />

anxious depression. Data from a genome-wide association study in<br />

the MARS samples show that a number <strong>of</strong> polymorphisms strongly<br />

interact with anxious depression to predict antidepressant therapy<br />

response. These results are currently being followed up in the<br />

STAR*D as well as GENDEP cohorts.<br />

Conclusions: Anxious depression is an important factor to be<br />

considered in antidepressant drug pharmacogenetic studies. Selected<br />

genetic predictors appear to be specific for this subtype <strong>of</strong> major<br />

depression.<br />

39<br />

S12.2 NTRK2 AND PDE11A PREDICT RESPONSE TO<br />

LITHIUM IN BIPOLAR DISORDER IN PATIENTS WITH<br />

ELATED MANIA AND POSITIVE FAMILY HISTORY<br />

J. Kelsoe*(1,2), S. Leckband(1,2), A. DeModena(1,2), T.<br />

Shekhtman(1,2), M. McCarthy(1,2)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California, San Diego 2.<br />

Department <strong>of</strong> Psychiatry, VA San Diego Healthcare System<br />

*jkelsoe@ucsd.edu<br />

Introduction: Lithium was the first mood stabilizer medication for<br />

bipolar disorder, and remains the gold standard with the strongest<br />

evidence for efficacy and reduction <strong>of</strong> suicide risk. A subset <strong>of</strong><br />

patients have a very robust response with virtual elimination <strong>of</strong><br />

episodes. Several clinical features have been shown to predict good<br />

response to lithium, in particular, elated mania and family history <strong>of</strong><br />

bipolar disorder, and it has been proposed that bipolar disorder with<br />

good lithium response may comprise a biologically distinct form <strong>of</strong><br />

illness. As some data suggest that this excellent lithium response is<br />

best obtained when treatment begins soon after onset <strong>of</strong> illness, there<br />

is a great clinical need for methods to predict response.<br />

Methodology: We have retrospectively assessed lithium response in<br />

a set <strong>of</strong> 288 bipolar patients using information from SCID or DIGS<br />

interview, lifechart and medical records. 632 SNPs were genotyped<br />

in 50 genes selected for their putative role in lithium’s mechanism <strong>of</strong><br />

action or bipolar disorder susceptibility. Positive SNPs from this<br />

analysis were examined in an independent prospective clinical trial<br />

sample <strong>of</strong> 77 subjects employing a monotherapy relapse prevention<br />

design with two year followup.<br />

Results: In the retrospective sample, SNPs in several genes showed<br />

nominal significant evidence <strong>of</strong> association to response. This<br />

evidence was substantially stronger in patients with a positive family<br />

history <strong>of</strong> bipolar disorder and a history <strong>of</strong> elated mania and in<br />

particular implicated NTRK2 (p=0.006) and PDE11A (p=0.0004).<br />

Association <strong>of</strong> NTRK2 was replicated in the independent prospective<br />

sample (rs1387923, p=0.028).<br />

Conclusions: To extend these studies in a larger sample, we have<br />

recently begun the Pharmacogenomics <strong>of</strong> Bipolar Disorder (PGBD)<br />

study which will use a similar prospective relapse prevention design<br />

at 10 international collaborative sites. Together these data support the<br />

idea that lithium responsive bipolar disorder may be genetically and<br />

clinically distinct, and that incorporating clinical covariates<br />

considerably improves the power to detect associated genes.


S12.3 CLINICAL INTERPRETATION OF WHOLE EXOME<br />

SEQUENCING DATA IN ANTIDEPRESSANT TREATMENT<br />

RESPONSE<br />

G. Laje*, N. Akula, F. McMahon<br />

NIMH<br />

*lajeg@mail.nih.gov<br />

Introduction: A wide range <strong>of</strong> markers associated with<br />

antidepressant response have been detected through candidate gene,<br />

genome-wide and meta-analyses. However, these common genetic<br />

variants have small effect and no current clinical utility. It is possible<br />

that rare variants that may <strong>of</strong>fer additional insight into the<br />

pathophysiology <strong>of</strong> this complex phenotype. We undertook a whole<br />

exome sequencing experiment to search for uncommon and rare<br />

variants in individuals selected from the Sequenced Treatment<br />

Alternatives to Relieve Depression study (STAR*D).<br />

Methodology: We selected 8 individuals with Caucasian Europeanancestry<br />

from the STAR*D sample (n =8) who represented<br />

treatment-resistant cases (extreme non-responders) and sustained<br />

treatment remitters. Extreme non-responders (NR), derived from<br />

Level 4, had failed 4 treatment strategies. Sustained remitters (SR)<br />

were matched on gender, anxious depression status, treatment<br />

adherence, and ancestry. Additionally, the controls, were remitters at<br />

Level 1 endpoint and did not relapse in the 6 months post Level 1<br />

completion (QIDS-SR 11). All samples were sequenced at EdgeBio<br />

using a SOLiD platform with ≥ 10X coverage.<br />

Results: Initial results pertaining to single nucleotide variants<br />

(SNVs) revealed that each participant had about 90,000 SNVs on<br />

initial annotation. To characterize the potential impact <strong>of</strong> the SNVs<br />

on the resulting protein, we used SIFT (Sorting Intolerant From<br />

Tolerant), an algorithm that predicts whether an amino acid<br />

substitution affects function, in both novel and known variants. There<br />

were no differences in the mean number <strong>of</strong> known variant s between<br />

groups. However, the mean number <strong>of</strong> novel variants, predicted as<br />

damaging, had a small but significant difference in non responders<br />

(89.8+/-7.2) and sustained remitters (100.8+/-3.8) (p 0.05).<br />

Conclusions: The detection <strong>of</strong> rare variants through high throughput<br />

sequencing may point to relevant genes involved in the<br />

pathophysiology <strong>of</strong> treatment-resistant depression.<br />

40<br />

S12.4 GENETICS OF ANTIPSYCHOTIC DRUG RESPONSE<br />

AND INDUCED WEIGHT GAIN<br />

D. Müller*(1), A. Tiwari(1), N. Chowdhury(1), T. Lett(1), E.<br />

Brandl(1), O. Likhodi(1), N. Freeman(1), J. Lieberman(2), S.<br />

Potkin(3), H. Meltzer(4), J. Kennedy(1)<br />

1. Dept. <strong>of</strong> Psychiatry, University <strong>of</strong> Toronto at CAMH 2. Columbia<br />

University 3. Department <strong>of</strong> Psychiatry and Human Behavior,<br />

University <strong>of</strong> California 4. Vanderbilt University<br />

*daniel_mueller@camh.net<br />

Introduction: The substantial inter-individual variability observed in<br />

response and side effects with antipsychotic drugs is likely to largely<br />

depend on genetic factors. Based on findings from our group pointing<br />

to an association between a putative functional SNP in the 3’UTR <strong>of</strong><br />

the neurexin-1 (NRXN1) gene and MRI white matter volume in<br />

frontal brain regions, we tested for an association between the<br />

NRXN1 gene and response to antipsychotic drugs.<br />

One <strong>of</strong> the most debilitating side effects, emerging with many newer<br />

antipsychotic drugs, is substantial weight gain associated with<br />

cardiovascular complications and metabolic syndrome. Since<br />

previous findings by our group and collaborators strongly implicated<br />

the leptin-melanocortin energy homeostasis system to be associated<br />

with antipsychotic-induced weight gain, we investigated four variants<br />

in each <strong>of</strong> the melanocortin-4 receptor and the neuropeptide Y (NPY)<br />

genes in our ongoing studies.<br />

Methodology: A total <strong>of</strong> 237 patients who underwent treatment for<br />

chronic schizophrenia or schizoaffective disorder were evaluated for<br />

antipsychotic response and induced weight gain for up to six months.<br />

We genotyped one SNP in the NRXN1 gene (rs1045881) for<br />

response to clozapine treatment in European-Americans (n = 169).<br />

For the weight gain phenotype, SNPs rs2229616, rs17782313,<br />

rs11872992, rs8087522 were tested in the MC4R gene while SNPs<br />

rs16147, rs16475, rs5573 and rs5574 were tested in the NPY gene.<br />

Results: The neurexin rs1045881 T-allele was over represented in<br />

the non-responder group, suggesting the variant may influence<br />

clozapine response (OR = 2.2; p = 0.01). As for the phenotype <strong>of</strong><br />

antipsychotic-induced weight gain and the MC4R gene, analysis<br />

showed that clozapine treated patients <strong>of</strong> European ancestry who<br />

were carriers <strong>of</strong> the rs8087522 A-allele (AG+AA) on average gained<br />

significantly more weight than non-carriers (p=0.027). Furthermore,<br />

a functional relevance <strong>of</strong> this SNP was revealed by electrophoretic<br />

mobility shift assay analyses, where the presence <strong>of</strong> the A-allele<br />

appears to create a transcription factor-binding site. With respect to<br />

the NPY gene, a significant association <strong>of</strong> rs16147 genotype with<br />

weight change was observed in clozapine treated patients <strong>of</strong><br />

European descent (p=.002). Our preliminary analyses revealed that<br />

carriers <strong>of</strong> the functionally relevant C-allele gained significantly more<br />

weight compared to individuals with TT-genotype (TC+CC vs. TT;<br />

5.61%±5.4% vs. 0.32%±4.8%). Similarly, two other polymorphisms<br />

(rs5573 and rs5574) were significantly associated with weight change<br />

(p=.009 and p=.022), although these three associated polymorphisms<br />

were found to be in high linkage disequilibrium.<br />

Conclusions: Our results tentatively suggest novel associations<br />

between functionally relevant markers with response (NRXN1) and<br />

induced weight gain (MC4R and NPY) in patients treated with<br />

antipsychotic medication for schizophrenia. However, replication and<br />

further work is needed before screening <strong>of</strong> these genes will provide<br />

more efficient treatment strategies through personalized medicine<br />

algorithms.


INDIVIDUAL ORAL<br />

PRESENTATIONS<br />

41


ORAL PRESENTATIONS SESSION 1: FUNCTIONAL<br />

GENOMICS & MODEL ORGANISMS I<br />

OPS1.1 INVESTIGATING THE ROLE OF MUTATIONS IN<br />

ZFP804A ON GENE EXPRESSION<br />

D. Knight*, L. Jones, L. Wilkinson, T. Al-Janabi, M. Owen, D.<br />

Blake, M. O'Donovan<br />

Cardiff University<br />

*knightdj2@cf.ac.uk<br />

Introduction: Genome-wide significant evidence for association<br />

between a polymorphism in the gene and schizophrenia followed by<br />

replication has identified ZNF804A as one <strong>of</strong> a few strongly<br />

implicated schizophrenia susceptibility genes. Determining the<br />

function <strong>of</strong> the ZNF804A protein, which is currently unknown, may<br />

provide a way <strong>of</strong> elucidating the pathophysiology <strong>of</strong> this common,<br />

complex disorder. Based on sequence homology ZNF804A is<br />

hypothesised to regulate gene expression. The main focus <strong>of</strong> this<br />

work was to identify in the brain tissues <strong>of</strong> mice in which the<br />

orthologue Zfp804a has been mutated to carry a nonsense variant,<br />

genes that exhibit altered expression using global assays <strong>of</strong> gene<br />

expression.<br />

Methodology: Using the Genechip Mouse Exon 1.0 ST Array<br />

(Affymetrix) and brain tissue from 9 wild type (WT) control mice<br />

and 7 mice homozygous for the mutation in Zfp804a global gene<br />

expression was investigated. Partek Genomics suite 6.5 (Partek Inc,<br />

St Louis, MO) was used for data pre-processing and expression<br />

analysis. In addition the mouse orthologues <strong>of</strong> genome wide<br />

significant genes from the PGC SZ GWAS were specifically<br />

investigated for differential expression.<br />

Results: Following a Bonferroni correction transcripts <strong>of</strong> 29 genes<br />

showed significant alterations in expression. After FDR correction,<br />

we identified no pathways that were significantly enriched for genes<br />

with nominally significantly (p


OPS1.3 ESTABLISHMENT OF COCAINE PREFERENCE IS<br />

DELAYED IN αCAMKII AUTOPHOSPHORYLATION-<br />

DEFICIENT MICE<br />

ECIP<br />

A. Easton*(1), W. Lucchesi(2), G. Schumann(1), C. Fernandes(3),<br />

K. Giese(2), C. Muller(1,4)<br />

1. Kings College London, MRC Social, Genetic and Developmental<br />

Psychiatry Centre 2. Kings College London, JBC MRC Centre for<br />

Neurodegeneration Research 3. Kings College London, Dept <strong>of</strong><br />

Psychosis Studies 4. Univ <strong>of</strong> Erlangen, Dept <strong>of</strong> Psychiatry and<br />

Psychotherapy<br />

*alanna.c.easton@kcl.ac.uk<br />

Introduction: Cocaine dependence has been classified by DSM-IV<br />

as a psychiatric disorder. Cocaine is the most widely used illicit<br />

psycho-stimulant drug in the UK with an estimated 1,000,000 users,<br />

and a prevalence rate <strong>of</strong> lifetime use between 3 % and 5% (UNODC<br />

2009). Despite this, the underlying processes by which cocaine<br />

dependence is established are not known but believed to involve<br />

molecular mechanisms <strong>of</strong> learning and memory. Alpha<br />

calcium/calmodulin dependent protein kinase II (αCaMKII) is a<br />

major synaptic kinase that is critical for memory formation. Upon<br />

autophosphorylation, αCaMKII switches to an autonomous state <strong>of</strong><br />

activity after calcium levels drop, preserving kinase activity. It has<br />

been reported that αCaMKII autophosphorylation-deficient mice (Mt)<br />

show severe one-trial learning impairments compared to<br />

heterozygous (Ht) and wild type (WT) counterparts. However, the<br />

requirement for the aCaMKII autophosphorylation can be overcome<br />

by repeated training. Accordingly, we hypothesised that the<br />

behavioural and motivational properties associated with drugs <strong>of</strong><br />

addiction will initially be attenuated in the Mt mice, diminishing after<br />

repeated treatment.<br />

Methodology: We tested cocaine’s effects on the learning curve<br />

established in a conditioned place preference (CPP) paradigm.<br />

Results: Mt mice were impaired in learning cocaine CPP, but after a<br />

7 day period with no treatment or conditioning (‘incubation’), Mt<br />

mice had the same level <strong>of</strong> preference as Ht and WT mice. Acute<br />

locomotor responses to cocaine and saline were identical in all<br />

groups. We can therefore rule out to a great extent metabolism<br />

differences in these mice. There was a significant conditioned<br />

hyperlocomotion effect seen in all genotypes with cocaine, but not<br />

saline. This suggests that the aCaMKII autophosphorylation is not<br />

required for this type <strong>of</strong> learning. Cocaine, produced considerable<br />

behavioural stimulating effects in all genotypes, but this sensitised<br />

over 7 treatments. Thus, this sensitisation is another type <strong>of</strong> learning<br />

that does not require aCaMKII autophosphorylation. Taken together,<br />

this study supports the role <strong>of</strong> learning processes in the development<br />

<strong>of</strong> addictive behaviours.<br />

Conclusions: Data suggest an important, specific role for αCaMKII<br />

autophosphorylation in the establishment <strong>of</strong> addiction related<br />

behavioural responses.<br />

43<br />

OPS1.4 LENTIVIRAL MEDIATED-MEDIATED GENE<br />

DELIVERY REVEALS DISTINCT ROLES OF NUCLEUS<br />

ACCUMBENS DOPAMINE D2 AND D3 RECEPTORS IN<br />

NOVELTY- AND LIGHT-INDUCED LOCOMOTOR<br />

ACTIVITY<br />

ECIP<br />

A. Fernandes*(1), A. Easton(1), M. De Souza Silva(2), G.<br />

Schumann(1), C. Müller(1,3), S. Desrivières(1)<br />

1. 1MRC-SGDP-Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London 2. Centre for Behavioural Neuroscience and Centre for<br />

Biological and Medical Research, University <strong>of</strong> Düsseldorf,<br />

Universitätsstr. 1, 40225 3. Section <strong>of</strong> Addiction Medicine,<br />

<strong>Psychiatric</strong> University Hospital, Friedrich-Alexander-University<br />

Erlangen-Nuremberg, Schwabachanlage 6,<br />

*alinda.fernandes@kcl.ac.uk<br />

Introduction: The importance <strong>of</strong> dopamine in brain function is<br />

shown by many functional studies investigating effects <strong>of</strong> alteration<br />

<strong>of</strong> the dopaminergic system leads to changes in cognitive and motor<br />

functions. Although the roles <strong>of</strong> specific dopamine receptors in<br />

behaviour have been extensively investigated using pharmacological<br />

agents and knockout mice, non-specificity <strong>of</strong> ligands and<br />

compensatory molecular adaptations in mutated animals restrict the<br />

interpretation <strong>of</strong> the results.<br />

Methodology: To explore the role <strong>of</strong> the dopamine D2 and D3<br />

receptors (D2R and D3R) in rats, we used lentivirus-mediated gene<br />

knockdown and overexpression to specifically manipulate expression<br />

levels <strong>of</strong> these genes in the rat nucleus accumbens (NAcc), a brain<br />

area important for motor response and motivation. Lentiviruses,<br />

inducing expression <strong>of</strong> rat D2R or D3R, or efficient knockdown <strong>of</strong><br />

either receptor by small hairpin (sh)RNAs knockdown were<br />

stereotaxically injected into the NAcc. Novelty- and light-induced<br />

locomotor activities as well as anxiety-related behaviours were<br />

measured.<br />

Results: In the present study we aimed at investigating the specific<br />

contributions <strong>of</strong> D2R and D3R, expressed in the NAcc, in novelty-<br />

and light-induced locomotor activity. Using lentiviral-mediated overexpression<br />

and gene knockdown to specifically and locally overexpress<br />

or repress these genes. We found that NAcc D2R and D3R<br />

share a similar function in spontaneous locomotor activity, thus<br />

regulating activity in a new environment, but not in a familiar<br />

environment. However, there was a dissociation <strong>of</strong> in the role <strong>of</strong><br />

NAcc D2R and D3R in visual stimulation-induced locomotor<br />

activity. While NAcc dopamine D2 receptors limited light-induced<br />

activity, the behaviour was facilitated by NAcc dopamine D3<br />

receptors. These findings are unlikely to reflect altered anxiety levels<br />

as no clear differences between groups were observed in wellestablished<br />

animal models <strong>of</strong> anxiety.<br />

Conclusions: In conclusion, this study demonstrates that lentivirus<br />

mediated gene delivery to a distinct region in the brain can provide<br />

insights into the brain region specific functionality <strong>of</strong> genes in<br />

behaviour. Our results suggest NAcc D2R and D3R are required for<br />

spontaneous locomotor activity in a new environment, but not in a<br />

familiar environment. There was little evidence for both receptors in<br />

anxiety-related behaviours. However, we were able to clearly<br />

distinguish opposite roles <strong>of</strong> Nac D2R and D3R in visual stimulationinduced<br />

locomotor activity.


OPS1.5 IN VIVO MICRORNA CSF PROFILING IN<br />

PATIENTS WITH SCHIZOPHRENIA<br />

ECIP<br />

J. Gallego*(1), T. Lencz(1, 2, 3, 4), M. Gordon(1, 2, 3, 4), J.<br />

Gentile(1), S. Ventura(1), C. Morell(1), N. Chitkara(1), A.<br />

Malhotra(1, 2, 3, 4)<br />

1. The Zucker Hillside Hospital 2. The Feinstein Institute for Medical<br />

Research 3. Albert Einstein College <strong>of</strong> Medicine 4. H<strong>of</strong>stra North<br />

Shore-LIJ school <strong>of</strong> medicine<br />

*jgallego@nshs.edu<br />

Introduction: MicroRNAs are small non-coding RNA molecules<br />

that are involved in post-transcriptional regulation <strong>of</strong> messenger<br />

RNAs. Recent studies have started to link alterations in miRNA<br />

expression to schizophrenia and other psychiatric disorders.<br />

However, most <strong>of</strong> these studies have examined a small number <strong>of</strong><br />

microRNAs and/or have used post-mortem brain tissue or whole<br />

blood as the source <strong>of</strong> transcript. By contrast, examination <strong>of</strong> a broad<br />

array <strong>of</strong> microRNAs in cerebrospinal fluid might provide an in vivo<br />

biomarker more directly reflecting functional changes in the brain.<br />

However, to our knowledge, no study has been conducted<br />

investigating the presence <strong>of</strong> microRNAs in CSF in schizophrenia or<br />

any other psychiatric disorder.<br />

Methodology: Four patients with chronic schizophrenia and four<br />

healthy volunteers underwent lumbar puncture and a standard blood<br />

draw. For each subject, total RNA was separately extracted from 3-<br />

5ml <strong>of</strong> CSF and one PaxGene tube <strong>of</strong> blood. Expression <strong>of</strong> 381<br />

validated microRNAs was assessed from each bi<strong>of</strong>luid type for each<br />

<strong>of</strong> the subjects with the Taqman Human MicroRNA A array (Applied<br />

Biosystems), which uses real-time RT-PCR to quantify the number <strong>of</strong><br />

amplification cycles (Ct) required to reach a given threshold.<br />

Results: A mean <strong>of</strong> 136.5 (SD=11) miRNAs were obtained from<br />

CSF in healthy volunteers (male: 100%, mean age: 38.8, black: 75%)<br />

and a mean <strong>of</strong> 180.5 (SD=11.2) miRNAs were obtained in patients<br />

with schizophrenia (male: 100%, mean age: 41, black: 75%).<br />

Approximately one-third <strong>of</strong> all CSF-expressed microRNAs<br />

demonstrated robust levels <strong>of</strong> expression (Ct


OPS2.2 GENOME WIDE STUDIES OF IMPRINTED GENES<br />

IN THE FRONTAL CORTEX: TRANSCRIPTOME AND<br />

METHYLOME ANALYSIS<br />

C. Barr*(1,2), W. Xie(3), K. Wigg(1), E. Dempster(1), C.<br />

Barbacioru(4), Y. Feng(1), L. Gomez(1), P. Monnier(1), R. Logan(1),<br />

J. Eubanks(1), B. Ren(3)<br />

1. The Toronto Western Research Institute 2. The Hospital for Sick<br />

Children 3. Ludwig Institute for Cancer Research 4. Life<br />

Technologies<br />

*cbarr@uhnres.utoronto.ca<br />

Introduction: Current data support approximately 90 validated<br />

imprinted genes in mice, but recent estimates indicate that this<br />

number may be well over 1,000. Many <strong>of</strong> the genes are imprinted in<br />

a tissue/cell type specific pattern and little is known <strong>of</strong> these<br />

genes. Previous evidence indicates that imprinted genes influence the<br />

development <strong>of</strong> specific brain regions and we sought to identify<br />

imprinted genes contributing to the development <strong>of</strong> the frontal cortex<br />

using crosses and reverse crosses <strong>of</strong> mouse strains to determine the<br />

parent-<strong>of</strong>-origin <strong>of</strong> expressed genes.<br />

Methodology: We used next generation sequencing <strong>of</strong> RNA (RNAseq),<br />

chromatin immunoprecipitation to histone modifications (ChiPseq),<br />

and bisulfite sequencing (MethylC-Seq) <strong>of</strong> DNA from the<br />

frontal cortex <strong>of</strong> the adult F1 mice to identity transcripts and<br />

epigenetic marks with parent-<strong>of</strong>-origin effects.<br />

Results: Using a FDR <strong>of</strong> .01, and requiring that the same parent-<strong>of</strong>origin<br />

bias was evident in 3 biological replicates, we identified<br />

known imprinted genes as well as novel patterns <strong>of</strong> expression for<br />

known imprinted genes. We also identified 109 novel transcripts<br />

with parent-<strong>of</strong>-origin effects. Using Methyl-seq we correctly<br />

identified 27 <strong>of</strong> 29 known differentially methylated regions (DMRs)<br />

associated with known imprinted genes as well as novel DMRs, most<br />

within known imprinted regions. However none <strong>of</strong> the novel<br />

imprinted transcripts that we identified using RNA-seq or were<br />

identified in a recent study, were marked by a DMR or by chromatin<br />

patterns that were indicative <strong>of</strong> imprinting.<br />

Conclusions: The finding <strong>of</strong> novel imprinted genes indicates that the<br />

number <strong>of</strong> imprinted genes with tissue specific effects is<br />

underestimated and requires further studies to identify the full<br />

complement <strong>of</strong> imprinted genes in brain. However the failure to find<br />

epigenetic patterns supporting these genes indicates either that these<br />

are false positive findings or an unknown mechanism confers the<br />

imprint. The understanding <strong>of</strong> parent-<strong>of</strong>-origin effects in specific<br />

brain regions will make a significant contribution to our<br />

understanding <strong>of</strong> the role <strong>of</strong> imprinting in brain function and behavior<br />

and the use <strong>of</strong> high throughput technologies now allows for the<br />

comprehensive study <strong>of</strong> mechanisms regulating imprinting.<br />

45<br />

OPS2.3 ANTIDEPRESSANT TREATMENT IS ASSOCIATED<br />

WITH EPIGENETIC ALTERATIONS IN THE PROMOTER<br />

OF P11 IN A GENETIC MODEL OF DEPRESSION<br />

ECIP<br />

P. Melas*(1), M. Rogdaki(1), A. Lennartsson(1), K. Björk(1), H.<br />

Qi(1), A. Witasp(1), M. Werme(1), G. Wegener(2), A. Mathé(1), P.<br />

Svenningsson(1), C. Lavebratt(1)<br />

1. Karolinska Institute 2. Aarhus University Hospital<br />

*philippe.melas@ki.se<br />

Introduction: P11 (S100A10) has been associated with the<br />

pathophysiology <strong>of</strong> depression both in humans and rodent models.<br />

Different types <strong>of</strong> antidepressants have been shown to increase P11<br />

levels in distinct brain regions and P11 gene therapy was recently<br />

proven effective in reversing depressive-like behaviors in mice.<br />

However, the molecular mechanisms that govern P11 gene<br />

expression in response to antidepressants still remain elusive.<br />

Methodology: Prefrontal cortical regions <strong>of</strong> a genetic rodent model<br />

<strong>of</strong> depression were examined. P11 mRNA and protein levels were<br />

measured by in situ hybridization, RT-PCR and Western blotting.<br />

P11 exons and the proximal promoter region <strong>of</strong> the gene were<br />

sequenced. Pyrosequencing technology was used to quantify the<br />

DNA methylation levels at the promoter region. Global DNA<br />

methylation levels were examined using the LUMA assay.<br />

Quantitative RT-PCR was used to investigate the mRNA levels <strong>of</strong><br />

three DNA methyltransferases (Dnmt1, Dnmt3a and Dnmt3b) and<br />

four candidate DNA demethylases (Mbd2, Mbd4, Aid and Gadd45b).<br />

Results: In this study we report decreased levels <strong>of</strong> P11, associated<br />

with higher DNA methylation in the promoter region, in the<br />

prefrontal cortex <strong>of</strong> the FSL (Flinders Sensitive Line) genetic rodent<br />

model <strong>of</strong> depression. This hypermethylated pattern was reversed to<br />

normal, as indicated by the control line, after chronic administration<br />

<strong>of</strong> escitalopram (a selective serotonin reuptake inhibitor SSRI). The<br />

escitalopram-induced hypomethylation was associated with both an<br />

increase in P11 gene expression and a reduction in the mRNA levels<br />

<strong>of</strong> two DNA methyltransferases that have been shown to maintain<br />

DNA methylation in adult forebrain neurons (Dnmt1 andDnmt3a).<br />

Conclusions: In conclusion, our data further support a role for P11 in<br />

depression-like states and suggest that this gene is controlled by<br />

epigenetic mechanisms that can be affected by antidepressant<br />

treatment.


OPS2.4 THE SLC6A4 VNTR GENOTYPE DETERMINES<br />

TRANSCRIPTION FACTOR BINDING AND EPIGENETIC<br />

VARIATION OF THIS GENE IN RESPONSE TO COCAINE<br />

IN VITRO<br />

J. Quinn*, V. Bubb, K. Haddley, P. Myers<br />

University <strong>of</strong> Liverpool<br />

*jquinn@liv.ac.uk<br />

Introduction: We have previously demonstrated that the variable<br />

number tandem repeats (VNTRs) termed LPR and Stin2 within the<br />

promoter and intron 2 <strong>of</strong> the serotonin transporter (SLC6A4) gene<br />

respectively, act as differential transcriptional regulators in reporter<br />

gene constructs both in vivo and in vitro. SLC6A4 regulation is in<br />

part regulated by the transcription factor CTCF via the Stin2<br />

domain. We now explore whether CTCF can also modify the<br />

transcriptional properties <strong>of</strong> the LPR. We demonstrate that CTCF<br />

will regulate an LPR directed reporter gene in JAr cells and that<br />

CTCF will bind to the endogenous LPR region. The binding <strong>of</strong> CTCF<br />

to the LPR is sensitive to challenge and the genotype determines<br />

CTCF binding and epigenetic variation at the locus in response to<br />

cocaine in vitro. Interestingly the loss <strong>of</strong> CTCF binding at the LPR in<br />

response to cocaine is correlated with the binding <strong>of</strong> the methylated<br />

DNA binding protein MeCP2 at the LPR. No variation <strong>of</strong> CTCF<br />

binding was observed at the Stin2 VNTR in this model although we<br />

had previously noted differential binding <strong>of</strong> CTCF to this VNTR in<br />

response to Lithium.<br />

Methodology: We measure the ability <strong>of</strong> the LPR and Stin2 VNTRs<br />

to support marker gene expression in JAr cells in response to cocaine<br />

and over expression <strong>of</strong> CTCF from an expression construct.We have<br />

used antibodies against CTCF, MeCP2 and various histone marks to<br />

address binding <strong>of</strong> these factors <strong>of</strong> the endogenous SLC6A4 VNTRs<br />

and its promoter in ChIP analysis.<br />

Results: Biochemically and functionally the LPR and Stin2 VNTRs<br />

were differentially regulated by the transcription factor CTCF. ChIP<br />

analysis allowed us to demonstrate differential CTCF binding to the<br />

LPR in response to cocaine. Prior to cocaine exposure both alleles <strong>of</strong><br />

the LPR bound CTCF, however CTCF only bound to the long allele<br />

following exposure. Significantly, this differential effect <strong>of</strong> cocaine<br />

on binding <strong>of</strong> CTCF was correlated with the binding <strong>of</strong> the<br />

transcriptional regulator MeCP2 specifically to the short allele and<br />

recruiting the histone deacetylase complex (HDAC). Concurrently,<br />

cocaine increased the association <strong>of</strong> positive histone marks over the<br />

SLC6A4 gene locus.<br />

Conclusions: The data demonstrated that exposure <strong>of</strong> JAr cells to<br />

cocaine can result in differential binding <strong>of</strong> CTCF, MeCP2 and<br />

HDAC to the LPR VNTR based on its specific genotype. This was<br />

correlated with modulation <strong>of</strong> histone marks that might alter<br />

epigenetic parameters affecting gene expression after the initial<br />

challenge. CTCF and MeCP2 are well characterised major<br />

modulators <strong>of</strong> epigenetic parameters and <strong>of</strong>ten converge on similar<br />

genes to modulate imprinting parameters. This could be a<br />

mechanism by which the genotype <strong>of</strong> SLC6A4 alters the expression<br />

<strong>of</strong> the gene in the medium to long term after challenge. Related<br />

VNTR domains populate the genome and can be both conserved in<br />

evolution and rapidly evolving. Extrapolation <strong>of</strong> our data on the<br />

SLC6A4 VNTRs suggests that such polymorphic VNTRs are<br />

potentially a major modulator <strong>of</strong> gene expression and therefore<br />

targets to mediate differential gene expression associated with<br />

psychiatric disorders.<br />

46<br />

OPS2.5 NEXT-GENERATION SEQUENCING IDENTIFIES<br />

MIRNAS ASSOCIATED WITH MOOD DISORDERS<br />

M. Carless*(1), J. Neary(1), M. Zlojutro(1), T. Dyer(1), J. Curran(1),<br />

L. Almasy(1), R. Duggirala(1), D. Glahn(2,3), J. Blangero(1)<br />

1. Texas Biomedical Research Institute, Department <strong>of</strong> <strong>Genetics</strong> 2.<br />

Yale University, Department <strong>of</strong> Psychiatry 3. Olin <strong>Psychiatric</strong><br />

Research Center, Institute <strong>of</strong> Living<br />

*mcarless@sfbrgenetics.org<br />

Introduction: miRNAs are potent regulators <strong>of</strong> gene expression that<br />

have recently been shown to be targets for the regulation <strong>of</strong> mood<br />

stabilizers. Although several miRNAs are suggested targets <strong>of</strong> mood<br />

stabilizers, their expression in individuals suffering from mood<br />

disorders and their biological role in related neuroanatomical and<br />

neurocognitive phenotypes are not well studied.<br />

Methodology: We have collected samples from approximately 1,300<br />

individuals within ~40 families that are part <strong>of</strong> the <strong>Genetics</strong> <strong>of</strong> Brain<br />

Structure and Function (GOBSF) study for which we have extensive<br />

neuroanatomical and neurocognitive data as well as measures <strong>of</strong><br />

depression, anxiety and substance abuse. In addition, we have<br />

collected samples from ~100 individuals (siblings) that are part <strong>of</strong> the<br />

<strong>Genetics</strong> <strong>of</strong> Bipolar Disorder (GOBD) study for which we have<br />

identical neuroanatomical and neurocognitive measures, as well as<br />

additional neuropsychological data. Using the newly-released TruSeq<br />

platform by Illumina, we have used multiplex-based sequencing<br />

analysis to identify miRNAs associated with depression in a subset<br />

(n=60) <strong>of</strong> the GOBSF population.<br />

Results: Several miRNAs were found to be associated with major<br />

depressive disorder (MDD), recurrent MDD (rMDD) and the Beck<br />

Depression Inventory (BDI) at a nominal p-value <strong>of</strong> 0.05. hsa-let-7e<br />

(p=0.045; p=0.024; p=0.029), hsa-miR-29c* (p=0.0.025; p=0.024;<br />

p=0.036) and hsa-miR-378* (p=0.006; p=0.021; p=0.031) were<br />

associated with all three measures <strong>of</strong> depression (MDD, rMDD and<br />

BDI, respectively). In addition, we identified associations with<br />

several miRNAs for which there is evidence <strong>of</strong> decreased expression<br />

in bipolar disorder (MDD: hsa-miR-22, p=0.021/BDI: hsa-miR-15a,<br />

p=0.036; hsa-miR-190, p=0.006; hsa-miR-27b, p=0.024) or which<br />

are suggested targets <strong>of</strong> mood stabilizers (rMDD: hsa-miR-30c,<br />

p=0.010; hsa-let-7b, p=0.015/BDI: hsa-miR-155, p=0.006; hsa-miR-<br />

34a, p=0.038; hsa-let-7b, p=0.026). Although no associations were<br />

seen that met correction for multiple testing, we believe that the<br />

consistency seen across multiple measures <strong>of</strong> depression, as well as<br />

prior evidence for a role in mood disorders, suggests that the<br />

miRNAs identified in this preliminary analysis may indeed play a<br />

role in depression and that analysis with our larger cohort will yield<br />

more powerful results.<br />

Conclusions: We are continuing to analyze sequencing data in the<br />

remaining GOBSF samples, as well as in our GOBD cohort to<br />

identify stronger associations <strong>of</strong> miRNAs with depression and bipolar<br />

disorder and also to gain sufficient power to detect associations with<br />

neuroanatomical and neurocognitive phenotypes. In addition, we are<br />

analyzing potentially novel miRNAs, <strong>of</strong> which we have identified<br />

145 candidates to date, to determine whether they are true miRNAs<br />

and to investigate their role in mood disorders.


ORAL PRESENTATIONS SESSION 3: PHENOTYPES &<br />

ENDOPHENOTYPES<br />

OPS3.1 GENOME-WIDE ASSOCIATION IMPLICATES<br />

FGF14 IN AMYGDALA VOLUME AND FEAR PROCESSING<br />

D. Glahn*(1,2), M. Carless(3), J. Kent(3), A. Winkler(1,2), T.<br />

Dyer(3), J. Curran(3), M. Johnson(3), H. Göring(3), E. Moses(3), R.<br />

Olvera(4), P. Kochunov(4), P. Fox(4), L. Almasy(3), R.<br />

Duggirala(3), J. Blangero(3)<br />

1. Olin Neuropsychiatric Research Center, Institute <strong>of</strong> Living 2. Yale<br />

University 3. Texas Biomedical Research Institute 4. UT Health<br />

Science Center, San Antonio<br />

*david.glahn@yale.edu<br />

Introduction: The amygdala has a preferential role in processing<br />

emotional stimuli and fear conditioning. Amygdala dysfunction has<br />

been documented in a number <strong>of</strong> mental illnesses, including mood<br />

disorders, schizophrenia, anxiety disorders, post-traumatic stress<br />

disorder and addiction. Determining the genetic factors that influence<br />

amygdala function and structure should provide empirically defined<br />

candidate genes for these mental illnesses.<br />

Methodology: To that end, we genotyped more than one million<br />

genetic variants throughout the genome <strong>of</strong> 605 Mexican American<br />

individuals from randomly selected extended pedigrees, for which we<br />

have extensive neuroanatomical and neurocognitive data.<br />

Results: We undertook a genome-wide association analysis <strong>of</strong><br />

amygdala volume and identified a single nucleotide polymorphism<br />

(SNP), rs1336722, which was significantly associated with amygdala<br />

volume (p=6.7x10 -8 ). Located within intron 1 <strong>of</strong> the FGF14 gene, the<br />

C allele (frequency=0.44) accounts for nearly 4% <strong>of</strong> the phenotypic<br />

variation seen in amygdala volume and is associated with a decline in<br />

volume. Three other SNPs (rs1415060, rs1336709, and rs9518703)<br />

within intron 1 <strong>of</strong> FGF14, which were in linkage disequilibrium with<br />

rs1336722, also showed genome wide significance. To determine if<br />

rs1336722 influences fearful facial perception in healthy individuals,<br />

subjects were asked to choose which <strong>of</strong> five words (e.g. happy, sad,<br />

angry, fear, or neutral) best represents the emotion portrayed on a<br />

series <strong>of</strong> 40 faces. While the identification <strong>of</strong> fearful facial<br />

expressions, relative to neutral faces, was significantly influenced by<br />

rs13366722 (p=0.002), task performance in general was not<br />

influenced by this SNP (p=0.766).<br />

Conclusions: FGF14codes for a fibroblast growth factor that is<br />

expressed in the developing and adult central nervous system.<br />

Mutations within this gene have been associated with dyskinesia,<br />

cerebellar ataxia and mild mental retardation. Further, animal models<br />

have suggested a function for FGF14 in neuronal signaling, axonal<br />

trafficking and synaptosomal function and indicate that reduced<br />

FGF14 may result in decreased response to dopamine agonists. We<br />

have identified a novel polymorphism that is associated with<br />

amygdala volume and are currently re-sequencing and genotyping the<br />

FGF14 gene more comprehensively to identify potential functional<br />

variation that may contribute to amygdala volume and the<br />

development <strong>of</strong> mental illnesses, particularly mood disorders<br />

47<br />

OPS3.2 FURTHER EVIDENCE FOR GENETIC<br />

ASSOCIATION BETWEEN SCHIZOPHRENIA AND<br />

BIPOLAR FROM NEW IMMUNOCHIP DATA ANALYSIS<br />

M. Hamshere*(1), E. Green(1), D. Grozeva(1), I. Jones(1), L.<br />

Jones(2), K. Gordon-Smith (2), L. Forty(1), J. Moran(3), J. Kranz(3),<br />

S. Purcell(3,4,5), P. Sklar(3,4,5), M. Owen(1), M. O'Donovan(1), N.<br />

Craddock(1)<br />

1. MRC Centre for Neuropsychiatric <strong>Genetics</strong> and Genomics, School<br />

<strong>of</strong> Medicine, Cardiff University, Heath Park, CF14 4XN 2.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Birmingham, National<br />

Centre for Mental Health, 25 Vincent Drive, B15 2FG 3. Stanley<br />

Centre for <strong>Psychiatric</strong> Research, Broad Institute 4. Department <strong>of</strong><br />

Psychiatry, Harvard Medical School 5. <strong>Psychiatric</strong> and<br />

Neurodevelopmental <strong>Genetics</strong> Unit, Centre for Human Genetic<br />

research, Massachusetts General Hospital<br />

*hamshereml@cardiff.ac.uk<br />

Introduction: We present Illumina ImmunoChip results data for<br />

bipolar disorder. The ImmunoChip contains fine mapping data for<br />

genes from autoimmune disorders, including extensive SNP data in<br />

the MHC region. Also <strong>of</strong> particular interest to bipolar is that the<br />

genes CACNA1C (alpha 1C subunit <strong>of</strong> the L-type voltage-gated<br />

calcium channel) and ANK3 (ankyrin G) are fine mapped. In<br />

addition, interesting SNPs from previous studies were included on the<br />

chip, including the top SNPs in CACNA1C and ANK3 from Ferreira<br />

et al (2008), plus a number <strong>of</strong> key schizophrenia SNPs identified in<br />

O’Donovan et al (2008), the MGS study (2009), the ISC study (2009)<br />

and the Sgene study (2009).<br />

Methodology: After extensive quality control <strong>of</strong> the data, genotypes<br />

were available for 2,630 bipolar and 4,322 control individuals at each<br />

<strong>of</strong> 127,124 autosomal SNPs. Each SNP was analysed with the<br />

Cochran Armitage Trend test, and systematic inflation <strong>of</strong> all the trend<br />

statistics was adjusted with the genomic control lambda statistic. A<br />

subset <strong>of</strong> the sample have not been analysed previously (1,388<br />

bipolar and 1,398 controls), allowing us to investigate independent<br />

evidence <strong>of</strong> association from previous studies. Here we describe<br />

results from SNPs previously associated with schizophrenia.<br />

Results: We will discuss our experiences <strong>of</strong> analysing data on the<br />

ImmunoChip and highlight some issues relevant to analysing nongenome-wide<br />

chip data with regions <strong>of</strong> extensive linkage<br />

disequilibium. Analysis <strong>of</strong> the schizophrenia defined SNPs <strong>of</strong> interest<br />

showed greater evidence for association in bipolar disorder than<br />

expected by chance. Loci <strong>of</strong> interest include rs17594721 (TCF4<br />

p=0.0103 in the independent sample), rs9922369 (RPGRIP1L<br />

p=0.0279) and rs13199775 (SLC17A1 p=0.0359).<br />

Conclusions: Our results provide us with an interesting overview the<br />

main candidate SNPs for schizophrenia analysed in our independent<br />

bipolar disorder sample (i.e. not previously included in any meta<br />

analyses). This adds to the evidence for genetic overlap between<br />

schizophrenia and bipolar disorder.


OPS3.3 NEUREXIN-1 AND SCHIZOPHRENIA<br />

SUBPHENOTYPES: BRAIN MORPHOLOGY AND<br />

CLOZAPINE RESPONSE<br />

ECIP<br />

T. Lett*(1), A. Voineskos(1), A. Tiwari(1), J. Lerch(2), S. Ameis(2),<br />

T. Rajji(1), B. Mulsant(1), H. Meltzer(3), J. Lieberman(4), S.<br />

Potkin(5), D. Müller(1), J. Kennedy(1)<br />

1. Centre for Addiction and Mental Health 2. Hospital for Sick<br />

Children 3. Vanderbilt University 4. Columbia University 5.<br />

University <strong>of</strong> California<br />

*tristram_lett<br />

Introduction: CNV structural variations in the neurexin-1 (NRXN1)<br />

gene increase the risk for both autism spectrum disorders (ASD) and<br />

schizophrenia. However, the effect in which NRXN1 gene variations<br />

may be related to the important subphenotypes <strong>of</strong> brain morphology<br />

and clozapine response has had little attention. Furthermore,<br />

schizophrenia is hypothesized to be at least in part, a disorder related<br />

to N-methyl D-aspartate (NMDA) receptor dysfunction.<br />

NRXN1modulates recruitment <strong>of</strong> NMDA receptors to the presynaptic<br />

membrane. Clozapine normalizes cortical hyperactivity <strong>of</strong><br />

NMDA receptors; thus, regulation <strong>of</strong> NRXN1 may mediate<br />

clozapine’s efficacy.<br />

Methodology: 53 healthy individuals between 18-59 years <strong>of</strong> age<br />

received structural MRI scans, which were processed to determine<br />

cortical gray and white matter lobar volumes, and volumes <strong>of</strong> striatal<br />

and thalamic structures. Each subject’s sensorimotor function was<br />

also assessed. All were genotyped at 11 single nucleotide<br />

polymorphisms <strong>of</strong> the NRXN1 gene. The general linear model was<br />

used to calculate the influence <strong>of</strong> NRXN1 variation on neural and<br />

cognitive phenotypes. Next, in silico analyses were conducted to<br />

assess potential functional relevance <strong>of</strong> any polymorphisms<br />

associated with brain measures. Finally, in an independent sample,<br />

rs1045881 was genotyped in 169 European-American schizophrenia<br />

patients prospectively assessed for clozapine response. Treatment<br />

response was defined as a 20% reduction in Brief <strong>Psychiatric</strong> Rating<br />

Scale (BPRS) from the time <strong>of</strong> enrolment into the study (baseline) to<br />

after 6 months <strong>of</strong> clozapine treatment.<br />

Results: A polymorphism located in the 3’ untranslated region <strong>of</strong><br />

NRXN1 significantly influenced white matter volumes in whole brain<br />

and frontal lobes after correcting for total brain volume, age and<br />

multiple comparisons (F2.25=5.498, p = 0.004; F1,49=8.231, p=0.006,<br />

respectively). Follow-up in silico analyses revealed that rs1045881 is<br />

a putative microRNA binding site that may regulate NRXN1<br />

expression. This variant also influenced sensorimotor performance, a<br />

neurocognitive function impaired in schizophrenia patients. We<br />

observed an association with clozapine response (20% reduction in<br />

BPRS scores; genotypic:p = 0.03, OR= 2.2 [1.08-4.30]). Post hoc<br />

analysis <strong>of</strong> quantitative assessment <strong>of</strong> positive and negative<br />

symptoms scores revealed a significant association between BPRS<br />

negative symptoms (but not positive symptoms) and genotype (F1,85<br />

between-subject = 4.69, p = 0.033)<br />

Conclusions: To the best <strong>of</strong> our knowledge, this is the first imaginggenetics<br />

study <strong>of</strong> NRXN1, and the first pharmacogenetic study <strong>of</strong> the<br />

putative functional rs1045881 polymorphism. Our findings<br />

demonstrate that the NRXN1 gene, a vulnerability gene for SCZ and<br />

ASD from both candidate gene studies and genome-wide CNV scans,<br />

influences brain structure and cognitive function. In conjunction with<br />

our in silico results, our findings provide evidence for<br />

neurobiological and cognitive susceptibility mechanisms by which<br />

the NRXN1 gene confers risk factors potentially for schizophrenia.<br />

Our pharmacogenetic findings suggest that the rs1045881 NRXN1<br />

polymorphism may influence clozapine response. NMDA receptor<br />

function may be a common factor for schizophrenia, and clozapine<br />

response.<br />

48<br />

OPS3.4 NRGN GENE, A GENOME-WIDE SUPPORTED<br />

SCHIZOPHRENIA RISK VARIANT, MODULATES<br />

PREFRONTAL CORTICAL INFORMATION PROCESSING<br />

DURING WORKING MEMORY<br />

ECIP<br />

A. Rauch*, R. Rasetti, J. Callicott, F. Zhang, B. Kolachana, Q. Chen,<br />

J. Apud, V. Mattay, D. Weinberger<br />

Genes, Cognition and Psychosis Program, Clinical Brain Disorders<br />

Branch, National Institute <strong>of</strong> Mental Health, National Institutes <strong>of</strong><br />

Health<br />

*rauchav@mail.nih.gov<br />

Introduction: A single nucleotide polymorphism (SNP)-based GWA<br />

study by Stefansson et al. (Nature, 2009) found a significant<br />

association <strong>of</strong> schizophrenia with a SNP (rs12807809) located<br />

upstream <strong>of</strong> the neurogranin gene (NRGN) on 11q24.2. The gene<br />

product <strong>of</strong> NRGN is a postsynaptic protein kinase substrate that binds<br />

calmodulin in the absence <strong>of</strong> calcium. NRGN may play a role as<br />

CaM reservoir and it is important for calmodulin's activation <strong>of</strong><br />

CaMKinase II which is part <strong>of</strong> NMDA receptor signaling. Animal<br />

studies showed that this gene product is involved in cognitive<br />

functions including memory processing. To date there have been no<br />

studies exploring the influence <strong>of</strong> the NRGN gene on memory related<br />

cortical information processing in humans. In the current study, we<br />

explored the effect <strong>of</strong> rs12807809 genotype on neural circuits<br />

underlying working memory (WM).<br />

Methodology: 219 healthy Caucasian subjects without any history <strong>of</strong><br />

psychiatric illness were studied with BOLD functional Magnetic<br />

Resonance Imaging (fMRI) while they performed a 2-back WM task.<br />

DNA isolation and analysiswere conducted on blood samples<br />

obtained from all subjects whogave informed consent according to<br />

NIMH Institutional ReviewBoard guidelines. Neuroimaging data<br />

were acquired on a GE Signa 3-T scanner (GE Systems, Milwaukee,<br />

Wisconsin) and were analyzed using SPM5. Given the low<br />

frequency <strong>of</strong> homozygous C allele carriers (6 subjects), they were<br />

combined with the heterozygotes (CT + CC, 70 subjects) and<br />

compared with subjects homozygous for the high frequency riskassociated<br />

T allele (TT, 149 subjects). Effect <strong>of</strong> NRGN genotype (TT<br />

vs C carriers) on brain activity during WM was assessed using a twosample<br />

t-test.<br />

Results: Age, gender, education, handedness, IQ, and task-related<br />

variables (2- Back accuracy and reaction time) were not significantly<br />

different between TT homozygotes and C carriers. There was<br />

significantly increased activation in the Dorsolateral Prefrontal<br />

Cortex (DLPFC) (BA9, xyz = 57 18 33, Z = 4.22, p = 0.008 and xyz<br />

= -48 3 36, Z = 4.03, p = 0.019, both FWE-corrected within DLPFC<br />

Region <strong>of</strong> interest) <strong>of</strong> the risk allele T homozygotes during 2Back<br />

WM in comparison to the non-risk allele C carriers (CT + CC).<br />

Conclusions: The relatively increased DLPFC activation observed in<br />

this study in the risk allele T homozygote healthy individuals<br />

implicates a phenomenon characterized as inefficiency in cortical<br />

information processing and shown to be a neuroimaging intermediate<br />

phenotype for genetic studies <strong>of</strong> schizophrenia. Our results suggest<br />

that NRGN, like a number <strong>of</strong> other genes implicated in<br />

schizophrenia, may be associated with clinical risk because <strong>of</strong><br />

modulating the efficiency <strong>of</strong> cortical memory circuitry.


ORAL PRESENTATIONS SESSION 4:<br />

PHARMACOGENOMICS<br />

OPS4.1 PHARMACOGENETICS OF ANTIDEPRESSANT<br />

TREATMENT OUTCOME IN DEPRESSION - ROLE OF HPA<br />

AXIS AND ADVERSE LIFE EVENTS<br />

M. Ising*, S. Lucae, E. Binder, F. Holsboer, B. Mueller-Myhsok<br />

Max Planck Institute <strong>of</strong> Psychiatry<br />

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

Introduction: While candidate gene studies suggested a strong<br />

genetic component <strong>of</strong> treatment outcome in major depression, recent<br />

genome-wide association studies (GWA) failed to identify consistent<br />

effects. It is likely that genetic variations contribute to treatment<br />

outcome in a concerted mode, which explains the low effect size if<br />

only a single variant at a time is considered. Furthermore, genetic<br />

factors explain only parts <strong>of</strong> the phenotype suggesting a role <strong>of</strong> other<br />

factors including biomarkers and environmental influences.<br />

Methodology: We conducted a GWA in patients from the Munich-<br />

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

replication sample. A set <strong>of</strong> 328 single nucleotide polymorphisms<br />

(SNPs) related to outcome in both GWA was genotyped in the<br />

Sequenced-Treatment-Alternatives-to-Relieve-Depression<br />

(STAR*D) study. We generated a multi-locus genetic variable<br />

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

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

antidepressant treatment outcome. Then, we further tested the<br />

moderating effects <strong>of</strong> adverse life events and <strong>of</strong> HPA axis measures<br />

as biomarkers for treatment outcome.<br />

Results: The multi-locus analysis revealed a significant contribution<br />

<strong>of</strong> a binary variable categorizing patients as carriers <strong>of</strong> a high vs. low<br />

number <strong>of</strong> response alleles in predicting antidepressant treatment<br />

outcome in MARS and STAR*D. In addition, we observed that the<br />

degree <strong>of</strong> normalization <strong>of</strong> a disturbed HPA axis regulation and the<br />

experienced adversity <strong>of</strong> past life events further contributes to<br />

treatment outcome depending on the individual risk genotype.<br />

Conclusions: Our results demonstrate the importance <strong>of</strong> multiple<br />

genetic factors in combination with HPA axis regulation and<br />

environmental exposure to predict antidepressant treatment outcome<br />

underscoring the multifactorial nature <strong>of</strong> this trait.<br />

49<br />

OPS4.2 GENOMICS OF SSRI TREATMENT OUTCOMES<br />

FOR MAJOR DEPRESSIVE DISORDER: GENOME-WIDE<br />

ASSOCIATIONS AND FUNCTIONAL GENOMICS<br />

ECIP<br />

Y. Ji*(1), J. Biernacka(1), S. Hebbring(1), Y. Chai(1), G. Jenkins(1),<br />

K. Snyder(1), M. Drews(1), Z. Desta(2), D. Flockhart(2), T.<br />

Mushiroda(3), M. Kubo(3), Y. Nakamura(3), N. Kamatani(3), D.<br />

Schaid(1), R. Weinshilboum(1), D. Mrazek(1)<br />

1. Mayo Clinic 2. Indiana University 3. RIKEN Center for Genomic<br />

Medicine<br />

*ji.yuan@mayo.edu<br />

Introduction: Functional genomics in pursuit <strong>of</strong> susceptibility loci<br />

for psychiatric disorders and response to therapy represents a<br />

complementary strategy to genetic association studies, which is<br />

especially important when genomic replication studies are<br />

challenging.<br />

Methodology: We performed a genome-wide association study<br />

(GWAS) for citalopram/escitalopram treatment outcomes (remission<br />

and response) in major depressive disorder (MDD) using DNA from<br />

529 patients recruited to an SSRI clinical trial. After GWA analyses<br />

<strong>of</strong> primary treatment outcomes, we functionally characterized 14<br />

SNPs selected from SNPs with the lowest p-values (1.04 × 10 -6 to<br />

8.33 × 10 -5 ) that were also near or within a gene. Two different<br />

experimental methods that assess the possible effect <strong>of</strong> a SNP on<br />

transcription, electrophoretic mobility shift (EMS) and reporter gene<br />

assays, were performed.<br />

Results: Functional genomic experiments showed that rs11144870<br />

(OR = 0.423, p = 1.04×10 -6 ) in intron 2 <strong>of</strong> the rib<strong>of</strong>lavin (vitaminB2)<br />

kinase (RFK) gene on chromosome 9 and rs915120 (OR = 0.497, p =<br />

1.15×10 -5 ) in intron 7 <strong>of</strong> the G protein-coupled receptor kinase<br />

(GRK5) gene on chromosome 10 appeared to be functionally<br />

significant based both on their ability to drive transcription and bind<br />

nuclear proteins. Increased transcription <strong>of</strong> RFK by the variant allele<br />

<strong>of</strong> rs11144870 may lead to reduced level <strong>of</strong> circulating vitamin B2, a<br />

phenomenon that has been previously linked to depressive symptoms<br />

in adults. Other potential candidate loci included a series <strong>of</strong> intronic<br />

SNPs in the glucosaminyl (N-acetyl) transferase 1(GCNT1) gene,<br />

and intergenic region close to the D-amino acid oxidase activator<br />

(DAOA) gene and the 5-hydroxytryptamine (serotonin) receptor 1B<br />

(HTR1B) gene.<br />

Conclusions: Our study represents an attempt to join functional<br />

genomics with GWAS data for SSRI treatment outcomes in MDD, a<br />

step that might help to provide biological plausibility for observed<br />

associations and to gain novel insights into underlying mechanisms.


OPS4.3 HTR2A VARIATION PREDICTS TREATMENT<br />

RESPONSE TO VENLAFAXINE XR IN GENERALIZED<br />

ANXIETY DISORDER<br />

F. Loh<strong>of</strong>f*, T. Richardson, S. Narasimhan, P. Multani, B. Etemad, K.<br />

Rickels<br />

UPenn<br />

*loh<strong>of</strong>f@mail.med.upenn.edu<br />

Introduction: Generalized Anxiety Disorder (GAD) is a chronic<br />

psychiatric disorder with significant morbidity and mortality.<br />

Antidepressants are the preferred choice for treatment; however,<br />

treatment response is <strong>of</strong>ten variable. Several studies in major<br />

depression have implicated a role <strong>of</strong> the serotonin receptor gene<br />

(HTR2A) in treatment response to antidepressants. We tested the<br />

hypothesis that the genetic polymorphism rs7997012 in the HTR2A<br />

gene predicts treatment outcome in GAD patients treated with<br />

venlafaxine XR.<br />

Methodology: Treatment response was assessed in 156 patients that<br />

participated in a 6-month open label clinical trial <strong>of</strong> venlafaxine XR<br />

for GAD. Primary analysis included HAM-A reduction at 6 months.<br />

Secondary outcome measure was the CGI-I score at 6 months.<br />

Genotype and allele frequencies were compared between groups<br />

using chi-square contingency analysis.<br />

Results: The frequency <strong>of</strong> the G-allele differed significantly between<br />

responders (70%) and non-responders (56%) at 6 months (p=0.05)<br />

using the HAM-A scale as outcome measure. Similarly, using the<br />

CGI-I as outcome, the G-allele was significantly associated with<br />

improvement (p=0.01). Assuming a dominant effect <strong>of</strong> the G-allele,<br />

improvement differed significantly between groups (p=0.001,<br />

OR=4.72). Similar trends were observed for remission although not<br />

statistically significant.<br />

Conclusions: We show for the first time a pharmacogenetic effect <strong>of</strong><br />

the HTR2A rs7997012 variant in anxiety disorders, suggesting that<br />

pharmacogenetic effects cross diagnostic categories. Our data<br />

document that individuals with the HTR2Ars7997012 SNP G-allele<br />

have better treatment outcome over time. Future studies with larger<br />

sample sizes are necessary to further characterize this effect in<br />

treatment response to antidepressants in GAD.<br />

50<br />

OPS4.4 A GENE EXPRESSION AND PROTEOMIC STUDY<br />

PROVIDE FURTHER EVIDENCE FOR THE INVOLVEMENT<br />

OF SPERMIDINE/SPERMINE N(1)-ACETYLTRANSFERASE<br />

(SSAT1) IN SUICIDE AND SUGGESTS NEW BIOLOGICAL<br />

DETERMINANTS OF SUICIDAL BEHAVIOUR IN BIPOLAR<br />

DISORDER PATIENTS<br />

ECIP<br />

A. Squassina*(1), M. Manchia(1,2), D. Congiu(1), M. Costa(1), G.<br />

Severino(1), C. Chillotti(3), R. Ardau(3), A. Soggiu(4), C. Piras(5),<br />

P. Roncada(6), P. Keddy(7), G. Robertson(7), M. Alda(2), M. Del<br />

Zompo(1,3)<br />

1. Laboratory <strong>of</strong> Molecular <strong>Genetics</strong>, Section <strong>of</strong> Clinical<br />

Pharmacology, Department <strong>of</strong> Neurosciences “B.B. Brodie”,<br />

University <strong>of</strong> Cagliari 2. Department <strong>of</strong> Psychiatry, Dalhousie<br />

University 3. Unit <strong>of</strong> Clinical Pharmacology <strong>of</strong> the University<br />

Hospital <strong>of</strong> Cagliari 4. Section <strong>of</strong> Neurophysiology and<br />

Neurochemistry, Department <strong>of</strong> Neurosciences “B.B. Brodie”,<br />

University <strong>of</strong> Cagliari 5. Department <strong>of</strong> Zootechnical Science,<br />

University <strong>of</strong> Sassari 6. Istituto Sperimentale L. Spallanzani 7.<br />

Laboratory <strong>of</strong> Molecular Neurobiology, Department <strong>of</strong><br />

Pharmacology, Sir Charles Tupper Medical Building, Dalhousie<br />

University<br />

*squassina@unica.it<br />

Introduction: Suicide is strongly associated with Major Affective<br />

Disorders with a risk for completed suicide 6 to 15 times higher in<br />

patients compared to the general population. Lithium is to date the<br />

most effective treatment for bipolar disorder (BD) with a well<br />

established protective effect against suicide. However, the<br />

mechanisms responsible for these clinical effects remain unclear. So<br />

far, several studies attempted to identify biological markers and genes<br />

involved in suicide. A gene expression microarray study performed<br />

using postmortem brain tissue by Sequeira et al. (2006) reported a<br />

significant reduction in expression <strong>of</strong> the gene encoding the enzyme<br />

spermidine/spermine N1-acetyltransferase 1 (SSAT1) in suicide<br />

completers with and without depression compared to controls. This<br />

result has been replicated by others.<br />

Methodology: To confirm and extend these findings, we measured<br />

SSAT1 gene expression levels in lymphoblastoid cell lines (LCLs)<br />

from two populations (Sardinian, n = 41 and Canadian, n = 24) <strong>of</strong> BD<br />

subjects characterized for suicidal behaviour and healthy controls. In<br />

order to test the effect <strong>of</strong> lithium on SSAT1, each LCL was divided<br />

into two lines cultured for 7 days in media with and without LiCl (1.0<br />

mmol/l).<br />

Results: SSAT1 expression was significantly increased by lithium in<br />

LCLs from controls (p< 0.001) and in subjects with low (p < 0.001)<br />

and high (p < 0.001) personal and genetic risk <strong>of</strong> suicide but not in<br />

LCLs from suicide completers (p > 0.05). These findings were<br />

replicated in the Canadian sample and the analysis on the pooled<br />

dataset increased the power and the significance <strong>of</strong> our findings. In<br />

contrast with the gene expression findings the protein study showed<br />

that SSAT1 levels were significantly decreased when comparing each<br />

<strong>of</strong> the 3 groups at different risk <strong>of</strong> suicide with controls. This could<br />

suggest a compensatory effect in the pathway <strong>of</strong> polyamine system.<br />

These differences were reduced or disappeared by lithium treatment<br />

in vitro. On the other hand, no differences were shown when<br />

comparing groups with different risk <strong>of</strong> suicide either with or without<br />

lithium treatment in vitro. The proteome analysis also highlighted<br />

several proteins differentially expressed in at least one <strong>of</strong> the<br />

comparisons or according to lithium treatment. These proteins are<br />

currently being identified and results will be presented at the<br />

conference.<br />

Conclusions: In conclusion, our findings suggest that suicide<br />

completers might represent a distinct group <strong>of</strong> patients with an altered<br />

SSAT1 function.


OPS4.5 BERBERINE AND EVODIAMINE INFLUENCE<br />

SEROTONIN TRANSPORTER (5-HTT) EXPRESSION VIA<br />

THE 5-HTT-LINKED POLYMORPHIC REGION<br />

ECIP<br />

Y. Hu*(1), E. Ehli(1,2), J. Hudziak(3), G. Davies(1,2)<br />

1. Avera Institute for Human Behavioral <strong>Genetics</strong>, Avera Behavioral<br />

Health Center 2. Department <strong>of</strong> Psychiatry, Sanford School <strong>of</strong><br />

Medicine, University <strong>of</strong> South Dakota, 3. Department <strong>of</strong> Psychiatry,<br />

College <strong>of</strong> Medicine, University <strong>of</strong> Vermont<br />

*yueshan.hu@avera.org<br />

Introduction: The serotonin transporter (5-HTT) regulates the<br />

serotonin transmission process via the modulation <strong>of</strong> extracellular<br />

fluid serotonin concentrations, which in turn regulates mood,<br />

emotion, and appetite. Studies including ours have demonstrated that<br />

the natural compounds berberine and evodiamine reduce food intake.<br />

Berberine also has antidepressant-like effects with the possible mode<br />

<strong>of</strong> action attributed to the regulation <strong>of</strong> serotonergic<br />

neurotransmission. Until recently the serotonin transporter genelinked<br />

polymorphic region (5-HTTLPR) which controls the<br />

transcriptional activity <strong>of</strong> 5-HTT, was reported to contain three<br />

alleles (S, LG, LA) and had been demonstrated to be associated with<br />

varying therapeutic responses to antidepressant treatment.<br />

Interestingly, our group has recently discovered and functionally<br />

characterized three novel 5-HTTLPR alleles: XS11, XL17, XL18.In this<br />

present study, the effect <strong>of</strong> berberine and/or evodiamine on 5-HTT<br />

expression and its promoter activity in a serotonergic neuronal cell<br />

line RN46A was investigated. The aim <strong>of</strong> the study was not only to<br />

elucidate the potential mode <strong>of</strong> action <strong>of</strong> berberine and evodiamine<br />

on the serotonergic system but also provide an example <strong>of</strong> the role <strong>of</strong><br />

pharmacogenetics in herbal medicine.<br />

Methodology: Real-time RT-PCR was used to measure the mRNA<br />

expression <strong>of</strong> 5-HTT in RN46A neuronal cells. Protein expression <strong>of</strong><br />

5-HTT was determined by western blotting. Promoter activity <strong>of</strong> 5-<br />

HTT was analyzed by dual-luciferase reporter (DLR) assay with<br />

plasmid constructs containing the six 5-HTTLPR alleles (S, XS11, LG,<br />

LA, XL17, and XL18 alleles).<br />

Results: The results showed that 100 µM berberine increased 5-HTT<br />

expression by 2.9 fold and treatment with 2 µM and 4 µM<br />

evodiamine increased 5-HTT expression by 0.7 fold and 1.5 fold<br />

respectively. Analysis <strong>of</strong> 5-HTT protein expression revealed that<br />

treatment with either 100 µM berberine or 2 µM evodiamine, alone or<br />

in combination increased 5-HTT protein expression by 0.6 fold, 0.9<br />

fold and 1.2 fold respectively. The treatment <strong>of</strong> the individual alleles,<br />

(S, XS11, LG, LA, XL17, and XL18 ), with 100 µM berberine increased<br />

5-HTT promoter activity by 67%, 128.7%, 106.9%, 100.4%, 26.2%,<br />

and 82%, and treatment with 2 µM evodiamine increased 5-HTT<br />

promoter activity by 216.7%, 81.6%, 305.6%, 181.5%, 175.3%, and<br />

102.2% respectively. Furthermore, treatment <strong>of</strong> S allele, with 100 µM<br />

berberine combined with 2 µM evodiamine increased promoter<br />

activity (315.3%) more than treatment <strong>of</strong> 100 µM berberine (62.8%),<br />

or 2 µM evodiamine (250.5%) alone.<br />

Conclusions: Both berberine and evodiamine alone and in<br />

combination increased 5-HTT mRNA and protein expression<br />

significantly. More interestingly, berberine and evodiamine increased<br />

5-HTT promoter activity differently depending on the genetic<br />

variation <strong>of</strong> the 5-HTTLPR polymorphism. This study has provided a<br />

convincing example <strong>of</strong> how herbal compounds influence the<br />

expression <strong>of</strong> one <strong>of</strong> the most intensively studied psychiatric<br />

candidate genes, the serotonin transporter.<br />

51<br />

ORAL PRESENTATIONS SESSION 5: GWAS/CANDIDATE<br />

GENES I<br />

OPS5.1 A GWAS OF PSYCHOSIS - DATA FROM THE<br />

PSYCHOSIS ENDOPHENOTYPES CONSORTIUM<br />

E. Bramon*, K. Lin, C. Lewis, M. Arranz, R. Murray, J. Powell<br />

Institute <strong>of</strong> Psychiatry - King's College London<br />

*elvira.bramon-bosch@kcl.ac.uk<br />

Introduction: Psychotic disorders including schizophrenia, bipolar<br />

and schizo-affective disorders affect approximately 2% <strong>of</strong> the general<br />

population. Epidemiological data show they are heritable and it is<br />

believed thousands <strong>of</strong> genes <strong>of</strong> small effect interacting with<br />

environmental risk factors are involved (McGuffin, 2007 Sullivan et<br />

al., 2003 Purcell et al, 2009). Through genome-wide association<br />

studies (GWAS) a number <strong>of</strong> promising loci are being identified for<br />

schizophrenia and bipolar disorder and there is growing evidence for<br />

a shared genetic architecture between diverse mental disorders<br />

(Rujescu et al., 2009 Stefansson et al., 2008 Huang et al., 2010 Owen<br />

et al., 2010 Owen et al., 2011 PGC, 2009 Sullivan et al., 2009<br />

Williams et al., 2010).We set out to do a GWAS looking at a broad<br />

phenotype <strong>of</strong> psychosis in a multi-centre sample from Europe and<br />

Australia.<br />

Methodology: All participants underwent a structured clinical<br />

interview with either the SADS, SCID or SCAN to ascertain a DSM-<br />

IV diagnosis <strong>of</strong> schizophrenia, schizo-affective disorder or bipolar<br />

disorder with a history <strong>of</strong> psychotic symptoms. Controls underwent<br />

the same assessments to rule out a personal or family history <strong>of</strong> the<br />

listed disorders. Family history was explored in all participants using<br />

the semi-structured FIGS interview. Participants were excluded if<br />

they had a history <strong>of</strong> neurological disease or head injury resulting in<br />

loss <strong>of</strong> consciousness. After quality control 4,661 participants (1,195<br />

cases, 797 unaffected relatives and 2,669 controls), 721,331 SNPs<br />

and 5 PCA covariate vectors were used for analysis. As part <strong>of</strong> the<br />

Wellcome Trust Case Control Consortium, these samples were<br />

genotyped using Affymetrix 6.0. A genome-wide association analysis<br />

was conducted in the program UNPHASED (Dudbridge, 2008). This<br />

package allows for the combined analysis <strong>of</strong> case-control and family<br />

study data, thus maximizing the statistical power <strong>of</strong> this sample.<br />

Results: We have identified several SNPs that cross genome-wide<br />

significance levels for association with psychosis as a broad<br />

phenotype. We found a promising SNP in the CUX2 gene reaching<br />

genome wide significance (p=10 -8 ), with further significant<br />

associations in multiple neighbouring proxy SNPs and with previous<br />

independent evidence <strong>of</strong> association with bipolar disorder and autism<br />

(Glaser et al, 2005 Kato 2007). As in previous research (Sullivan,<br />

2010 Williams et al, 2011), we have identified loci in the HLA region<br />

in chromosome 6 showing association with psychosis (p=10 -5 - 10 -7 ).<br />

Our current findings are preliminary and await replication in an<br />

independent sample.<br />

Conclusions: These data contribute to the international effort to<br />

produce a large enough sample to investigate psychosis. We intend to<br />

study a selection <strong>of</strong> biological markers <strong>of</strong> brain function and structure<br />

that are relevant in psychosis, which should increase the power to<br />

detect associations.


OPS5.2 THE NEURONAL TRANSPORTER GENE SLC6A15<br />

CONFERS RISK TO MAJOR DEPRESSION<br />

S. Lucae*(1), M. Kohli(1), P. Saemann(1), M. Schmidt(1), M.<br />

Alexander(2), S. Ripke(1), F. Holsboer(1), B. Müller-Myhsok(1), E.<br />

Binder(1)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry 2. Institute <strong>of</strong> Human <strong>Genetics</strong>,<br />

Department <strong>of</strong> Genomics, Life&Brain Center, University <strong>of</strong> Bonn<br />

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

Introduction: Major depression (MD) is one <strong>of</strong> the most prevalent<br />

psychiatric disorders and a leading cause <strong>of</strong> loss in work productivity.<br />

A combination <strong>of</strong> genetic and environmental risk factors likely<br />

contributes to MD. We present data from a genome-wide association<br />

study revealing a neuron-specific neutral amino acid transporter<br />

(SLC6A15) as a novel susceptibility gene for MD.<br />

Methodology: We performed a genome-wide case-control study in a<br />

stringently selected sample <strong>of</strong> MD inpatients <strong>of</strong> a tertiary clinic in<br />

Munich, Germany, and matched controls devoid <strong>of</strong> any lifetime<br />

psychiatric diagnoses. We performed replication <strong>of</strong> the results <strong>of</strong> the<br />

GWAS in six additional independent samples <strong>of</strong> German, Dutch,<br />

United Kingdom and African American origin. The herein reported<br />

association results are based on an overall sample size <strong>of</strong> 15,089<br />

unrelated individuals. To further characterize the functional relevance<br />

<strong>of</strong> the identified locus, we analyzed genotypic influences <strong>of</strong><br />

associated SNPs on pre-mortem human hippocampus and<br />

lymphoblastoid cell line expression pr<strong>of</strong>iles. We also employed in<br />

vivo high resolution structural magnetic resonance imaging (MRI)<br />

with a focus on the hippocampal formation.<br />

Results: A meta-analysis conducted across all 7 samples resulted in a<br />

genome-wide significant association with a p-value <strong>of</strong> 1.41e-09 for<br />

the recessive model <strong>of</strong> rs1545843 and an estimate <strong>of</strong> an OR = 1.398<br />

(95% CI 1.254-1.557). Homozygote carriers <strong>of</strong> the A-allele <strong>of</strong> this<br />

SNP had a higher risk to suffer from depression and depressive<br />

symptoms compared to carriers <strong>of</strong> the two other genotypes. Risk<br />

allele carrier status in humans and chronic stress in mice were<br />

associated with a downregulation <strong>of</strong> the expression <strong>of</strong> this gene in the<br />

hippocampus, a brain region implicated in the pathophysiology <strong>of</strong><br />

MD. The same polymorphisms also showed associations with<br />

alterations in hippocampal volume and neuronal integrity.<br />

Conclusions: Decreased SLC6A15 expression, due to genetic or<br />

environmental factors might alter neuronal circuits related to the<br />

susceptibility for MD. Our convergent data from human genetics,<br />

expression studies, brain imaging and animal models suggest a novel<br />

pathophysiological mechanism for MD that may be accessible to<br />

drug targeting.<br />

52<br />

OPS5.3 AKAPS INTEGRATE GENOME-WIDE<br />

ASSOCIATION STUDY FINDINGS FOR AUTISM<br />

SPECTRUM DISORDERS<br />

ECIP<br />

G. Poelmans*(1), B. Franke(2,3), J. Glennon(1), D. Pauls(4), J.<br />

Buitelaar(1)<br />

1. Department <strong>of</strong> Cognitive Neuroscience, Donders Institute for<br />

Brain, Cognition and Behaviour, Radboud University Nijmegen<br />

Medical Centre 2. Department <strong>of</strong> Human <strong>Genetics</strong>, Radboud<br />

University Nijmegen Medical Centre 3. Department <strong>of</strong> Psychiatry,<br />

Donders Institute for Brain, Cognition and Behaviour, Radboud<br />

University Nijmegen Medical Centre 4. <strong>Psychiatric</strong> and<br />

Neurodevelopmental <strong>Genetics</strong> Unit, Center for Human Genetic<br />

Research, Massachusetts General Hospital, Harvard Medical School<br />

*g.poelmans@psy.umcn.nl<br />

Introduction: Autism spectrum disorders (ASDs) are highly<br />

heritable. In recent years, six genome-wide association studies<br />

(GWASs) <strong>of</strong> ASDs were published. Integration <strong>of</strong> the findings <strong>of</strong><br />

such studies can provide further insight into the genetic etiology <strong>of</strong><br />

ASDs.<br />

Methodology: In order to identify genetic networks, bioinformatics<br />

and systematic literature analyses were conducted for 200 genes from<br />

five published GWASs that yielded association with ASDs through<br />

single nucleotide polymorphisms (SNPs) at P < 0.0001. The sixth<br />

published GWAS was used to validate the findings. We also searched<br />

for overlap between the identified candidate genes and genes located<br />

in copy number variations (CNVs) in people with ASDs and we<br />

identified microRNAs that downregulate the expression <strong>of</strong> ASD<br />

candidate genes and are dysregulated in ASDs.<br />

Results: A total <strong>of</strong> 117 <strong>of</strong> the 200 ASD candidate genes encode<br />

proteins fitting into three signalling networks regulating<br />

steroidogenesis, neurite outgrowth and synaptic function, and we<br />

were able to place 35 other ASD candidate genes - including strong<br />

ASD candidates such as CNTNAP2, MET and NRXN1 - in the<br />

identified networks. Importantly, the proteins encoded by 10 ASDlinked<br />

AKAP (A-kinase anchor protein) genes functionally integrate<br />

signalling cascades within and between these networks. Lastly, we<br />

found that 79 <strong>of</strong> the 200 ASD candidate genes were found in CNVs<br />

in people with ASDs and 100 genes were targets <strong>of</strong> ASD-implicated<br />

microRNAs.<br />

Conclusions: We have identified three signalling networks for ASDs<br />

that provide an important contribution to our understanding <strong>of</strong> the<br />

molecular basis <strong>of</strong> the disorder and that are functionally integrated by<br />

AKAPs, ‘druggable' proteins that deserve further attention as possible<br />

targets for psychopharmacological treatment <strong>of</strong> ASDs.


OPS5.4 GENOME-WIDE GENE EXPRESSION STUDY OF A<br />

SCHIZOPHRENIA DATASET<br />

A. Sanders*(1), H. Göring(2), E. Drigalenko(2), J. Shi(3), W.<br />

Moy(1), J. Freda(1), J. Jacobi(1), D. He(1), M. Collaboration(4), J.<br />

Duan(1), P. Gejman(1)<br />

1. NorthShore University HealthSystem, and University <strong>of</strong> Chicago<br />

2. Texas Biomedical Research Institute 3. National Cancer Institute 4.<br />

Molecular <strong>Genetics</strong> <strong>of</strong> Schizophrenia (MGS) Collaboration<br />

*alan.sanders.md@gmail.com<br />

Introduction: Large schizophrenia case-control GWAS have found<br />

common-variant low-effect loci with most signals being intergenic or<br />

intronic, rare large-effect CNVs, and also evidence for polygenic<br />

contributions to schizophrenia. Each type <strong>of</strong> variant may regulate<br />

mRNA expression as their functional mechanism, but previous work<br />

examining expression has been hampered by underpowered samples.<br />

Methodology: We selected EBV-transformed LCLs derived from<br />

968 European ancestry case samples and 968 screened controls with<br />

pre-existing GWAS data from MGS. We grew the selected LCLs<br />

with intermixed cases and controls, used standardized conditions, and<br />

extracted total RNA. We have assayed mRNA from the first half <strong>of</strong><br />

the sample with Illumina HT-12v4 microarrays, and are now assaying<br />

the remainder using RNAseq. We have studied and controlled for<br />

known confounding effects (e.g., age, sex, ancestry, growth rate,<br />

energy status, transformation site, viral load), as well as unknown<br />

confounders with an expression principal components analysis (PCA)<br />

approach. We used regression analysis to identify transcripts<br />

differentially expressed by affection status and will be performing<br />

eQTN analyses.<br />

Results: We present here the results from our gene expression<br />

experiment with 446 <strong>of</strong> 968 cases and 457 <strong>of</strong> 968 controls, i.e. the<br />

portion <strong>of</strong> the sample studied with microarray. After stringent quality<br />

control, we retained 27,118 transcripts and no sample was identified<br />

as an outlier (all were retained). Examining carriers for<br />

schizophrenia risk CNVs at the 1q21.1, 15q13.3, 16p11.2, and<br />

22q11.21 loci showed the predicted copy number effects for welldetected<br />

RefSeq genes: deletions showing lower expression than the<br />

normal diploid copy number in 78% <strong>of</strong> the transcripts within the<br />

CNV boundaries, and <strong>of</strong> duplications with higher expression 49% <strong>of</strong><br />

the time. After a genome-wide Bonferroni correction, we found 569<br />

differentially expressed transcripts representing 509 genes,<br />

distributed genome-wide, especially at the chromosome 6p histone<br />

cluster located within the MHC region, where the cases showed<br />

consistent histone downregulation. An alternative analytical strategy<br />

incorporating only known confounders (i.e., not using the expression<br />

PCA approach) resulted in fewer genome-wide significant findings,<br />

but similarly converged on histones <strong>of</strong> the MHC region.<br />

Conclusions: It is highly provocative that the most prominent<br />

expression result here is also the most significant schizophrenia<br />

GWAS locus (i.e., MHC), a result which strongly suggests that LCLs<br />

are a useful cell model for studying expression in schizophrenia, and<br />

suggest this may be the case for some other psychiatric<br />

disorders. We will present interim analyses for the remaining half <strong>of</strong><br />

the sample (studied by RNAseq), including assessing inter-method<br />

consistency and for artifacts <strong>of</strong> either method, both for overall results<br />

and in ~50 samples assayed with both microarrays and RNAseq. We<br />

will also present our eQTNs analyses, and discuss their relevance for<br />

explaining genetic contributions to schizophrenia.<br />

53<br />

OPS5.5 A RIGOROUS FAMILY-BASED REPLICATION<br />

STUDY OF SCHIZOPHRENIA GWAS FINDINGS<br />

K. Åberg(1), Y. Liu(2), J. Bukszár(1), J. McClay JL(1), S.<br />

consortium(1), P. Sullivan(2), E. van den Oord*(1)<br />

1. VCU 2. UNC<br />

*ejvandenoord@vcu.edu<br />

Introduction: The aim <strong>of</strong> this study was to identify genetic variants<br />

involved in Schizophrenia (SCZ).<br />

Methodology: We first performed a meta-analysis <strong>of</strong> 18 SCZGWAS<br />

(~22,000 subjects from European ancestry). Using a novel<br />

statisticalframework we developed, we then identified six data bases<br />

(a linkage meta-analysis, brain transcriptome meta-analysis,<br />

schizophrenia candidate gene data base, OMIM, expression QTL<br />

database) that appeared informative about SCZgenetics. These<br />

informative data bases were integrated in the meta-analysis to<br />

identify the most promising SCZcandidate SNPs and genes. Finally,<br />

9,400 selected SNPs were genotyped in 6,300 independent samples<br />

from 1,900 nuclear families. Prior to analyses, we subdivided<br />

subjects into ancestralgroups based on IBS sharing between founders.<br />

Family-based association tests were performed within each group and<br />

a “meta-analysis” was performed by combining test statistics across<br />

all groups.<br />

Results: Results showed enrichment <strong>of</strong> small p-values in subjects<br />

from European, Asian, and African ancestry (average fold increase <strong>of</strong><br />

medium p-value equaled 1.19, 1.12, and 1.07 respectively) as well as<br />

the meta-analysis involving all three groups (fold increase medium pvalue<br />

1.15). Even though some <strong>of</strong> the SNPs had p-values with ranks<br />

up to 100,000 in the GWAS meta-analysis, SNPs selected after data<br />

integration replicated equally well or even better compared to SNPs<br />

with the smallest p-values but that were not implicated by multiple<br />

data sources. The top SNPs from our replication study had p-values<br />

in the 10 -4 -10 -6 range and included severalcoding region SNPs. We<br />

were also able to replicate variants in TCF4 and NOTCH4 that have<br />

previously been associated with SCZ. Forty-eight SNPs (3 times<br />

more than expected) from the MHCregion had p-values < 0.01 in<br />

patients from European ancestry but did not replicate in the other<br />

ethnic groups. This pattern <strong>of</strong> replication can be explained by the fact<br />

that our GWAS meta-analysis involved subjects from European<br />

ancestry only and that LD structure and disease architecture is known<br />

to be highly population specific for the MHCregion.<br />

Conclusions: Our results supported a polygenic architecture for<br />

SCZas multiple genes with small effects replicated.


ORAL PRESENTATIONS SESSION 6: NEXTGEN<br />

SEQUENCING<br />

OPS6.1 WHOLE-EXOME SEQUENCING OF FIVE<br />

FAMILIES WITH BIPOLAR DISORDER<br />

F. Goes*(1), M. Pirooznia(1), J. Parla(2), M. Kramer(2), E.<br />

Elhaik(3), A. Chakravarti(3), R. Karchin(4), P. Zandi(5), W.<br />

McCombie(2), J. Potash(1)<br />

1. Johns Hopkins University School <strong>of</strong> Medicine, Dept. <strong>of</strong> Psychiatry<br />

2. Cold Spring Harbor Laboratory 3. Johns Hopkins University<br />

School <strong>of</strong> Medicine, Institute <strong>of</strong> Genetic Medicine 4. Johns Hopkins<br />

University, Dept. <strong>of</strong> Biomedical Engineering 5. Johns Hopkins<br />

Bloomberg School <strong>of</strong> Public Health, Department <strong>of</strong> Mental Health<br />

*fgoes1@jhmi.edu<br />

Introduction: Complex disorders such as bipolar disorder (BP) are<br />

likely to harbor both common and rare susceptibility alleles. While<br />

common variation has been widely studied in the last decade,<br />

identification <strong>of</strong> rare variants has only recently become feasible with<br />

second-generation sequencing. In this study, we employ wholeexome<br />

sequencing to identify rare coding variants that segregate in<br />

five families with BP.<br />

Methodology: Exome sequencing was performed in 30 individuals<br />

from five multiplex BP families using solution-based capture and<br />

paired-end sequencing on the Illumina GA II. Alignment and variant<br />

calling were performed with BWA, SAM tools and GATK. SNVs<br />

were annotated with SeattleSeq and SWISS-PROT databases, and the<br />

SCHISM (Supervised CHaracterization <strong>of</strong> Inherited SNPs and<br />

Mutations) algorithm. Each family was analyzed individually by<br />

selecting novel variants that were predicted to be damaging and<br />

segregated with disease.<br />

Results: Mean coverage for the targeted exome sequence was 97% at<br />

1X coverage, 91% at 10X and 62% at 40X. A total <strong>of</strong> 41,154<br />

variants were identified, including 3,340 novel variants. Of these, 52<br />

were nonsense and 2,027 were missense variants, <strong>of</strong> which 504 were<br />

predicted to be damaging to protein function. In the within-family<br />

analysis, we found 16 novel damaging missense variants that<br />

segregated with BP.<br />

Conclusions: Our initial results show that a family-based analysis<br />

with multiplex pedigrees can successfully winnow novel variants to a<br />

relatively small number that segregate within families. These<br />

preliminary analyses provide evidence for 16 novel variants that may<br />

be associated with familial BP. These variants are currently being<br />

genotyped in an additional 1000 BP cases and 1000 controls, and the<br />

results will be presented at the WCPG.<br />

54<br />

OPS6.2 FAMILY-BASED EXOME SEQUENCING IN<br />

FAMILIES MULTIPLY-AFFECTED FOR SCHIZOPHRENIA<br />

J. Rosenfeld*(1,2), A. Malhotra(1,2), T. Lencz(1,2)<br />

1. Zucker Hillside Hospital 2. Feinstein Institute for Medical<br />

Research<br />

*jrosenfeld@nshs.edu<br />

Introduction: Schizophrenia is a highly heritable disease, yet recent<br />

genome-wide association studies have demonstrated that common<br />

polygenic alleles cannot fully account for the observed inheritance.<br />

Recently, the exomes (the entire gene-coding region <strong>of</strong> the genome)<br />

<strong>of</strong> individuals affected with a variety <strong>of</strong> Mendelian diseases have<br />

been sequenced to identify rare, highly penetrant mutations<br />

underlying the pathology <strong>of</strong> those particular cases. Since<br />

schizophrenia (like other complex disorders) is likely to have a subset<br />

<strong>of</strong> cases caused by rare protein-coding changes, exome sequencing<br />

may be a useful approach, particularly in multiply-affected families<br />

in which several individuals may share a single causative mutation.<br />

Methodology: We have completely sequenced the exomes <strong>of</strong> two<br />

families that are multiply affected with schizophrenia. Each family<br />

consisted <strong>of</strong> two affected individuals and either two or four<br />

unaffected first degree relatives. The exome <strong>of</strong> each individual was<br />

captured using the Agilent SureSelect technique and then sequenced<br />

on a single lane <strong>of</strong> the Illumina HiSeq sequencer with 100 bp pairedend<br />

reads. The exomes were analyzed to determine many different<br />

kinds <strong>of</strong> variations including SNPs, rearrangements and copy-number<br />

variations (CNV). We utilized existing computational tools for the<br />

SNP detection, while the rearrangement and CNV detection were<br />

performed using our own newly created tools.<br />

Results: High-quality, high-depth reads were obtained on 75% <strong>of</strong> the<br />

exome, with a median <strong>of</strong> 80x depth. Initial analysis <strong>of</strong> the first family<br />

indicates that there were an average <strong>of</strong> 20,000 SNPs per<br />

individual. After filtering the variants, ~200 missense mutations and<br />

3 nonsense mutations shared by cases and absent in unaffected family<br />

members. Results from all 10 individuals will be presented at the<br />

meeting.<br />

Conclusions: We have utilized exome sequencing to identify<br />

mutations underlying the schizophrenia in two families. The overall<br />

genetic similarity <strong>of</strong> family members enhances the ability to pinpoint<br />

the variations by eliminating family-specific mutations that are<br />

irrelevant to disease status. Candidate mutations will then be tested in<br />

a larger panel <strong>of</strong> cases and controls to eliminate false positives and<br />

determine frequency and penetrance <strong>of</strong> potentially causative<br />

mutations.


OPS6.3 BRAIN TRANSCRIPTOME ANALYSIS BY RNA-SEQ<br />

REVEALS NOVEL DIFFERENTIALLY EXPRESSED GENES<br />

IN BIPOLAR DISORDER<br />

N. Akula*(1), J. Wendland(1), G. Laje(1), K. Choi(2), M.<br />

Webster(3), H. Bravo(4), S. Detera-Wadleigh(1), F. McMahon(1)<br />

1. Mood and Anxiety Disorders Section,Human <strong>Genetics</strong> Branch,<br />

National Institute <strong>of</strong> Mental Health, National Institutes <strong>of</strong> Health 2.<br />

Department <strong>of</strong> Psychiatry and Program in Neuroscience, Uniformed<br />

Services University <strong>of</strong> the Health Sciences, Center for the Study <strong>of</strong><br />

Traumatic Stress 3. Stanley Lab <strong>of</strong> Brain Research 4. Center for<br />

Bioinformatics and Computational Biology, University <strong>of</strong> Maryland<br />

*akulan@mail.nih.gov<br />

Introduction: RNA-seq is a relatively new technique that leverages<br />

high-throughput sequencing technologies to provide estimates <strong>of</strong><br />

transcript abundance at a precision not previously realized in<br />

hybridization-based microarray assays. RNA-seq also enables the<br />

detection <strong>of</strong> novel, low abundance transcripts, allele-specific<br />

expression, alternative splicing, and post-transcriptional<br />

modifications, such as RNA editing. This technique thus provides a<br />

fundamental context in which risk alleles can be evaluated for their<br />

impact on gene expression and co-regulation. In a preliminary study,<br />

we used RNA-seq to characterize the brain transcriptome in bipolar<br />

disorder.<br />

Methodology: We performed deep-sequencing (~130M paired-end<br />

reads) <strong>of</strong> high quality total RNA (RNA-integrity number ≥ 7)<br />

extracted from prefrontal cortex obtained post-mortem from 4 cases<br />

diagnosed with bipolar I disorder and 5 age- and sex-matched,<br />

psychiatrically-healthy controls, provided by the Stanley Medical<br />

Institute. Library preparation, fragmentation and PCR enrichment <strong>of</strong><br />

the target RNA was followed by paired-end sequencing on the<br />

Illumina GA-IIx system. The resulting reads were mapped and<br />

aligned to the reference genome (hg18) using TopHat (Trapnell et al.<br />

2009). Preliminary analysis <strong>of</strong> differential expression <strong>of</strong> known<br />

(NCBI-RefSeq and Ensemble) and alternate transcripts was<br />

performed using Cufflinks (Trapnell et al. 2010), in parallel with<br />

analysis with HTSeq (http://wwwhuber.embl.de/users/anders/HTSeq/)<br />

and DESeq (Anders and Huber<br />

2010) analysis. Transcripts with raw counts ≤ 50 were excluded from<br />

downstream analysis. Significant differentially expressed transcripts<br />

were identified based on a p-value ≤ 0.05, adjusted for multiple<br />

testing. These transcripts were further analyzed in DAVID (Dennis et<br />

al. 2003; Huang da et al. 2009) in order to detect enriched functional<br />

groups <strong>of</strong> genes.<br />

Results: A total <strong>of</strong> 172 transcripts were differentially expressed by as<br />

much as tenfold; <strong>of</strong> these 154 transcripts were down-regulated in<br />

bipolar disorder. Differentially-expressed transcripts showed<br />

significant enrichment <strong>of</strong> the Gene Ontology terms “neuronal<br />

development,” “apoptosis,” “G-protein coupled receptors,” and<br />

“calcium ion homeostasis.”<br />

Conclusions: Further analyses to identify novel alternate transcripts<br />

that are differentially expressed in bipolar disorder brain are<br />

underway. These preliminary results demonstrate that RNA-seq may<br />

reveal gene expression changes in biologically-relevant pathways that<br />

were not detected consistently in previous microarray-based<br />

expression pr<strong>of</strong>iling studies.<br />

55<br />

OPS6.4 ROBUST DISCOVERY OF COPY NUMBER<br />

VARIATION IN MATCHED SWEDISH SCHIZOPHRENIA<br />

CASE-CONTROL WHOLE-EXOME TARGETED<br />

SEQUENCING<br />

ECIP<br />

M. Fromer*(1), J. Moran(1), K. Chambert(1), P. Sullivan(2), C.<br />

Hultman(3), P. Sklar(1,4,5), S. Purcell(1,4)<br />

1. Stanley Center for <strong>Psychiatric</strong> Research, Broad Institute <strong>of</strong><br />

Harvard and MIT 2. Departments <strong>of</strong> <strong>Genetics</strong>, Psychiatry, and<br />

Epidemiology, University <strong>of</strong> North Carolina at Chapel Hill 3.<br />

Department <strong>of</strong> Medical Epidemiology and Biostatistics, Karolinska<br />

Institutet 4. Center for Human Genetic Research, Massachusetts<br />

General Hospital 5. Division <strong>of</strong> <strong>Psychiatric</strong> Genomics, Mount Sinai<br />

School <strong>of</strong> Medicine<br />

*fromer@broadinstitute.org<br />

Introduction: There has been much recent debate regarding the<br />

genetic architecture underlying susceptibility to schizophrenia and<br />

other neuropsychiatric diseases. Nonetheless, it is clear that rare<br />

copy number variations (CNV) play a role in the pathology <strong>of</strong><br />

schizophrenia. Most work on this topic has been done using predefined<br />

single-nucleotide polymorphism (SNP) arrays. In order to<br />

more fully elucidate this phenomenon at higher genomic resolution<br />

and identify rare CNV (both deletions and duplications), we have<br />

undertaken high-coverage whole-exome targeted resequencing <strong>of</strong> 550<br />

matched Swedish schizophrenia cases and controls. Specifically, we<br />

use the high-coverage sequencing read depth as a proxy for copy<br />

number, so that we would like to identify regions where a particular<br />

sample has significant depletion or enrichment in sequenced reads<br />

and infer a deletion or duplication CNV, respectively.<br />

However, this effort is confounded by multiple known factors,<br />

including GC content <strong>of</strong> the targeted exome intervals, the size and<br />

sequence complexity <strong>of</strong> these intervals, sequencing batch, proximity<br />

to segmental duplications, and nucleotide-level population variation,<br />

all <strong>of</strong> which may lead to differential capture <strong>of</strong> exomic sequence or<br />

sequencing efficiency. Moreover, there are presumably multiple<br />

unknown factors and interactions between all such factors that lead to<br />

sample-specific variability in read depth that does not directly result<br />

from genomic copy number variation.<br />

Methodology: In order to overcome these issues, we have performed<br />

rigorous statistical, data-driven normalization using a principal<br />

component analysis (PCA) approach. We detected and removed read<br />

depth variation driven by these factors, resulting in a de-noised data<br />

set with which we then proceeded to make CNV calls using a hidden<br />

Markov model (HMM) algorithm for segmentation <strong>of</strong> the exome into<br />

diploid, deletion, and duplication regions for each sample.<br />

Results: Overall, we achieved high sensitivity in recovering CNV<br />

calls based on Affymetrix 6.0 SNP array data for these same<br />

samples. Moreover, we were able to detect a large number <strong>of</strong><br />

previously unknown rare CNVs with nominally significant<br />

association with susceptibility to schizophrenia, where a number <strong>of</strong><br />

these CNV were in brain-expressed genes.<br />

Conclusions: We conclude that high-coverage exome sequencing<br />

and careful analysis <strong>of</strong> sequencing depth can provide valuable<br />

insights into the location and character <strong>of</strong> rare CNV conferring risk<br />

for schizophrenia, and, more generally, provides a broadly applicable<br />

tool for both sensitive and specific CNV discovery.


OPS6.5 IDENTIFICATION AND ANALYSIS OF RARE<br />

VARIATION IN 503 PUTATIVE SCHIZOPHRENIA RISK<br />

LOCI IN 1876 SAMPLES BY TARGETED RESEQUENCING<br />

C. O'Dushlaine(1,2), J. Moran(2), D. Ruderfer(1,2), K. Chambert(2),<br />

P. Lichtenstein(3), C. Hultman(3), P. Sullivan(4), S. Purcell(1,2),<br />

P. Sklar(1,2,5)<br />

1. <strong>Psychiatric</strong> and Neurodevelopmental <strong>Genetics</strong> Unit, Massachusetts<br />

General Hospital 2. Stanley Center for <strong>Psychiatric</strong> Research, Broad<br />

Institute <strong>of</strong> Harvard and MIT 3. Department <strong>of</strong> Medical<br />

Epidemiology, Karolinska Institutet 4. Departments <strong>of</strong> <strong>Genetics</strong>,<br />

Psychiatry, and Epidemiology, University <strong>of</strong> North Carolina at<br />

Chapel Hill 5. Division <strong>of</strong> <strong>Psychiatric</strong> Genomics, Department <strong>of</strong><br />

Psychiatry, Mount Sinai School <strong>of</strong> Medicine<br />

Introduction: We performed array-based sequence capture followed<br />

by next generation sequencing to analyze 503 genes identified via<br />

genomic studies. These genes were chosen from regions that contain<br />

highly penetrant copy number variants in autism, schizophrenia, and<br />

bipolar disorder (1q21.1, 15q11.2, 15q13-13.3, 16p11.2, 16p12.1,<br />

17p12 and 22q11.21; n=197 genes). Genes were also selected for the<br />

following reasons: within CNVs identified as highly connected<br />

through GRAIL analysis (n=32), presence in a gene locus strongly<br />

associated with schizophrenia (n=17), and bipolar disorder (n=6),<br />

identified as potentially enriched in rare variants through exome<br />

sequencing (n=117), identified as enriched in pathway analyses <strong>of</strong><br />

GWAS data (n=20), or found in a region <strong>of</strong> excess runs <strong>of</strong><br />

homozygosity (n=11).<br />

Methodology: Case and control DNA pools were carefully<br />

constructed from a Swedish schizophrenia sample collection to<br />

decrease the likelihood <strong>of</strong> population stratification. Pools contained<br />

only a single sex to allow for sequencing <strong>of</strong> X-chromosome genes.<br />

Blood-derived DNAs were normalized and pooled with 20 DNA<br />

samples per pool. An Agilent SureSelect custom-designed<br />

oligonucleotide array was synthesized and used for solution-based<br />

hybrid selection on each pool. Captured products were then<br />

sequenced by Illumina GA using 76 bp paired-end reads. The hybrid<br />

selection array design included exons as well as microRNAs within<br />

CNV regions or in genes. In total 1.56Mb, encompassing 7095 exons<br />

from 503 RefSeq genes (including 12 microRNAs and 18 miR137regulated<br />

genes) were sequenced in 1876 samples (938 cases, 938<br />

controls).<br />

Results: We present findings from this study for single variant and<br />

gene-based (variable threshold) association tests. In particular, we<br />

find evidence for association for the calcium channel genes<br />

CACNA1H, CACNA1S and for the 3p21 genes ITIH3 and NEK4. We<br />

address analytic challenges inherent in the analysis <strong>of</strong> rare variants.<br />

Conclusions: *The authors acknowledge the support <strong>of</strong> an<br />

Anonymous Foundation, the Stanley Medical Research Institute and<br />

the National Institutes <strong>of</strong> Mental Health.<br />

56<br />

ORAL PRESENTATIONS SESSION 7: STATISTICAL<br />

GENETICS, GENETIC EPIDEMIOLOGY<br />

OPS7.1 CNR1 GENOTYPE MODERATES THE EFFECTS OF<br />

CHILDHOOD ABUSE ON ANHEDONIA<br />

A. Agrawal*(1), E. Nelson(1), A. Littlefield(2), K. Bucholz(1), L.<br />

Degenhardt(3), A. Henders(4), P. Madden(1), N. Martin(4), G.<br />

Montgomery(4), M. Pergadia(1), K. Sher(2), A. Heath(1), M.<br />

Lynskey(1)<br />

1. Washington University School <strong>of</strong> Medicine, Psychiatry 2.<br />

University <strong>of</strong> Missouri in Columbia 3. Burnet Institute 4. Queensland<br />

Institute for Medical Research<br />

*arpana@wustl.edu<br />

Introduction: Endocannabinoids, primarily, Narachidonoylethanolamine<br />

(anadamide) and 2-arachidonoylglycerol<br />

(2-AG) are naturally occurring molecules that have putative<br />

anxiolytic, analagesic and anti-depressant effects. Their influence is<br />

partly mediated by the CB1 receptors to which they bind. Recent<br />

studies have found that rodents with deficiencies in CB1 receptor<br />

activity (due to targeted knock-out or antagonist action) show<br />

anhedonia and depressed mood. In addition, there is accumulating<br />

evidence that persistent stress directly contributes to differential<br />

fluctuations in anadamide and 2-AG. In rodents experiencing<br />

repeated stress exposure, decline in anadamide can modify<br />

corticosteroid levels, indicating the endocannabinoid signaling may<br />

be critical in adaptation to stress. While the theories linking<br />

endocannabinoids to stress adaptation and mood have been validated<br />

in rodent models, there are no human studies that clearly demonstrate<br />

the nature <strong>of</strong> this relationship. We hypothesized that a variant in the<br />

gene encoding the human endocannabinoid receptor (CNR1) -<br />

rs1049353 - interacts with childhood exposure to serious physical<br />

abuse to produce anhedonia.<br />

Methodology: Logistic regression was used to examine the<br />

association between anhedonia and the main effects <strong>of</strong> and<br />

interaction between rs1049353 and childhood physical abuse in four<br />

independent samples. Sample A consisted <strong>of</strong> 1140 female twins from<br />

Missouri, Sample B consisted <strong>of</strong> 1934 heroin dependent cases and<br />

controls from Australia, Sample C consisted <strong>of</strong> 203 Missouri college<br />

aged students with and without a family history <strong>of</strong> alcoholism and<br />

Sample D consisted <strong>of</strong> 1740 Australian general population twins. All<br />

subjects were <strong>of</strong> European-American descent. Anhedonia was<br />

defined as self-reported loss <strong>of</strong> interest in activities considered<br />

enjoyable and childhood physical abuse was also defined using a<br />

variety <strong>of</strong> self-report items.<br />

Results: In Sample A, there was a strong main effect <strong>of</strong> childhood<br />

physical abuse on anhedonia but not main effect <strong>of</strong> genotype.<br />

However, the interaction was significant (P=0.03). While 22% <strong>of</strong> the<br />

full sample reported anhedonia, 58% <strong>of</strong> individuals who were<br />

homozygous for the major allele (GG) and experienced abuse<br />

reported anhedonia compared with 28% <strong>of</strong> carriers <strong>of</strong> the minor allele<br />

(AA/AG) experiencing abuse with no differences between AA and<br />

AG individuals. When stratified by genotype, there was no main<br />

effect <strong>of</strong> abuse on anhedonia in AA/AG individuals indicating that<br />

rs1049353 buffers the potent impact <strong>of</strong> childhood physical abuse on<br />

anhedonia. This interaction was systematically replicated in Samples<br />

B, C and D.<br />

Conclusions: This is one <strong>of</strong> few replicated examples <strong>of</strong> genotype x<br />

environment interaction that further underscores the role <strong>of</strong> the<br />

endogenous cannabinoid system in buffering the pathogenic effects<br />

<strong>of</strong> childhood abuse on mental health. Consistent with the preclinical<br />

literature, our association directly replicated evidence from rodent<br />

models and validates the important role <strong>of</strong> cannabinoid receptors in<br />

modulation <strong>of</strong> mood.


OPS7.2 ASSESSING THE CONTRIBUTION FAMILY DATA<br />

CAN MAKE TO CASE-CONTROL STUDIES OF RARE<br />

VARIANTS<br />

D. Curtis*<br />

Barts and the London School <strong>of</strong> Medicine and Dentistry<br />

*david.curtis@qmul.ac.uk<br />

Introduction: When pathogenic variants are rare then even among<br />

cases the proportion <strong>of</strong> subjects possessing a variant might be low,<br />

meaning that very large samples might be required to conclusively<br />

demonstrate evidence <strong>of</strong> an effect.Relatives <strong>of</strong> subjects within a casecontrol<br />

sample might provide useful additional information.<br />

Methodology: The method <strong>of</strong> model-free linkage analysis<br />

implemented in MFLINK was adapted to incorporate linkage<br />

disequilibrium (LD) parameters in order to model a putative<br />

pathogenic variant in complete LD with a disease locus. The<br />

performance <strong>of</strong> the method was tested against other methods applied<br />

to case-control samples. The effect <strong>of</strong> adding in to the analysis<br />

relatives <strong>of</strong> cases and controls found to carry the variant was<br />

investigated.<br />

Results: The method produced similar results to other methods when<br />

applied to case-control samples. When affected siblings or cousins <strong>of</strong><br />

cases possessing the variant were incorporated they had a large effect<br />

on the results obtained. The evidence for involvement increased or<br />

reduced as expected, depending on whether or not the relatives<br />

themselves were found to possess the variant. The size <strong>of</strong> the effect<br />

was large relative to that expected from just increasing the size <strong>of</strong> a<br />

standard case-control sample.<br />

Conclusions: Affected relatives <strong>of</strong>fer a valuable resource to assist the<br />

interpretation <strong>of</strong> case-control studies <strong>of</strong> rare variants. The method is<br />

capable <strong>of</strong> including other relative types and can deal with complex<br />

pedigrees.<br />

57<br />

OPS7.3 A POISSON REGRESSION APPROACH FOR<br />

ASSOCIATION MAPPING OF COUNT PHENOTYPES<br />

S. Ghosh*, A. Chakrabortty<br />

Indian Statistical Institute<br />

*saurabh@isical.ac.in<br />

Introduction: Clinical end-point traits are <strong>of</strong>ten characterized in<br />

terms <strong>of</strong> quantitative endophenotypes. Since these phenotypes carry<br />

more information on within-genotype variability, it has been argued<br />

that analyzing quantitative precursors may be a more powerful<br />

strategy to identify novel genetic factors underlying a complex<br />

disorder. The statistical methods <strong>of</strong> choice for association mapping <strong>of</strong><br />

quantitative phenotypes based on population-level data have been<br />

Analysis <strong>of</strong> Variance (ANOVA) or the Kruskal Wallis test. However,<br />

both these methods are strictly suited to analyze continuous traits. For<br />

psychiatric disorders, traits such as symptom counts <strong>of</strong>ten serve as<br />

endophenotypes <strong>of</strong> interest for understanding the genetic basis <strong>of</strong> the<br />

clinical end-point trait. Since such traits are discrete in nature, it may<br />

not be optimal to use ANOVA or the Kruskal Wallis test to detect<br />

association.<br />

Methodology: For population level data, we propose a Poisson<br />

regression approach that computes the likelihood <strong>of</strong> the count<br />

phenotype conditional on an additive allele count at a SNP. The test<br />

statistic is asymptotically distributed as chi-squares with one degree<br />

<strong>of</strong> freedom under no association between the SNP and the<br />

phenotype. For family-based data involving trios with at least one<br />

heterozygous parent at a SNP, we use a similar Poisson regression<br />

model conditional on two indicator variables: the marker allele<br />

transmitted by the heterozygous parent and the marker allele<br />

transmitted by the other parent. A one degree <strong>of</strong> freedom test based<br />

only on the coefficient <strong>of</strong> the first indicator is protected against<br />

population stratification as it tests for association in the presence <strong>of</strong><br />

linkage. A two degrees <strong>of</strong> freedom test based on both the indicators is<br />

also a valid test for association, but is susceptible to population<br />

stratification.<br />

Results: We have performed extensive simulations under different<br />

genetic models to compare the relative performance <strong>of</strong> the proposed<br />

Poisson regression with ANOVA and the Kruskal Wallis test for<br />

population level data. We find that while the asymptotic tests for<br />

ANOVA and Kruskal Wallis yield an inflated rate <strong>of</strong> false positives,<br />

especially when there is heteroskedasticity in the distribution <strong>of</strong> the<br />

trait across the QTL genotypes, our proposed method maintains the<br />

correct size. Moreover, our method yields uniformly more power (for<br />

equivalent empirical levels) compared to the other methods for the<br />

different genetic parameters considered in our simulations. For a trio<br />

design, we find that the powers are lower than a population-level<br />

design where the number <strong>of</strong> sampled trios is equal to the number <strong>of</strong><br />

random individuals sampled. We applied our method to analyze an<br />

endophenotype defined as the number <strong>of</strong> externalizing symptoms<br />

associated with alcoholism using data generated in the Collaborative<br />

Study On the <strong>Genetics</strong> <strong>of</strong> Alcoholism (COGA) project. We found<br />

significant evidence <strong>of</strong> association in the class 1 alcohol<br />

dehydrogenase subunit ADH1C in the 4q22.3 region.<br />

Conclusions: The proposed Poisson regression model is ideally<br />

suited for association analyses <strong>of</strong> count phenotypes and is more<br />

powerful compared to existing approaches such as ANOVA or the<br />

Kruskal Wallis test.


OPS7.4 EVALUATION OF PERMUTATION-BASED<br />

APPROACHES FOR MULTI-MARKER ASSOCIATION<br />

V. Moskvina*(1), K. Schmidt(2), A. Vedernikov(1), M. Owen(1), N.<br />

Craddock(1), P. Holmans(1), M. O'Donovan(1)<br />

1. School <strong>of</strong> Medicine, Cardiff University 2. School <strong>of</strong> Mathematics,<br />

Cardiff University<br />

*moskvinav1@cf.ac.uk<br />

Introduction: Genome-wide association studies have revealed a<br />

small number <strong>of</strong> genome-wide significant associations for psychiatric<br />

disorders. Since there is clear evidence for a substantial contribution<br />

to risk from variants that are tagged by GWAS arrays (ISC, 2009),<br />

the most likely explanation for this is limited power <strong>of</strong> the studies to<br />

date. Until larger studies are conducted, one possible approach to<br />

increasing power is to conduct set-based analyses to test the<br />

collective effect <strong>of</strong> larger groups <strong>of</strong> SNPs, for example a set <strong>of</strong><br />

markers within a single gene (Moskvina et al, 2009), the hypothesis<br />

being that under a polygenic model, there may be multiple<br />

independent signals within a single gene. Several set-based methods<br />

have been proposed, among which the most widely used are the<br />

product <strong>of</strong> all p-values (ProdP) and the average <strong>of</strong> the chi-square<br />

statistics (as in PLINK, Purcell et al, 2007), but the most powerful<br />

method has yet to be determined. The major challenge for set based<br />

analyses is to allow for marker-marker correlations (LD) and<br />

therefore typically, the significance <strong>of</strong> set-based analyses is assessed<br />

by permutations to adjust for this.<br />

Methodology: To compare the performance <strong>of</strong> the permutation based<br />

ProdP test (which gives similar results to the average chi-square) with<br />

those <strong>of</strong> the Hotelling's T 2 test (HT 2 ) proposed by Chapman &<br />

Whittaker (2008) and logistic regression analysis, neither <strong>of</strong> which<br />

require permutations, in simulated and real data. We also investigated<br />

the effects <strong>of</strong> LD on the results obtained, as an appropriate method<br />

would give identical results when additional markers in perfect LD<br />

are included to those obtained when such markers are excluded.We<br />

compared ProdP and HT 2 tests for simulated and real data. The HT 2<br />

test is practically identical to logistic regression analysis without<br />

covariates with all the markers included in the model. We therefore<br />

focused on the HT 2 test for comparison with the ProdP method. We<br />

applied both methods to simulated data with varying numbers <strong>of</strong><br />

SNPs in tight LD, and also to real GWAS data comprising 11791<br />

genes with two or more SNPs (the WTCCC bipolar dataset). We<br />

tested the effect <strong>of</strong> pruning SNPs by LD i.e. selecting markers at<br />

random and removing those which are in LD with the selected ones<br />

above a certain threshold value <strong>of</strong> r 2 .<br />

Results: In the presence <strong>of</strong> even modestly correlated markers, the<br />

HT 2 and ProdP tests are broadly correlated but give very different<br />

individual gene-wide p-values. As markers become less dependent<br />

(i.e. more aggressive LD pruning), the results <strong>of</strong> the two tests<br />

converge, but this occurs at quite a low r 2 threshold for pruning<br />

(r 2 =0.05). We also show that in a real dataset, when markers are LD<br />

pruned at this threshold, there is considerable loss in power, as would<br />

be expected if there are multiple signals within gene sets arising from<br />

SNPs that are partially correlated, or single true signals that are<br />

partially tagged by multiple SNPs, each conferring additional<br />

partially independent information.<br />

Conclusions: We show that in the presence <strong>of</strong> marker-marker LD at<br />

a level typical in GWAS datasets, gene-wide tests statistics derived<br />

from the HT 2 approach are more valid and more powerful than those<br />

derived from permutations. Unless markers are aggressively LD<br />

pruned, which risks loss <strong>of</strong> power, the HT 2 (and regression based<br />

approaches) are recommended over the customary approaches that<br />

rely on permutations.<br />

58<br />

OPS7.5 UNDERESTIMATED EFFECT SIZES IN GWAS:<br />

FUNDAMENTAL LIMITATIONS OF SINGLE SNP<br />

ANALYSIS FOR DISEASE STATUS<br />

ECIP<br />

S. Stringer*(1), N. Wray(2), R. Kahn(1), E. Derks(1)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience 2. Queensland Institute<br />

<strong>of</strong> Medical Research<br />

*s.stringer@umcutrecht.nl<br />

Introduction: <strong>Psychiatric</strong> traits are <strong>of</strong>ten highly heritable. Only a<br />

small proportion <strong>of</strong> the heritability can be explained by observed<br />

genetic variants in traditional genome-wide association (GWA)<br />

studies. We propose an additional explanation for the missing<br />

heritability in disease status by showing that effect sizes can be<br />

severely underestimated with single SNP analysis.<br />

Methodology: We defined a model <strong>of</strong> complex disease based on the<br />

multi-locus odds model with four model parameters: prevalence,<br />

effect size, risk allele frequency, and number <strong>of</strong> effect SNPs. We<br />

used this model to simulate the odds ratios that would be obtained<br />

with single SNP analysis and studied the relationship between true<br />

effect size and estimated effect size. We derived the same<br />

relationship for a model <strong>of</strong> continuous disease trait based on a multilocus<br />

linear model. Finally, we derived the relationship between<br />

biased effect size estimates and missing heritability.<br />

Results: Moderate to large effect sizes <strong>of</strong> causal variants may be<br />

estimated as relatively small effect sizes in single SNP association<br />

testing. This underestimation effect is most severe for diseases<br />

influenced by multiple risk variants and results in an increased<br />

missing heritability. Continuous disease traits generated with linear<br />

genetic models are not affected by the underestimation effect.<br />

Conclusions: Single SNP association methods test for association at<br />

a single SNP at a time, and thus test across an averaged genetic<br />

background. The non-linearity <strong>of</strong> the relationship between genetic<br />

risk and disease risk results in biased estimates <strong>of</strong> effect size. As<br />

many GWA studies <strong>of</strong> psychiatric traits apply single SNP analysis to<br />

disease status, their results could underestimate true effect sizes and<br />

reduce the explained variance. Therefore, when a multi-locus model<br />

<strong>of</strong> disease risk is assumed, single SNP analysis is ill-suited for<br />

identifying large effect sizes. Instead, we recommend analyzing all<br />

SNPs simultaneously.


ORAL PRESENTATIONS SESSION 8: GWAS/CANDIDATE<br />

GENES II<br />

OPS8.1 META-ANALYSIS OF EUROPEAN AND ASIAN-<br />

ANCESTRY SAMPLES IDENTIFIES 3 NOVEL LOCI<br />

ASSOCIATED WITH BIPOLAR DISORDER<br />

ECIP<br />

D. Chen*(1), X. Jiang(1), N. Akula(1), J. Wendland(1), C. Steele(1),<br />

L. Kassem(1), J. Park(2), N. Chatterjee(2), S. Jamain(3), A.<br />

Cheng(4), M. Leboyer(3), P. Muglia(5), T. Schulze(1,6), S.<br />

Cichon(7), M. Rietschel(8), F. McMahon(1)<br />

1. Human <strong>Genetics</strong> Branch, National Institute <strong>of</strong> Mental Health<br />

Intramural Research Program, National Institutes <strong>of</strong> Health, U.S.<br />

Dept <strong>of</strong> Health and Human Services 2. Division <strong>of</strong> Cancer<br />

Epidemiology and <strong>Genetics</strong>, National Cancer Institute, National<br />

Institutes <strong>of</strong> Health, US Department <strong>of</strong> Health and Human Services 3.<br />

Inserm U955, Department <strong>of</strong> Psychiatry, Groupe Hospitalier Henri<br />

Mondor-Albert Chenevier, AP-HP, Université Paris Est, Fondation<br />

FondaMental 4. Institute <strong>of</strong> Biomedical Sciences, Academia Sinica 5.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Toronto & NeuroSearch 6.<br />

Section on <strong>Psychiatric</strong> <strong>Genetics</strong>, Department <strong>of</strong> Psychiatry and<br />

Psychotherapy, University Medical Center, Georg-August-<br />

Universität 7. Institute <strong>of</strong> Neuroscience and Medicine (INM-1),<br />

Structural and Functional Organization <strong>of</strong> the Brain, Genomic<br />

Imaging, Research Center Juelich, D-52425 Juelich, Germany<br />

Department <strong>of</strong> Genomics, Life & Brain Center, University <strong>of</strong> Bonn,<br />

D-53127 Bonn, Germany 8. Department <strong>of</strong> Genetic Epidemiology in<br />

Psychiatry, Central Institute <strong>of</strong> Mental Health, University <strong>of</strong><br />

Mannheim, D-68159<br />

*chend4@mail.nih.gov<br />

Introduction: Meta-analyses <strong>of</strong> bipolar disorder (BD) genome-wide<br />

association studies (GWAS) have identified several genome-wide<br />

significant signals in European-ancestry samples, but so far these<br />

seem to account for little <strong>of</strong> the inherited disease risk.<br />

Methodology: We performed a meta-analysis <strong>of</strong> over 750,000 highquality<br />

genetic markers on a discovery sample <strong>of</strong> about 15,000<br />

subjects <strong>of</strong> European and Asian ancestry. The most significant<br />

findings were further tested in a replication sample <strong>of</strong> 3,800 cases and<br />

controls. We then assessed the expression <strong>of</strong> associated gene(s) after<br />

exposing cells in culture to pharmacological treatments for<br />

BD. Based on these results and other established risk loci, we<br />

estimated the number <strong>of</strong> susceptibility loci and effect size distribution<br />

in BD, according to the method described by Park and colleagues<br />

(2010).<br />

Results: The results suggest novel association findings near the<br />

genes TRANK1 (LBA1), LMAN2L, and PTGFR. The most significant<br />

SNP, rs9834970 near TRANK1, was significant at the p=2.4 x 10 -11<br />

level under a fixed-effects model. Supportive evidence for prior<br />

association findings near ANK3 and a locus on chromosome 3p21<br />

was also observed. After addition <strong>of</strong> the replication samples to the<br />

meta-analysis, the overall picture was similar, although the<br />

heterogeneity test became significant for several SNPs. In cultured<br />

cells, valproate markedly increased TRANK1 expression in a dose-<br />

and time-dependent manner.<br />

Conclusions: These results support previous GWAS findings and<br />

add new candidate genes to a growing list <strong>of</strong> bipolar disorder risk<br />

loci, confirming the critical importance <strong>of</strong> sample size in GWAS<br />

methods.<br />

59<br />

OPS8.2 ASSOCIATION WITH SYNE1 VARIANT IN BOTH<br />

BIPOLAR DISORDER AND RECURRENT MAJOR<br />

DEPRESSION<br />

E. Green*(1), D. Grozeva(1), I. Jones(1), L. Jones(2), L. Forty(1), K.<br />

Gordon-Smith(1,2), A. Farmer(3), P. McGuffin(3), J. Moran(4), J.<br />

Kranz(4), S. Purcell(4), P. Sklar(4), M. Owen(1), M. O'Donovan(1),<br />

N. Craddock(1)<br />

1. MRC Centre for Neuropsychiatric <strong>Genetics</strong> and Genomics, Dept.<br />

<strong>of</strong> Psychological Medicine, Cardiff University 2. Dept. <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Birmingham 3. MRC SGDP Centre, Institute <strong>of</strong><br />

Psychiatry, King's College London 4. Stanley Centre for <strong>Psychiatric</strong><br />

Research, Broad Institute<br />

*greenek@cf.ac.uk<br />

Introduction: The bipolar disorder psychiatric GWAS consortium<br />

reported last year a combined analysis <strong>of</strong> 16,737 individuals<br />

including 7,481 individuals affected with bipolar disorder (BD) and<br />

9,250 control individuals. In addition to confirming the association <strong>of</strong><br />

SNPs (single nucleotide polymorphisms) in ankyrin 3 (ANK3), a<br />

genome-wide significant association was found in the region <strong>of</strong><br />

SYNE1 and its brain-expressed splice variant CPG2 (Pgc = 4.3 x 10 -8<br />

OR = 1.15)<br />

Methodology: We have genotyped a total <strong>of</strong> 2592 control individuals<br />

(1579 independent <strong>of</strong> the WTCCC (Wellcome trust case control<br />

consortium) BD GWAS), 1542 Independent BD cases and 1194<br />

unipolar recurrent major depression cases (UP) in the SNP rs9371601<br />

using the Sequenom iPLEX Gold technology.<br />

Results: An association was seen with rs9371601 in SYNE1 with the<br />

same risk in both the independent BD case control sample (1542 BD<br />

and 1579 controls) with a Trend P value <strong>of</strong> 0.008, OR = 1.15, and<br />

the UP samples (1194 UP and all controls 2592) Trend P value =<br />

0.02, OR = 1.11.<br />

Conclusions: Our study supports the association <strong>of</strong> SYNE1 in an<br />

independent BD sample. We also found that the risk allele conferred<br />

increased risk for recurrent major depression with similar effect size<br />

to bipolar disorder.


OPS8.3 GWAS ANALYSIS ACROSS FIVE PSYCHIATRIC<br />

ILLNESSES: RESULTS OF A COMBINED DATASET<br />

POWER OF 56,867 INDIVIDUALS<br />

S. Ripke*(1,2,3), B. Neale(1,2,3)<br />

1. MGH 2. Broadinstitute 3. PGC - CrossDisorderGroup<br />

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

Introduction: One <strong>of</strong> the primary goals <strong>of</strong> the <strong>Psychiatric</strong> GWAS<br />

Consortium (PGC) was to explore the genetic overlap across<br />

psychiatric phenotypes. Specifically, we began with 5 core<br />

psychiatric illnesses: autism (AUT), attention deficit/hyperactivity<br />

disorder (ADHD), bipolar disorder (BPD), major depression disorder<br />

(MDD), and schizophrenia (SCZ). We amassed an unprecedented<br />

sample size with power equivalent to 56,867 independent individuals<br />

<strong>of</strong> European ancestry (N = 31,035 cases / 25,832 controls), derived<br />

from five distinct diseases: MDD (9,229 / 7,347), BIP (6,988 /<br />

4,859), SCZ (9,372 / 7,815), AUT (3,499 / 3,864), ADD (1,947 /<br />

1,947). For AUT and ADHD, the majority <strong>of</strong> samples are familybased,<br />

but we have represented the effective case-control sample size.<br />

Methodology: After technical and genetic quality control and<br />

successive imputation using Hapmap - Phase 3 as reference a<br />

common dataset <strong>of</strong> 56,867 individuls with dosages on 1.3 million<br />

SNPs was obtained. We conducted two primary analyses to assess<br />

genetic overlap across diseases. To identify sharing at a composite<br />

level, we applied polygene-analysis as described by the ISC 1 . After<br />

demonstrating significant sharing across these phenotypes, we then<br />

proceeded to conduct the primary single locus meta-analysis using a<br />

SE-weighted approach combining evidence from each <strong>of</strong> the distinct<br />

diseases. Beyond these two primary analyses, we conducted a range<br />

<strong>of</strong> post hoc tests to dissect the top signals and understand the<br />

structure <strong>of</strong> the risk for each <strong>of</strong> the individual diseases. We have also<br />

endeavored to identify a replication set, even though identifying such<br />

a cohort that reflects the construction <strong>of</strong> our initial set is challenging.<br />

1 International Schizophrenia Consortium: Common polygenic<br />

variation contributes to risk <strong>of</strong> schizophrenia and bipolar<br />

disorder.Nature. 2009 Aug 6460(7256):748-52.<br />

Results: Here we confirmed multiple previously described genes<br />

from either single-disease studies or previously performed crossdisorder<br />

studies. Specifically, we identify genome-wide significant pvalues<br />

within ITIH3/4 (3p21), MIR-137 (1p21), MHC<br />

(6p21), CACNA1C (12p13), CACNB2 (10p12), CNNM2 (10p24)<br />

and TCF4 (18q21). Many genes within regions <strong>of</strong> suggestive pvalues<br />

are under further investigation. Some results <strong>of</strong> the mentioned<br />

secondary analyses will also be presented here.<br />

Conclusions: Cross-disorder analyses are providing promising<br />

results for the understanding <strong>of</strong> psychiatric illness. Such work,<br />

however, is only successful in the face <strong>of</strong> careful combination <strong>of</strong><br />

datasets. To achieve this the Cross-Disorder-Subgroup <strong>of</strong> the PGC<br />

has identified and solved many <strong>of</strong> these issues.<br />

60<br />

OPS8.4 FIVE NOVEL SUSCEPTIBILITY GENES<br />

IDENTIFIED FOR ALZHEIMER'S DISEASE<br />

D. Harold*, P. Hollingworth, R. Sims, A. Gerrish, J. Chapman, M.<br />

Owen, J. Williams, G. Consortia<br />

Cardiff University<br />

*harolddh@cardiff.ac.uk<br />

Introduction: In 2009 we published a genome-wide association<br />

study (GWAS) <strong>of</strong> Alzheimer's disease (AD) in which we identified<br />

two new genome-wide significant susceptibility loci: CLU and<br />

PICALM. However we also observed more variants with P< 1×10 -5<br />

than expected by chance. These included SNPs at CR1 and BIN1, loci<br />

which have subsequently shown genome-wide significant association<br />

with AD. In an attempt to identify new common susceptibility<br />

variants for AD we have undertaken a three-stage association study<br />

comprising 19,870 cases and 39,846 controls. We also genotyped<br />

these samples at loci showing suggestive evidence for association in<br />

the American Alzheimer's Disease Genetic Consortium (ADGC)<br />

GWAS.<br />

Methodology: In Stage 1, we performed a meta-analysis <strong>of</strong> four AD<br />

GWAS datasets (6,688 cases and 13,685 controls). SNPs showing<br />

association at P


OPS8.5 IDENTIFICATION OF COMMON VARIANTS<br />

INFLUENCING PROTEIN ABUNDANCE LEVELS IN<br />

ALZHEIMER’S DISEASE<br />

A. Lourdusamy*(1), K. Lunnon(1), P. Proitsi(2), J. Powell(2), A.<br />

Stewart(3), S. Williams(3), S. Lovestone(1,2), R. Dobson(1)<br />

1. NIHR Biomedical Research Centre for Mental Health, South<br />

London and Maudsley NHS Foundation Trust & Institute <strong>of</strong><br />

Psychiatry, Kings College London 2. MRC Centre for<br />

Neurodegeneration, King's College London, De Crespigny Park 3.<br />

SomaLogic, Inc. 2945 Wilderness Place, 80301<br />

*anbarasu.lourdusamy@kcl.ac.uk<br />

Introduction: Alzheimer’s disease (AD) is a common<br />

neurogenerative disorder with a substantial genetic component.<br />

Recent genome-wide association studies (GWAS) have identified<br />

many gene variants that alter the risk <strong>of</strong> AD but the effects <strong>of</strong> these<br />

variants are likely to be small 1,2,3 . Plasma protein abundance levels in<br />

AD may be biologically relevant endophenotypes and the analysis <strong>of</strong><br />

genetic variants that influence protein levels may provide a<br />

complimentary approach to GWAS leading to a greater<br />

understanding <strong>of</strong> disease mechanisms.<br />

Methodology: We measured protein abundance levels <strong>of</strong> 822<br />

proteins in the plasma <strong>of</strong> 303 subjects from AddNeuroMed study<br />

comprising cases, normal elderly controls and well as those suffering<br />

mild cognitive impairment (MCI) approximately half <strong>of</strong> which<br />

convert to AD within 2 years. Using the genotypes from these<br />

subjects, we tested both the main SNP effects (genetic) and<br />

diagnosis-SNP interaction (diagnostic) effects on the association <strong>of</strong><br />

486,402 SNPs with protein abundance levels as quantitative traits.<br />

Results: We identified many local (cis) genetic effects including<br />

SNPs in or near the SIGLEC9 (p = 7.23 x 10 -93) , FCGR2A (p = 1.50<br />

x 10 -88 ), PDGFRB (p = 8.99 x 10 -79 ), IL6R (p = 2.62 x 10 -48 ), and<br />

TNC (p = 4.23 x 10 -40 ) genes. Interestingly, we identified several<br />

distant (trans) genetic effects <strong>of</strong> the NPS gene, 9 SNPs in or near<br />

NPS are associated with different proteins, the most significant<br />

association is with PSMA1 (p = 1.69 x 10 -13 ), and two SNPs <strong>of</strong> ABO<br />

gene associated with the protein levels <strong>of</strong> SELE (p = 3.07 x 10 -18)<br />

and CD209 (p = 8.21 x 10 -17 ). Many SNP – protein associations<br />

were identified for the AD diagnostic effects including a SNP from<br />

TIMP3 associated with EPHA5 protein levels (rs137484, p = 9.58 x<br />

10 -18 ), SNP from FTO associated with DMP1 (p = 1.20 x 10 -16 ), and<br />

a SNP in APBB2 associated with MMP10 (rs10003861, p = 2.62 x<br />

10 -12 ).<br />

Conclusions: These results suggest that the use <strong>of</strong> protein abundance<br />

levels as intermediate quantitative traits in GWAS may provide<br />

powerful approach for the identification <strong>of</strong> AD disease loci.<br />

61<br />

ORAL PRESENTATIONS SESSION 9: MIXED TOPICS<br />

OPS9.1 GENES TO BIOLOGY…AND BACK TO<br />

BEHAVIOR: UNDERSTANDING TRAJECTORIES OF RISK<br />

FOR ALCOHOL DEPENDENCE<br />

D. Dick*<br />

Virginia Commonwealth University<br />

*ddick@vcu.edu<br />

Introduction: As the field makes progress in identifying genes<br />

involved in complex psychiatric and substance use outcomes, the<br />

next challenge is to better understand how identified genes contribute<br />

to the manifestation <strong>of</strong> complex clinical disorders. This talk will<br />

present an overview <strong>of</strong> the integration <strong>of</strong> gene identification efforts<br />

with twin studies and longitudinal community-based projects in order<br />

to understand and map the complex pathways from gene to clinical<br />

disorder, with focus on the development <strong>of</strong> alcohol-related problems.<br />

Methodology: Data from the Finnish twin studies, which are<br />

population-based epidemiological samples with assessments<br />

spanning ages 12 to 26, demonstrate the changing influence <strong>of</strong><br />

genetic effects on alcohol use and related phenotypes across<br />

adolescence into young adulthood, and as a function <strong>of</strong> the<br />

environment. We use this information to test hypotheses about the<br />

risk associated with specific, identified genes (emerging from the<br />

Collaborative Study <strong>of</strong> the <strong>Genetics</strong> <strong>of</strong> Alcoholism COGA) using<br />

community-based samples <strong>of</strong> individuals studied longitudinally. To<br />

this end, data will be presented from the Child Development Project,<br />

a community-based study <strong>of</strong> ~500 children followed annually from<br />

age 5-25 the Mobile Youth Study, an on-going community-based<br />

sample <strong>of</strong> children ages 10-18 from high-risk, impoverished<br />

neighborhoods in Mobile, Alabama and the Avon Longitudinal Study<br />

<strong>of</strong> Parents and Children, an epidemiological cohort <strong>of</strong> ~10,000<br />

children enrolled when their mothers were pregnant and assessed<br />

yearly -- prenatally through young adulthood.<br />

Results: We find that genes associated with adult alcohol<br />

dependence in COGA (GABRA2, CHRM2) are associated with<br />

behavior problems earlier in development, and that these associations<br />

are moderated by environmental characteristics. Predisposing genes<br />

show a stronger association with problematic outcomes under<br />

conditions <strong>of</strong> poor parental monitoring and higher peer<br />

deviance. Further, there appear to be different pathways to risk for<br />

alcohol dependence, through a variety <strong>of</strong> impulsive and externalizing<br />

characteristics.<br />

Conclusions: Characterizing the risk associated with identified genes<br />

in population and community-based samples will be necessary in<br />

order to understand how genetic influences result in the eventual<br />

alteration <strong>of</strong> risk for psychiatric and substance use outcomes. This<br />

talk will demonstrate how, through interdisciplinary research, we are<br />

gaining a greater understanding <strong>of</strong> how genetic and environmental<br />

influences contribute to the risk for alcohol related problems, and<br />

how that risk manifests across development.


OPS9.2 EXAMINING THE ASSOCIATION BETWEEN<br />

GPSM1 AND HUMAN ALCOHOL DEPENDENCE USING<br />

ASSOCIATION, BIOINFORMATICS, AND EXPRESSION<br />

STUDIES<br />

A. Adkins*(1,2,4), F. Aliev(1,3,4), O. McMichael(1,4), D.<br />

Thiselton(1,3,4), C. Prescott(5), D. Dick(1,3,4), V.<br />

Vladimirov(1,3,4), J. Bettinger(4,6), S. Bowers(1,3,4), K.<br />

Kendler(1,3,4), B. Riley(1,3,4)<br />

1. Virginia Institute for <strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong> 2. Dept<br />

<strong>of</strong> Human and Molecular <strong>Genetics</strong> 3. Dept <strong>of</strong> Psychiatry 4. Virginia<br />

Commonwealth University 5. Dept <strong>of</strong> Psychology, University <strong>of</strong><br />

Southern California 6. Dept <strong>of</strong> Pharmacology and Toxicology<br />

*adkinsae@vcu.edu<br />

Introduction: Relapse, a hallmark <strong>of</strong> addiction, may result from<br />

neuroadaptations in the brain during chronic drug use resulting in the<br />

brain’s inability to function normally during periods <strong>of</strong> withdrawal.<br />

Identifying genes within these pathways, and the biological basis <strong>of</strong><br />

their action, is <strong>of</strong> critical therapeutic interest. Model organism<br />

studies can manipulate experimental conditions to mimic certain<br />

aspects <strong>of</strong> the process <strong>of</strong> human withdrawal, craving and relapse. Gprotein<br />

signaling modulator 1 (GPSM1) expression in rat nucleus<br />

accumbens (NAc) increases following withdrawal from chronic<br />

ethanol exposure and results in an increased, compulsive motivation<br />

to seek ethanol (Bowers et al., 2008). The carefully constructed<br />

experimental paradigm used in the study suggests results can be<br />

compared to similar human phenotypes <strong>of</strong> craving and motivation to<br />

drink alcohol. We investigated the role <strong>of</strong> GPSM1 in human Alcohol<br />

Dependence (AD) and alcohol-related phenotypes using association<br />

data and expression studies.<br />

Methodology: Tag SNPs for the LD block surrounding GPSM1<br />

(chr9: 138261561-138455700) were selected using HAPMAP and<br />

Haploview. The Irish Affected Sib Pair Study <strong>of</strong> Alcohol<br />

Dependence (IASPSAD) case-control sample (N=562 cases, N=569<br />

controls) was genotyped and association analyses performed using<br />

Haploview and Plink. Data for N=776 AD cases and affected siblings<br />

from the IASPSAD and N=2000 Irish controls will be available<br />

shortly. Standard primers were designed to uniquely amplify all 4<br />

Refseq GPSM1 transcripts and real-time PCR run to assess RNA<br />

levels in commercial RNA tissues (N=26). Human alcoholic (N=41)<br />

and control (N=41) brain samples from the TRC in Sydney, Australia<br />

will be examined for RNA expression <strong>of</strong> the 4 transcripts in the NAc<br />

and 5 other brain regions.<br />

Results: 2 <strong>of</strong> the 6 tag SNPs (rs10781510, rs3124994) were<br />

significantly associated with AD (P=0.01, P=0.003) and DSM-IV AD<br />

symptom 3 (P=0.009, P=0.004). Symptom 3 gauges if the subject<br />

has ever had a drink when they promised they wouldn’t or have drank<br />

more than intended. This phenotype suggests an increased or<br />

uncontrollable motivation to drink (or craving) for alcohol that may<br />

relate to the observed phenotype in rats. To further understand the<br />

mechanism <strong>of</strong> action <strong>of</strong> GPSM1, expression levels <strong>of</strong> RNA were<br />

assessed in multiple tissues. RNA levels <strong>of</strong> 4 different GPSM1<br />

transcripts were measured in a commercial tissue panel. Work<br />

measuring RNA levels <strong>of</strong> the transcripts in the TRC brain samples is<br />

ongoing. C. elegans orthologs <strong>of</strong> GPSM1 will be knocked down and<br />

mutants assessed for alcohol-related phenotypes.<br />

Conclusions: Initial findings show an association between SNPs 5’<br />

to GPSM1 and AD as well as a craving-like phenotype. This supports<br />

studies in rats and suggests a role for GPSM1 in the neuroadaptations<br />

contributing to relapse. Completion <strong>of</strong> RNA expression studies and<br />

work in C. elegans will lead to a better understanding <strong>of</strong> the biology<br />

<strong>of</strong> the system in which GPSM1 operates.<br />

62<br />

OPS9.3 COPY NUMBER VARIATION IN SCHIZOPHRENIA<br />

AND BIPOLAR DISORDER IN A SWEDISH POPULATION<br />

ECIP<br />

S. Bergen*(1,3), C. O'Dushlaine(1,2,3), D. Ruderfer(1,2,3), S.<br />

Akterin(4), J. Moran(3), K. Chambert(3), M. Schalling(5), U.<br />

Ösby(5), M. Landen(4), E. Scolnick(3), P. Lichtenstein(4), C.<br />

Hultman(4), P. Sullivan(6), P. Sklar(1,3,7), S. Purcell(1.2.3)<br />

1. <strong>Psychiatric</strong> and Neurodevelopmental <strong>Genetics</strong> Unit, Massachusetts<br />

General Hospital 2. Analytic and Translational <strong>Genetics</strong> Unit,<br />

Massachusetts General Hospital 3. Stanley Center for <strong>Psychiatric</strong><br />

Research, Broad Institute <strong>of</strong> Harvard and MIT 4. Department <strong>of</strong><br />

Medical Epidemiology and Biostatistics, Karolinska Institutet 5.<br />

Department <strong>of</strong> Molecular Medicine and Surgery, Karolinska Institutet<br />

6. Departments <strong>of</strong> <strong>Genetics</strong>, Psychiatry, and Epidemiology,<br />

University <strong>of</strong> North Carolina at Chapel Hill 7. Division <strong>of</strong> <strong>Psychiatric</strong><br />

Genomics, Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong><br />

Medicine<br />

*sbergen@broadinstitute.org<br />

Introduction: Several large, rare copy number variations (CNVs)<br />

have now been associated with schizophrenia, but a clear picture has<br />

yet to emerge for CNV involvement in bipolar disorder.<br />

Methodology: We investigated CNVs in these disorders in a new<br />

Swedish sample consisting <strong>of</strong> 1,506 individuals with schizophrenia<br />

(SCZ), 834 with bipolar disorder (BD), and 2,089 controls. All<br />

subjects were genotyped using the Affymetrix 6.0 array, and CNVs<br />

were called using Birdsuite.<br />

Results: In keeping with prior reports, deletions were enriched to a<br />

greater extent in the SCZ sample. Deletions observed only once and<br />

those <strong>of</strong> intermediate size, 200-500kb, were more frequent in both<br />

case groups compared to controls (SCZ: p=.013, p=.019; BD: p=.017,<br />

p=.046), while the largest CNVs, >500kb, were significantly enriched<br />

only in the SCZ subjects (p=.0018). Duplications did not show<br />

significant overrepresentation for either case group for any size or<br />

frequency examined. Generally, more CNVs intersected genes in<br />

SCZ than in controls, but this observation had less statistical support<br />

in BD. Many large CNVs previously associated with SCZ and other<br />

psychiatric and neurological conditions were observed in this sample<br />

and, for these regions, duplications were observed more <strong>of</strong>ten than<br />

deletions (N=88 vs. 47). SCZ subjects possessed significantly more<br />

duplications at the 16p11.2 locus (p=.0012), and the 9q12-13 locus<br />

showed similar CNV enrichment in SCZ (p=.0027). Other nominally<br />

significant (p


OPS9.4 INVESTIGATING THE GENETIC BASIS OF<br />

TOURETTE SYNDROME IN EUROPEAN POPULATIONS: A<br />

MULTINATIONAL INITIATIVE<br />

P. Paschou*, J. Karagiannidis, P. Aslanidou, E. Grigoriou, S.<br />

Papasotiriou, V. Stathias,. The TSGeneSEE Initia<br />

Dept. <strong>of</strong> Molecular Biology and <strong>Genetics</strong>, Democritus University <strong>of</strong><br />

Thrace<br />

*ppaschou@mbg.duth.gr<br />

Introduction: Tourette Syndrome (TS) is a neuropsychiatric disorder<br />

<strong>of</strong> childhood onset, that is marked by multiple motor and vocal tics<br />

and high comorbidity rates with obsessive compulsive disorder and<br />

attention deficit and hyperactivity disorder. Although it was once<br />

considered rare, a prevalence <strong>of</strong> 0.4-1% is currently reported. The<br />

pathophysiological pathways leading to the onset <strong>of</strong> symptoms<br />

remain poorly understood. However, it is thought that a complex<br />

genetic background is involved, with multiple genes interacting with<br />

environmental factors in order to lead to the onset <strong>of</strong> symptoms.<br />

Multiple chromosomal loci have so far been implicated in the<br />

etiology <strong>of</strong> the disorder. However, studies have been marked by the<br />

difficulty to consistently replicate original positive findings.<br />

Methodology: Undertaking a multinational effort, we have collected<br />

one <strong>of</strong> the largest samples for the study <strong>of</strong> TS genetics that is<br />

available to date. A total <strong>of</strong> 250 trios (one child affected with TS and<br />

their parents) <strong>of</strong> Greek, Albanian, Italian, Polish, Hungarian, and<br />

German origin were analyzed in order to attempt to replicate the<br />

genetic association with regions that have been previously implicated<br />

in TS etiology. Variation across SLITRK1, DAT1, MAOA, TPH2,<br />

and the 17q25 region was investigated for association with the TS<br />

phenotype. Furthermore, two novel candidates for TS susceptibility,<br />

were also tested in our sample. The transmission test for linkage<br />

disequilibrium was used in order to test for over- or undertransmission<br />

<strong>of</strong> studied alleles to affected individuals. Both single<br />

markers and haplotypic tests were performed.<br />

Results: Our results reveal multiple positive signals <strong>of</strong> genetic<br />

association. Over the past few years, association <strong>of</strong> TS etiology with<br />

the SLITRK1 gene has become a subject <strong>of</strong> intense debate, with<br />

many replication studies being unable to support original positive<br />

findings <strong>of</strong> association with particular variants. Studying variation<br />

across the gene, we show that the SLITRK1 gene may indeed be<br />

involved in TS etiology in European populations, and even at a level<br />

that was previously unappreciated. Meta-analysis <strong>of</strong> our results with<br />

existing datasets, provides strong support for this hypothesis. On the<br />

other hand, genes in the dopaminergic and serotonergic pathways, are<br />

not significantly associated with the disorder in our sample.<br />

Conclusions: Our results provide novel insights into the pathogenesis<br />

<strong>of</strong> TS and support the hypothesis that different genetic factors may be<br />

implicated in TS susceptibility across different populations, even<br />

within Europe.<br />

63<br />

OPS9.5 POTENTIAL BENEFITS, RISKS, AND ETHICAL<br />

APPLICATIONS OF PSYCHIATRIC GENETIC TESTING: A<br />

PSYCHIATRIC GENETIC RESEARCHERS PERSPECTIVE<br />

ECIP<br />

J. Erickson*, M. Cho<br />

Stanford University<br />

*jessdae8@stanford.edu<br />

Introduction: As genetic research rapidly advances, promising to<br />

transform health care services, it is crucial to address the potential<br />

social and ethical implications <strong>of</strong> this innovation. Mental health is<br />

one <strong>of</strong> the fields that will be affected by these advancing<br />

technologies. Although studies have explored potential benefits, risks<br />

and ethical concerns regarding psychiatric genetic testing among<br />

mental health pr<strong>of</strong>essionals and potential users, there has been limited<br />

focus on perspectives on these matters among the group who is<br />

actively generating this knowledge, the psychiatric genetic<br />

researchers. This work presents the results <strong>of</strong> an exploratory study<br />

interviewing world-leading experts in psychiatric genetics research<br />

who are conducting studies in depression, schizophrenia, and bipolar<br />

disorder. Their input provides a needed addition to the ongoing<br />

debate on how to incorporate psychiatric genetics research into<br />

evolving health care settings in a realistic, safe, and ethical manner.<br />

Methodology: Top experts in the field <strong>of</strong> genetics research for<br />

bipolar disorder, major depression and/or schizophrenia were<br />

contacted for a one-hour, audio-recorded interview. Twenty-five<br />

psychiatric genetic researchers from 5 countries were interviewed<br />

using a semi-structured interview guide focusing on participants’<br />

beliefs about benefits, harms and ethical concerns that could result<br />

from their work, and on suggested strategies for the ethical<br />

implementation <strong>of</strong> their research into clinical practice. Interview<br />

transcripts were imported into NVivo 9, and 3 trained researchers<br />

coded the transcripts to assure a high coefficient <strong>of</strong> rater reliability<br />

(Scott’s Pi > .90).<br />

Results: The majority <strong>of</strong> respondents believed that the current state<br />

<strong>of</strong> genetic research for BPD, schizophrenia, or depression is far away<br />

from clinical utility. Three main barriers to the progression <strong>of</strong> this<br />

research were identified as: lack <strong>of</strong> funding, the complexity <strong>of</strong> each<br />

disorder, and the difficulty <strong>of</strong> following psychiatric patients. Subjects<br />

classified the top 3 anticipated benefits from their research as: more<br />

individualized medicine, earlier diagnosis, and de-stigmatization <strong>of</strong><br />

mental illness. Primary concerns voiced by interviewees about ethical<br />

issues that could result from advancements in their research were the<br />

premature commercialization <strong>of</strong> study results, a revival <strong>of</strong> eugenics,<br />

and an increasing demand for prenatal psychiatric genetic testing.<br />

Most interviewees opposed direct-to-consumer genetic testing for<br />

psychiatric disorders and believed that psychiatrists should provide<br />

patients with results. However, all participants agreed that physicians<br />

are currently unprepared to interpret genetic results and suggested<br />

strategies to prepare psychiatrists to use genetics research in a clinical<br />

practice.<br />

Conclusions: This preliminary analysis <strong>of</strong> psychiatric genetic<br />

researchers’ views contributes a unique perspective to the growing<br />

literature <strong>of</strong> stakeholder views on psychiatric genetic testing. Overall,<br />

researchers expressed frustration with the progress <strong>of</strong> psychiatric<br />

genetics research. They identified barriers to the progress <strong>of</strong> their<br />

research and risks and ethical concerns resulting from this type <strong>of</strong><br />

work. However, they emphasized the need to continue with this line<br />

<strong>of</strong> research due to the significant benefits genetics information will<br />

provide. Interviewees also provided insights on the type <strong>of</strong> training<br />

necessary to interpret tests. Being at the forefront <strong>of</strong> psychiatric<br />

genetics research, these experts provided a well-informed and<br />

realistic perspective regarding genetic testing for mental illness.


ORAL PRESENTATIONS SESSION 10: FUNCTIONAL<br />

GENOMICS & MODEL ORGANISMS II<br />

OPS10.1 HUMAN INDUCED PLURIPOTENT STEM CELL-<br />

TECHNOLOGY TO STUDY NEURAL DIFFERENTIATION<br />

ECIP<br />

L. D'Aiuto*(1), A. Watson(1), M. Bamne(1), B. Heath(2), R. Di<br />

Maio(3), G. Raimondi(4), V. Nimgaonkar(1)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Pittsburgh 2. Department<br />

<strong>of</strong> Human <strong>Genetics</strong>, University <strong>of</strong> Pittsburgh 3. Department <strong>of</strong><br />

Neurology, University <strong>of</strong> Pittsburgh 4. Department <strong>of</strong> Immunology,<br />

University <strong>of</strong> Pittsburgh<br />

*daiutol@upmc.edu<br />

Introduction: Neuronal Stem/Progenitor (NS/P) cells derived from<br />

human induced pluripotent stem cells (iPSC) provide an<br />

unprecedented opportunity to study human brain development and<br />

model neurodegenerative and neurodevelopmental diseases. We have<br />

initiated neurodevelopmental studies using human cytomegalovirus<br />

(HCMV) as a tool to perturb normal neural differentiation. HCMV<br />

infects neural stem cells and neuroprogenitor cells localized the in<br />

ventricular and subventricular zones. It is a major cause <strong>of</strong> prenatal<br />

encephalitis and mental retardation. HCMV has been investigated<br />

using neurospheres prepared using forebrain tissues from fetal<br />

abortuses. These models provide important information, but have<br />

obvious limitations. Hence we have investigated CMV effects on<br />

human iPSC-derived NS/P cells<br />

Methodology: We generated iPSCs from adult human fibroblasts<br />

obtained through adult skin biopsies. iPSCs were differentiated into<br />

neurospheres that were expanded as monolayer cultures <strong>of</strong> NS/Ps.<br />

The neurospheres were further differentiated into neurons stainable<br />

with Tuj1, tyrosine hydroxylase, and NR4A2. Functional competency<br />

was confirmed by live imaging <strong>of</strong> intracellular calcium influx. NS/P<br />

cells and neurons were infected with human cytomegalovirus<br />

(HCMV) at multiplicity <strong>of</strong> infection (MOI=3). Cell viability was<br />

assessed by FACS analysis.<br />

Results: Cytopathic effects <strong>of</strong> HCMV were observed on the 10th day<br />

post infection in neuroprogenitor cells. Earlier, the adherence <strong>of</strong> these<br />

cells to the underlying matrix was reduced. Neurons were much more<br />

refractory to infection. Reduced cell density and shortening <strong>of</strong><br />

neuritic processes was only observed at day 15 post-infection. We are<br />

now examining intracellular gene expression.<br />

Conclusions: Human iPS cells can efficiently generate neurospheres,<br />

which can be expanded as monolayer cultures <strong>of</strong> NS/P cells or<br />

differentiated into neurons. iPSC-derived NS/P cells and neurons<br />

<strong>of</strong>fer powerful cellular models to investigate the effects <strong>of</strong><br />

neurotropic viral agents on human neurodevelopment.<br />

64<br />

OPS10.2 SYSTEMS GENETICS IN MOOD DISORDERS:<br />

INTEGRATION OF GENOME-WIDE ASSOCIATION<br />

RESULTS WITH HUMAN BRAIN EXPRESSION DATA<br />

S. Detera-Wadleigh*, N. Akula, F. McMahon<br />

National Institutes <strong>of</strong> Health<br />

*deteras@mail.nih.gov<br />

Introduction: Meta-analytic studies <strong>of</strong> genome-wide association<br />

study (GWAS) data have shown significant association between<br />

major mood disorders and genetic variants in the ANK3, CACNA1C<br />

and PBRM1 gene regions (Ferreira et al. 2008 McMahon et al 2010<br />

Liu et al. 2011). Modest statistical support for association has been<br />

reported also for several genes in a meta-analysis <strong>of</strong> major depression<br />

GWA data (Wray et al. 2010). In addition, a meta-analysis <strong>of</strong> 183<br />

smaller studies that focused on 20 SNPs in 18 candidate genes<br />

supported association in 6 genes. Differential gene expression in<br />

brains from patients would provide an important functional correlate<br />

to these findings. Since genes or their gene products act as<br />

components <strong>of</strong> biological pathways, defects in these molecules would<br />

cause cumulative and extensive system-wide perturbations that<br />

impact on networks <strong>of</strong> interacting pathways, leading to pathology.<br />

Methodology: We assembled a list <strong>of</strong> associated genes ranked by<br />

level <strong>of</strong> support in GWAS and identified those that displayed<br />

differential expression in postmortem brain obtained from cases with<br />

a major mood disorder. We used several microarray-based<br />

transcriptome datasets representing various brain regions from<br />

patients with bipolar disorder or major depression as well as<br />

psychiatrically-healthy controls (The Stanley Neuropathology<br />

Consortium Integrative Database, http://sncid.stanleyresearch.org/).<br />

To capture a glimpse <strong>of</strong> the pathways involved we used genes that<br />

passed the association-expression integration analysis as seed for<br />

network analysis. Networks were generated using the Michigan<br />

Molecular Interaction (MiMI) plugin tool (Gao et al. 2009 Tarcea et<br />

al. 2009) in Cytoscape (http://www.cytoscape.org/).<br />

Results: The most stringent group, including ANK3, CACNA1C,<br />

and PBRM1, formed two small networks. ANK3 and CACNA1C<br />

node clusters were linked through 6 different nodes. (A node refers to<br />

a protein or a gene in a network). PBRM1 formed a separate node<br />

cluster. Functional annotation in DAVID Bioinformatics Resource<br />

(http://david.abcc.ncifcrf.gov/home.jsp, NIAID/NIH) (Denis et al.<br />

2003 Huang et al. 2009) revealed that ANK3 and CACNA1C nodes<br />

were enriched for MAPK and beta adrenergic receptor pathways,<br />

while the PBRM1 cluster was enriched for chromatin remodeling and<br />

transcriptional regulation, consistent with its known function.<br />

Lowering the stringency to include other genes led to the recruitment<br />

<strong>of</strong> signaling pathways for neurotransmitter and hormone receptors,<br />

neurotrophin, ErbB and the cell cycle.<br />

Conclusions: Network analysis <strong>of</strong> genes that are implicated by both<br />

GWAS and differential brain expression is a promising strategy for<br />

identifying functional gene clusters and biological pathways involved<br />

in the genetic risk for mood disorders.


OPS10.3 RNA-SEQ ANALYSIS OF DIFFERENTIATING<br />

HUMAN NEURONS DERIVED FROM INDUCED<br />

PLURIPOTENT STEM CELLS (IPSCS)<br />

M. Lin(1), E. Pedrosa(2), A. Shah(2), D. Zheng(1,3,4),<br />

H. Lachman*(2,5)<br />

1. Department <strong>of</strong> <strong>Genetics</strong> Albert Einstein College <strong>of</strong> Medicine 2.<br />

Department <strong>of</strong> Psychiatry Albert Einstein College <strong>of</strong> Medicine 3.<br />

Department <strong>of</strong> Neurology Albert Einstein College <strong>of</strong> Medicine 4.<br />

Department <strong>of</strong> Neuroscience Albert Einstein College <strong>of</strong> Medicine 5.<br />

Department <strong>of</strong> Medicine Albert Einstein College <strong>of</strong> Medicine<br />

*herb.lachman@einstein.yu.edu<br />

Introduction: The development <strong>of</strong> patient-specific iPSCs for in vitro<br />

disease modeling is a promising approach for studying the molecular<br />

basis <strong>of</strong> SZ and other neuropsychiatric disorders. An especially useful<br />

application <strong>of</strong> this technology is gene expression pr<strong>of</strong>iling using<br />

patient-specific differentiating neurons. In the past, this analysis was<br />

carried out on autopsy samples using microarrays. Live,<br />

differentiating, patient-specific neurons has many advantages over<br />

autopsy specimens and deep sequencing (RNA-Seq) is becoming the<br />

method <strong>of</strong> choice for expression pr<strong>of</strong>iling over microarrays.<br />

Methodology: iPSCs are being developed from controls and patients<br />

with SZ with and without 22q11.2 deletions using standard protocols.<br />

Neuronal differentiation was carried out using a protocol that<br />

primarily yields glutamatergic neurons. RNA was extracted from<br />

iPSCs and a small cluster <strong>of</strong> neurons. cDNA was synthesized and<br />

amplified, and the samples were subjected to paired-end deep<br />

sequencing using an Illumina GA2 next generation sequencing<br />

platform.<br />

Results: Our findings show that early differentiating neurons<br />

undergo an extraordinary series <strong>of</strong> changes in gene expression and in<br />

the generation <strong>of</strong> splicing is<strong>of</strong>orms. More than 2x10 8 reads were<br />

obtained for both iPSCs and day 10 neurons. The expression <strong>of</strong> 9733<br />

genes was significantly altered as the iPSCs differentiated into<br />

neurons (p


OPS10.5 CONVERGENT FUNCTIONAL GENOMICS OF<br />

ANXIETY DISORDERS: TRANSLATIONAL<br />

IDENTIFICATION OF GENES, BIOMARKERS, PATHWAYS<br />

AND MECHANISMS<br />

A. Niculescu*(1), H. Le-Niculescu(1), Y. Balaraman(1), S. Patel(1),<br />

M. Ayalew(1), J. Gupta(1), R. Kuczenski(2), A. Shekhar(1), M.<br />

Geyer(2), N. Schork(3)<br />

1. Indiana University School <strong>of</strong> Medicine 2. UC San Diego 3. Scripps<br />

Research Institute<br />

*anicules@iupui.edu<br />

Introduction: Anxiety disorders are prevalent and disabling yet<br />

understudied from a genetic standpoint, compared to other major<br />

psychiatric disorders such as bipolar disorder and schizophrenia. The<br />

fact that they are more common, diverse, and perceived as embedded<br />

in normal life may explain this relative oversight. In addition, as for<br />

other psychiatric disorders, there are technical challenges related to<br />

the identification and validation <strong>of</strong> candidate genes and peripheral<br />

biomarkers. Human studies, particularly genetic ones, are susceptible<br />

to the issue <strong>of</strong> being underpowered, due to genetic heterogeneity, the<br />

effect <strong>of</strong> variable environmental exposure on gene expression, and<br />

difficulty <strong>of</strong> accrual <strong>of</strong> large well phenotyped samples. Animal model<br />

gene expression studies, in a genetically homogeneous and<br />

experimentally tractable setting, can avoid artifacts and provide<br />

sensitivity <strong>of</strong> detection. Subsequent translational integration <strong>of</strong> the<br />

animal model datasets with human genetic and gene expression<br />

datasets can ensure cross-validatory power and specificity for illness.<br />

Methodology: We have used a pharmacogenomic mouse model<br />

(involving treatments with an anxiogenic drug- yohimbine, and an<br />

anti-anxiety drug—diazepam) as a discovery engine for identification<br />

<strong>of</strong> anxiety candidate genes as well as potential blood<br />

biomarkers. Gene expression changes in key brain regions for<br />

anxiety (prefrontal cortex, amygdala, hippocampus) and blood were<br />

analyzed using a Convergent Functional Genomics (CFG) approach,<br />

which translationally integrates our new data with published human<br />

and animal model data, as a translational strategy <strong>of</strong> cross-matching<br />

and prioritizing findings.<br />

Results: Our work identifies top candidate genes(such as FOS,<br />

GABBR1, NR4A2, DRD1, ADORA2A, QKI, RGS2, PTGDS,<br />

HSPA1B, DYNLL2, CCKBR and DBP), brain-blood biomarkers<br />

(such as FOS, QKI and HSPA1B), pathways (such as cAMP<br />

signaling) and mechanisms for anxiety disorders -notably signal<br />

transduction and reactivity to environment.<br />

Conclusions: Overall, this work complements our previous similar<br />

work (on bipolar mood disorders and schizophrenia) conducted over<br />

the last decade. It concludes our programmatic first pass mapping <strong>of</strong><br />

the genomic landscape <strong>of</strong> the triad <strong>of</strong> major psychiatric disorders<br />

using CFG, and permitted us to uncover the significant genetic<br />

overlap between anxiety and these other major psychiatric disorders,<br />

notably the under-appreciated overlap with schizophrenia. PDE10A,<br />

TAC1 and other genes uncovered by our work provide a molecular<br />

basis for the frequently observed clinical co-morbidity and<br />

interdependence between anxiety and other major psychiatric<br />

disorders, and suggest schizo-anxiety as a possible new nosological<br />

entity.<br />

66


POSTER SESSION I<br />

ABSTRACTS<br />

67


GENOMICS (GWAS, SEQUENCING)<br />

PP1 THE PI3K-AKT-GSK3 SIGNALING PATHWAY AND<br />

OVERLAPS BETWEEN SCHIZOPHRENIA AND BIPOLAR<br />

DISORDER<br />

F. Karege(1), A. Meary(2), N. Perroud(1), A. Carrard(1), F.<br />

Schur<strong>of</strong>f(2), A. Malafosse*(1)<br />

1. Geneva University Hospitals 2. Hopital Chenevrier-Mondor<br />

*alain.malafosse@hcuge.ch<br />

Introduction: Schizophrenia (SCZ) and Bipolar disorder (BPD) are<br />

considered as different diseases in the diagnostic manuals. Genetic<br />

studies suggest however, an overlap <strong>of</strong> these disorders. The PI3K-<br />

Akt-GSK3ß signaling pathway is involved in cellular process such as<br />

neurodevelopment. Its dysfunction could impact SCZ and BPD. We<br />

aimed to investigate the PI3KC, PKB/AKT and GSK3 gene variants<br />

and protein levels both in SCZ and BPD.<br />

Methodology: This case-control study included 830 subjects <strong>of</strong><br />

European ancestry: 400 unrelated SCZ 230 unrelated BPD, and 200<br />

unrelated healthy controls. Molecular genotyping <strong>of</strong> the following<br />

genes was performed: Akt1, PI3KC3 and GSK3. SNPs and other<br />

genetic variants were assessed. Additionally, postmortem brain<br />

protein levels were assayed in deceased SCZ, BPD and matchedcontrols.<br />

Lastly, an epigenetic study <strong>of</strong> DNA methylation on CpG<br />

islands was assessed in Akt1 and GSK3 genes.<br />

Results: For the Akt1 gene, the analysis identified a significant<br />

global distortion in SCZ (P=0.0026) and in BPD (p=0.046). A<br />

sliding window procedure showed a 5-SNP haplotype (TCGAG) to<br />

be associated with SCZ (p=0.0001) and BPD (p=0.0041). Other<br />

multi-SNP haplotypes also showed significant association with both<br />

diseases. When SCZ and BPD were combined, the global p-value<br />

improved (p=0.00001). For the GSK3ß gene, one SNP (rs6438552)<br />

was found associated with the risk <strong>of</strong> SCZ (p=0.0008) and weakly<br />

with BPD (p=0.06). With respect to controls, the PI3KC3 gene also<br />

showed an association with one SNP (p=0.01) in SCZ and with a 2variants<br />

(p=0.004) and a 3-variants (p=0.001) haplotype in both<br />

disorders combined. In the epigenetic study, differential methylation<br />

was found in CpG islands <strong>of</strong> GSK3ß genes from SCZ. On the basis <strong>of</strong><br />

selected Akt1 genotype, significant difference in protein expression<br />

emerged between risk allele carriers and non carriers subjects<br />

(p=0.02).<br />

Conclusions: A number <strong>of</strong> genetic variants was found in both SCZ<br />

and BPD. Furthermore, AKT and PI3KC protein levels were<br />

decreased in brain (PFC) <strong>of</strong> SCZ and BPD. Preliminary data also<br />

indicated a different GSK3ß gene methylation status in SCZ patients<br />

compared to control subjects. This convergent evidence suggest an<br />

alteration in the PI3K-AKT-GSK3 signaling pathway in both SCZ<br />

and BPD. These findings support the role <strong>of</strong> this pathway in the<br />

etiology <strong>of</strong> SCZ and BPD. The study is consistent with the overlap<br />

model <strong>of</strong> these diseases.<br />

68<br />

PP2 A GENETIC ASSOCIATION STUDY AND COGNITIVE<br />

FUNCTION ANALYSIS OF THE NEUROPILIN AND<br />

TOLLOID-LIKE 1 GENE AND SCHIZOPHRENIA IN<br />

JAPANESE POPULATION<br />

M. Banno*(1), B. Aleksic(1,2), T. Koide(1), T. Kikuchi(1,3), K.<br />

Kohmura(1), Y. Adachi(1), M. Ikeda(2), T. Inada(4), T. Iidaka(1), N.<br />

Iwata(2), N. Ozaki(1)<br />

1. Department <strong>of</strong> Psychiatry, Nagoya University Graduate School <strong>of</strong><br />

Medicine 2. Department <strong>of</strong> Psychiatry, Fujita Health University<br />

School <strong>of</strong> Medicine 3. Matsuzaki Hospital 4. Department <strong>of</strong><br />

Psychiatry, Seiwa Hospital, Institute <strong>of</strong> Neuropsychiatry<br />

*solvency@med.nagoya-u.ac.jp<br />

Introduction: One <strong>of</strong> the major hypotheses for the pathophysiology<br />

<strong>of</strong> schizophrenia is that numerous risk factors converge on the<br />

hyp<strong>of</strong>unction <strong>of</strong> the neurotransmitter glutamate resulting from the<br />

antagonism <strong>of</strong> the N-methyl-D-aspartate (NMDA) receptor. This<br />

hypothesis arises from observations that the NMDA receptor<br />

antagonist phencyclidine can produce a psychotic condition similar to<br />

the positive, negative, and cognitive symptoms <strong>of</strong> schizophrenia.<br />

Methodology: We examined the association between schizophrenia<br />

and neuropilin and tolloid-like 1 gene (NETO1), a postsynaptic<br />

protein associated with the NMDAR complex. We selected 7 single<br />

nucleotide polymorphisms (SNPs) in NETO1 region, which showed<br />

nominal evidence for statistical significance based on the first GWAS<br />

<strong>of</strong> schizophrenia conducted in Japanese population (JGWAS)<br />

(case560, control548). Additionally, we included a SNP that was<br />

shown to be associated with schizophrenia in the previous study (Shi,<br />

J et al, Nature, 2009). We genotyped the 8 SNPs in the independent<br />

replication sample comprised <strong>of</strong> 963 patients and 919 controls.To<br />

complement genetic association findings we evaluated the effect <strong>of</strong> 8<br />

SNPs on the cognitive performance measured by Continuous<br />

Performance Test (CPT) and Wisconsin Card Sorting Test (WCST)<br />

(case118, control104).<br />

Results: There was no genetic association signal detected in the<br />

independent replication sample after Bonferroni correction was<br />

applied (lowest P=0.26). Furthermore, we did not detect any <strong>of</strong> the<br />

common SNPs selected for our association study could affect scores<br />

in CPT or WCST.<br />

Conclusions: We conclude that common SNPs within NETO1 may<br />

not increase the risk for schizophrenia in the Japanese subjects.<br />

However, we cannot exclude the possibility that common SNPs <strong>of</strong><br />

smaller effect size or rare variants in NETO1 may increase the<br />

susceptibility to schizophrenia.


PP3 AN ASSOCIATION STUDY OF TCF4<br />

POLYMORPHISMS WITH SCHIZOPHRENIA IN JAPANESE<br />

POPULATION<br />

T. Kaneko*(1), T. Kanazawa(1), S. Kawashige(2), A. Tsutsumi(1),<br />

J. Koh(1), H. Yoneda(1)<br />

1. Osaka Medical College 2. Ozone Hospital<br />

*psy078@poh.osaka-med.ac.jp<br />

Introduction: TCF4 gene (transcription factor 4, on 18q21.2) is a<br />

member <strong>of</strong> the basic helix-loop-helix (bHLH) transcription factor<br />

family. TCF4 regulates gene expression by hetero-dimerization with<br />

other transcription factors in the brain during development, while it<br />

regulates the development <strong>of</strong> lymphoid progenitors to the B- and Tcell<br />

lineages and plasmacytoid dendritic cell (PDC) differentiation.<br />

Mutation inTCF4 cause an autosomal-dominant neurodevelopmental<br />

disorder, Pitt-Hopkins Syndrome. Recently, meta-analysis<br />

<strong>of</strong> GWAS reported rs9960767 on intron 4 <strong>of</strong> TCF4 was associated<br />

with schizophrenia in Caucasians (p=4.1×10 -9 , Stefansson et<br />

al., Nature, 2009). In Asian population, only rs2958182 in the<br />

TCF4 gene was reported an association with schizophrenia in Han<br />

Chinese (p=3.64×10 -6 , Li et al., Biological Psychiatry, 2010).<br />

Methodology: In the current study, we analyzed two SNPs,<br />

rs2958182 and rs2924336 in 200 schizophrenia patients and sex- and<br />

age-matched 200 normal control subjects, because rs9960767 in<br />

Stefansson's report was not polymorphic in Japanese Hapmap data.<br />

Selected two SNPs is located near rs9960767.<br />

Results: The detailed result is shown at the congress.<br />

69<br />

PP4 TRANSCRIPTOMIC ANALYSIS OF POSTMORTEM<br />

BRAIN IDENTIFIES DYSREGULATED SPLICE ISOFORMS<br />

OF NOVEL CANDIDATE GENES FOR SCHIZOPHRENIA<br />

O. Cohen*(1), S. Bialosuknia(1), M. Tsuang(2), F. Middleton (1), S.<br />

Faraone(1), S. Glatt(1)<br />

1. <strong>Psychiatric</strong> Genetic Epidemiology & Neurobiology Laboratory<br />

(PsychGENe Lab) Departments <strong>of</strong> Psychiatry and Behavioral<br />

Sciences SUNY Upstate Medical University 2. Center for Behavioral<br />

Genomics Department <strong>of</strong> Psychiatry University <strong>of</strong> California, San<br />

Diego<br />

*coheno@upstate.edu<br />

Introduction: The diverse expression <strong>of</strong> certain alternatively spliced<br />

transcript variants contributes to the fine-tuning that controls<br />

neurodevelopment. Alternative splicing, however, can be altered by<br />

many different environmental and genetic variables and therefore,<br />

result in unbalanced circuitry. Identifying dysregulated splice variants<br />

in schizophrenia, which is known to have neurodevelopmental and<br />

genetic origins, may shed light on the developmental mechanism(s)<br />

<strong>of</strong> this disorder.<br />

Methodology: Postmortem brain tissue samples were obtained from<br />

20 individuals with schizophrenia (SZ) and 20 neuropsychiatrically<br />

normal comparison subjects included in the Harvard Brain Tissue<br />

Resource Center. Gray matter tissue samples were extracted from<br />

two brain regions implicated in the disorder: Brodmann area 10<br />

(BA10) and caudate head. Tissue from four subjects per group was<br />

lysed, and total mRNA in each sample was isolated, reverse<br />

transcribed, and hybridized to Affymetrix Human Gene 1.0 ST arrays<br />

(exon array) to identify genes with dysregulated exons. Dysregulation<br />

<strong>of</strong> exons identified on the exon array was confirmed by qPCR in each<br />

<strong>of</strong> the eight tested subjects. Subsequent to confirmation, exons were<br />

tested for replication in the remaining sample <strong>of</strong> 16 SZ and 16 (12<br />

caudate) normal control tissues using the same primer sets.<br />

Results: The exon array revealed numerous dysregulated exons and<br />

genes <strong>of</strong> interest. Based on their pr<strong>of</strong>iles <strong>of</strong> exon dysregulation,<br />

regional distribution, and involvement in neurodevelopment, two<br />

genes were selected for further analysis, including the enabled<br />

homolog <strong>of</strong> Drosophila (ENAH) and Kelch-like protein 5 (KLHL5).<br />

Both genes are associated with cytoskeleton function, which<br />

orchestrates dynamic aspects <strong>of</strong> cell activity such as axon guidance,<br />

and cell migration, and are therefore attractive candidates in the study<br />

<strong>of</strong> the pathological development that leads to SZ. In BA10 <strong>of</strong> SZ<br />

brains, ENAH showed significantly lower exon 12 expression<br />

(suggestive <strong>of</strong> its exclusion by alternative splicing) compared to<br />

control brains by microarray however, ENAH exons were not<br />

dysregulated in SZ in the caudate head. Analysis by qPCR confirmed<br />

the exon-array results in both brain regions. Results were replicated<br />

in BA10 while, contrary to the discovery sample, a significant<br />

difference in exon 12 expression was observed in caudate head in the<br />

larger replication sample. Exon 10 was found to be included in<br />

KLHL5 is<strong>of</strong>orms more <strong>of</strong>ten in SZ than control in both BA10 and<br />

caudate head. Confirmation and replication by qPCR showed the<br />

same trend but were not significant in either brain region.<br />

Conclusions: The differential exclusion or inclusion <strong>of</strong> specific<br />

exons may result in different functions <strong>of</strong> the translated proteins. A<br />

disturbance in the alternative splicing mechanism due to mutations in<br />

the genes, malfunctioning alternative splicing regulators, or<br />

environmental cues, may cause an imbalanced expression <strong>of</strong> is<strong>of</strong>orms<br />

that, in turn, might contribute to the development <strong>of</strong> SZ.


PP5 ASSOCIATION STUDY AND EXPRESSION ANALYSIS<br />

BETWEEN MAGI2 AND SCHIZOPHRENIA<br />

T. Koide*(1,9), B. Aleksic(1,9), A. Yoshimi(2), I. Kushima(1,9), Y.<br />

Nakamura(1,9), M. Ikeda(3), K. Ohi(4,9), Y. Yasuda(4,9), R.<br />

Hashimoto(4,5,9), T. Inada(6), U. Hiroshi(7), M. Suzuki(8), M.<br />

Takeda(4,5), N. Iwata(3,9), N. Ozaki(1,9)<br />

1. Departments <strong>of</strong> Psychiatry, Nagoya University Graduate School <strong>of</strong><br />

Medicine 2. Departments <strong>of</strong> Neuropsychopharmacology and Hospital<br />

Pharmacy 3. Department <strong>of</strong> Psychiatry, Fujita Health University<br />

School <strong>of</strong> Medicine 4. Department <strong>of</strong> Psychiatry, Osaka University<br />

Graduate School <strong>of</strong> Medicine 5. Molecular Research Center for<br />

Children's Mental Development, United Graduate School <strong>of</strong> Child<br />

Development, Osaka University, Kanazawa University, and<br />

Hamamatsu University Graduate School <strong>of</strong> Medicine 6. Seiwa<br />

Hospital, Institute <strong>of</strong> Neuropsychiatry 7. Ujike nishiguchi Clinic 8.<br />

Department <strong>of</strong> Neuropsychiatry, University <strong>of</strong> Toyama 9. CREST,<br />

Japan Science and Technology Agency<br />

*t-koide@med.nagoya-u.ac.jp<br />

Introduction: Schizophrenia is a complex, heritable psychiatric<br />

disorder, affecting approximately 1% <strong>of</strong> the general population.<br />

Although the heritability <strong>of</strong> schizophrenia was estimated to be about<br />

64%, the genetic basis <strong>of</strong> schizophrenia is unclear. Recently, genes<br />

related to schizophrenia or genetic variants that may modulate risk<br />

for this disease have been identified using both linkage and candidate<br />

based or whole genome association studies. The first schizophrenia<br />

GWAS in the Japanese population known as homogeneous (JGWAS)<br />

implied many associations in several loci, including the same SNPs<br />

already reported in schizophrenia GWAS in the European population.<br />

However, the sample size <strong>of</strong> JGWAS was relatively smaller (case<br />

575 and control 564). Thus, the results <strong>of</strong> JGWAS should be<br />

replicated in a larger independent population. From the possible<br />

association signals <strong>of</strong> JGWAS, we selected membrane-associated<br />

guanylate kinase, WW and PDZ domain-containing 2 gene (MAGI2)<br />

region in terms <strong>of</strong> its biological evidence and strong intensity <strong>of</strong><br />

association signals. MAGI2 is located in 7q21 and ranging over about<br />

1.44 mega base pairs. Linkage disequilibrium structure within the<br />

MAGI2 locus is coarse with the multiple recombination hot spots.<br />

MAGI2 interacts with ERBB4, which is related to NRG1 mediated<br />

developmental signaling pathways. There was the only one<br />

association study focusing on MAGI2 due to the gene length and LD<br />

pattern. Therefore, in order to explore the association between<br />

MAGI2 and schizophrenia, we conducted association study based on<br />

locus signals implied in JGWAS within MAGI2 region and<br />

expression analysis.<br />

Methodology: Based on the result <strong>of</strong> first schizophrenia GWAS<br />

(JGWAS) in Japanese population, known as homogeneous, we<br />

selected 5 SNPs based on p-value from MAGI2 region and carried out<br />

the association study using an independent large Japanese sample set<br />

(case 1624, control 1621). To support our findings on genetic<br />

association, we performed expression analysis <strong>of</strong> MAGI2 at mRNA<br />

level in the sample consisted <strong>of</strong> lymphoblastoid cell lines derived<br />

from 30 schizophrenic patients and the same number <strong>of</strong> age and<br />

gender matched healthy controls.<br />

Results: In the association study, using independent replication<br />

sample we found trend for genetic association (best p=0.06), and<br />

when the data are combined across two sample sets evidence for<br />

association had strengthen (p=0.0033) by meta analysis using<br />

PLINK. In the expression study, we couldn't observe significant<br />

difference in expression between cases and controls.<br />

Conclusions: Our results suggested possibility <strong>of</strong> genetic association<br />

between common SNP within MAGI2 locus and schizophrenia in the<br />

Japanese population. However, further studies involving larger<br />

samples are needed.<br />

70<br />

PP6 WHOLE EXOME SEQUENCING REVEALS THE<br />

GENETIC BASIS OF A CASE OF IDIOPATHIC HEMOLYTIC<br />

ANEMIA AND SUGGESTS CANDIDATE RARE VARIANTS<br />

FOR ADHD IN A UTAH PEDIGREE<br />

G. Lyon*(1,3), T. Jiang(2), R. Robison(3), H. Hakonarson(1), M.<br />

Yandell(3), R. VanWijk(5), L. Yang(2), P. Zhang(2), R. Wu(2), Y.<br />

Liu(2), X. Yang(2), J. Xing(3), B. Moore(3), J. Glessner(1), W.<br />

McMahon(3), K. Wang(4)<br />

1. Center for Applied Genomics, CHOP 2. BGI-Shenzhen 3.<br />

University <strong>of</strong> Utah 4. USC 5. University Medical Center Utrecht<br />

*lyong1@email.chop.edu<br />

Introduction: Exome sequencing has identified the causes <strong>of</strong> several<br />

Mendelian diseases, although it has rarely been used in a clinical<br />

setting to diagnose the genetic cause <strong>of</strong> a disorder in a single patient.<br />

Methodology: Exome capture was carried out using a commercially<br />

available Agilent SureSelect in solution method as per the<br />

manufacturer guidelines. Each captured library was loaded on the<br />

Illumina GAIIx platform, and GA sequencing was performed for<br />

each captured library independently to ensure each sample had at<br />

least 20x coverage on the exome. Variants are functionally annotated<br />

using the ANNOVAR s<strong>of</strong>tware using a SIFT cut<strong>of</strong>f <strong>of</strong> 0.05 and the<br />

allele frequency measure from the 1000 Genomes Project.<br />

Additionally, the variants reduction pipeline, with some differences<br />

for recessive model and dominant model, was utilized in trimming<br />

down the potential functional candidates.<br />

Results: We performed exome sequencing on a pedigree with a<br />

father and two sons with attention deficit/hyperactivity disorder<br />

(ADHD), to identify candidate genes for this complex disease. Over<br />

the course <strong>of</strong> the study, one subject was discovered to have an<br />

idiopathic hemolytic anemia (IHA) with subsequent spleen removal,<br />

which was suspected to be genetic in origin. Analysis <strong>of</strong> his exome<br />

readily identified him to be a compound heterozygote for two rare<br />

non-synonymous mutations in PKLR, likely causing the IHA. We<br />

confirmed the deficiency by functional biochemical testing,<br />

consistent with a diagnosis <strong>of</strong> red blood cell pyruvate kinase<br />

deficiency. Using a new s<strong>of</strong>tware program (VAAST) developed by<br />

Mark Yandell at the University <strong>of</strong> Utah, we prioritized many<br />

candidate variants that might be causative for the ADHD in this<br />

family. We are studying these variants in case-control cohorts and<br />

other pedigrees, along with conducting functional studies.<br />

Conclusions: We present our results explaining the cause <strong>of</strong><br />

idiopathic hemolytic anemia, along with many possible candidate<br />

rare variants that could be causative for ADHD. Our study has<br />

clinical and ethical implications for exome sequencing in a research<br />

setting. How to handle unrelated findings <strong>of</strong> clinical significance in<br />

the context <strong>of</strong> originally planned complex disease research remains a<br />

largely uncharted area for clinicians and researchers. We specifically<br />

discuss our experience with this dilemma and <strong>of</strong>fer our views on how<br />

to make the most out <strong>of</strong> exome sequencing data.


PP7 POSTMORTEM HIPPOCAMPAL GABAERGIC GENE<br />

EXPRESSION IN ALCOHOLICS AND COCAINE ADDICTS:<br />

TRANSLATIONAL FINDINGS IN ALCOHOL NAIVE P AND<br />

NP RATS<br />

M. Enoch*(1), Z. Zhou(1), M. Kimura(1), D. Mash(2), Q. Yuan(1),<br />

D. Goldman(1)<br />

1. NIAAA, NIH 2. University <strong>of</strong> Miami School <strong>of</strong> Medicine<br />

*maenoch@niaaa.nih.gov<br />

Introduction: Addiction to alcohol and cocaine results in neuronal<br />

adaptation. Equally, neurobiological variation, for example in the<br />

GABAergic system that is strongly implicated in the acute and<br />

chronic effects <strong>of</strong> alcohol, may predispose to the development <strong>of</strong><br />

addiction. In this study we attempted to distinguish between trait and<br />

state effects using a translational approach by comparing the relative<br />

expression <strong>of</strong> GABAergic genes in postmortem hippocampus <strong>of</strong><br />

alcoholics, cocaine addicts and controls and in postmortem<br />

hippocampus <strong>of</strong> alcohol naïve P and NP rats, selectively bred for<br />

extremes <strong>of</strong> alcohol seeking behavior.<br />

Methodology: Samples <strong>of</strong> postmortem hippocampus were obtained<br />

from the University<br />

<strong>of</strong> Miami Brain Endowment Bank from 8<br />

alcoholics, 8 cocaine addicts and 8 controls, all men. There were no<br />

group differences in mean age, ethnicity or postmortem interval.<br />

Total RNA was extracted, mRNA was isolated and high-throughput<br />

parallel sequencing was performed across the whole genome using<br />

the Illumina Genome Analyzer. Approximately 346 million bases<br />

were mapped per sample. The read counts were then log2<br />

transformed and normalized using quantile normalization before<br />

group comparisons were performed. Selectively bred alcoholpreferring<br />

(P) and non-preferring (NP) male rats, 8 in each group, all<br />

sacrificed at 90 days, were obtained from the Indiana University<br />

School <strong>of</strong> Medicine. The identical RNA Seq method was used to<br />

perform whole genome sequencing <strong>of</strong> mRNA transcripts. Following<br />

global analyses <strong>of</strong> the 16,008 genes in humans and 11,406 genes in<br />

rats that had identifiable transcripts in the hippocampus we then<br />

focused on 25 candidate genes that encode proteins implicated in<br />

GABA synthesis, metabolism, synaptic transmission and re-uptake.<br />

Results: In human hippocampus there were both common and<br />

specific effects in GABAergic pathway genes. In both cocaine<br />

addicts and alcoholics, genes encoding GAT1 (GABA transporter)<br />

and GABBR1 (presynaptic autoreceptor) were down-regulated,<br />

potentially resulting in increased GABA in the synaptic cleft.<br />

GABRG2, encoding the gamma2 subunit, a component <strong>of</strong> nearly all<br />

GABA A postsynaptic receptors, was downregulated in both<br />

alcoholics and cocaine addicts. GABRA2 and GABRG1 were only<br />

down-regulated in alcoholics, mirroring the strong associations with<br />

alcoholism in case-control studies in humans. Cocaine addicts had<br />

reduced expression <strong>of</strong> genes implicated in GABA synthesis. Robust,<br />

overlapping findings between alcoholics, cocaine addicts and P rats<br />

included changes in expression <strong>of</strong> GABBR1 and GABRG2 and genes<br />

encoding GABA A receptor-associated trafficking proteins.<br />

Conclusions: Our study has demonstrated that, at least within the<br />

hippocampus, chronic alcohol and cocaine exposure results in both<br />

common and specific changes in expression <strong>of</strong> GABAergic genes<br />

that are directionally consistent and biologically plausible. In<br />

particular, the key finding <strong>of</strong> significant down-regulation <strong>of</strong><br />

GABBR1 in alcoholics, cocaine addicts and alcohol naïve P rats<br />

indicates that down-regulation <strong>of</strong> this gene that encodes GABAB<br />

receptors, potentially resulting in increased GABA in the synaptic<br />

cleft, may be a risk factor for the development <strong>of</strong> addiction in<br />

humans. Indeed, preclinical studies have shown that activation <strong>of</strong><br />

GABAB receptors blocks the rewarding effects <strong>of</strong> drugs <strong>of</strong> abuse.<br />

Our findings may therefore have possible therapeutic potential.<br />

71<br />

PP8 ASSOCIATION OF TPH2 GENE WITH SUICIDAL<br />

BEHAVIOR IN THE CHINESE POPULATION<br />

B. Liu(1,2,3), S. Chen(1,2,3), X. Li(1,2,3), Y. Wang(1,2,3), Q.<br />

Zhao(1,2,3), X. Li(1,2,3), Q. Shen(1,2,3), L. He(1,2,3), G. He*(1,2,3)<br />

1. Bio-X Center, Key Laboratory for the <strong>Genetics</strong> <strong>of</strong> Developmental<br />

and Neuropsychiatric Disorders (Ministry <strong>of</strong> Education), Shanghai<br />

Jiao Tong University 2. Institutes <strong>of</strong> Biomedical Sciences, Fudan<br />

University, 138 Yixueyuan Road 3. Institute for Nutritional Sciences,<br />

Shanghai Institutes <strong>of</strong> Biological Sciences, Chinese Academy <strong>of</strong><br />

Sciences, 320 Yueyang Road<br />

*heguang@sjtu.edu.cn<br />

Introduction: Suicidal behavior (SB) has been a devastating and<br />

challenging worldwide public-health problem. SB is the fifth leading<br />

cause <strong>of</strong> overall death and the primary cause <strong>of</strong> adolescent death in<br />

China. Evidence from family, twin, and adoption studies does suggest<br />

that genetic factors might be involved in the etiology <strong>of</strong> SB. The<br />

TPH2 gene, located on 12q21.1, is crucial for brain serotonin<br />

synthesis and predominantly expressed in the brain stem. In order to<br />

clarify the role <strong>of</strong> TPH2 gene in Chinese SB, we undertook an<br />

association study <strong>of</strong> this gene with Chinese subjects. Our results<br />

support that TPH2 gene may contribute to the etiology <strong>of</strong> SB.<br />

Methodology: Six polymorphisms <strong>of</strong> TPH2 Gene (rs10784941,<br />

rs1386494, rs2171363, rs476816, rs1386486, and rs1872824) were<br />

tested in a total <strong>of</strong> 297 suicide attempt patients (SA) (126 female, age<br />

46.10±13.14 years) and 329 non-suicide attempt patients (NSA) (112<br />

female, age 43.22±12.27 years), 67% <strong>of</strong> SA experienced at least once<br />

violent suicide. All the samples were Han Chinese in origin from<br />

Shanghai. Diagnostic and Statistical Manual <strong>of</strong> Mental Disorder,<br />

Third Revised Edition (DSM-III-R), was taken as the diagnosis<br />

criteria. Each subject was given a standardized interview and<br />

diagnosis was confirmed by clinical interviews conducted by at least<br />

two independent experienced psychiatrists. Written informed consent<br />

was obtained from either the participants or the participants' legal<br />

representatives after the research aims and procedures were fully<br />

explained. The study protocol was reviewed and approved by the<br />

Shanghai Ethical Committee <strong>of</strong> Human Genetic Resources.<br />

Peripheral blood samples for DNA extraction were obtained from the<br />

subjects with the classical phenol-chlor<strong>of</strong>orm method.<br />

Results: Between 297 SA and 329 NSA individuals, SNP<br />

rs10784941 showed statistically difference in both allele frequencies<br />

(p=0.0259, OR=0.76, 95% CI [0.60-0.97]) and genotype frequencies<br />

(p=0.0169). Visibly, the frequency <strong>of</strong> G allele is higher in SA (45.4<br />

%) than that in NSA (38.8%). SNP rs4760816 exhibited difference in<br />

only allele frequencies (p=0.0291, OR=0.77, 95%CI [0.61-0.97]).<br />

Haplotype "GCGTAA" is more prevalent in SA than in NSA, which<br />

accounted for 9.2% and 4.1% respectively, and showed the<br />

significant difference between SA and NSA (p=0.001, OR=2.35,<br />

95%CI [1.38-4.01]).<br />

Conclusions: Both the single point and multi-locus haplotype<br />

analysis strongly support that TPH2 may be a susceptibility gene for<br />

SB in Chinese. In conclusion, our results forcefully support that<br />

TPH2 may be an important gene in the predisposition to SB.


PP9 ASSOCIATION STUDY OF NEUROTROPHIC<br />

FACTORS AND RECEPTORS GENES WITH SERUM BDNF<br />

A. Terracciano*(1), M. Piras(2), M. Lobina(2), O. Meirelles(1), A.<br />

Mulas(2), W. Chan(1), A. Sutin(1), L. Crisponi(2), D.<br />

Schlessinger(1)<br />

1. National Institute on Aging NIH 2. Istituto di Ricerca Genetica e<br />

Biomedica, CNR<br />

*terraccianoa@mail.nih.gov<br />

Introduction: The purpose <strong>of</strong> this research is to identify genetic<br />

variants associated with levels <strong>of</strong> Brain Derived Neurotrophic Factor<br />

(BDNF) in serum. BDNF plays a key role in the survival, growth,<br />

and differentiation <strong>of</strong> neurons. Levels <strong>of</strong> BDNF in serum are lower in<br />

mood disorder patients and increase in response to antidepressant<br />

treatment. Lower serum BDNF is also found in subjects scoring high<br />

on neuroticism, a stable tendency to experience negative emotions<br />

and a predisposing factor for mood disorders.This study estimates the<br />

heritability <strong>of</strong> serum BDNF and investigates whether SNPs in genes<br />

encoding neurotrophic factors and their receptors are associated with<br />

serum BDNF. We examine the BDNF Val66Met functional variant,<br />

which has been investigated in previous studies with mixed results<br />

(Cattaneo et al., 2010, Jiang et al., 2009, Lang et al., 2009, Minelli et<br />

al., in press, Ozan et al., 2010, Terracciano et al., 2010a, Trajkovska<br />

et al., 2007). In addition, we test for associations between serum<br />

BDNF and SNPs in four neurotrophic factor genes (NGF,<br />

BDNF,NTF3, and NTF4) and their receptors (NTRK1, NTRK2,<br />

NTRK3, and NGFR).<br />

Methodology: We performed the association test on a family-based<br />

sample (N = 2054) from Sardinia, Italy. We measured serum BDNF<br />

concentration and conducted a genome-wide association scan<br />

(GWAS).<br />

Results: From a set <strong>of</strong> 428 sib-pairs, one pair from each nuclear<br />

family with available data, we estimated a heritability <strong>of</strong> 0.48 for<br />

serum BDNF.There was no association between serum BDNF and<br />

Val66Met (rs6265 p > 0.05). A meta-analysis that combined the<br />

current and published studies also found no association between<br />

peripheral levels <strong>of</strong> BDNF and the BDNF Val66Met variant. The<br />

analyses <strong>of</strong> SNPs from 8 genes encoding neurotrophic factors and<br />

their receptors identified several SNPs with nominally significant<br />

associations, including SNPs in the BDNF gene (e.g., rs11030102, p<br />

= .001). The strongest effect was found for the genotyped SNP<br />

rs7170215 (P = 4.8x10 -5 ), which maps within the neurotrophic<br />

tyrosine kinase receptor, type 3 (NTRK3). The A allele (frequency<br />

56%) was associated with lower serum BDNF concentrations (Beta =<br />

-0.51, SE = 0.13), an effect that explained approximately 1% <strong>of</strong> the<br />

variance in serum BDNF. An additional association signal was<br />

present at the 5' <strong>of</strong> NTRK3, with rs11073742 (P = 1.2x10 -5 T allele:<br />

frequency 65%, Beta = -0.56, SE = 0.13).<br />

Conclusions: NTRK3 is a neurotrophic receptor that mediates the<br />

neurotrophic/TRK signal transduction pathway. This finding suggests<br />

that NTRK3 receptor might play a role in regulative feedback on the<br />

expression and storage <strong>of</strong> BDNF. This gene has also been proposed<br />

as a candidate gene for a number <strong>of</strong> psychiatric disorders, including<br />

major depressive disorder (Feng et al., 2008, Verma et al., 2008) and<br />

bipolar disorder (Athanasiu et al.). In conclusion, our study and a<br />

meta-analysis <strong>of</strong> the literature indicate that the BDNF Val66Met<br />

variant is not associated with serum BDNF. Our study further<br />

indicates that a common variant in NTRK3 might regulate the level <strong>of</strong><br />

BDNF in serum, one <strong>of</strong> the best characterized biomarker <strong>of</strong> mood<br />

disorders.<br />

Acknowledgments: This research was supported in part by the<br />

Intramural Research Program <strong>of</strong> the NIH, National Institute on<br />

Aging.<br />

72<br />

PP10 ASSOCIATION BETWEEN NEGATIVE MOOD<br />

DELUSIONS IN BIPOLAR DISORDER AND GENETIC<br />

VARIATIONS IN 3Q26.1<br />

ECIP<br />

S. Meier*(1), J. Treutlein(1), J. Frank(1), J. Strohmaier(1), R.<br />

Breuer(1), C. Schmäl(1), E. Vassos(2), M. Mattheisen(3,4,5), F.<br />

Degenhardt(3,4), T. Mühleisen(3,4), B. Hänisch(3,4), L. Priebe(3,4),<br />

T. Schulze(6), M. Nöthen(3,4), S. Cichon(3,7), 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 2. Social,<br />

Genetic and Developmental Psychiatry Centre, Institute <strong>of</strong><br />

Psychiatry, King's College London 3. Department <strong>of</strong> Genomics, Life<br />

& Brain Center, University <strong>of</strong> Bonn 4. Institute <strong>of</strong> Human <strong>Genetics</strong>,<br />

University <strong>of</strong> Bonn 5. Institute for Medical Biometry, Informatics,<br />

and Epidemiology, University <strong>of</strong> Bonn 6. Department <strong>of</strong> Psychiatry<br />

and Psychotherapy, University Medical Center, Georg-August-<br />

University 7. Institute <strong>of</strong> Neuroscience and Medicine (INM-1),<br />

Research Center Juelich<br />

*sandra.meier@zi-mannheim.de<br />

Introduction: It has previously been suggested that clinical symptom<br />

dimensions in bipolar disorder (BD) may prove more useful for<br />

delineating the genetics <strong>of</strong> the disorder than standard diagnostic<br />

models. This concept, however, has not yet been applied to data from<br />

genome-wide association studies (GWAS) in BD.<br />

Methodology: We performed a GWAS <strong>of</strong> factor dimensions in 637<br />

clinically well-characterized BD patients all <strong>of</strong> German ancestry. In a<br />

subsequent follow-up study <strong>of</strong> our top GWAS results, we included an<br />

additional 290 BD patients <strong>of</strong> German ancestry.<br />

Results: Rs9875793 located in an intergenic region on 3q26.1was<br />

significantly associated with the factor dimension Negative Mood<br />

Delusions in the combined BD sample (n= 927 P= 4.65 x 10 -8 , OR=<br />

2.66). In a subsequent case-control analysis the G allele <strong>of</strong> rs9875793<br />

in was overrepresented in patients with Negative Mood Delusions<br />

symptoms (P= 0.0007, OR= 1.74), currently we aim to replicate this<br />

finding in an independent sample <strong>of</strong> BD patients. Further we<br />

explored whether the association <strong>of</strong> rs9875793 with Negative Mood<br />

Delusions can be extended to patients with schizophrenia and major<br />

depression disorder (MDD) (n=554) and (SCZ) (n= 977). We<br />

observed a trend for association <strong>of</strong> rs9875793 with Negative Mood<br />

Delusions in MDD patients, and allele frequencies <strong>of</strong> rs9875793<br />

differentiated schizoaffective patients <strong>of</strong> depressive subtype from<br />

other SCZ patients.<br />

Conclusions: In summary,we have demonstrated that refining<br />

phenotypes in genetic associations studies can facilitate the search for<br />

more valid nosological entities. Further our results suggested that the<br />

association <strong>of</strong> the chromosome 3 variant rs9875793 with a specific<br />

clinical symptom pattern compiled <strong>of</strong> negative mood and delusions is<br />

observable across diagnostic borders.


PP11 EXPRESSION DATA FROM MULTIPLE BRAIN<br />

REGIONS AND POLYGENIC SCORE ANALYSIS<br />

A. Richards*, L. Jones, P. Holmans, M. O'Donovan<br />

Cardiff University<br />

*wmdalr@cf.ac.uk<br />

Introduction: Variation in gene expression is a possible mechanism<br />

by which genetic polymorphisms may confer susceptibility to<br />

schizophrenia. The International Schizophrenia Consortium (ISC)<br />

found that schizophrenia risk alleles are enriched among SNPs<br />

selected for marginal evidence for association (p < 0.5) from genome<br />

wide association analyses. We have previously demonstrated that<br />

marginally associated alleles with a greater effect on brain gene<br />

expression are superior predictors <strong>of</strong> schizophrenia affected status<br />

through polygenic score analysis than those with a lesser effect. Here,<br />

we examine whether using gene expression data from multiple brain<br />

areas can further enhance this prediction.<br />

Methodology: We used an eQTL dataset containing 163 adult<br />

control samples, each with data for four brain regions (frontal and<br />

temporal cortices, pons and cerebellum). We assigned high or low<br />

eQTL status to SNPs separately for each brain region, and also based<br />

upon a combination <strong>of</strong> the four regions. SNPs associated (p < 0.5)<br />

with schizophrenia status were then identified using the large PGC<br />

meta-analysis <strong>of</strong> schizophrenia GWAS studies, excluding the ISC<br />

dataset. The high eQTL and low eQTL SNP sets were then used to<br />

predict schizophrenia disease status <strong>of</strong> samples in the ISC GWAS<br />

dataset using the polygenic score method <strong>of</strong> analysis reported by the<br />

ISC.<br />

Results: We observed that higher probability cis-eQTLs predicted<br />

schizophrenia status significantly better in independent datasets than<br />

those with a lower probability for being a cis-eQTL. This effect<br />

became several orders <strong>of</strong> magnitude more significant when eQTL<br />

data based upon a combination <strong>of</strong> brain regions was used (p=7.5e -7 ).<br />

Conclusions: These results further support our hypothesis that some<br />

schizophrenia risk alleles affect the progress <strong>of</strong> the disorder via their<br />

effects on expression with a high level <strong>of</strong> statistical significance.<br />

They also demonstrate the utility <strong>of</strong> combining expression data from<br />

multiple brain regions.<br />

73<br />

PP12 A GENOME WIDE SURVEY SUPPORTS THE<br />

INVOLVEMENT OF LARGE COPY NUMBER VARIANTS IN<br />

MENTAL RETARDATION AND SCHIZOPHRENIA<br />

E. Derks*(1), S. MacGregor(2), A. Maclean(3), A. McKechanie(3),<br />

A. McRae(2), B. Pickard(4), S. Purcell(5), P. Sklar(5), D. StCLair(6),<br />

N. Wray(2), P. Visscher(2), D. Blackwood (3)<br />

1. University Medical Center Utrecht 2. Queensland Institute <strong>of</strong><br />

Medical Research 3. University <strong>of</strong> Edinburgh 4. University <strong>of</strong><br />

Strathclyde 5. Center for Human Genetic Research 6. Aberdeen<br />

University<br />

*e.m.derks@umcutrecht.nl<br />

Introduction: Mental retardation is associated with high rates <strong>of</strong><br />

psychiatric disorders (e.g., schizophrenia, autism). Previous studies<br />

have shown increased rates <strong>of</strong> Copy Number Variants (CNVs) both<br />

for mental retardation and psychiatric disorders with large overlap in<br />

candidate regions. The aim <strong>of</strong> this study is to study the role <strong>of</strong> large<br />

(>100 kb and >1Mb) CNVs in a sample <strong>of</strong> 170 Scottish patients with<br />

mental retardation.<br />

Methodology: Samples were genotyped using the Affymetrix 6.0<br />

array.A psychiatric condition was present in 119 subjects, including<br />

schizophrenia (N=64), autism (N=19), bipolar disorder (N=21), and<br />

depression (N=15) as primary diagnoses. Subjects were divided into<br />

two groups: subjects with mental retardation and a clinical diagnosis<br />

for schizophrenia (MR+SCZ N=64), and subjects with mental<br />

retardation and no clinical diagnosis except for depression (MR only<br />

N=66).We selected only those CNVs with a LOD>10 and physical<br />

length greater than 100 kb and restricted analysis to the remaining<br />

1,791 segments.<br />

Results: Including all CNVs larger than 100kb, CNV burden (total,<br />

deletions, and duplications) was similar in “MR+SCZ” and “MR<br />

only”. Restricting analysis to CNVs larger than 1Mb, total CNV<br />

burden was higher in “MR+SCZ” (rate=.25) compared to “MR only”<br />

(rate=.09 empirical p=.03) which was mainly explained by an<br />

increased number <strong>of</strong> large duplications (rates are .16 and .02,<br />

respectively, empirical p=.01). Seven <strong>of</strong> the 33 large CNVs were<br />

located at candidate regions 1q21.1, 15q13.1, 16p13.1, and 22q11.21<br />

which is a significant enrichment <strong>of</strong> the candidate regions (p=.002).<br />

Of the 26 CNVs located outside candidate regions, ten CNVs (9<br />

schizophrenia patients and 1 bipolar patient) were located at 15q11.2<br />

(18.5-20.1Mb). This region does not overlap with the previously<br />

reported candidate region at 15q11.2 (20.3-20.8Mb). The CNV<br />

burden in this region is significantly higher in “MR+SCZ” subjects<br />

(rate=.14) compared to “MR only” subjects (rate=0.0 empirical<br />

p=.001). The finding was replicated in the Scottish International<br />

Schizophrenia Consortium (ISC) samples as we detected a higher<br />

CNV rate in 1130 schizophrenia cases (rate=.088) compared to 984<br />

controls (rate=.066 Empirical p=.038).<br />

Conclusions: The CNV burden for large (>1Mb) CNVs is increased<br />

in patients with mental retardation and schizophrenia compared to<br />

patients with mental retardation only. Furthermore, candidate regions<br />

for neurodevelopmental disorders are significantly enriched for<br />

CNVs larger than 1Mb. We replicated the role <strong>of</strong> CNVs in regions<br />

1q21.1, 15q13.1, 16p13.1, and 22q11.21 in causing mental<br />

retardation with or without psychiatric diagnosis and in addition<br />

identified a novel region <strong>of</strong> interest for schizophrenia. This region is<br />

located at chromosome 15q11.2 (18.5 to 20.1Mb) and includes the<br />

POTEB (POTE ankyrin domain family, member B) gene. The<br />

function <strong>of</strong> this gene might be related to the function <strong>of</strong> ANK3<br />

(ankyrin 3, node <strong>of</strong> Ranvier) which has been found to be associated<br />

with bipolar disorder.


PP13 INVESTIGATING THE CONTRIBUTION OF<br />

COMMON GENETIC VARIANTS TO THE RISK AND<br />

PATHOGENESIS OF ADHD<br />

E. Stergiakouli*(1), M. Hamshere(1), P. Holmans(1), K. Langley(1),<br />

I. Zaharieva(1), A. Pocklington(1), Z. Hawi(2), L. Kent(3), M.<br />

Gill(2), N. Williams(1), M. Owen(1), M. O'Donovan(1), A.<br />

Thapar(1)<br />

1. MRC Centre in Neuropsychiatric <strong>Genetics</strong> and Genomics, Cardiff<br />

University 2. Department <strong>of</strong> Psychiatry, Trinity Centre for Health<br />

Sciences 3. Bute Medical School<br />

*stergiakoulie@cardiff.ac.uk<br />

Introduction: Although rare copy number variants (CNVs) appear to<br />

contribute to ADHD, the role <strong>of</strong> common risk variants (SNPs) in<br />

ADHD is still unclear. However, ADHD GWAS have not been<br />

adequately powered up to date. In our study, we performed a GWAS<br />

<strong>of</strong> ADHD with the aim being to identify associated variants and also<br />

examine whether SNPs, including those below conventional levels <strong>of</strong><br />

significance, impact on the same biological pathways hit by CNVs.<br />

Methodology: 727 children with ADHD and 5,081 controls were<br />

analysed using Illumina arrays. Single locus tests were performed as<br />

well as ALIGATOR pathway analysis (Holmans et al. 2009) on a<br />

large set <strong>of</strong> pathways from a variety <strong>of</strong> public databases. The gene<br />

sets enriched in pathway analysis <strong>of</strong> our GWAS data were tested for<br />

an excess <strong>of</strong> genes hit by large, rare CNVs (>500kb,


PP15 WHOLE GENOME ASSOCIATION SCAN OF HIGH<br />

RESOLUTION REGIONAL MRI VOLUME AND THICKNESS<br />

MEASURES FOR LINKING GENETICS OF BRAIN<br />

CHANGES WITH ALZHEIMER'S DISEASE<br />

M. Khondoker*<br />

*mizanur.khondoker@kcl.ac.uk<br />

Introduction: Genome-wide association studies has been an<br />

effective tool for understanding the genetic basis <strong>of</strong> many complex<br />

diseases. However, apart from the APOE, comparatively little is<br />

known about the contribution <strong>of</strong> genetic risk alleles to Alzheimer's<br />

disease (AD). APOE ε 4 allele accounts for only a tiny fraction <strong>of</strong> the<br />

heritability <strong>of</strong> AD and many more genetic risk alleles are thought to<br />

be undiscovered. Imaging genetics, where associations between<br />

quantitative brain imaging measures and genetic variants are<br />

investigated, has recently emerged as an interesting research area for<br />

linking genetics <strong>of</strong> brain changes with AD.<br />

Methodology: In this study, we undertake a genome wide association<br />

scan <strong>of</strong> high resolution whole brain automated magnetic resonance<br />

imaging (MRI) regional cortical volume and thickness measures<br />

combining the genetics and imaging data obtained from the<br />

Alzheimer's Neuroimaging Initiative and the AddNeuroMed study.<br />

Results: Imaging genetics analysis has been done previously with a<br />

lower coverage <strong>of</strong> the brain image (e.g., only AD-related brain<br />

region). In the current analysis with a better coverage <strong>of</strong> the whole<br />

brain image and bigger sample size, we anticipate to detect novel<br />

genetic susceptivity loci for AD.<br />

Conclusions: Imaging genetics analysis has been done previously<br />

with a lower coverage <strong>of</strong> the brain image (e.g., only AD-related brain<br />

region). In the current analysis with a better coverage <strong>of</strong> the whole<br />

brain image and bigger sample size, we anticipate to detect novel<br />

genetic susceptivity loci for AD.<br />

75<br />

PP16 RESEQUENCING AND FOLLOW-UP OF VAV3<br />

GUANINE NUCLEOTIDE EXCHANGE FACTOR IN<br />

SCHIZOPHRENIC PATIENTS<br />

B. Aleksic(1), I. Kushima(1), M. Ikeda(2), A. Yoshimi(1), H.<br />

Ujike(3), M. Suzuki(4), T. Inada(5), M. Takeda(6), K. Kaibuchi(7),<br />

N. Iwata(2), N. Ozaki(1)<br />

1. Department <strong>of</strong> Psychiatry, Nagoya University Graduate School <strong>of</strong><br />

Medicine 2. Department <strong>of</strong> Psychiatry, Fujita Health University<br />

School <strong>of</strong> Medicine 3. Department <strong>of</strong> Neuropsychiatry, Okayama<br />

University Graduate School <strong>of</strong> Medicine, Dentistry and<br />

Pharmaceutical Sciences 4. Department <strong>of</strong> Neuropsychiatry,<br />

University <strong>of</strong> Toyama Graduate School <strong>of</strong> Medicine and<br />

Pharmaceutical Sciences 5. Seiwa Hospital, Institute <strong>of</strong><br />

Neuropsychiatry 6. Department <strong>of</strong> Psychiatry, Osaka University<br />

Graduate School <strong>of</strong> Medicine 7. Department <strong>of</strong> Cell Pharmacology,<br />

Nagoya University Graduate School <strong>of</strong> Medicine<br />

Introduction: In recently completed Japanese genome wide<br />

associated study <strong>of</strong> schizophrenia one <strong>of</strong> the top association signals<br />

(within top 10 hits) was detected in the region <strong>of</strong> VAV3, a gene that<br />

maps to the chromosome 1p13.3. To follow-up this association<br />

signal, we performed resequencing <strong>of</strong> VAV3 and evaluated effect <strong>of</strong><br />

associated common variant on expression pattern <strong>of</strong> VAV3.<br />

Methodology: Four independent samples were used in the present<br />

study: (1) 575 unrelated patients with schizophrenia and 564 subjects<br />

with no personal or family history <strong>of</strong> psychiatric disorders were<br />

selected for genome wide screening analysis, (2) for the replication<br />

analysis we used sample comprising 1511 cases and 1517 healthy<br />

controls (3) mutation analysis was performed on a total <strong>of</strong> 321<br />

patients suffering from schizophrenia and (4) Lymphoblastoid cell<br />

lines sample derived from 30 schizophrenic patients and same<br />

number age and gender matched healthy controls was used for<br />

expression analysis. As basis for a more detailed functional<br />

interpretation <strong>of</strong> the novel rare variants, we performed ab-initio<br />

structure predictions and comprehensive in-silico analysis.<br />

Results: Four rare novel missense variants were detected in the<br />

present study. The mutations were followed-up in large independent<br />

case controls sample (>4000 subjects) and one <strong>of</strong> the novel variants<br />

was associated with schizophrenia. Moreover, three different<br />

bioinformatics algorithms had predicted functional effect <strong>of</strong> rare<br />

variant that was associated with schizophrenia. We couldn't detect<br />

that expression <strong>of</strong> VAV3 is influenced by associated common variant.<br />

Conclusions: We conclude that these results are suggestive <strong>of</strong> an<br />

association with schizophrenia at VAV3 locus, but that they could<br />

not be considered conclusive without further replications.


PP17 DEEP RESEQUENCING AND ASSOCIATION<br />

ANALYSIS OF SCHIZOPHRENIA CANDIDATE GENES<br />

P. Sullivan(1), C. Hilliard*(1), J. Crowley(1), Y. Kim(1), M.<br />

Morgan(2), P. Sklar(3), S. Purcell(3,4), J. Lieberman(5), R. Gibbs(2)<br />

1. Departments <strong>of</strong> <strong>Genetics</strong>, University <strong>of</strong> North Carolina 2. Human<br />

Genome Sequencing Center and Department <strong>of</strong> Molecular and<br />

Human <strong>Genetics</strong>, Baylor College <strong>of</strong> Medicine 3. Center for Human<br />

Genetic Research, Massachusetts General Hospital 4. The Broad<br />

Institute <strong>of</strong> Harvard and MIT 5. Department <strong>of</strong> Psychiatry, Columbia<br />

University<br />

*chillia@email.unc.edu<br />

Introduction: In 2006, there were ~10 genes for which there was<br />

reasonable evidence for involvement in the etiology <strong>of</strong> schizophrenia<br />

(COMT, DAOA, DISC1, DRD2, DRD3, DTNBP1, HTR2A, NRG1,<br />

SLC6A3, SLC6A4). The pattern <strong>of</strong> results for these genes suggests<br />

that there may be sequence variants that have not yet been<br />

discovered, and re-sequencing has been limited.<br />

Methodology: To address this limitation, we re-sequenced the<br />

coding regions, 5-prime and 3-prime UTRs, splice sites, promoters<br />

and conserved intronic regions <strong>of</strong> these 10 genes in 727 cases with<br />

schizophrenia from the CATIE clinical trial and 729 matched<br />

controls.<br />

Results: A total <strong>of</strong> 778 single nucleotide polymorphisms (SNPs)<br />

were identified by Sanger re-sequencing, including 620 novel<br />

variants. DISC1 showed a significantly enriched total SNP count<br />

relative to controls and relative to the other nine genes, primarily in<br />

the form <strong>of</strong> novel non-synonomous variants. A total <strong>of</strong> 92 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: association<br />

with schizophrenia in CATIE, novelty, SNP type (non-sense and<br />

missense mutations) and minor allele frequency in cases <strong>of</strong> European<br />

ancestry. These 92 variants have now been genotyped in an<br />

independent sample <strong>of</strong> 3,391 schizophrenia cases and 3,181 controls,<br />

all <strong>of</strong> European ancestry, a subset <strong>of</strong> the International Schizophrenia<br />

Consortium (ISC). Analyses examining association <strong>of</strong> these variants<br />

with schizophrenia are now being finalized.<br />

Conclusions: These data are expected to provide a clearer picture <strong>of</strong><br />

the role that variation in traditional candidate genes plays in<br />

schizophrenia etiology.<br />

76<br />

PP18 SUPPORT FOR INVOLVEMENT OF GLUTAMATE<br />

DECARBOXYLASE 1 AND NEUROPEPTIDE Y IN ANXIETY<br />

DISORDER SUSCEPTIBILITY<br />

J. Donner(1,2,3), T. Sipilä(1,2,3), S. Ripatti(4), L. Kananen(1,2), J.<br />

Lönnqvist(3,5), S. Pirkola(5), I. Hovatta*(1,2,3,4)<br />

1. Research Programs Unit, Molecular Neurology, Biomedicum-<br />

Helsinki, University <strong>of</strong> Helsinki 2. Department <strong>of</strong> Medical <strong>Genetics</strong>,<br />

Haartman Institute, University <strong>of</strong> Helsinki 3. Department <strong>of</strong> Mental<br />

Health and Substance Abuse Services, National Institute for Health<br />

and Welfare 4. Institute <strong>of</strong> Molecular Medicine Finland (FIMM),<br />

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

University Central Hospital<br />

*iiris.hovatta@helsinki.fi<br />

Introduction: Anxiety disorders, including panic disorder,<br />

generalized anxiety disorder, obsessive-compulsive disorder, posttraumatic<br />

stress disorder, and phobias, are one <strong>of</strong> the most common<br />

mental illnesses characterized by excessive, prolonged and<br />

debilitating levels <strong>of</strong> anxiety. Their onset is influenced by both<br />

genetic and environmental factors, with heritability estimates<br />

averaging 30-40 %. Given the small effect sizes <strong>of</strong> risk genotypes<br />

indentified in anxiety disorders, it has become increasingly apparent<br />

that understanding <strong>of</strong> interplay between genetic and environmental<br />

factors is needed to better explain the underlying etiology.<br />

Methodology: We carried out a genetic association analysis <strong>of</strong> two<br />

genes previously implicated in anxiety susceptibility, glutamate<br />

decarboxylase 1 (GAD1), and neuropeptide Y (NPY). Our sample<br />

consisted <strong>of</strong> the Finnish population-based Health 2000 sample<br />

including 282 anxiety disorder cases and 575 matched controls. We<br />

first tested 8 single nucleotide polymorphisms (SNPs) from GAD1<br />

and 10 from NPY for association to anxiety disorders. Second, we<br />

examined the interaction <strong>of</strong> these SNPs and the number <strong>of</strong> childhood<br />

adverse life events, one the strongest known risk factors for anxiety<br />

disorders, on the development <strong>of</strong> the anxiety disorder.<br />

Results: In a subgroup <strong>of</strong> patients with phobias, we observed<br />

evidence for association to GAD1 (P = 0.0001). In addition, we found<br />

that individuals having experienced more childhood adverse life<br />

events had a greater risk <strong>of</strong> developing an anxiety disorder if they<br />

carried specific risk genotypes within the NPY locus (P = 0.004).<br />

Conclusions: Genetic variation in GAD1 and NPY may affect<br />

susceptibility to anxiety disorders in the Finnish population. The<br />

effect <strong>of</strong> NPY was only seen in an interaction with childhood adverse<br />

life events. This finding highlights the importance <strong>of</strong> environmental<br />

risk factors in influencing predisposition to anxiety disorders.


PP19 INVESTIGATION OF THE INVOLVEMENT OF DE<br />

NOVO CODING POINT MUTATIONS IN EHMT1 IN<br />

SCHIZOPHRENIA<br />

S. Dwyer*, G. Kirov, M. Owen, M. O'Donovan<br />

MRC Centre in Neuropsychiatric <strong>Genetics</strong> and Genomics,<br />

Department <strong>of</strong> Psychological Medicine and Neurology, Cardiff<br />

University School <strong>of</strong> Medicine<br />

*dwyersl@cf.ac.uk<br />

Introduction: In the largest study <strong>of</strong> de novo copy number variants<br />

in schizophrenia to date, we found 2 de novo CNVs that disrupt<br />

EHMT1 (presented at this meeting). EHMT1 encodes for<br />

euchromatin histone methyltransferase which is involved in<br />

epigenetic regulation through histone methylation. A recent study<br />

has shown that EHMT is a key epigenetic regulator <strong>of</strong> neuronal genes<br />

and processes which regulate simple learning and complex memory<br />

in Drosophila (Kramer et al 2011). CNVs and point mutations at this<br />

locus are known to cause 9q subtelomere deletion syndrome (9qSDs),<br />

a neurodevelopmental disorder that is characterised by severe mental<br />

retardation and hypotonia. Behavioural problems have also been<br />

reported in a proportion <strong>of</strong> 9qSDs cases (Stewart et al 2007). Given<br />

that de novo CNVs at EHMT1 have been shown to occur in a small<br />

proportion <strong>of</strong> patients with schizophrenia, the aim <strong>of</strong> this study was to<br />

identify if de novo coding point mutations exist at the same locus in<br />

690 cases.<br />

Methodology: The sample used consisted <strong>of</strong> 690 parent-proband<br />

trios with schizophrenia from Bulgaria. A total <strong>of</strong> 29 fragments<br />

which encompassed the coding region <strong>of</strong> EHMT1 were screened in<br />

the probands using the LightScanner and Sanger sequencing. The<br />

parents <strong>of</strong> individuals harbouring rare coding mutations were<br />

subsequently sequenced to assess whether variants were de novo. The<br />

possible effects <strong>of</strong> the variants on the protein and exon splicing was<br />

determined using PolyPhen, SIFT and SKIPPY.<br />

Results: We did not find any de novo point mutations in our sample.<br />

We did find several mutations predicted to be damaging, 4 <strong>of</strong> which<br />

occurred in multiple individuals<br />

Conclusions: Within the constraints <strong>of</strong> sample size, we find no<br />

evidence that de novo point mutations at EHMT1 occur in probands<br />

with schizophrenia. We are currently sequencing the gene in a set <strong>of</strong><br />

control probands to test whether rare transmitted damaging mutations<br />

occur at an increased rate in people with schizophrenia.<br />

77<br />

PP20 NEXT GENERATION SEQUENCING ANALYSIS OF<br />

SPLICING PATTERNS AND EXONIC VARIATION IN THE<br />

SCHIZOPHRENIA CANDIDATE GENE DPYSL2<br />

C. Winchester*(1,3,4), G. Hamilton(2), B. Morris(1,3), J. Pratt(1,4),<br />

R. Hunter(1,5), M. Bailey(6)<br />

1. <strong>Psychiatric</strong> Research Institute <strong>of</strong> Neuroscience in Glasgow<br />

(PsyRING), Universities <strong>of</strong> Glasgow and Strathclyde 2. Institute <strong>of</strong><br />

Molecular, Cell and Systems Biology, College <strong>of</strong> Medical,<br />

Veterinary and Life Sciences, University <strong>of</strong> Glasgow 3. Institute <strong>of</strong><br />

Neuroscience and Psychology, College <strong>of</strong> Medical, Veterinary and<br />

Life Sciences, University <strong>of</strong> Glasgow 4. Strathclyde Institute <strong>of</strong><br />

Pharmacy and Biomedical Sciences, Centre for Neuroscience<br />

University <strong>of</strong> Strathclyde (CeNsUS), University <strong>of</strong> Strathclyde 5.<br />

Gartnavel Royal Hospital 6. School <strong>of</strong> Life Sciences, College <strong>of</strong><br />

Medical, Veterinary and Life Sciences, University <strong>of</strong> Glasgow<br />

*catherine.winchester@glasgow.ac.uk<br />

Introduction: Next generation sequencing is a powerful<br />

technologyrevolutionising our analysis and understanding <strong>of</strong><br />

genomes. It is particularly useful for deep re-sequencing <strong>of</strong> the<br />

human genome to elucidate the genetic basis <strong>of</strong> complex polygenic<br />

disorders such as schizophrenia that arise from influences <strong>of</strong> multiple<br />

common and rare variants in many genes. Dihydropyrimidinase-like<br />

2 (DPYSL2) is an emerging candidate risk gene for the disorder. We<br />

and others have shown association <strong>of</strong> DPYSL2 gene variants with<br />

schizophrenia in case-control association studies and our data show<br />

that DPYSL2 transcripts are differentially expressed in the prefrontal<br />

cortex from schizophrenia patients and from a rodent PCP model <strong>of</strong><br />

cognitive deficits and that DPYSL2 SNP alleles have effects on<br />

DPYSL2 transcript expression. Recently it emerged that differential<br />

expression <strong>of</strong> DPYSL2 protein in brain regions with clinical<br />

relevance to schizophrenia is one <strong>of</strong> the most reproducible findings<br />

across proteomic studies.<br />

Methodology: DPYSL2 transcripts were sequenced from overlapping<br />

RT-PCR products generated from RNA isolated from the prefrontal<br />

cortex <strong>of</strong> 36 schizophrenia patients and healthy controls (n=18).<br />

Indexed libraries were generated from each sample and paired-end<br />

sequencing was performed on the Illumina Genome Analyzer II. The<br />

data underwent quality control measures using an automated pipeline,<br />

then were aligned to the reference genome sequence using TOPHAT<br />

and further analysed using custom scripts. Exonic variants were<br />

identified usingBCFTools andVCFTools. Splice site variation was<br />

identified by custom scripts and visualised in the UCSC Genome<br />

Browser.<br />

Results: This approach resulted in a massive depth <strong>of</strong> nucleotide<br />

coverage (>10,000x) with approximately 500,000 reads <strong>of</strong> 85<br />

nucleotides per sample, enabling confidence estimates to be made for<br />

any findings that differed from the reference sequence. We have<br />

identified, several novel coding variants, alternate splicing patterns at<br />

the 5' end <strong>of</strong> the gene and novel transcript is<strong>of</strong>orms with skipped<br />

exons.<br />

Conclusions: Next generation sequencing for deep re-sequencing is<br />

facilitating the elucidation <strong>of</strong> the genetic basis <strong>of</strong> schizophrenia by<br />

identifying novel common and rare variants in schizophrenia risk<br />

genes. We demonstrate the utility <strong>of</strong> this approach with DPYSL2.


PP21 A GENOME-WIDE ASSOCIATION STUDY AND<br />

FOLLOW-UP ANALYSIS OF DANISH SCHIZOPHRENIA<br />

CASES AND CONTROLS<br />

D. Demontis*(1), J. Pallesen(1), J. Grove(1), C. Pedersen(2), M.<br />

Hollegaard(3), A. Hedemand(1), M. Nordent<strong>of</strong>t(4), M.<br />

Mattheisen(5), S. Cichon(5), G. Consortium(1,5), S. Steinberg(6), H.<br />

Stefansson(6), D. Hougaard(3), P. Mortensen(2), O. Mors(7), A.<br />

Børglum(1,7)<br />

1. Institute <strong>of</strong> Biomedicine, Dept. <strong>of</strong> Human <strong>Genetics</strong>, Aarhus<br />

University 2. National Centre for Register-based Research, Aarhus<br />

University 3. Dept. <strong>of</strong> Clinical Biochemistry and Immunology,<br />

Section <strong>of</strong> Neonatal Screening and Hormones, Statens Serum Institut<br />

4. <strong>Psychiatric</strong> Centre Copenhagen, Copenhagen University Hospital<br />

5. Institute <strong>of</strong> Human <strong>Genetics</strong>, University <strong>of</strong> Bonn 6. deCODE<br />

<strong>Genetics</strong> 7. Centre for <strong>Psychiatric</strong> Research, Aarhus University<br />

Hospital<br />

*ditte@humgen.au.dk<br />

Introduction: In order to investigate genetic risk variants implicated<br />

with schizophrenia in the Danish population we have performed a<br />

genome-wide association study <strong>of</strong> cases and controls all born in<br />

Denmark (stage 1) and subsequently genotyped the most significantly<br />

associated SNPs in a follow up sample (stage 2).<br />

Methodology: Stage 1: The investigated sample consisted <strong>of</strong> cases<br />

diagnosed with schizophrenia according to ICD-10 and controls all<br />

obtained from the Danish Newborn Screening Biobank. DNA was<br />

extracted from dried blood spots and each sample was wholegenome-amplified<br />

in three separate reactions which were pooled<br />

before genotyping on the Illumina Human 610-quad beadchip. 1774<br />

individuals (882 controls, 888 cases) and 541,148 SNPs passed<br />

stringent quality control. Stage 2: We followed up on the strongest<br />

signals from stage 1 (171 SNPs) in an independent Danish sample<br />

which consisted <strong>of</strong> 1369 cases and 1771 controls after quality control.<br />

Results: Association analysis <strong>of</strong> the follow-up sample found ten<br />

SNPs with nominal P-values < 0.05, nine <strong>of</strong> them demonstrating a<br />

directionally consistent replication (lowest P-value = 0.006,<br />

rs4757144). In the combined analysis <strong>of</strong> stage 1 and stage 2<br />

individuals, rs4757144 on chromosome 11 demonstrated the<br />

strongest association with schizophrenia (P = 1.77 x 10 -6 ). Among<br />

the ten SNPs demonstrating nominal significant association in the<br />

follow-up sample were two moderately independent SNPs<br />

(rs10828623 and rs10741058, r 2 = 0.6) located in the gene CACNB2<br />

on chromosome 10. These two SNPs were also among the 15 most<br />

associated SNPs in the combined analysis. A region-wise association<br />

analysis <strong>of</strong> stage 1 data summarizing permutation P-values <strong>of</strong> SNPs<br />

in a region <strong>of</strong> 100kbp into one P-value, found significant association<br />

<strong>of</strong> a region placed on chromosome 10 upstream the gene ZEB1 (P =<br />

7.04 x 10 -7 ). The SNP rs3123688 located in this region was the<br />

second strongest in the combined analysis (P = 3.1 x 10 -6 ). In a metaanalysis<br />

analysis including stage 1 and stage 2 individuals and<br />

additional 464 schizophrenia cases and 1,272 controls from a German<br />

sample rs4757144 remained the most significantly associated SNP<br />

(P-value = 3.41x10 -6 ).<br />

Conclusions: rs4757144 is located in the circadian rhythmassociated<br />

gene ARNTL. Circadian-rhythm abnormalities in<br />

schizophrenia and bipolar disorder patients have been reported and<br />

smaller candidate gene studies have investigated the association <strong>of</strong><br />

ARNTL with bipolar disorder and schizophrenia with inconsistent<br />

results. Our analyses indicate the implication <strong>of</strong> two loci on<br />

chromosome 10, one close to the gene ZEB1, which has not<br />

previously been reported in relation to psychiatric diseases and one<br />

containing the gene CACNB2. The region containing this gene was<br />

reported as one <strong>of</strong> the eight regions demonstrating the strongest<br />

signals in a genome-wide association study <strong>of</strong> bipolar I disorder in<br />

the Han Chinese population.<br />

78<br />

PP22 COPY NUMBER VARIATION ON CHROMOSOMES 1P<br />

IS ASSOCIATED WITH SCHIZOPHRENIA<br />

D. Tsuang*(1,2), C. Yu(1,2), S. Kim(1), S. Millard(2), L. Leong(2),<br />

S. Meichle(1), J. Silverman(3), L. Siever(3, 4), A. Radant(1,2), E.<br />

Wijsman(1)<br />

1. University <strong>of</strong> Washington 2. VAPSHCS 3. Mount Sinai School <strong>of</strong><br />

Medicine 4. Bronx VA Medical Center<br />

*dwt1@uw.edu<br />

Introduction: Multiple recent studies have used whole-genome<br />

genotyping methods to discover novel structural variations in the<br />

DNA <strong>of</strong> complex genetic disorders. In schizophrenia, the<br />

identification <strong>of</strong> novel, rare copy number variations (CNVs) has<br />

generated much excitement. The majority <strong>of</strong> these CNVs range in<br />

size from a kb to several Mb and are thought to be rare, highly<br />

penetrant, and found in only a small number <strong>of</strong> individuals (e.g.,


PP23 GENETIC ASSOCIATION STUDIES OF SUICIDE<br />

ATTEMPTS IN MOOD DISORDER PATIENTS<br />

R. Perlis*<br />

Massachusetts General Hospital and Harvard Medical School<br />

*rperlis@partners.org<br />

Introduction: Risk for suicide attempt appears to be in part heritable.<br />

Previous investigations <strong>of</strong> common variation in psychiatric<br />

phenotypes suggest large sample sizes are required to reliability<br />

identify such variations, and meta-analysis may facilitate this process.<br />

Methodology: Fixed and random-effects meta-analysis were used to<br />

examine data from multiple large mood disorder cohorts. Primary<br />

comparisons considered lifetime suicide attempting individuals<br />

contrasted with non-attempting individuals. In addition to examining<br />

single variants, we investigated aggregate risk scores and pathways<br />

for association with suicide attempt.<br />

Results: No single locus reached a threshold for genomewide<br />

significance. However, polygenic and pathway-based analyses<br />

suggest some shared risk across cohorts. These results will be<br />

discussed in the context <strong>of</strong> follow-up <strong>of</strong> genomewide association<br />

results.<br />

Conclusions: These results indicate that individual loci <strong>of</strong> very large<br />

effect do not appear to contribute to suicide liability in these mood<br />

disorder cohorts. However, they suggest the utility <strong>of</strong> further<br />

investigation using alternatives to single-variant association tests.<br />

79<br />

PP24 EXPRESSION QUANTITATIVE TRAIT<br />

NUCLEOTIDES (EQTNS) IN THE 15Q25 NICOTINIC<br />

ACETYLCHOLINERGIC RECEPTORS (NACHRS) GENE<br />

CLUSTER ARE ASSOCIATED WITH SMOKING<br />

J. Duan*(1), A. Sanders(1), E. Drigalenko(2), W. Moy(1), L.<br />

Chen(3), L. Bierut(3), J. Freda(1), J. Jacobi(1), D. He(1), M.<br />

Collaboration(4), C. Consortium(5), H. Göring(2), P. Gejman(1)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Sciences, NorthShore<br />

University HealthSystem and University <strong>of</strong> Chicago 2. Department <strong>of</strong><br />

<strong>Genetics</strong>,Texas Biomedical Research Institute 3. Department <strong>of</strong><br />

Psychiatry, Washington University 4. Molecular <strong>Genetics</strong> <strong>of</strong><br />

Schizophrenia (MGS) Collaboration 5. Cross-Population Meta-<br />

Analysis (CROSSPOP) Consortium<br />

*jduan@uchicago.edu<br />

Introduction: Common variants at the 15q25 nAChRs gene cluster<br />

(CHRNA5-CHRNA3-CHRNB4) have been found associated with<br />

smoking quantity and nicotine dependence. There are two<br />

independent SNP association signals here across different continental<br />

populations (1) rs16969968, a nonsynonymous SNP at CHRNA5<br />

(Asp398Asn) associated with reduced receptor function, and (2) an<br />

intronic SNP at CHRNA3, rs6495308, without evidence for a<br />

biological mechanism yet.<br />

Methodology: We performed an eQTN mapping on 15q25 nAChR<br />

gene cluster with gene expression data from lymphoblastoid cell lines<br />

(LCLs) from European ancestry (EA) MGS subjects, 446<br />

schizophrenia cases and 457 controls.<br />

Results: We identified eight SNPs associated with CHRNA5<br />

expression (p≤0.05; Bonferroni corrected) but none with CHRNA3 or<br />

CHRNB4. These associations are tagged by two SNPs: (1)<br />

rs12899940 (p=3.0 x 10 -6 ), located within an enhancer sequence ~114<br />

kb downstream from CHRNA5, and (2) rs569207 (p=1.8 x10 -4 ), an<br />

intronic SNP which may affect splicing as it is located 34 bp away<br />

from exon 2 <strong>of</strong> CHRNA5. The effect <strong>of</strong> rs569207 on CHRNA5<br />

expression is mainly driven by schizophrenia cases (p=6.48 x 10 -<br />

4 ). Furthermore, while rs569207 is in strong LD with rs6495308<br />

(r 2 =0.90), the SNP tagging the cross-population association with<br />

smoking, LD is weaker for rs12899940 (r 2 =0.22). Next, we tested<br />

rs569207 and rs12899940 for association with smoking in 3,189 EA<br />

and 1,362 African American (AA) daily smokers from MGS. Both<br />

eQTNs were nominally associated with cigarettes per day (CPD) in<br />

the EA sample (rs569207 p=0.013 and rs12899940 p=0.027), while<br />

only rs569207 showed association with CPD in the AA sample<br />

(p=0.007).<br />

Conclusions: The putative eQTNs identified here provide a potential<br />

biological explanation for one <strong>of</strong> the common variation associations<br />

with smoking in the 15q25 nAChRs gene cluster.


PP25 AN INVESTIGATION OF INHERITANCE PATTERNS<br />

IN CNVS VALIDATES KNOWN LOCI AND SUGGESTS<br />

NOVEL CANDIDATE SCHIZOPHRENIA GENES<br />

ECIP<br />

E. Rees*(1), D. Ivanov(1), M. Ikeda(2), D. Ruderfer(3,4,5), J.<br />

Moran(5), K. Chambert(5), L. Georgieva(1), D. Grozeva(1), S.<br />

Purcell(3,4,5), P. Sklar(3,4,5), M. O'Donovan(1), M. Owen(1), G.<br />

Kirov(1)<br />

1. Cardiff University 2. Fujita Health University 3. Massachusetts<br />

General Hospital and Harvard Medical School 4. Massachusetts<br />

General Hospital 5. Broad Institute<br />

*reeseg@cf.ac.uk<br />

Introduction: Copy number variations (CNVs) at several loci have<br />

an important role within the complex aetiology <strong>of</strong> Schizophrenia<br />

(SZ). These are deletions at 1q21.1, 3q29, 2p16.3 (NRXN1),<br />

15q11.2, 15q13.3, 17p12, 22q11.2 and duplications at 16p11.2 and<br />

16p13.1. Recent research has also revealed an increased risk <strong>of</strong><br />

developing SZ with exonic duplications in VIRP2 and C16orf72.<br />

Through investigating the inheritance patterns <strong>of</strong> CNVs in 662<br />

Bulgarian <strong>of</strong>fspring-parent trios we aim to assess whether there is an<br />

overrepresentation <strong>of</strong> transmitted to non-transmitted CNVs at known<br />

SZ loci and identify novel CNV associations.<br />

Methodology: Through Mendelian inheritance, parents heterozygous<br />

for a CNV should transmit it fifty per cent <strong>of</strong> the time to their<br />

<strong>of</strong>fspring. If CNVs are called incorrectly we would expect an overall<br />

preferential non-transmission and a high false positive rate <strong>of</strong> de<br />

novos. We tested 662 Bulgarian trios for transmission <strong>of</strong> CNVs. For<br />

replication we used the publicly available MGS dataset (2903 cases<br />

and 2738 controls), 521 Bulgarian controls, and 143 unrelated cases<br />

from Bulgaria, that passed filtering. All samples were genotyped at<br />

the BROAD Institute on Affymetrix 6.0 arrays. All datasets<br />

underwent the same vigorous quality control procedures, which<br />

included CNVs >= 15kb, >=15 probe coverage and subsequent<br />

filtering with a modified Z-score calling algorithm. Using the<br />

Bulgarian trios as our discovery sample, we selected potential genes<br />

<strong>of</strong> interest if they were hit by CNVs with a transmission to nontransmission<br />

ratio <strong>of</strong> 2:0 or better. These genes were then further<br />

investigated in a case-control analysis <strong>of</strong> trios, cases and controls,<br />

using PLINK.<br />

Results: Before filtering <strong>of</strong> the trios with the Z-score, there was an<br />

apparent preferential non-transmission <strong>of</strong> rare (


PP27 ANALYSIS OF ASSOCIATION BETWEEN A GENETIC<br />

MARKER LOCATED IN THE ZNF804A GENE (RS1344706)<br />

AND SCHIZOPHRENIA IN A CASE-CONTROL SAMPLE<br />

FROM INDONESIA<br />

ECIP<br />

N. Dai*(1,2), W. Qin(1,2), M. Wildenauer(1,2), A. Kusumawardhani<br />

and the Indonesian Schizophrenia <strong>Genetics</strong> Consortium (3), D.<br />

Wildenauer(1,2,5), S. Schwab(1,4)<br />

1. School <strong>of</strong> Psychiatry and Clinical Neurosciences, The University<br />

<strong>of</strong> Western Australia, 2. Western Australian Institute for Medical<br />

Research, Centre for Medical Research, The University <strong>of</strong> Western<br />

Australia 3. University <strong>of</strong> Indonesia 4. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Erlangen-Nuremberg 5. Centre for Clinical Research in<br />

Neuropsychiatry, Graylands Hospital<br />

*20683868@student.uwa.edu.au<br />

Introduction: ZNF804Acodes for a zinc finger protein with so far<br />

not well defined function. Only recently, reports have shown that<br />

ZNF804A is expressed in mouse embryos, and that expression <strong>of</strong><br />

ZNF804A might be up-regulated due to interaction with Hoxc8 as a<br />

downstream target (Chung et al, Journal <strong>of</strong> Biomedicine and<br />

Biotechnology, 2010). In 2008, O'Donovan and Colleagues reported<br />

strong evidence <strong>of</strong> association <strong>of</strong> a single nucleotide polymorphism<br />

located in the ZNF804A genomic region, rs1344706, with<br />

schizophrenia based on a genome wide association scan (O'Donovan<br />

et al., (2008) Nature Genet 40:1053-1055). The initial screen<br />

included 479 cases with schizophrenia and 2937 control individual.<br />

Evidence for association was considerably strengthened by two<br />

subsequent replication phases, as well as by a meta-analysis, included<br />

in the first report. Since then, a number <strong>of</strong> reports became available<br />

confirming the original finding. Most <strong>of</strong> these reports are based on<br />

Caucasian samples. Our aim was to study rs1344706, which has<br />

been found to be associated with schizophrenia, but also with<br />

depressive disorders, in a large Asian case control sample from<br />

Indonesia for association with schizophrenia.<br />

Methodology: Cases and controls were recruited in the area <strong>of</strong><br />

Jakarta and Bogor through collaboration with psychiatrists from the<br />

University <strong>of</strong> Indonesia. DSM-IV and ICD-10 diagnosis <strong>of</strong><br />

schizophrenia was established using structured interviews (DIPS,<br />

translated into Bahasa Indonesia). Controls were screened for major<br />

psychiatric disorders using a shortened interview version. DNA was<br />

isolated from blood samples using standard procedures. Overall, 1117<br />

control subjects and 1097 cases were included in our studies. A<br />

commercially available TaqMan assay (Applied Biosystems) was<br />

used for genotyping the single nucleotide polymorphism rs1344706.<br />

For quality control, samples were re-genotyped for this specific DNA<br />

variant using an RFLP assay.<br />

Results: No deviation from Hardy-Weinberg equilibrium was seen in<br />

cases or controls. Controls revealed a T-allele frequency <strong>of</strong> 0.488,<br />

while frequency in cases for the T-allele was 0.529. This resulted in a<br />

P-value <strong>of</strong> 0.006 (chi2 test). Genotype distribution was also<br />

statistically significantly different between cases and controls<br />

(P=0.027). The odds ratio was 1.179 (95% confidence interval 1.047<br />

– 1.327).<br />

Conclusions: We have been able to show association <strong>of</strong> rs1344706<br />

with schizophrenia in a large sample from Indonesia. The type <strong>of</strong> the<br />

associated allele (T-allele) was consistent with previous reports. It is<br />

interesting to note that the Indonesian samples revealed similar allele<br />

frequencies for the T-allele as recently reported for a sample from the<br />

Han Chinese population (Zhang et al, Mol Psych.2011,T -allele<br />

frequency in cases: 0.53, T-allele frequency in controls: 0.46,). Our<br />

results support a potential role <strong>of</strong> ZNF804A in the etiology <strong>of</strong><br />

schizophrenia and show that this is apparently not population<br />

specific.<br />

81<br />

PP28 NEXT-GENERATION ASSOCIATION STUDY OF<br />

UNCOMMON, PUTATIVELY FUNCTIONAL VARIANTS<br />

WITH MAJOR PSYCHOSES<br />

A. Takata*(1,2,3), Y. Iwayama(2), K. Yamada(2), E. Hattori(2), T.<br />

Toyota(2), A. Sano(4), M. Nakamura(4), M. Kato(4), S. Kanba(3), T.<br />

Yoshikawa2(2), T. Kato(1)<br />

1. Laboratory for Molecular Dynamics <strong>of</strong> Mental Disorders, RIKEN<br />

Brain Science Institute 2. Laboratory for Molecular Psychiatry,<br />

RIKEN Brain Science Institute 3. Department <strong>of</strong> Psychiatry, Kyushu<br />

University, School <strong>of</strong> Medicine 4. Department <strong>of</strong> Psychiatry,<br />

Kagoshima University Graduate School <strong>of</strong> Medical and Dental<br />

Sciences<br />

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

Introduction: Genome-wide association studies have successfully<br />

identified several common SNPs that are associated with psychiatric<br />

disorders with genome-wide significance. However, all <strong>of</strong> these SNPs<br />

discovered so far have only weak effects (OR 1.1-1.5). Therefore,<br />

increasing attention is now being paid to uncommon (MAF < 5%)<br />

variants, which may have relatively large effects. In this study, we<br />

tested association between uncommon, putatively functional variants<br />

and major psychoses (schizophrenia and bipolar disorder) based on<br />

two strategies, both <strong>of</strong> which were driven by the next-generation<br />

sequencing technology.<br />

Methodology: 1) Discovery <strong>of</strong> candidate variant(s) by exome<br />

sequencing <strong>of</strong> a patient born <strong>of</strong> a consanguineous marriage followed<br />

by association study using a case-control cohort. We performed<br />

exome sequencing <strong>of</strong> a schizophrenic patient born <strong>of</strong> a<br />

consanguineous marriage using SureSelect Human All Exon Kit and<br />

Illumina Genome Analyzer II. The patient also manifested proximal<br />

limb weakness and muscle biopsy revealed multiple deletions <strong>of</strong><br />

mitochondrial DNA. His parents did not have any specific psychiatric<br />

and neurological symptoms. Therefore, recessive inheritance <strong>of</strong> these<br />

phenotypes was suspected. 2) Association study <strong>of</strong> uncommon,<br />

putatively functional variants discovered in the 1000 Genomes<br />

Project. We selected candidate variants from the 1000 Genomes<br />

Project data released in December 2010 with these criter ia, 1)<br />

Specific in Asian, 2) in the databases <strong>of</strong> schizophrenia or bipolar<br />

genes (SZGene or Bipolar Disorder Gene Database), 3) nonsynonymous<br />

and splice site variants predicted to be damaging, 4)<br />

Phred score >10, 5) MAF < 5%.<br />

Results: Results <strong>of</strong> 1). By exome sequencing, total 20317 <strong>of</strong> coding<br />

variants were identified. Among them, 65 variants satisfied these<br />

criteria, 1) not in dbSNP130, 2) not in HapMap exomes, 3) not in inhouse<br />

data <strong>of</strong> healthy Japanese, 4) non-synonymous or splice site<br />

variants, 5) homozygous (variant frequency > 75%). When we sorted<br />

these remaining variants by conservation score (PhyloP score), a<br />

missence variant in CNTNAP2 (V786L), whose copy number<br />

variations were known to be associated with autistic spectrum<br />

disorders and schizophrenia, ranked at the top. This variant was not<br />

private and detected in other Japanese with low frequency. Then, we<br />

tested association <strong>of</strong> this variant with schizophrenia using a Japanese<br />

case-control cohort (2011 cases and 2169 controls). However, four <strong>of</strong><br />

controls, as well as five <strong>of</strong> cases, carried this variant in a homozygous<br />

state and we could not observe significant difference. Results <strong>of</strong> 2).<br />

Total 47 variants in genes including ADRA1A, ANK3, ERBB3,<br />

GRIN3A and HTR1E remained.<br />

Conclusions: Conclusions <strong>of</strong> 1). This result indicates that even<br />

putatively functional variants in strong candidate genes are not<br />

necessarily associated with disease. Conclusions <strong>of</strong> 2). These<br />

variants are good candidates for further investigations. Associations<br />

<strong>of</strong> these variants with major psychoses were, and will be studied<br />

using previously described schizophrenia case-control cohort and<br />

bipolar case-control samples.


PP29 ALTERATIONS IN WNT/β-CATENIN PATHWAY<br />

MRNA EXPRESSION LEVELS IN MODIFIED<br />

ELECTROCONVULSIVE THERAPY<br />

M. Nishiguchi*, A. Tsutsumi, T. Kanazawa, H. Kikuyama, J. Koh,<br />

H. Yoneda<br />

Osaka Medical College<br />

*psy062@poh.osaka-med.ac.jp<br />

Introduction: Modified electroconvulsive therapy (m-ECT) is one <strong>of</strong><br />

the treatment options with high efficacy and safety, for drug<br />

treatment-resistant depression or schizophrenia, although the<br />

mechanism <strong>of</strong> m-ECT in the human brain is not fully understood. As<br />

for the mechanism <strong>of</strong> antidepressant treatment, the series <strong>of</strong> research<br />

on the postmortem brain pointed out the involvement <strong>of</strong> BDNF<br />

(brain-derived neurotrophic factor) gene through the hippocampal<br />

neurogenesis. Moreover, increased BDNF gene expression was also<br />

found in the treatment <strong>of</strong> m-ECT. Therefore the explanation <strong>of</strong> both<br />

treatments is possible only on BDNF gene, however the gene<br />

influences too many organs and illnesses.<br />

Wnt signaling pathway has received a lot <strong>of</strong> attention <strong>of</strong><br />

neurogenesis. GSK3β can be suppressed by binding Wnt to frizzled<br />

receptor, and β-catenin migrate to the nucleus with accumulating in<br />

cells as free form without being GSK3β phosphorylated. β-catenin<br />

begins the transcription <strong>of</strong> Wnt target genes by composing complex<br />

with T cell factor, which biological reaction is essential for cell-cycle<br />

rotation.The proliferation <strong>of</strong> neural stem cells rises by the<br />

accumulation <strong>of</strong> β-catenin in the nucleus. Especially the Yamanaka<br />

four transcription factor (sox2, oct-4, klf-4, c-myc) in Wnt signaling<br />

pathway is indispensable for the first-generation iPS(induced<br />

pluripotent stem cell). We aimed to the reveal association between<br />

the pre- and post- m-ECT with analyzing these four transcription<br />

factors mRNA expression changing.<br />

Methodology: 15 patients with treatment resistant depression were<br />

enrolled for the current study. Gene expression was carried out by<br />

RT-PCR method.<br />

Results: We will report the effect <strong>of</strong> m-ECT and four genes in Wnt<br />

signaling pathway.<br />

Conclusions: Further evaluations <strong>of</strong> this relationship in larger<br />

samples should help clarify the nature relationship between m-ECT<br />

and neurogenesis.<br />

82<br />

PP30 INVESTIGATION OF DNA POLYMORPHISMS IN A<br />

LARGE SAMPLE OF HEROIN ADDICTED INDIVIDUALS<br />

FROM WESTERN AUSTRALIA TREATED WITH<br />

NALTREXONE IMPLANTS FOR ASSOCIATION WITH<br />

ADDICTION SUSCEPTIBILITY AND TREATMENT<br />

RESPONSE<br />

S. Schwab*(1,2), N. Morris(2), A. Scott(2), D. Wildenauer(2,3), G.<br />

Hulse(2)<br />

1. University <strong>of</strong> Erlangen-Nuremberg 2. University <strong>of</strong> Western<br />

Australia 3. Centre for Clinical Research in Neuropsychiatry<br />

*sibylle.schwab@uk-erlangen.de<br />

Introduction: The population prevalence <strong>of</strong> heroin addiction in the<br />

adult Australian population aged between 15 and 54 is estimated to<br />

be in the range <strong>of</strong> 7 per 1000. It has been shown that susceptibility to<br />

heroin dependence is strongly influenced by genetic factors with<br />

heritability estimates as high as 0.7 and most likely a small number <strong>of</strong><br />

genes as well as environmental factors contributing. Pharmacological<br />

treatment options are based on interaction with the u-opioid receptor.<br />

One <strong>of</strong> the most common treatments includes the substitution <strong>of</strong> the<br />

stimulating agent heroin with methadone. Another treatment option is<br />

based on blocking the u-opioid receptor with naltrexone, making it<br />

unavailable for heroin. For our study we had access to a heroin<br />

addicted population from Western Australia who underwent<br />

treatment with naltrexone implants.<br />

Methodology: We have ascertained 896 individuals with DSMIV<br />

confirmed diagnosis <strong>of</strong> heroin dependence in Western Australia. All<br />

<strong>of</strong> the individuals have been treated using naltrexone implants.<br />

Plasma levels <strong>of</strong> naltrexone and its primary metabolite, naltrexol, are<br />

available. In addition, data on first use <strong>of</strong> heroin, switch to regular<br />

use <strong>of</strong> heroin, and co-morbidity with other drug abuse have been<br />

collected. DNA <strong>of</strong> these samples had been isolated for genotyping<br />

candidate genes and for GWAS.<br />

Results: Overall, 768 markers from 108 candidate genes were<br />

successfully genotyped using a GoldenGate assay (Illumina). These<br />

polymorphisms are currently being analysed for association with<br />

susceptibility to addiction as well as for a possible impact on<br />

response to naltrexon implants.<br />

Conclusions: A large sample <strong>of</strong> heroin addicted individuals with<br />

naltrexone treatment is available for genetic studies into addiction<br />

and treatment outcome.


PP31 GENOME-WIDE ASSOCIATION META-ANALYSIS OF<br />

TOTAL BRAIN VOLUME: RESULTS FROM THE ENIGMA<br />

CONSORTIUM<br />

A. Arias-Vásquez*<br />

On behalf <strong>of</strong> the ENIGMA consortium:<br />

http://enigma.loni.ucla.edu/publications/ohbm2011/<br />

*a.ariasvasquez@psy.umcn.nl<br />

Introduction: There is evidence for shared genetic factors explaining<br />

part <strong>of</strong> the variance <strong>of</strong> brain structure, regional brain volumes and<br />

psychiatric phenotypes. The rationale behind this approach is that<br />

differences <strong>of</strong> brain volume, either global or regional, are related to<br />

their specific functioning and to more global cognitive processes. Our<br />

overall aim is to identify common genetic variants that modify the<br />

risk for psychiatric disorders (in particular attention<br />

deficit/hyperactivity disorder (ADHD)) based on the identification <strong>of</strong><br />

genetic variants involved in the variance <strong>of</strong> the volume <strong>of</strong> brain<br />

structures in healthy individuals.<br />

Methodology: The mean total brain volume was automatically<br />

segmented in T1-weighted MRI scans from all contributing groups<br />

using the s<strong>of</strong>tware package FreeSurfer. 16 groups worldwide have<br />

now pooled their imaging genomics data from a total <strong>of</strong> >7000<br />

subjects, boosting the power to detect effects that no single study<br />

could identify. Genetic homogeneity <strong>of</strong> the samples was assessed via<br />

multi-dimensional scaling plots. Genotypes were imputed using the<br />

HapMap III reference panel, and associations were tested via dosage<br />

<strong>of</strong> each imputed SNP (accounting for kinship in family-based<br />

samples). Imputed SNPs were quality controlled, via an automated<br />

system, to filter out SNPs with low frequency or poorimputation<br />

quality. Results files and summary statistics from each group were<br />

pooled for meta-analysis which was conducted at each SNP across all<br />

groups.<br />

Results: By February 2011, 17 groups had uploaded GWAS results<br />

in >7000 subjects. Association conducted at ~1.3 million autosomal<br />

SNPs showed that none <strong>of</strong> the individual sites contained genomewide<br />

significant GWAS results (p


PP33 A GENOME-WIDE CNV ASSOCIATION STUDY<br />

ON AUTISM SPECTRUM DISORDER (ASD) IN THE<br />

JAPANESE POPULATION<br />

X. Liu*(1), K. Tokunaga(1), T. Sasaki(2)<br />

1. Department <strong>of</strong> Human <strong>Genetics</strong>, Graduate School <strong>of</strong> Medicine,<br />

The University <strong>of</strong> Tokyo 2. Laboratory <strong>of</strong> Health Education Graduate<br />

School <strong>of</strong> Education and Office for Mental Health The University <strong>of</strong><br />

Tokyo<br />

*liuxiaoxi@m.u-tokyo.ac.jp<br />

Introduction: Family and twin studies have strong evidence that<br />

autism spectrum disorder has a strong genetic background<br />

however,all know variation only accounts for a small fraction for<br />

known patitens. We conducted a genome-wide copy number<br />

variation (CNV) association study on ASD in the Japanese<br />

population.<br />

Methodology: Participants consisted <strong>of</strong> 500 ASD trio Japanese<br />

CNVs were detected using Genome-Wide Human SNP array<br />

6.0 and determined by Penncnv. CNVs with length


PP35 GENOME-WIDE ASSOCIATION STUDY IDENTIFIES<br />

GENETIC LOCI ASSOCIATED WITH BODY MASS INDEX<br />

AND HDL-CHOLESTEROL LEVELS DURING<br />

PSYCHOPHARMACOLOGICAL TREATMENT<br />

L. Athanasiu*(1,2,3), A. Brown(1,4), A. Birkenaes(1), M.<br />

Mattingsdal(3,5), I. Agartz(1,6), I. Melle(1,3), V. Steen(7,8), O.<br />

Andreassen(1,3), S. Djurovic(1,2,3)<br />

1. Institute <strong>of</strong> Clinical Medicine, University <strong>of</strong> Oslo 2. Department <strong>of</strong><br />

Medical <strong>Genetics</strong>, Oslo University Hospital 3. Division <strong>of</strong> Mental<br />

Healt and Addiction, Oslo University Hospital 4. Department <strong>of</strong><br />

Biostatistics, University <strong>of</strong> Oslo 5. Research Unit, Sorlandet Hospital<br />

6. Department <strong>of</strong> <strong>Psychiatric</strong> Research, Diakonhjemmet Hospital 7.<br />

Dr.Einar Martens Research Group for Biological Psychiatry,<br />

Department <strong>of</strong> Clinical Medicine, University <strong>of</strong> Bergen 8. Center for<br />

Medical <strong>Genetics</strong> and Molecular Medicine, Haukeland University<br />

Hospital<br />

*lavinia.athanasiu@medisin.uio.no<br />

Introduction: Metabolic and cardiovascular side effects are <strong>of</strong> major clinical<br />

importance because <strong>of</strong> their relevance for the increased mortality seen in<br />

severe mental disorders, which is mainly due to increased somatic morbidity.<br />

A better understanding <strong>of</strong> the mechanisms underlying metabolic and<br />

cardiovascular adverse effects associated with current psychopharmacological<br />

drugs is therefore imperative, and can lead to development <strong>of</strong> more tolerable<br />

drugs and personalized treatment based on the patient's genetic variants, as<br />

well. We performed a genome-wide association study <strong>of</strong> metabolic and<br />

cardiovascular risk factors during pharmacological therapy in patients with<br />

severe mental disorders<br />

Methodology: Our sample consisted <strong>of</strong> 594 patients with a severe mental<br />

disorder (schizophrenia or bipolar disorder) from the Thematically Organized<br />

Psychosis (TOP) Study and were successfully genotype on Affymetric<br />

Genome-Wide Human SNP array 6.0 (Affymetrix Inc., Santa Clara, CA,<br />

USA) The patients were examined for twelve indicators <strong>of</strong> metabolic side<br />

effects (body mass index, waist circumference, total cholesterol, HDLcholesterol,<br />

LDL-cholesterol, HDL-cholesterol/total cholesterol ratio,<br />

triglycerides, glucose, and C-reactive protein), and cardiovascular variables<br />

(blood pressure and heart rate) were measured. The patients used<br />

antipsychotics, mood stabilizers and/or antidepressants. We analyzed<br />

interactions between gene variants and three categories <strong>of</strong><br />

psychopharmacological agents based on their reported potential for side<br />

effects and defined genome-wide significance based on false discovery rate<br />

(FDR) <strong>of</strong> 0.1. We investigated these interactions between SNP and the<br />

medication with respect to differences in the level <strong>of</strong> metabolic- and<br />

cardiovascular side effects, using linear regression models implemented in<br />

PLINK.<br />

Results: The analyses revealed 14 significant interactions (FDR


PP37 SCHIZOPHRENIA TWO-HIT HYPOTHESIS IN VELO-<br />

CARDIO FACIAL SYNDROME<br />

H. Williams*, M. O'Donovan, M. Owen<br />

MRC centre for neuropsychiatric genetics and genomics, Department<br />

<strong>of</strong> psychological medicine, Cardiff University, CF14 4XN<br />

*williamshj1@cf.ac.uk<br />

Introduction: Velo-cardio facial syndrome (VCFS) is a common<br />

microdeletion syndrome associated with a number <strong>of</strong> wide ranging<br />

phenotypes. Of major interest is the observation that VCFS patients<br />

exhibit an elevated rate <strong>of</strong> schizophrenia that is some 30 times that <strong>of</strong><br />

the general population. Although much work has been conducted<br />

within the 22q11.2 deletion region to elucidate the source <strong>of</strong> this<br />

elevated risk there are no genetic markers that can distinguish<br />

between VCFS subjects that develop schizophrenia and those that do<br />

not. This observation has led to the suggestion that a schizophrenia<br />

risk locus may reside outside the deleted region and may modulate<br />

the risk via a two-hit model.<br />

Methodology: We sought to test whether secondary copy number<br />

variants (CNVs) could be responsible for the development <strong>of</strong><br />

schizophrenia in a proportion <strong>of</strong> the sample <strong>of</strong> VCFS patients (n=48)<br />

where half (n=24) had a diagnosis <strong>of</strong> psychosis. We used genomewide<br />

microarrays to identify large (>100kb), rare (


PP39 META-ANALYSIS OF SIX GENOME-WIDE<br />

ASSOCIATION STUDIES OF ADHD IN ADULTS<br />

D. Boomsma*(1), V. Saviouk(1), A. Arias-Vásquez(2), N. Amin(3),<br />

J. Hottenga(1), B. Penninx(4), J. Smit(4), E. de geus(1), S. Kooij(5),<br />

C. van Duijn(3), B. Franke(2)<br />

1. Biol Psychol, VU Univ 2. Human <strong>Genetics</strong>, Radboud University<br />

Nijmegen Medical Center 3. Dept Epidemiology, Erasmus Univ<br />

Medical Center 4. Dept Psychiatry, VU Univ Medical Center 5.<br />

PsyQ, Dept adult ADHD<br />

*di.boomsma@psy.vu.nl<br />

Introduction: Although apparently lower than in children, the<br />

heritability <strong>of</strong> ADHD in adults is significant. In the Netherlands we<br />

have collected genome-wide SNPs and phenotype data on adult<br />

ADHD in four population based samples, one sample <strong>of</strong> parents <strong>of</strong><br />

children ascertained for childhood ADHD and in one cohort which<br />

was ascertained for major depressive disorder (MDD).<br />

Methodology: The samples derive from 3 projects in Nijmegen<br />

(IMAGE, BIG and NBS, total N = 2190), from the Netherlands Twin<br />

Register (NTR, N = 2143) and from the Erasmus Rucphen Family<br />

study (ERF, N = 1054). The clinical cohort comes from the<br />

Netherlands Study <strong>of</strong> Depression and Anxiety (NESDA, N = 1001).<br />

In all studies ADHD was assessed by self-report: the Conners Adult<br />

ADHD Rating Scales (CAARS) were used in NTR and ERF and the<br />

23 item ADHD Rating Scale questionnaire was employed in the<br />

Nijmegen samples. Phenotypic scores were obtained for Inattention<br />

and Hyperactivity domains, as well as for ADHD symptoms. After<br />

thorough genotyping quality control and imputation using MACH<br />

and HAPMAP European Reference Set, ~2.3 mln SNPs were<br />

analyzed for each study/phenotype using MACH2QTL with an<br />

additive model and sex/age as covariates. Currently, a meta-analysis<br />

<strong>of</strong> the genome-wide association scans is being performed using all<br />

samples.<br />

Results: First results from the NTR group demonstrate a genomewide<br />

significance for the cumulative ADHD Index in CAARS at<br />

rs6119272 (20q11, p=2.4E-10) within a putative protein. Another two<br />

significant hits were within ALDH1A2 (rs4646591, p=3.57E-9) and<br />

RORA (rs12324535, p=3.44E-8) genes. Other top genes in<br />

preliminary results included: ADHD Index - CYP2A6, SCGN,<br />

CSMD1, ATM, UBE2R2, UBAP2, RBMS3, WWOX, GALNT14;<br />

Inattention - ALDH18A1, PROX1, UHRF1BP1, SNRPC, GATM,<br />

HPS5, NAV3, APOB, CMPK1, MUC5AC; Hyperactivity -<br />

ADAMTS17, HYAL3, ACPL2, RASSF1, GPC5, GCN1L1,<br />

SLC6A2, NLRP12, ZNF234, PRKD1. Cadherin 13, or CDH13, one<br />

<strong>of</strong> the top candidates for ADHD, gave a p-value 9.79E-5 at<br />

rs6563978 and was among top 100 genes for Inattention in a cohort<br />

with MDD. Interestingly, other cadherins were also seen among top<br />

genes for the ADHD index (PCDH7, p=0.0001 and CDH2,<br />

p=0.0002) in the MDD cohort. Meta-analysis <strong>of</strong> NTR and NESDA<br />

samples also point to PCDH15 and CDH28 as possible influencing<br />

factors for ADHD index and PCDH9 and CDH12 for<br />

inattention. Among other overrepresented gene families in the results<br />

were Usher syndrome genes and CUB and Sushi multiple domains<br />

genes (CSMD) that recently were shown to be involved in addiction<br />

phenotypes, known to be associated with ADHD.<br />

Conclusions: Results <strong>of</strong> GWAS <strong>of</strong> ADHD symptoms assessed by<br />

self-report in the population overlap with findings from clinical<br />

samples. If confirmed in the final meta-analysis in the current study,<br />

this would open up new routes towards the identification <strong>of</strong> new<br />

ADHD genes.<br />

87<br />

PP40 GENOME-WIDE SNP-SNP INTERACTION ANALYSIS<br />

IN BIPOLAR DISORDER<br />

T. Mühleisen*(1), M. Mattheisen(2), F. Degenhardt(1), T.<br />

Becker(3), M. Steffens(3), S. Herms(1), B. Haenisch(1), J.<br />

Strohmaier(4), B. Bohn(14), T. Gerstner(14), M. Griebel(14), W.<br />

Maier(15), P. Propping(1), M. Rietschel(4), M. Nöthen(1), S.<br />

Cichon(1,16)<br />

1. Inst Hum Genet, Dept Genomics, Life & Brain Ctr, Univ Bonn 2.<br />

Dept Biostatistics, Harvard School <strong>of</strong> Public Health 3. IMBIE, Univ<br />

Bonn 4. Dept Genet Epidemiol in Psychiatry, Central Inst Mental<br />

Health 5. IMIBE, Univ Duisburg-Essen 6. Inst Epidemiol, Helmholtz<br />

Zentrum München, German Res Ctr for Environmental Health 7. Inst<br />

Clinical Mol Biol, Univ Kiel 8. MPI Psychiatry 9. <strong>Psychiatric</strong> Ctr<br />

Nordbaden 10. Dept Psychiatry, Univ Würzburg 11. Dept Psychiatry<br />

and Psychotherapy, Univ Hospital 12. Dept Clinical and<br />

Developmental Psychology, Inst Psychology, Univ Tübingen 13.<br />

Dept Psychiatry and Psychotherapy, Univ Göttingen 14. Inst for<br />

Numerical Simulation, Univ Bonn 15. Dept Psychiatry, Univ Bonn<br />

16. INM-1, Res Ctr Juelich<br />

*thomas.muehleisen@uni-bonn.de<br />

Introduction: In recent years, genome-wide association studies<br />

(GWAS) have identified many common single-nucleotide<br />

polymorphisms (SNPs) that contribute to complex traits. However,<br />

there is still missing heritability in genetically-complex traits,<br />

including bipolar disorder (BD) and other common neuropsychiatric<br />

diseases. Although this might be explained by overestimation <strong>of</strong><br />

heritability, it could be explained in part by yet unidentified common<br />

variants with very small genetic effect sizes, or by epistatic effects<br />

(interaction) between genetic variants. Due to the high number <strong>of</strong><br />

possible SNP-SNP interaction pairs arising from typical microarraybased<br />

GWAS datasets (about 500K SNPs), genome-wide interaction<br />

analysis (GWIA) are computationally challenging (about 1.0E11 SNP<br />

pairs).<br />

Methodology: In the present study, we applied a novel approach for<br />

GWIA to a dataset <strong>of</strong> 473,227 SNPs from 1,158 patients with a<br />

DSM-IV diagnosis <strong>of</strong> BD and 2,172 population-based controls, all <strong>of</strong><br />

German ancestry. This approach has been implemented in the latest<br />

version <strong>of</strong> the program INTERSNP (Herold et al. 2009).<br />

Results: Given the genome-wide significance at the 0.05 level for<br />

genotypic interaction and a marker set with 500K SNPs (P=1.0E-12<br />

Becker et al. 2010), we found a genome-wide significant interaction<br />

between a SNP in the gene B3GALTL on chromosome 13q12 and a<br />

intergenic SNP on chromosome 11q24 (P=9.49E-13). The B3GALTL<br />

marker also participates in the second most significant pair by<br />

interacting with another SNP on chromosome 11q24, which is in<br />

strong linkage disequilibrium with the 11q24 SNP <strong>of</strong> the top finding<br />

(r-squared greater than 0.8).<br />

Conclusions: This supportive result suggest that there is a truepositive<br />

interaction between the loci 13q12 and 11q24 in bipolar<br />

disorder. Our main GWIA finding, however, requires evidence for<br />

replication. For this purpose, we are currently following-up our top<br />

results in large independent samples <strong>of</strong> bipolar disorder. The results<br />

will be presented at the conference.


PP41 FUNCTIONAL REGULATION OF THE AHI1 GENE IN<br />

SCHIZOPHRENIA VIA A DISTANT METHYLATION OF AN<br />

ALTERNATIVE (TSS) PROMOTER: A HYPOTHESIS<br />

IMPLICATING THE ROLE OF LINE ELEMENTS<br />

F. Macciardi*(1,3), T. Zemojtel(2), F. Torri(1,3), E. Osimo(3), B.<br />

Lerer(4), S. Gaudi(5)<br />

1. Dept <strong>of</strong> Psychiatry and Human Behavior, UCI 2. Max Plank<br />

Institute for Molecular <strong>Genetics</strong> 3. Universita' degli Studi di Milano<br />

4. Hadassah Medical Organization 5. Department <strong>of</strong> Infectious,<br />

Parasitic and Immune-Mediated Diseases, Italian National Institute <strong>of</strong><br />

Health<br />

*fmacciar@uci.edu<br />

Introduction: The Abelson Helper Insertion virus 1 (AHI1) is a<br />

schizophrenia-associated gene that we originally discovered (Lerer et<br />

al, 2003) and later confirmed (Amann et al, 2005). Since then, many<br />

other investigators also confirmed the association <strong>of</strong> AHI1 to<br />

schizophrenia in other populations (Ingason et al, 2005 and 2010).<br />

More recently, we fine-mapped the entire 1M bp genomic region <strong>of</strong><br />

AHI1, extending our analyses to the population-specific evolution <strong>of</strong><br />

the gene and its regulatory network (Torri et al, FASEBJ, 2010).<br />

This initial annotation was a first step to characterize the functional<br />

role and regulation <strong>of</strong> AHI1 in schizophrenia. Intriguingly, one <strong>of</strong> the<br />

highest associated SNP, which has been also extensively replicated<br />

by others in several papers as well as in a recent meta-analysis, is<br />

rs1475069 (A/C) that lies at a great molecular distance (~ 250,000<br />

bp) from the 5' end <strong>of</strong> the gene.<br />

Methodology: We have re-sequenced the 1M bp region around the<br />

AHI1 gene in extremely different subjects (those who do and those<br />

who do not present the risk alleles at SNPs associated to the disease)<br />

in our initial discovery population. We also sequenced wholegenomes<br />

from additional 25 schizophrenic patients from other<br />

populations, looking for SNPs and other genomic signatures in and<br />

around AHI1. In other subjects, we have performed a MeDIP-Seq in<br />

post-mortem brain tissues.<br />

Results: Preliminary annotation from one <strong>of</strong> us (T. Z.) has shown<br />

that the SNP is located in a pretty well conserved region <strong>of</strong> ~40nt.<br />

The "C" variant creates a "CpG" dinucleotide and the latter might be<br />

subject to methylation. Another "CpG" dinucleotide -- which is<br />

directly upstream <strong>of</strong> the SNP-- is methylated. The methylation data<br />

was generated by application <strong>of</strong> MeDIP-seq in the context <strong>of</strong> brain<br />

tissue (postmortem human frontal cortex gray matter). Also, a TAF1<br />

binding signal is present immediately downstream the SNP (see also<br />

a small cluster <strong>of</strong> DNaseI hypersensitivity signal). The latter might<br />

indicate that the region containing the SNP could be serving as a<br />

functional promoter/enhancer (TSS). In fact, a CAGE tag (identified<br />

in HepG2) maps in this region, starting just one nucleotide (nt)<br />

upstream the SNP. The tag contains a mutation at the 2nd nt, which<br />

corresponds to the location <strong>of</strong> the SNP<br />

Conclusions: The above evidence suggests a functional-promoterregion<br />

scenario. Given the unusually wide-spreading distribution and<br />

large number <strong>of</strong> close to very distant eQTLs regulating the expression<br />

<strong>of</strong> AHI1, SNP rs1475069 could be another eQTL for the gene.<br />

Alternatively, this hypothetical promoter may be functionally related<br />

to the L1PA3 Transposable Element lying nearby, regulating AHI1<br />

via specific transcripts.<br />

88<br />

PP42 GENOME-WIDE ASSOCIATION STUDY OF<br />

ANXIETY-SPECTRUM DISORDERS IN TWO CAUCASIAN<br />

COHORTS<br />

T. Otowa*(1), S. Aggen(1), E. Castelao(2), Z. Kutalik(2), B.<br />

Maher(3), E. van den Oord(1), M. Preisig(2), J. Hettema(1)<br />

1. Virginia Commonwealth University 2. University <strong>of</strong> Lausanne 3.<br />

Johns Hopkins University<br />

*totowa@vcu.edu<br />

Introduction: Studies <strong>of</strong> anxiety disorders (ADs) in families and<br />

twins have demonstrated that genetic factors account for about 50%<br />

<strong>of</strong> their pathophysiology, while the susceptibility genes <strong>of</strong> ADs have<br />

not been elucidated. Considering high comorbidity <strong>of</strong> ADs with each<br />

other, there may be common genetic risk factors among ADs. To<br />

identify genetic variants contributing to AD susceptibility, we<br />

performed genome-wide association studies (GWAS) in two<br />

independent cohorts with anxiety-related phenotypes.<br />

Methodology: The first sample consisted <strong>of</strong> 2725 subjects from the<br />

Molecular <strong>Genetics</strong> <strong>of</strong> Schizophrenia (MGS) study control sample.<br />

The second sample comprised 2027 subjects from a population-based<br />

cohort (PsyCoLaus). These Caucasian samples were genotyped on<br />

the Affymetrix 6.0 platform and 500K GeneChip, respectively. We<br />

applied factor analysis to generate factor scores for a common AD<br />

phenotype as a quantitative trait in association analysis. Logistic<br />

regression for the factor score was carried out in each sample,<br />

correcting for population ancestry. These results were compared to<br />

those from more traditional case-control analyses.<br />

Results: After the data cleaning and quality control <strong>of</strong> genotype data,<br />

626,833 SNPs (MGS) and 331,798 SNPs (PsyCoLaus) on autosomal<br />

chromosomes were further analyzed. Neither GWAS identified any<br />

SNP posessing genome-wide significance. A preliminary metaanalysis<br />

<strong>of</strong> the two GWAS also did not produce any genome-wide<br />

significance results. The strongest association in the meta-analysis<br />

was observed at the SNP rs2947344 near ARHGAP22 on 10q11 (P min<br />

= 5.8 x 10 -6 ). Several moderate associations <strong>of</strong> SNPs in prior<br />

candidate genes were also identified (eg. CNTNAP2 and PDE4B).<br />

Conclusions: The present study identifies several SNPs associated<br />

with ADs which are worthy <strong>of</strong> further investigation. However, our<br />

results suggest that SNPs with major effects on ADs are unlikely to<br />

exist. Meta-analysis <strong>of</strong> larger sample sizes will be needed to identify<br />

novel candidate genes for ADs.


PP43 HIGH RESOLUTION ASSESSMENT OF COPY<br />

NUMBER VARIATION IN EIGHT PAIRS OF<br />

MONOZYGOTIC TWINS DISCORDANT FOR<br />

SCHIZOPHRENIA OR BIPOLAR DISORDER<br />

R. Bloom*(1), A. Kähler(1,2,3), A. Collins(1), G. Chen(4), T.<br />

Cannon(5), C. Hultman(2), P. Sullivan(1)<br />

1. Department <strong>of</strong> <strong>Genetics</strong>, University <strong>of</strong> North Carolina at Chapel<br />

Hill 2. Department <strong>of</strong> Medical Epidemiology and Biostatistics,<br />

Karolinska Institutet 3. Department <strong>of</strong> Psychiatry, Oslo Univeristy<br />

Hospital - Ulleval 4. Department <strong>of</strong> Biostatistics, University <strong>of</strong> North<br />

Carolina at Chapel Hill 5. Departments <strong>of</strong> Psychology, Psychiatry,<br />

and Human <strong>Genetics</strong>, University <strong>of</strong> California at Los Angeles School<br />

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

*rbloom@email.unc.edu<br />

Introduction: Schizophrenia and bipolar disorder have a<br />

concordance rate <strong>of</strong> about 50% in monozygotic twins, suggesting a<br />

complicated combination <strong>of</strong> genetic, epigenetic and environmental<br />

factors contributing to the etiology <strong>of</strong> these psychiatric diseases.<br />

Copy number variation (CNVs) is an important source <strong>of</strong> genetic<br />

variation, and CNVs play a clear role in the etiology <strong>of</strong> many<br />

psychiatric diseases, including schizophrenia and bipolar disorder.<br />

Methodology: Using the Swedish Twin Registry, we ascertained and<br />

clinically evaluated eight pairs <strong>of</strong> monozygotic twins where one twin<br />

has schizophrenia or bipolar disorder and the other is unaffected.<br />

Monozygosity was confirmed using genotyping. Using DNA from<br />

peripheral blood, we are using array-based comparative genomic<br />

hybridization (aCGH) to detect CNVs that arose in mitosis. Using<br />

two-channel arrays, we compare the affected to the unaffected twin in<br />

order to enhance specificity. Moreover, we have used two different<br />

aCGH platforms (Nimblegen's 2.1M probe array and Agilent's<br />

SurePrint G3 1x1M array) in order to conduct an unbiased genome<br />

screen and to enhance accuracy. Segments <strong>of</strong> copy number variation<br />

were called with genoCN, and we are currently verifying copy<br />

number changes by additional methods.<br />

Results: Preliminary results identified no large CNVs but varying<br />

CNV burden across the different twin pairs. We observed CNV<br />

differences between twins in regions known to undergo mitotic<br />

rearrangements (e.g., areas <strong>of</strong> T cell receptor recombination).<br />

Conclusions: Genetic mosaicism may make it harder to detect CNVs<br />

and we are therefore developing techniques to work around these<br />

issues. Further analysis will provide an in-depth look at genetic<br />

differences between monozygotic twins, as well as provide insight<br />

into different aCGH technologies.<br />

89<br />

PP44 ADDITIONAL SUPPORT FOR CACNA1C AND<br />

CHROMOSOME 15Q14 AS SUSCEPTIBILITY LOCI FOR<br />

BIPOLAR DISORDER<br />

E. Green*(1), M. Hamshere(2), D. Grozeva(1), I. Jones(1), L.<br />

Jones(3), K. Gordon-Smith(1,3), L. Forty(1), J. Moran(4), J.<br />

Kranz(4), S. Purcell(4), P. Sklar(4), M. Owen(1), M. O'Donovan(1),<br />

N. Craddock(1)<br />

1. MRC Centre for Neuropsychiatric <strong>Genetics</strong> and Genomics, Dept.<br />

<strong>of</strong> Psychological Medicine, Cardiff University 2. Biostatistics and<br />

Bioinformatics Unit, MRC Centre for Neuropsychiatric <strong>Genetics</strong> and<br />

Genomics, Cardiff University 3. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Birmingham 4. Stanley Centre for <strong>Psychiatric</strong><br />

Research, Broad Institute<br />

*greenek@cf.ac.uk<br />

Introduction: A recent meta-analysis <strong>of</strong> genome-wide association<br />

studies on bipolar disorder (BD) (4,387 cases and 6,209 controls) by<br />

Ferreira et al. 2008 identified a strong association with ANK3<br />

(ankyrin G), provided independent support for the association with<br />

CACNA1C (alpha 1C subunit <strong>of</strong> the L-type voltage-gated calcium<br />

channel) and identified a third region <strong>of</strong> association on chromosome<br />

15q14.<br />

Methodology: We have genotyped new samples consisting <strong>of</strong> 1238<br />

BD and 2913 controls on the Illumina platform using the<br />

immunochip. The most associated directly genotyped and imputed<br />

SNPs for the 3 regions (ANK3, CACAN1C and chromosome 15q14)<br />

from the meta-analysis with a p value less than 10 -6 were<br />

genotyped. Fine mapping was also performed genotyping SNPs<br />

within a recombination region around the most highly significant<br />

SNPs in ANK3 and CACAN1C (rs10994336 and rs1006737<br />

respectively).<br />

Results: An association was seen with rs1006737 in CACNA1C with<br />

the same risk allele. Fine mapping <strong>of</strong> the recombination region<br />

around rs1006737 did not refine the signal, rs1006737 remained the<br />

most associated SNP (Trend P = 5.8x10 -4 ). We also noted support for<br />

the previously identified SNPs on chromosome 15q14 (rs12899449 &<br />

rs2172835, Trend P = 0.055 and 0.035 respectively). We saw no<br />

replication for the 2 SNPs that previously showed strongest<br />

association in ANK3, nor in the SNPs genotyped in the<br />

recombination region around rs10994336.<br />

Conclusions: In summary, our study supports the findings that<br />

CACAN1C and the chromosomal region on chromosome 15q14 as<br />

bipolar disorder susceptibility loci.


PP45 DE NOVO CNVS AFFECTING MULTIPLE<br />

GENES/PATHWAYS MAY EXPLAIN DISCORDANCE OF<br />

MONOZYGOTIC TWINS FOR SCHIZOPHRENIA<br />

C. Castellani*, R. O'Reilly, S. Maiti, S. Singh<br />

The University <strong>of</strong> Western Ontario<br />

*ccastel3@uwo.ca<br />

Introduction: Schizophrenia is a common (1%) psychiatric disorder<br />

<strong>of</strong> high heritability (80%) and low concordance (48%) between<br />

monozygotic twins. Such epidemiological features are best explained<br />

by involvement <strong>of</strong> multifactorial genetic, epigenetic and<br />

environmental interactions. Given the multifactorial heterogeneity<br />

implicated in this disease we feel that an explanation for the<br />

discordance <strong>of</strong> MZ twins (MZD) using genome-wide comprehensive<br />

analysis <strong>of</strong> twins may <strong>of</strong>fer genetic insight into the causation <strong>of</strong> this<br />

disease. Specifically, we report on genome-wide copy number<br />

changes (CNCs) between six MZD twin pairs that allow<br />

identification <strong>of</strong> pair specific de novo CNCs that may explain their<br />

discordance.<br />

Methodology: We (O’Reilly, Psychiatrist) identified six pairs <strong>of</strong><br />

monozygotic twins discordant for schizophrenia and collected their<br />

blood and cheek swab samples following informed consent and ethics<br />

approval for this research by the Committee on Research Involving<br />

Human Subjects <strong>of</strong> the University <strong>of</strong> Western Ontario. This DNA<br />

was hybridized on Affymetrix Human SNP Array 6.0 that includes<br />

1.8 million markers. CNVs were called by Affymetrix Genotyping<br />

Console 4.0 as well as Partek Genotyping Suite s<strong>of</strong>tware suites.<br />

Results: Twin pairs differed for a minimum <strong>of</strong> one to thirteen CNVs.<br />

The best explanation for the observed differences in CNVs between<br />

MZ twins is de novo mutation (DNM) occurring during early<br />

embryogenesis. This explanation was confirmed in two <strong>of</strong> the six<br />

pairs using parental genotypes. Conceptually, DNMs may also lead to<br />

varying degrees <strong>of</strong> mosaicism, an unrecognized form <strong>of</strong> genomic<br />

variability affecting genetic individuality. Our analysis shows that<br />

each pair <strong>of</strong> MZD differs for a number <strong>of</strong> genetic features including<br />

SNP and CNVs. More important, the SNP and CNV differences<br />

between ill and well twin invariably affect a set <strong>of</strong> genes that are<br />

implicated in neurodevelopment and already reported as potential<br />

contributors to schizophrenia by family and population studies. The<br />

results argue for extensive heterogeneity and <strong>of</strong>fer independent<br />

support for the involvement <strong>of</strong> these genes in the development <strong>of</strong><br />

schizophrenia. We found that each <strong>of</strong> the six MZD pairs is different<br />

with respect to the schizophrenia related gene(s) affected. The only<br />

common pathway affected in more than one pair involves olfactory<br />

receptors that have been implicated in the development <strong>of</strong><br />

schizophrenia. It is likely that the genetic differences across twins<br />

represent the heterogeneous manifestation <strong>of</strong> the disease including<br />

age <strong>of</strong> onset.<br />

Conclusions: The results show genome wide de novo changes are<br />

common during ontogeny. Depending on the nature <strong>of</strong> changes and<br />

the genes involved they may contribute to discordance <strong>of</strong> MZD twins<br />

for any feature including disease. The fact that different MZD pairs<br />

have identified different genes <strong>of</strong>fers support for the extensive<br />

genetic heterogeneity in schizophrenia and may argue for the need for<br />

“patient specific” etiology and treatment strategies for schizophrenia.<br />

90<br />

PP46 FAMILY-BASED WHOLE GENOME SEQUENCING<br />

OF SCHIZOPHRENIA<br />

S. McCarthy*(1), J. Badner(2), W. McCombie(1), W. Byerley(3)<br />

1. Cold Spring Harbor Laboratory 2. University <strong>of</strong> Chicago Medical<br />

Center 3. University <strong>of</strong> California San Francisco<br />

*mccarthy@cshl.edu<br />

Introduction: Schizophrenia is a severe neuropsychiatric disorder<br />

that affects approximately 1% <strong>of</strong> most populations. Despite evidence<br />

for a large heritable component in schizophrenia, the identification <strong>of</strong><br />

genetic risk factors has been very difficult due the genetic and<br />

phenotypic complexity <strong>of</strong> the disorder. Genome-wide association<br />

data suggest that common variants with small effect sizes may<br />

explain 2-30% <strong>of</strong> the genetic liability to schizophrenia. Although the<br />

global contribution <strong>of</strong> rare variants to schizophrenia susceptibility is<br />

unclear, the association <strong>of</strong> rare recurrent CNVs and an elevated CNV<br />

burden suggests that a spectrum <strong>of</strong> disease variants below the<br />

resolution <strong>of</strong> array-CGH may be family and individual specific.<br />

Methodology: We are combining the power <strong>of</strong> whole genome<br />

sequencing with the advantages <strong>of</strong> family based designs to identify<br />

rare genetic variants <strong>of</strong> large effect that significantly predispose<br />

carriers to schizophrenia in large multiplex families. We are in the<br />

process <strong>of</strong> sequencing the whole genome <strong>of</strong> 10 individuals from two<br />

families <strong>of</strong> European ancestry on Illumina's HiSeq2000. At least<br />

three affected members and one unaffected representing several<br />

family branches are being sequenced.<br />

Results: We recently completed sequencing all members from one<br />

family and data analysis is underway. Although we expect to<br />

discover a large number <strong>of</strong> variants genomewide, we aim to narrow<br />

the search for risk variants by first focusing our attention on shared<br />

rare variants and CNVs disrupting coding exons, UTRs and<br />

promoters in regions <strong>of</strong> linkage previously detected in these<br />

pedigrees. Variants discovered using our approach will be followed<br />

up in additional available family members<br />

Conclusions: Whole genome sequencing represents a unique and<br />

long desired opportunity to gain signficant insights into the genetics<br />

and biology <strong>of</strong> all psychiatric conditions. Coupling whole genome<br />

sequencing with family based study designs will be an important<br />

strategy to rapidly identify risk variants and genes that elevate the<br />

liability to schizophrenia thereby promoting improved diagnosis and<br />

potentially better theraputics at the personal and family level.


PP47 IDENTIFYING MOOD DISORDER SUSCEPTIBILITY<br />

LOCI IN A DENSELY AFFECTED PEDIGREE FROM THE<br />

ANDALUCIAN REGION OF SPAIN<br />

A. Collins*(1), R. Bloom(1), Y. Kim(1), M. Nöthen(2), S. Cichon(2),<br />

M. Rietschel(3), P. Sullivan(1)<br />

1. Department <strong>of</strong> <strong>Genetics</strong>, University <strong>of</strong> North Carolina at Chapel<br />

Hill 2. Institute <strong>of</strong> Human <strong>Genetics</strong>, Department <strong>of</strong> Genomics,<br />

University <strong>of</strong> Bonn 3. Department <strong>of</strong> Genetic Epidemiology in<br />

Psychiatry, Central Institute <strong>of</strong> Mental Health Mannheim, University<br />

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

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

Introduction: Mood disorders have a lifetime prevalence <strong>of</strong><br />

approximately 20% in adults in the United States, with a 3.9%<br />

prevalence for bipolar disorder. Both major depressive disorder and<br />

bipolar disorder are thought to have a strong genetic component<br />

however, both disorders display complex genetics and high degrees<br />

<strong>of</strong> heterogeneity. The use <strong>of</strong> large pedigrees with high prevalence<br />

may reduce heterogeneity and select for variants <strong>of</strong> large effect,<br />

allowing easier mapping <strong>of</strong> disease loci. We have therefore identified<br />

a large multigenerational pedigree densely affected with bipolar<br />

disorder. The pedigree has five generations descended from a single<br />

founder pair. There was limited in-migration into the region where<br />

the pedigree members lived. The pedigree contains 18 individuals<br />

with bipolar disorder (including a sibship with six <strong>of</strong> eleven<br />

individuals affected) and seven individuals with recurrent major<br />

depression.<br />

Methodology: We have applied multiple complementary genomic<br />

technologies to this pedigree, including SNP genotyping using the<br />

Illumina Omni 1M chip, copy number variation mapping using<br />

Nimblegen aCGH tiling arrays, and exome sequencing <strong>of</strong> affected<br />

individuals.<br />

Results: Shared segment analysis using SNP genotypes from eleven<br />

individuals affected with bipolar disorder have yielded regions <strong>of</strong> the<br />

genome as potential candidates, including a 4Mb region on chr18<br />

shared by seven bipolar I cases. Copy number analysis did not reveal<br />

any large, novel, and potentially causative CNVs. Exome sequencing<br />

results will be prioritized by potential candidate regions.<br />

Conclusions: This family appears to carry a genetic factor increasing<br />

risk for mood disorders, including bipolar disorder. While we have<br />

identified regions <strong>of</strong> potential interest, our current analyses have not<br />

explained the incidence <strong>of</strong> mood disorders seen in this densely<br />

affected pedigree. Careful evaluation <strong>of</strong> these regions within<br />

additional family members and additional genetic follow-up will be<br />

necessary to identify the causative loci in this pedigree.<br />

91<br />

PP48 INTEGRATING BIAS CORRECTION IN READ-DEPTH<br />

ANALYSIS OF WHOLE GENOME SEQUENCING DATA TO<br />

IDENTIFY COPY NUMBER GAIN AND LOSS<br />

ECIP<br />

J. Szatkiewicz*(1,2), W. Wang(4), W. Sun(1,3), W. Wang(4), D.<br />

Lin(3), P. Sullivan(1,2)<br />

1. Department <strong>of</strong> <strong>Genetics</strong> 2. Department <strong>of</strong> Psychiatry 3.<br />

Department <strong>of</strong> Biostatistics 4. Department <strong>of</strong> Computer Science<br />

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

Introduction: Copy number variation (CNV) has been shown to play<br />

an important role in the etiology <strong>of</strong> schizophrenia and autism as well<br />

as in developmental delay, changes in brain size, and epilepsy. Highthroughput<br />

sequencing (HTS) technologies have opened new and<br />

exciting opportunities for CNV discovery. Huge amounts <strong>of</strong><br />

sequencing data are emerging for the study <strong>of</strong> psychiatric disorders.<br />

However, the detection and analysis <strong>of</strong> CNVs from HTS data are<br />

challenging. Some <strong>of</strong> the statistical challenges stem from the<br />

experimental biases in HTS that have direct effects on pairing the<br />

reads and estimating read depth. We present a novel statistical<br />

method and s<strong>of</strong>tware tool for CNV discovery from read-­‐‑depth (RD)<br />

analysis <strong>of</strong> genome sequencing.<br />

Methodology: Our method uses a hidden Markov model and<br />

negative binomial regression framework to identify copy number<br />

gains and losses while simultaneously accounting for the effects <strong>of</strong><br />

multiple confounding factors. Known confounding factors are<br />

corrected explicitly (e.g., GC content and mappability). Unknown<br />

experimental biases, such as ambiguous reads mapped to the regions<br />

due to misalignment and sequencing errors, are tolerated by the overdispersion<br />

parameter <strong>of</strong> the negative binomial distribution. Given the<br />

predicted CNV regions, additional refinements are carried out by<br />

applying a hidden semi-Markov model and by integrating paired-end<br />

mapping, split-read mapping, and breakpoint-junction analysis to<br />

redefine CNV-boundaries.<br />

Results: To calibrate our method, we analyzed the high-coverage<br />

data from the 1000 Genomes Project (1000GP) and compared the<br />

CNV calls to a gold standard CNV dataset and to the extensive<br />

validation performed by 1000GP. Our read-depth-based integrative<br />

method is complementary to split-­‐‑read and read-­‐‑pair approaches. We<br />

have obtained whole genome sequencing data from two cases with<br />

schizophrenia sequenced at 40X coverage and will apply our method<br />

to these data.<br />

Conclusions: Our method has been efficiently implemented in C++.<br />

Due to the highly parallel structure <strong>of</strong> the problem, we plan to<br />

parallelize the analysis process to further accelerate the computation.<br />

A user-friendly implementation <strong>of</strong> the complete analytic protocol will<br />

be freely available.


PP49 QUANTITATIVE TRANSCRIPTOME SEQUENCING<br />

REVEALS ALTERED PRESYNAPTIC VESICLE<br />

TRAFFICKING AND MYELINATION IN THE TEMPORAL<br />

CORTEX IN SCHIZOPHRENIA<br />

J. Wu(1,2,3), P. Tooney(1,2,3), X. Wang(2,3), M. Cairns*(1,2,3)<br />

1. Schizophrenia Research Institute 2. University <strong>of</strong> Newcastle 3.<br />

Hunter Medical Research Institute<br />

*murray.cairns@newcastle.edu.au<br />

Introduction: While genome wide transcription analysis <strong>of</strong><br />

postmortem tissue using hybridization has provided important insight<br />

into the neuropathology <strong>of</strong> schizophrenia, it represents a restricted<br />

view <strong>of</strong> disease-associated transcriptional variation based on a<br />

predetermined probe set. Deep sequencing technology now provides<br />

the opportunity to make an un-biased analysis <strong>of</strong> mRNA transcription<br />

and processing down to nucleotide resolution. In this study we used a<br />

sequencing approach to investigate whole genome expression in<br />

postmortem cortical grey matter from the superior temporal gyrus<br />

(BA22) in 9 cases <strong>of</strong> schizophrenia and 9 non-psychiatric controls<br />

matched for gender, age, brain hemisphere, postmortem interval, and<br />

pH.<br />

Methodology: The data was generated from 76 bp reads using an<br />

Illumina Genome Analyser II, and aligned using TopHat, arranged<br />

into exons and splice<strong>of</strong>orms using Bowtie and digitally quantified<br />

using Cufflinks s<strong>of</strong>tware.<br />

Results: The output, amounting to more than 38 Gb <strong>of</strong> sequence,<br />

revealed significant alteration <strong>of</strong> gene expression including many<br />

genes and pathways previously shown to be associated with<br />

schizophrenia. Genes in the presynaptic vesicle trafficking and<br />

myelination pathways were particularly striking and broadly<br />

consistent with previous analysis using microarray. There was also<br />

substantial overlap with target genes predicted to interact with<br />

microRNA shown previously to be differentially expressed in this<br />

tissue 1 . We also identified differences in the abundance <strong>of</strong><br />

transcriptional and splicing is<strong>of</strong>orms between the groups, which<br />

could have important functional significance for schizophrenia. We<br />

are currently investigating functionally significant coding and noncoding<br />

polymorphism in the context <strong>of</strong> transcripts altered in<br />

schizophrenia.<br />

Conclusions: This analysis provides a rich source <strong>of</strong> data, with detail<br />

way beyond the scope <strong>of</strong> hybridization, that will continue to provide<br />

clues about the underlying causes <strong>of</strong> cortical gene dysregulation in<br />

schizophrenia as new questions arise.<br />

1. Beveridge NJ, Gardiner E, Carroll AP, Tooney PA, Cairns MJ.<br />

Schizophrenia is associated with an increase in cortical microRNA<br />

biogenesis. Molecular Psychiatry 2010 15(12): 1176-1189.<br />

92<br />

PP50 NEXT-GENERATION SEQUENCING OF KNOWN AND<br />

PUTATIVE SUSCEPTIBILITY GENES FOR<br />

SCHIZOPHRENIA AND AUTISM SPECTRUM DISORDERS<br />

TO DETECT RARE HIGH-PENETRANT RISK VARIANTS<br />

D. Morris*, S. Furlong, E. Kenny, P. Cormican, C. Fahey, R. Anney,<br />

G. Donohoe, A. Corvin, L. Gallagher, M. Gill<br />

Trinity College Dublin<br />

*morrisdw@tcd.ie<br />

Introduction: Schizophrenia (SZ) and autism spectrum disorders<br />

(ASD) are complex neurodevelopmental disorders that share certain<br />

phenotypes including cognitive deficits and some behavioural<br />

characteristics. Such similarities suggest that these disorders may<br />

share an underlying pathology and thus may share some genetic risk<br />

variants. This study involves next-generation sequencing <strong>of</strong> the<br />

exonic regions <strong>of</strong> 215 potential susceptibility genes in an Irish sample<br />

<strong>of</strong> 150 cases <strong>of</strong> ASD, 300 cases <strong>of</strong> SZ and 300 controls, in order to<br />

identify single nucleotide polymorphisms, indels and structural<br />

variants contributing to one or both disorders.<br />

Methodology: A multiplex target enrichment method is used<br />

whereby DNA samples are multiplexed together using DNA<br />

indexes/barcodes and enriched for the exonic regions <strong>of</strong> these genes<br />

using the Agilent SureSelect target enrichment method. This is<br />

followed by 80bp paired-end sequencing in a single lane <strong>of</strong> an<br />

Illumina GAII. Gene selection comprised <strong>of</strong> five categories: 1)<br />

Interactors <strong>of</strong> NRXN1, 2) Interactors <strong>of</strong> DISC1, 3) Genes within the<br />

Glutamate Receptor Complexes NMDA, mGluR5 and AMPA, 4)<br />

Cell adhesion molecules and 5) Functional and Positional Candidates.<br />

Results: Analysis <strong>of</strong> the pilot set <strong>of</strong> samples indicates that the<br />

approach undertaken is successful with an even spread <strong>of</strong> sequence<br />

information for 24 indexed samples per lane, >8X coverage for 84%<br />

<strong>of</strong> target regions and overall SNP concordance with previous GWAS<br />

data (Affymetrix 6.0) <strong>of</strong> 99.3%.<br />

Conclusions: In addition to description <strong>of</strong> this novel sequencing<br />

method, data will be presented on the rare variant analysis in the SZ<br />

and ASD samples.


PP51 GENOME-WIDE ASSOCIATION ANALYSIS OF<br />

COGNITIVE ABILITIES IN A HEALTHY NORWEGIAN<br />

SAMPLE WITH REPLICATION IN A LARGE UK-BASED<br />

SAMPLE<br />

ECIP<br />

A. Christ<strong>of</strong>orou*(1), T. Espeseth(2), G. Davies(3), C. Fernandes(1),<br />

A. Tenesa(3), N. Pendleton(4), A. Lundervold(1), S. Cichon(5), I.<br />

Reinvang(2), V. Steen(1), I. Deary(3), S. Le Hellard(1)<br />

1. University <strong>of</strong> Bergen 2. University <strong>of</strong> Oslo 3. University <strong>of</strong><br />

Edinburgh 4. University <strong>of</strong> Manchester 5. University <strong>of</strong> Bonn<br />

*andrea.christ<strong>of</strong>orou@med.uib.no<br />

Introduction: Cognitive abilities vary greatly among individuals and<br />

are known to be valid predictors <strong>of</strong> life outcomes, including<br />

education and occupation. Family, twin and adoption studies confirm<br />

that differences in cognitive abilities are heritable, with genetic<br />

factors accounting for approximately 50% <strong>of</strong> the variance. In<br />

addition, it is well-established that cognitive deficits are shared by<br />

common psychiatric disorders, such as schizophrenia and bipolar<br />

disorder, and that variation in these impairments is genetically<br />

associated with these disorders. Thus, understanding the genetic<br />

factors underlying cognition may help to elucidate the mechanisms<br />

underlying psychiatric illness. To that end, we have undertaken a<br />

genome-wide approach <strong>of</strong> a well-characterised, homogeneous sample<br />

<strong>of</strong> healthy adults, the Norwegian Cognitive Neuro<strong>Genetics</strong> (NCNG)<br />

sample, with replication in the larger, well-characterised Cognitive<br />

Ageing <strong>Genetics</strong> in England and Scotland (CAGES) sample.<br />

Methodology: We performed a genome-wide association study<br />

(GWAS), using the Illumina HumanHap610-Quad Genotyping<br />

BeadChip, in the NCNG (N=670), who were tested for various<br />

neuropsychological measures <strong>of</strong> cognition. Focusing on two factors<br />

<strong>of</strong> general intelligence – fluid (Gf) and crystallized intelligence (Gc)<br />

– both single-marker and gene-based analyses were performed. In<br />

silico replication was performed using the CAGES sample<br />

(N=~3500), which was also genotyped on the HumanHap610. The<br />

top SNPs and genes from the NCNG were directly mined and whole<br />

genome rank correlation analyses were performed. In addition, gene<br />

set enrichment analysis (GSEA) was applied, querying the top hits<br />

from the NCNG dataset for enrichment <strong>of</strong> significant signal in the<br />

CAGES dataset.<br />

Results: We identified one genome-wide significant SNP (Gc:<br />

P=9.82x10 -9 ) and 148 SNPs with P≤10 -4 . Preliminary gene ontology<br />

analysis <strong>of</strong> the genes implicated by the top 1000 SNPs from the<br />

NCNG study identified synapse organization as the top biological<br />

process for Gf (raw P=8.00x10 -4 adjusted P=0.28) and neuron<br />

differentiation for Gc (raw P=5.86x10 -6 adjusted P=0.0051).<br />

Replication at the SNP level in the CAGES sample was marginal<br />

with only 17 <strong>of</strong> the 148 SNPs meeting the nominal significance level<br />

(P≤0.05). Similarly, negligible genome-wide correlation was<br />

observed (Spearman's rho


PP53 EBV COPY NUMBER IN LCLS<br />

W. Moy*(1), J. Duan(1), A. Sanders(1), J. Freda(1), J. Jacobi(1), D.<br />

He(1), M. Collaboration(2), P. Gejman(1)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Sciences, NorthShore<br />

University HealthSystem and University <strong>of</strong> Chicago 2. Molecular<br />

<strong>Genetics</strong> <strong>of</strong> Schizophrenia (MGS) Consortium<br />

*wmoy@northshore.org<br />

Introduction: Transformation <strong>of</strong> B-lymphocytes with EBV produces<br />

LCLs, which are the largest renewable source <strong>of</strong> DNA for genotyping<br />

and sequencing, e.g., the NIMH collections <strong>of</strong> LCLs at Rutgers<br />

University Cell and DNA Repository. Furthermore, due largely to<br />

limitations <strong>of</strong> available living brain tissue, LCLs are commonly used<br />

as a cellular model to study the effects <strong>of</strong> genetic variation in<br />

neurologic and in psychiatric disorders, such as for the study <strong>of</strong> gene<br />

expression pr<strong>of</strong>iles (e.g., see Sanders et al. WCPG 2011 abstract for<br />

such a study in schizophrenia). The determinants <strong>of</strong> EBV copy<br />

number in LCLs, whether genetic or environmental (e.g., epigenetic,<br />

transformation methods), or both, remain unknown. However, EBV<br />

copy number (viral load) varies across LCLs and strongly influences<br />

the transcriptional abundances <strong>of</strong> many genes (and <strong>of</strong> many other<br />

cellular phenotypes). Circulating B-lymphocytes in most adults<br />

(>90%) carry a latent EBV infection, though whether EBV copy<br />

number is influenced by past infection remains unknown. Some<br />

researchers have hypothesized that acute EBV infection may play a<br />

role in the etiology <strong>of</strong> major psychiatric disorders, for which<br />

definitive evidence remains unavailable. We report here a systematic<br />

analysis <strong>of</strong> determinants <strong>of</strong> EBV copy number in LCLs.<br />

Methodology: To detect loci influencing EBV copy number in<br />

LCLs, we performed a GWAS using the MGS schizophrenia casecontrol<br />

collection (European ancestry, EA, N=5,334; African<br />

American, AA, N=2,259) with transformation site, age, sex, caseness,<br />

and ancestry PCs as covariates.<br />

Results: In the EA sample we found suggestive association at<br />

12q24.21 with rs708831 (p=1.2×10 -7 ) and at 17q21.31 with<br />

rs1043284 (p=7.8×10 -7 ), and in the AA sample at 7q33 with<br />

rs273961 (p=4.1×10 -7 ). rs708831 is ~140 kb from MED13L<br />

(mediator complex subunit 13-like), a widely-expressed gene<br />

including fetal and adult brain. rs1043284 is in the 3’-UTR <strong>of</strong><br />

RUNDC1 (RUN domain containing 1), a gene which may play a role<br />

as an inhibitor <strong>of</strong> the tumor suppressor gene, p53. It is known that<br />

EBV impairs the p53 pathway, similar to other DNA tumor viruses.<br />

We note that rs273961 is in an intronic region <strong>of</strong> CREB3L2 (cAMP<br />

responsive element binding protein 3-like 2), a gene involved in<br />

protein trafficking (ER à Golgi). We found a significant association<br />

with transformation site (p


PP55 CASE-CONTROL ANALYSIS OF BIPOLAR<br />

DISORDER GENETICS USING EXOME SEQUENCE<br />

CAPTURE DATA<br />

J. Parla*(1), I. Iossifov(1), D. Lewis(1), S. Mavruk(1), M.<br />

Pirooznia(2), F. Goes(2), P. Zandi(2,3), R. Karchin(4), J. Potash(2),<br />

W. McCombie(1)<br />

1. Cold Spring Harbor Laboratory 2. Department <strong>of</strong> Psychiatry, Johns<br />

Hopkins School <strong>of</strong> Medicine 3. Department <strong>of</strong> Mental Health, Johns<br />

Hopkins Bloomberg School <strong>of</strong> Public Health 4. Department <strong>of</strong><br />

Biomedical Engineering and Institute for Computational Medicine,<br />

Johns Hopkins University<br />

*parla@cshl.edu<br />

Introduction: Bipolar disorder I (BP) is a major mental illness with a<br />

significant genetic component, as demonstrated by family, twin, and<br />

adoption studies. Molecular investigation into the genetics <strong>of</strong> BP has<br />

included linkage analyses, and, more recently, several genome-wide<br />

association studies (GWAS). While GWAS has yielded a couple <strong>of</strong><br />

genome-wide significant results, the “missing heritability” problem<br />

suggests that other approaches are necessary to uncover BP<br />

susceptibility variants. We are applying exome capture and<br />

resequencing towards finding rare genetic variants that contribute to<br />

BP.<br />

Methodology: The initial stage <strong>of</strong> case-control data production<br />

involved 105 BP I probands and 51 matched controls. This study was<br />

enabled by SeqCap EZ Human Exome SR v1 kits (26.2 Mb target<br />

size) from NimbleGen and advanced DNA sequencers from Illumina.<br />

Each exome capture sample was initially sequenced on one lane <strong>of</strong> a<br />

paired-end 76-cycle GAIIx run, with up to one lane <strong>of</strong> additional<br />

sequencing if the initial data were not enough to provide<br />

approximately 80% <strong>of</strong> the exome target at 20X sequencing depth.<br />

The sequence data generated from each capture were analyzed using<br />

a bioinformatic pipeline consisting <strong>of</strong> the alignment program BWA,<br />

the SAMtools s<strong>of</strong>tware package, and custom scripts.<br />

Results: On average, 36.5 million pass-filter sequence read pairs (5.5<br />

billion bases) were produced per captured sample, with 99.7% <strong>of</strong><br />

bases attributed to sample DNA (rather than sequencing adapter),<br />

93.5% <strong>of</strong> read pairs properly mapped with unique genomic<br />

coordinates, 57.5% <strong>of</strong> properly mapped read pairs mapped to exome<br />

probe sequences, and 31.3% <strong>of</strong> the total input pass-filter sequence<br />

data mapped to the exome target. The sequence data provided 81.6%<br />

<strong>of</strong> the exome covered at 20X depth, 13,694 coding SNPs, 7,545<br />

synonymous SNPs, and 6,149 nonsynonymous SNPs per sample, on<br />

average. From these results, we have identified 275 genes with<br />

possibly damaging, non-synonymous SNPs found in cases, but not in<br />

controls. There also appears to be an enrichment <strong>of</strong> non-synonymous<br />

SNPs in BP GWAS-identified genes in cases compared to controls.<br />

Conclusions: We generated significant exome coverage for 156 casecontrol<br />

samples and called several thousand coding variants per<br />

individual, including some found in cases but not controls. Additional<br />

case-control analyses will be completed soon. We have recently<br />

initiated a custom capture design with NimbleGen that will include<br />

their updated exome target (v2), along with the promoters and exons<br />

<strong>of</strong> 1,517 genes that encode post-synaptic proteins and genes<br />

identified in BP GWAS. The integration <strong>of</strong> synapse genes will allow<br />

us to investigate the relationship <strong>of</strong> synapse function to BP.<br />

95<br />

PP56 NO EVIDENCE OF EXCESS REGIONS OF<br />

HOMOZYGOSITY IN SCHIZOPHRENIA OR BIPOLAR<br />

DISORDER<br />

J. Pallesen*(1), J. Grove(1,2), D. Demontis(1), N. Lassen(1), L.<br />

Foldager(2), M. Hollegaard(3), C. Pedersen(4), T. Ørnt<strong>of</strong>t(5,6), M.<br />

Didriksen(7), D. Hougaard(3), C. Wiuf(2), P. Mortensen(4), O.<br />

Mors(8), A. Børglum(1,8)<br />

1. Inst. <strong>of</strong> Biomedicine, Depts. <strong>of</strong> Human <strong>Genetics</strong> 2. Bioinformatics<br />

Research Centre 3. Dept. <strong>of</strong> Clinical Biochemistry and Immunology,<br />

Statens Serum Institut 4. National Centre for Register-based Research<br />

5. Dept. <strong>of</strong> Molecular Medicine 6. AROS Applied Biotechnology A/S<br />

7. Synaptic Transmission, H. Lundbeck A/S 8. Centre for <strong>Psychiatric</strong><br />

Research, Aarhus University Hospital<br />

*jonatanpallesen@hotmail.com<br />

Introduction: Regions <strong>of</strong> homozygosity (ROHs) are stretches <strong>of</strong><br />

DNA sequence where each location contains two identical alleles.<br />

ROHs are caused by extended haplotypes, and are hypothesized to be<br />

indicative <strong>of</strong> positive selection pressure. The presence <strong>of</strong> ROHs has<br />

been found associated to a number <strong>of</strong> diseases and traits, among<br />

others late onset Alzheimer’s disease and schizophrenia (SZ). Lencz<br />

et al (PNAS, 2007) defines a common ROH (cROH) as a region<br />

where a preset number <strong>of</strong> individuals share 100 or more homozygous<br />

calls. In a SZ case-control study they found an association between<br />

ROHs and SZ, with nine cROHs being individually more frequent in<br />

cases than in controls, and cROHs being more frequent overall in<br />

cases compared to controls. A different study by Vine et al (Psychiatr<br />

Genet, 2009) applied the same method to a bipolar (BP) data set, and<br />

found no significant association. We apply here the methods used in<br />

those studies to a different SZ data set and a different BP data set.<br />

Methodology: Subjects in the SZ study were obtained from the<br />

Danish Newborn Screening biobank.Genotyping was performed<br />

using an Illumina Human 610-quad beadchip. After quality control,<br />

the study included 1,774 individuals (888 cases, 888 controls) and<br />

541,148SNPs. Subjects in the BP study was obtained from WTCCC.<br />

Genotyping was performed using theGeneChip 500K Mapping Array<br />

Set (Affymetrix chip). After quality control, the study included4,806<br />

individuals (1,868 cases, 2,938 controls) and 402,910 SNPs.ROHs<br />

and cROHs were defined as in the mentioned publications. Analyses<br />

were carried out using plink and Python code produced by the<br />

primary author.<br />

Results: With the appropriate cROH threshold, we found a number<br />

and coverage <strong>of</strong> cROHs similar to that <strong>of</strong> the two previous studies.<br />

For neither SZ nor BP, did we find any individual cROH with a<br />

significant difference in the distribution between cases and controls<br />

after adjusting for multiple testing, nor were there any systematic<br />

majority <strong>of</strong> either cases or controls among the top results. Seven out<br />

<strong>of</strong> the nine cROHs that were found by Lencz et al to be significant for<br />

SZ, were also present in our SZ study, but none <strong>of</strong> them differed<br />

significantly in the distribution between cases and controls. We did<br />

not replicate the finding <strong>of</strong> overall greater number <strong>of</strong> cROHs among<br />

cases, but instead found a significant overall greater number <strong>of</strong><br />

cROHs among controls (P = 0.0022). In BP, we found as Vine et al<br />

that there was no significant difference in the overall number <strong>of</strong><br />

cROHs among cases compared to controls.<br />

Conclusions: We could not replicate the results <strong>of</strong> Lencz et al <strong>of</strong><br />

individual cROHs being associated to SZ. But we did find a<br />

significant difference in the overall number <strong>of</strong> cROHs, although in<br />

contrast to Lencz et al we found them to be more frequent in controls.<br />

We replicated the results <strong>of</strong> Vine et al <strong>of</strong> no difference in the overall<br />

number <strong>of</strong> cROHs in BP. The results suggest that cROHs do not have<br />

a large role to play in the case <strong>of</strong> BP. But there are indications that<br />

the distribution <strong>of</strong> cROHs is different in SZ patients.


PP57 GENOME-WIDE ASSOCIATION STUDY OF<br />

MIGRAINE AND COMORBID DEPRESSION<br />

M. Rivera*(1), Z. Samaan(2), S. Cohen-Woods(1), G. Breen(1), R.<br />

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

Farmer(1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London 2. Department <strong>of</strong> Psychiatry and Behavioural Neurosciences,<br />

McMaster University<br />

*margarita.rivera_sanchez@kcl.ac.uk<br />

Introduction: Migraine is a common neurological disorder that is<br />

highly comorbid with psychiatric disorders such as depression.<br />

Epidemiological and clinical studies have consistently reported a<br />

positive correlation between depression and migraine. These studies<br />

show a higher risk for depression among individuals with migraine<br />

and also a higher risk for migraine among individuals with<br />

depression. The aim <strong>of</strong> this genome-wide association study (GWAS)<br />

is to identify genetic variants that contribute to the development <strong>of</strong><br />

migraine comorbid with depression.<br />

Methodology: The sample consists <strong>of</strong> approximately 2,500<br />

depressive cases and healthy controls sourced from the Depression<br />

Case-Control (DeCC) study and the Depression Network (DeNT)<br />

study. ICD-10 depression diagnoses were made using the SCAN<br />

interview. The controls were screened for lifetime absence <strong>of</strong> any<br />

psychiatric disorders. Within each group, the different categories <strong>of</strong><br />

migraine were established using the Structured Migraine Interview<br />

(SMI) designed using International Headache Society criteria.<br />

Depression cases and controls were genotyped using the Illumina<br />

HumanHap610-Quad BeadChip by the Centre National de<br />

Génotypage.<br />

Results: After applying stringent quality control criteria for missing<br />

genotypes, departure from Hardy-Weinberg equilibrium, and low<br />

minor allele frequency, logistic regression corrected for population<br />

stratification was performed to test for association to migraine in the<br />

whole sample, that is in depressive cases, and controls. Strong<br />

evidence for association was found for several SNPs located on<br />

chromosomes 2, 3 and 17 (p=7.6 x10 -6 , p=5.6 x10 -6 , p=6.5 x10 -6 ,<br />

respectively).<br />

Conclusions: This is the first GWAS study investigating the genetic<br />

contribution to migraine comorbid with depression in a large clinical<br />

sample <strong>of</strong> depressive patients and controls individuals. This study has<br />

identify evidence for association with several genetic variants that<br />

merit further investigation.<br />

96<br />

PP58 GENOME-WIDE COPY NUMBER VARIATION (CNV)<br />

ANALYSIS FOR SCHIZOPHRENIA IN A LARGE<br />

ASHKENAZI COHORT<br />

S. Guha*(1), J. Rosenfeld(1), A. Malhotra(1,2,3,4,5), A. Darvasi(6),<br />

T. Lencz(1,2,3,4,5)<br />

1. Department <strong>of</strong> Psychiatry, Division <strong>of</strong> Research, The Zucker<br />

Hillside Hospital Division <strong>of</strong> the North Shore – Long Island Jewish<br />

Health System 2. Center for <strong>Psychiatric</strong> Neuroscience, The Feinstein<br />

Institute for Medical Research 3. Department <strong>of</strong> Psychiatry and<br />

Behavioral Science, Albert Einstein College <strong>of</strong> Medicine <strong>of</strong> Yeshiva<br />

University 4. Department <strong>of</strong> Psychiatry, H<strong>of</strong>stra University School <strong>of</strong><br />

Medicine 5. Dept. <strong>of</strong> Molecular Medicine, H<strong>of</strong>stra University School<br />

<strong>of</strong> Medicine 6. Department <strong>of</strong> <strong>Genetics</strong> The Institute <strong>of</strong> Life Sciences,<br />

The Hebrew University <strong>of</strong> Jerusalem<br />

*sauravguha@gmail.com<br />

Introduction: Copy number variations (CNVs), are recognized to<br />

contribute to normal human genomic variability and also associated<br />

with the risk <strong>of</strong> human diseases. Recent genome-wide studies have<br />

provided evidence that CNVs may also be associated with<br />

schizophrenia and other psychiatric disorders. Ethnically<br />

homogeneous founder population may increase power for detection<br />

<strong>of</strong> rare and common CNVs associated with human disease.<br />

Methodology: To identify copy number variations (CNVs) that<br />

increase the risk <strong>of</strong> schizophrenia, we performed a whole-genome<br />

CNV analysis on a cohort <strong>of</strong> 1233 schizophrenia and schizoaffective<br />

cases and 2279 controls drawn from the Ashkenazi Jewish (AJ)<br />

population from Israel. Ethnically homogeneous samples were<br />

selected from a larger AJ cohort and genotyped for 1.4 million probes<br />

using Illumina HumanOmni1-Quad arrays. After stringent quality<br />

filtering for both samples and markers, we performed PCA with AJ<br />

specific AIMs and MDS correction in PLINK to correct for any<br />

population stratification effects.<br />

Results: Currently, we are analyzing all the samples after QC were<br />

using multiple CNV calling algorithms to overcome the false positive<br />

CNV calls, <strong>of</strong>ten associated with CNV analysis. The final CNV<br />

identification would be based on common consensus calls from all<br />

the used algorithms with minimum 5 probes and 20 kb in size and<br />

quality score for the respective algorithms.<br />

Conclusions: The resulting CNV calls would be subsequently<br />

analyzed at PLINK for rare and common CNVs associated with<br />

schizophrenia.


PP59 GENOME-WIDE ANALYSIS OF RARE COPY<br />

NUMBER VARIANTS IN ATTENTION<br />

DEFICIT/HYPERACTIVITY DISORDER (ADHD)<br />

CONFIRMS THE ROLE OF RARE VARIANTS AND<br />

IMPLICATES DUPLICATIONS AT 15Q13.3 IN DISEASE<br />

ETIOLOGY<br />

B. Franke*(1), N. Williams(2), E. Mick(3), S. Faraone(4), O.<br />

IMAGE II Consortium(4)<br />

1. Departments <strong>of</strong> Human <strong>Genetics</strong> and Psychiatry, Donders Institute<br />

for Brain, Cognition and Behavior, Radboud University Nijmegen<br />

Medical Centre 2. Department <strong>of</strong> Psychological Medicine, School <strong>of</strong><br />

Medicine, Cardiff University 3. Department <strong>of</strong> Psychiatry, Harvard<br />

Medical School, Massachusetts General Hospital 4. Departments <strong>of</strong><br />

Psychiatry and <strong>of</strong> Neuroscience and Physiology, State University <strong>of</strong><br />

New York Upstate Medical University<br />

*b.franke@antrg.umcn.nl<br />

Introduction: ADHD is a common neurodevelopmental psychiatric<br />

disorder with a strong impact on patients’ lives. ADHD is highly<br />

heritable, but due to its multifactorial etiology, identifying the genes<br />

involved has been difficult. Recent findings suggest that rare copy<br />

number variants (CNVs) might be <strong>of</strong> importance for ADHD etiology.<br />

Methodology: The current study performed a genome-wide analysis<br />

<strong>of</strong> rare CNVs (less than 1% population frequency) larger than 100 kb<br />

in size in a large sample <strong>of</strong> children with DSM-IV ADHD (n=896)<br />

and controls (n=2455) from the IMAGE II study.<br />

Results: A total <strong>of</strong> 1562 individually rare CNVs >100 kb were<br />

observed, which segregated into 912 independent loci. Overall, the<br />

rate <strong>of</strong> rare CNVs larger than 100 kb was increased by 1.15-fold in<br />

the ADHD cases compared to controls (p=0.014) with duplications<br />

spanning known genes showing 1.2 fold enrichment (p=0.01).<br />

Moreover, CNVs identified in our ADHD cases were significantly<br />

enriched for loci previously implicated in autism (p=0.009) and<br />

schizophrenia (p=0.03). In accordance with a previous study, rare<br />

CNVs larger than 500 kb showed the greatest level <strong>of</strong> enrichment<br />

(1.28-fold) in ADHD cases compared to controls (p=0.032). Single<br />

locus analysis revealed that a duplication on chromosome 15q13.3<br />

showed a nominally significant association with ADHD (p=0.011)<br />

and this was replicated in a total <strong>of</strong> 3455 ADHD cases and 11105<br />

controls obtained from four independent cohorts from the UK, the<br />

USA and Canada (combined p=0.0018). Exploratory analyses in a<br />

subset <strong>of</strong> patients suggested that the presence <strong>of</strong> the duplication at<br />

15q13.3 increases disease severity and is associated with the presence<br />

<strong>of</strong> conduct disorder.<br />

Conclusions: Our findings support an earlier finding <strong>of</strong> CNV<br />

enrichment in ADHD and implicate duplications at the 15q13.3 locus<br />

as a novel candidate risk factor for ADHD. Due to its frequency <strong>of</strong><br />

0.6% in the populations investigated and its relatively large effect<br />

size (O.R.=1.94 (C.I. 1.28-2.94)), this locus could contribute<br />

substantially to ADHD's etiology.<br />

97<br />

PP60 A GENOME-WIDE APPROACH TO INVESTIGATE<br />

SCHIZOPHRENIA WITH COMORBID EPILEPSY<br />

A. Kähler*(1,2,3), S. Akterin(2), P. Magnusson(2), S. Purcell(4,5,6),<br />

P. Sklar(4,5,6,7), P. Lichtenstein(2), C. Hultman(2), P. Sullivan(1,2)<br />

1. Department <strong>of</strong> <strong>Genetics</strong>, University <strong>of</strong> North Carolina 2.<br />

Department <strong>of</strong> Medical Epidemiology and Biostatistics, Karolinska<br />

Institutet 3. Department <strong>of</strong> Psychiatry, Oslo University Hospital -<br />

Ulleval 4. <strong>Psychiatric</strong> and Neurodevelopmental <strong>Genetics</strong> Unit,<br />

Department <strong>of</strong> Psychiatry, Massachusetts General Hospital and<br />

Harvard Medical School 5. Center for Human <strong>Genetics</strong> Research,<br />

Massachusetts General Hospital 6. Stanley Center for <strong>Psychiatric</strong><br />

Research, Broad Institute 7. Division <strong>of</strong> <strong>Psychiatric</strong> Genomics,<br />

Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong> Medicine<br />

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

Introduction: Schizophrenia is a complex disease, with a heritability<br />

estimate <strong>of</strong> 81%. The last three years have been a success for the field<br />

<strong>of</strong> schizophrenia genetics, with reports <strong>of</strong> robust associations <strong>of</strong> rare<br />

copy number variations (CNVs) and common single nucleotide<br />

polymorphisms (SNPs). Evidence has been provided for a polygenic<br />

architecture for schizophrenia with a risk pr<strong>of</strong>ile significantly<br />

predicting caseness in independent schizophrenia samples but not<br />

non-psychiatric disorder samples. However, schizophrenia is<br />

clinically heterogeneous, and pinpointing more homogeneous<br />

subgroups might lead to new discoveries. There is an increased risk<br />

for epilepsy in patients with schizophrenia and vice versa, and<br />

specific large CNVs have been associated with risk for both diseases.<br />

In the present study we approach the clinical heterogeneity by<br />

assessing schizophrenia patients with and without comorbid epilepsy,<br />

to search for genetic differences.<br />

Methodology: A sample <strong>of</strong> 5,346 schizophrenia cases and 6,491<br />

controls has been collected from a national epidemiological sampling<br />

frame in Sweden. All cases were identified via the Swedish Hospital<br />

Discharge Register, and had been hospitalized ≥2 times with a core<br />

schizophrenia discharge diagnosis. GWAS genotyping is now<br />

available for 4,922 subjects (Affymetrix 6.0) and genotyping will be<br />

completed on the rest <strong>of</strong> the sample by June 2011 (Illumina Omni<br />

Express). Subjects with comorbid epilepsy were identified based on<br />

questionnaire data (~5% <strong>of</strong> schizophrenia cases) and were verified<br />

using discharge and outpatient care diagnoses (ICD8-9: 345; ICD10:<br />

G40-G41). CNV calling was performed using PennCNV. GWAS and<br />

CNV analyses are ongoing.<br />

Results: We will present results evaluating the hypothesis that<br />

epilepsy defines a genetically distinct subgroup <strong>of</strong> cases with<br />

schizophrenia. The assessment will be based on: schizophrenia risk<br />

pr<strong>of</strong>ile scores, CNV burden, and association analyses for specific<br />

SNP and CNV loci.<br />

Conclusions: In our large population-based case-control GWAS<br />

sample we confirm that there is an increased presence <strong>of</strong> epilepsy in<br />

patients with schizophrenia. Our sample will enable us to further<br />

investigate the genetics <strong>of</strong> the subgroup <strong>of</strong> patients with both<br />

disorders which might lead to new insights regarding the etiology <strong>of</strong><br />

schizophrenia.


PP61 INCREASED FREQUENCY OF A COMMON<br />

DELETION IN ERBB4 IN THE IRISH STUDY OF HIGH-<br />

DENSITY SCHIZOPHRENIA FAMILIES<br />

E. Loken*(1), R. Elves(1), B. Webb(2), D. Walsh(3), F. O'Neill(4),<br />

K. Kendler(1,2), B. Riley(1,2)<br />

1. Department <strong>of</strong> Human <strong>Genetics</strong>, Virginia Commonwealth<br />

University 2. Department <strong>of</strong> Psychiatry, Virginia Commonwealth<br />

University 3. Health Research Board 4. Department <strong>of</strong> Psychiatry,<br />

Queens University<br />

*lokenek@vcu.edu<br />

Introduction: Rare copy number variants (CNVs) (eg, in NRXN1)<br />

and recurrent, low frequency CNVs (eg, on 22q) are associated with<br />

schizophrenia. Common CNVs, by contrast, have not been assessed,<br />

although there is clear potential for such CNVs to act as common,<br />

small effect-size variants in the etiology <strong>of</strong> common complex<br />

traits. In CNV calls from a genome-wide association study (GWAS)<br />

<strong>of</strong> Irish multiplex pedigrees, we observed an apparent excess <strong>of</strong> a<br />

common deletion in the first intron <strong>of</strong> ERBB4 when in affected<br />

family members compared to unaffected ones. The total ISHDSF<br />

sample is made <strong>of</strong> 270 families and 1,425 individuals, 854 <strong>of</strong> which<br />

were genotyped on the Illumina 610 Quad array. This common<br />

5,000bp ERBB4 deletion has been identified in several<br />

populations. This is the first study <strong>of</strong> its relationship to<br />

schizophrenia. We directly compared the frequency <strong>of</strong> this common<br />

CNV in 1) genetically independent members <strong>of</strong> the Irish multiplex<br />

pedigrees and population estimates and 2) Irish singleton cases<br />

compared to Irish population controls.<br />

Methodology: Family CNVs were called from Illumina 610 Quad<br />

array probe intensity data using PennCNV. A population sample <strong>of</strong><br />

N=1320 North Americans <strong>of</strong> European descent estimated the ERBB4<br />

CNV frequency at 4.5%. To increase the number <strong>of</strong> such samples,<br />

independent pedigree member identity was determined by using all<br />

possible members <strong>of</strong> the families with independent biologic<br />

relationships. We selected 277 genetically independent pedigree<br />

members for comparison with the population frequency and directly<br />

assessed their copy number status by real-time PCR. Because<br />

differences between the population frequency and the frequency<br />

observed in our sample could be due to ethnic differences, we are<br />

also testing the deletion frequency in an independent sample <strong>of</strong> Irish<br />

cases and Irish controls. We used RNase P amplification for highly<br />

accurate quantitation <strong>of</strong> DNA concentration. We called CNV<br />

genotypes using TaqMan copy number assays from ABI and<br />

CopyCaller S<strong>of</strong>tware v1.0.<br />

Results: Preliminary validation data in the first 111 independent<br />

family members yields a deletion frequency 10.4% compared with<br />

4.5% in the North American population sample (chi-sq=15.17,<br />

p


PP63 A GENOME-WIDE SEARCH FOR GENETIC<br />

MODIFIERS OF ADAPTIVE BEHAVIOR IN MALES WITH<br />

FRAGILE X SYNDROME<br />

A. Kähler*(1,2,3), A. Collins(1), J. Szatkiewicz(1), M. Losh(4), D.<br />

Hatton(5), A. Taylor(6), J. Piven(7), P. Sullivan(1,2)<br />

1. Department <strong>of</strong> <strong>Genetics</strong>, University <strong>of</strong> North Carolina 2.<br />

Department <strong>of</strong> Medical Epidemiology and Biostatistics, Karolinska<br />

Institutet 3. Department <strong>of</strong> Psychiatry, Oslo University Hospital -<br />

Ulleval 4. Department <strong>of</strong> Communication Sciences and Disorders,<br />

Northwestern University 5. Department <strong>of</strong> Special Education,<br />

Vanderbilt University 6. Kimball <strong>Genetics</strong> 7. Department <strong>of</strong><br />

Psychiatry, University <strong>of</strong> North Carolina<br />

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

Introduction: Autism is a severe neurodevelopmental disorder<br />

defined by social and communication deficits and ritualisticrepetitive<br />

behaviors. In ~10% <strong>of</strong> cases, autism co-occurs with a<br />

Mendelian genetic disorder such as Fragile X Syndrome (FXS). FXS<br />

is the most frequent form <strong>of</strong> inherited mental retardation, and is<br />

caused by loss-<strong>of</strong>-function mutations in Fragile X mental retardation<br />

1 (FMR1). Although FXS subjects all have FMR1 mutations, the<br />

degree <strong>of</strong> autism varies dramatically from minimal to moderate or<br />

severe. In the present study, we use FMR1 as a “toehold” for the<br />

identification <strong>of</strong> genetic modifiers <strong>of</strong> adaptive behavior. Given that<br />

all subjects have the same predisposing genetic lesion, searches for<br />

genetic modifiers influencing adaptive behaviors could be<br />

particularly powerful as heterogeneity is minimized.<br />

Methodology: Males with FXS were recruited from volunteer<br />

registries. Phenotypic data were collected using the Vineland<br />

Adaptive Behavior Scales-II (VABS), measuring adaptive behavior<br />

skills (communication, daily living skills, socialization, and motor<br />

skills). GWAS genotyping was conducted using Illumina 1M SNP<br />

chips. The GWAS SNPs were partitioned into a hypothesis-driven set<br />

consisting <strong>of</strong> SNPs in or near genes that code for molecules known to<br />

interact with FMR1 or its protein product and a discovery set (all<br />

other SNPs). Association tests were performed using linear<br />

regression in PLINK with VABS as the dependent variable and age<br />

and ancestry principal components as covariates. CNV calling was<br />

carried out with PennCNV, and pathway analyses with Aligator.<br />

Results: The final GWAS data set consisted <strong>of</strong> 88 FXS males and<br />

733,183 SNPs. There were 16,178 SNPs in 519 genes encoding<br />

molecules that interact with FMR1 or its protein product, and none <strong>of</strong><br />

these reached the significance level for the hypothesis-driven SNP<br />

set. Similarly, no SNP in the discovery set reached genome-wide<br />

significance. However, based on pathway analysis, there was a<br />

significant enrichment <strong>of</strong> genes with small p-values in 16p11.2, a<br />

region containing a CNV previously associated with autism and<br />

schizophrenia. Moreover, around a third <strong>of</strong> subjects had predicted<br />

CNVs covering regions and genes associated with autism, including a<br />

subject with a large 16p11.2 duplication and the largest degree <strong>of</strong><br />

social impairment in the sample. Verification genotyping is in<br />

progress and will be presented.<br />

Conclusions: Based on these preliminary and as yet unverified<br />

findings, these data may be consistent with a “two-hit” model – the<br />

initial hit is the FMR1 mutation and phenotypes <strong>of</strong> clinical<br />

importance are then determined by mutations elsewhere in the<br />

genome (e.g., common variation on 16p11.2 and pathogenic CNVs).<br />

Thus, even “simple” Mendelian disorders have unexpected<br />

complexity in relation to behavioral outcomes.<br />

99<br />

PP64 A STATISTICAL METHOD FOR DETECTING RARE<br />

VARIANTS IN PEDIGREES WITH PSYCHIATRIC<br />

DISORDERS<br />

Y. Shugart*(1), M. Xiong(2)<br />

1. IRP, Stat Genomics, NIMH/NIH 2. Department <strong>of</strong> Epidemiology,<br />

University <strong>of</strong> Texas<br />

*kay1yao@mail.nih.gov<br />

Introduction: The hyphothesis <strong>of</strong> common disorder rare variants<br />

assumes that complex disorders such as psychiatric disorders are<br />

causes by multiple rare variants (assuming MAF less than 5%) with<br />

modest effect. These rare variants are expected to affect protein<br />

function and many <strong>of</strong> them are missense mutations in protein coding<br />

regions. We hypothesize that these rare variants are likely to<br />

modulate gene expression level or change amino acids sequence, and<br />

can sometime lead to human disorders.<br />

Methodology: Recently, a few new statistical methods have emerged<br />

in analyzing exome sequencing data in case-control samples. For<br />

instance, the well-developed CMC method focus on the comparison<br />

<strong>of</strong> number <strong>of</strong> non-synonymous alleles between cases and controls.<br />

However, this type <strong>of</strong> approach can be applied to families data. Here<br />

we propose a simplistic statistical method, which compares the<br />

average <strong>of</strong> rare variants counts in affected relative pairs and unrelated<br />

relative pairs. The estimate from each group is then weighted by<br />

variance calculated from the IBD estimates for various types <strong>of</strong><br />

related pairs. Further, we use the actual IBD sharing to take into<br />

account the dependent relatioship from multiple affected pairs within<br />

the sample pedigree.<br />

Results: Simulation based results indicated that our method worked<br />

reaonably well under a variety <strong>of</strong> genetic models. S<strong>of</strong>tware<br />

development is underway.<br />

Conclusions: It is feasible to use affected relatives and unaffected<br />

relative pairs from the same population to detect rare variants<br />

underlying complex human disorders such as schizophrenia and<br />

bipolar disorder.


PP65 WHOLE GENOME SEQUENCING IN LARGE<br />

PAKISTANI FAMILIES WITH MENTAL ILLNESS<br />

M. Ayub*(1), D. Blackwood(2), K. Mahmood(3), R. Taj(4), A.<br />

Maclean(2), A. Mir(5), F. Naeem(6), J. Knowles(7)<br />

1. University <strong>of</strong> Durham 2. University <strong>of</strong> Edinburgh 3. Arrahma<br />

Hospital 4. Pakistan Institute <strong>of</strong> Medical Sciences 5. International<br />

Islamic University 6. Lahore Institute <strong>of</strong> Research and Development<br />

7. University <strong>of</strong> Southern California<br />

*mohammad.ayub@tewv.nhs.uk<br />

Introduction: Genome wide association studies have successfully<br />

identified many genes which are likely to be associated with<br />

Affective Disorders and Schizophrenia. However a significant<br />

proportion <strong>of</strong> variance remains unexplained. One possible<br />

explanation may be involvement <strong>of</strong> multiple rare variants. Over last<br />

two years a few studies have been published which utilized either<br />

whole genome or exome sequencing to find disease causing<br />

mutations mainly in Mendalian disorders. This approach can<br />

potentially be useful in families with complex disorders. Multiply<br />

affected single families may be a useful resource for study <strong>of</strong> rare<br />

variants utilizing whole genome sequencing.<br />

Methodology: We have collected a number <strong>of</strong> such families from<br />

Pakistan. In these families either Affective Disorder or Schizophrenia<br />

is segregated in apparently Mendalian fashion. We initially<br />

performed genome wide linkage analysis in some <strong>of</strong> these families<br />

and found significant linkage. We are currently performing whole<br />

genome sequencing in selected individuals in some <strong>of</strong> these families.<br />

Results: We shall present initial findings from the sequencing data<br />

from this study.<br />

Conclusions: We shall explore the challenges in interpretation <strong>of</strong><br />

sequencing data in this context.<br />

100<br />

PP66 PHENOTYPING, PREVALENCE, AND GENETIC<br />

SIGNATURE OF POSTPARTUM DEPRESSION IN THE<br />

NESDA GWAS STUDY<br />

S. Meltzer-Brody*(1), B. Penninx(2), I. Jones(3), P. Sullivan(1)<br />

1. University <strong>of</strong> North Carolina at Chapel Hill 2. VU University<br />

Medical Center 3. Cardiff University School <strong>of</strong> Medicine<br />

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

Introduction: Efforts to identify the genetic basis <strong>of</strong> major<br />

depression (MDD) have proven challenging. It is possible that the<br />

highly heterogenous nature <strong>of</strong> MDD has complicated efforts to find<br />

biomarkers and genetic associations. Decreasing heterogeneity by<br />

focusing on a specific sub-type <strong>of</strong> MDD <strong>of</strong>fers significant promise.<br />

Postpartum depression (PPD) is an MDD sub-type occurring within<br />

the first 3 months postpartum and has devastating consequences for<br />

the woman, her children and family. PPD is common with a<br />

prevalence <strong>of</strong> 10-20% and provides a more homogenous subset <strong>of</strong><br />

MDD (females only, age-banded, all exposed to the same<br />

biopsychosocial event). Objectives: To perform phenotyping for PPD<br />

cases in the NESDA GWAS cohort study and to perform an unbiased<br />

screen for a genetic signature <strong>of</strong> PPD.<br />

Methodology: The parent project that supplied subjects for this<br />

GWAS is the Netherlands Study <strong>of</strong> Depression and Anxiety<br />

(NESDA). NESDA is an ongoing longitudinal cohort study designed<br />

to be representative <strong>of</strong> individuals with depressive and anxiety<br />

disorders. Initial recruitment <strong>of</strong> NESDA participants was from<br />

09/2004-02/2007. Participants were aged 18 to 65 years at enrollment<br />

with current DSM-IV MDD diagnosis ascertained by CIDI. Within<br />

these studies, 1992 MDD cases had GWAS genotyping completed.<br />

To determine PPD status, we developed a modified lifetime version<br />

<strong>of</strong> the Edinburgh Postnatal Depression Scale (EPDS) which was<br />

administered during the four year follow-up interview <strong>of</strong> NESDA<br />

(09/2008-02/2011). The modified EPDS contains questions about<br />

lifetime history <strong>of</strong> mood or anxiety changes during pregnancy and<br />

postpartum, timing <strong>of</strong> initial onset <strong>of</strong> symptoms, and history and type<br />

<strong>of</strong> mental health treatment. We defined a positive screen for lifetime<br />

history <strong>of</strong> PPD based on a standard cut-<strong>of</strong>f (total score >12). Based<br />

on PPD phenotyping, we performed a hypothesis generating GWAS<br />

<strong>of</strong> PPD based on 435,291 SNPs.<br />

Results: In the NESDA cohort, 2396 study participants have been<br />

phenotyped for PPD. Data analysis is currently in progress and will<br />

be formally completed by July 2011 and ready to present in<br />

September 2011. We will report: 1) the prevalence <strong>of</strong> lifetime history<br />

<strong>of</strong> PPD; 2) prevalence <strong>of</strong> history <strong>of</strong> a positive screen on the EPDS; 3)<br />

risk factors for PPD that are distinct from MDD such as age cohort<br />

effect, marital status, SES, severity <strong>of</strong> illness, parity status, clinical<br />

outcome, and history <strong>of</strong> trauma and abuse; 4) timing <strong>of</strong> onset <strong>of</strong><br />

perinatal mood symptoms (pregnancy vs postpartum); 5) GWAS<br />

analyses based on PPD phenotyping.<br />

Conclusions: PPD is a severe and persistent form <strong>of</strong> MDD that is one<br />

<strong>of</strong> the most frequent complications <strong>of</strong> childbirth. Understanding the<br />

phenotypic characteristics and risk factors that distinguish PPD from<br />

MDD as well as elucidating the genomic signature <strong>of</strong> PPD has<br />

important and far reaching implications that could provide insight<br />

into trait-related vulnerabilities to MDD and potentially lead to future<br />

prospective identification <strong>of</strong> women at risk for developing PPD.


PP67 SEQUENCING HUNDREDS OF EXOMES IN SWEDISH<br />

SCHIZOPHRENIA PATIENTS AND CONTROLS<br />

D. Ruderfer(1,2), M. Fromer(1,2), J. Moran(2), K. Chambert(2), P.<br />

Lichtenstein(3), P. Sullivan(4), C. Hultman(3), P. Sklar(1,2,5), S.<br />

Purcell(1,2)<br />

1. Massachusetts General Hospital 2. Stanley Center, Broad Institute<br />

3. Karolinska Institutet 4. University <strong>of</strong> North Carolina 5. Mount<br />

Sinai School <strong>of</strong> Medicine<br />

Introduction: Schizophrenia is common, complex disease with a<br />

strong genetic basis. Although recent genome-wide studies have<br />

indicated important roles for both common single nucleotide<br />

polymorphisms (SNPs) and large, rare genic deletions and<br />

duplications, the majority <strong>of</strong> the heritability remains unexplained.<br />

Methodology: Next-generation sequencing (NGS) technologies can<br />

interrogate genetic variation that has previously eluded detection in<br />

genome-wide studies, namely rare SNPs and small<br />

insertions/deletions. We aim to relate this class <strong>of</strong> rare genetic<br />

variation to risk for schizophrenia by applying NGS to large,<br />

carefully-selected samples <strong>of</strong> cases and controls. We have selected<br />

959 samples for whole-exome sequencing from a larger samples <strong>of</strong><br />

over 5,000 Swedes on whom whole-genome SNP data are available.<br />

Results: To date, we have sequenced 559 individuals at high depth,<br />

targeting CCDS and RefSeq genes with the Agilent SureSelect target<br />

sets. Solexa/Illumina GAII 76-base paired-end reads were aligned to<br />

hg19 using BWA and the Picard pipeline. Mean coverage was 140x<br />

and 97% <strong>of</strong> target bases were covered at 10x or more. Using the<br />

Genome Analysis Toolkit (GATK) we called a total <strong>of</strong> 173,298<br />

variant sites passing all quality-control filters, with a mean<br />

transition/transversion ratio <strong>of</strong> 3.1. Comparison with GWAS calls at<br />

~5000 exonic sites shows very high genotypic concordance<br />

(>99.9%). Per individual, ~94.4% <strong>of</strong> variants with a non-reference<br />

genotype were previously observed in the 1000 Genomes Project.<br />

Using a second variant-calling approach (Samtools), 87% <strong>of</strong> sites<br />

overlapped with GATK calls; the consensus set demonstrated a<br />

higher Ti/Tv (3.23) than the calls specific to a single method (1.66<br />

and 1.68 for GATK and Samtools, respectively).<br />

Conclusions: We have developed PLINK/Seq, an analytic toolset to<br />

handle downstream QC and association analysis <strong>of</strong> the genetic<br />

variation data from NGS studies. Genotype-phenotype association<br />

analysis is ongoing, at the single variant, gene and gene-set<br />

(pathway/network) levels. The main focus <strong>of</strong> this presentation will be<br />

to present the primary association results from this study and discuss<br />

the some <strong>of</strong> the analytic challenges inherent in these studies.<br />

101<br />

PP68 MICRODELETIONS OF PROXIMAL 15Q11.2<br />

ASSOCIATED WITH SCHIZOPHRENIA<br />

T. Wassink*, D. Rudd, M. Axelsen, N. Andreasen<br />

University <strong>of</strong> Iowa<br />

*thomas-wassink@uiowa.edu<br />

Introduction: Genomic copy number variation has been found to<br />

underlie a significant number <strong>of</strong> cases <strong>of</strong> neuropsychiatric disorders.<br />

Interestingly, most neuropsychiatric CNVs increase risk for multiple<br />

disorders. Chromosome 15q11-13 is emblematic <strong>of</strong> this, with CNVs<br />

across the interval being associated with Prader-Willi/Angelman<br />

syndrome, autism, and other psychiatric phenotypes. The region<br />

contains four canonical segmental duplications that predispose to<br />

CNV formation, but only recently have disease-associated CNVs<br />

been reported between the two most proximal breakpoints (BP1 and<br />

BP2). We report here a CNV study <strong>of</strong> schizophrenia in which we<br />

identified multiple 15q11.2 BP1-BP2 deletions. These findings are<br />

supplemented by additional cases from our clinical cytogenetics<br />

laboratory.<br />

Methodology: 181 individuals with schizophrenia and 69 controls<br />

were studied with the Affymetrix Array 6.0. Array data was scanned<br />

for CNVs using two s<strong>of</strong>tware programs: CNAG and Genotyping<br />

Console. CNVs identified by both programs were carried forward for<br />

analysis. Participants had also undergone extensive neuropsychiatric<br />

testing and structural brain magnetic resonance imaging. CNVs were<br />

compared between affecteds and controls and also with CNVs<br />

recorded in the Database <strong>of</strong> Genomic Variants. CNVs unique to<br />

schizophrenia were confirmed by qPCR and were then examined for<br />

relationships with our various quantitative phenotypic measures.<br />

Results: Amongst the numerous CNVs that were called, we<br />

identified three schizophrenia cases with 15q11.2 BP1-BP2 deletions;<br />

these have not been previously reported in schizophrenia. These cases<br />

were supplemented by findings from our clinical cytogenetics<br />

laboratory. Using the Nimblegen 385,000 probe array, we identified<br />

two individuals with both 15q11.2 and 22q11 deletions. One <strong>of</strong> these<br />

had childhood onset schizophrenia, while the other was diagnosed<br />

with developmental delay. Examination <strong>of</strong> quantitative phenotypic<br />

data from these individuals identified numerous interesting brain and<br />

cognitive features.<br />

Conclusions: We report a high rate (3/181=1.7%) <strong>of</strong> 15q11.2<br />

deletions in individuals with schizophrenia. This CNV, like others<br />

associated with neuropsychiatric phenotypes, appears to underlie<br />

multiple disorders. The CNV may also be associated with distinct<br />

brain and cognitive features.


PP69 PARENT OF ORIGIN EFFECT (POE) ANALYSIS OF<br />

SEROTONERGIC AND NORADRENERGIC<br />

POLYMORPHISMS IN MAJOR PSYCHOSES PATIENTS<br />

WITH SUICIDAL BEHAVIOUR<br />

C. Teo, C. Zai, J. Strauss, J. Kennedy, V. De Luca<br />

CAMH<br />

Introduction: Suicidality is a major health concern worldwide<br />

particularly in major psychoses patients. Attempted suicide is<br />

familiar. There is strong neurobiological evidence showing that<br />

serotonergic and noradrenergic dysfunction is implicated in suicidal<br />

behaviours.<br />

Methodology: By the mean <strong>of</strong> ETDT and QTDT, we performed a<br />

POE analysis on 471 families and we analyzed six genes in regards <strong>of</strong><br />

paternal and maternal transmissions. Then we investigated<br />

polymorphisms in serotonergic and noradrenergic genes, considering<br />

suicidal behaviour severity and as dichotomous phenotype (presence<br />

<strong>of</strong> suicide attempt).<br />

Results: The six polymorphisms (HTTLPR, rs25531, VNTR-2,<br />

COMT rs4680, TH tetranucleotide repeat and TPH2 rs 1078941)<br />

were not associated with suicide in paternal and maternal<br />

transmissions. On the other hand, the analysis <strong>of</strong> the POE markers in<br />

the adrenergic and serotonergic genes revealed that the TH allele 6<br />

tended toward high severity in suicidal behaviour (p=0.060).<br />

Conclusions: The marginal finding <strong>of</strong> association between TH and<br />

severe suicidal behaviour are convergent with previous reports. On<br />

the other hand, our sample does not have enough power to exclude<br />

the other polymorphisms investigated as major candidate for<br />

suicidality in major psychoses.<br />

102<br />

PP70 SEQUENCING BASED COMPREHENSIVE GENOME<br />

AND TRANSCRIPTOME ANALYSES OF<br />

VELOCARDIOFACIAL SYNDROME<br />

A. Urban*(1), Y. Zhang(2), X. Zhu(1), D. Levinson(1), S.<br />

Weissman(2)<br />

1. Stanford University School <strong>of</strong> Medicine 2. Yale University School<br />

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

*aeurban@stanford.edu<br />

Introduction: Velocardi<strong>of</strong>acial syndrome (VCFS) is a relatively<br />

common genomic disorder, typically caused by the heterozygous<br />

deletion <strong>of</strong> 3 million basepairs <strong>of</strong> genomic DNA sequence on<br />

chromosome 22q11. There are various developmental defects<br />

including neurodevelopmental disorders such as early onset<br />

schizophrenia, autism spectrum disorders (ASD) and learning<br />

disabilites; however, the combinations and severity <strong>of</strong> symptoms vary<br />

widely between patients with no clear genetic basis to such<br />

variability, and for most symptoms there is no clear candidate gene.<br />

Earlier [Urban, Korbel et al., PNAS 2006], we developed very-high<br />

resolution array-CGH and used it to determine the exact breakpoints<br />

<strong>of</strong> a panel <strong>of</strong> VCFS deletions, even in regions <strong>of</strong> segmental<br />

duplication (SD). We discovered that even ‘typical 3 Mbp’ deletions<br />

can differ in their endpoints by up to several hundred thousand bp in<br />

length, affecting the copy number <strong>of</strong> many genes between patients.<br />

Then we developed a method to use the new massively parallel<br />

sequencing platforms (2 nd -generation sequencing, “next-generation<br />

sequencing”) for the mapping <strong>of</strong> structural variation in the human<br />

genome (paired-end mapping, PEM, now a standard component <strong>of</strong><br />

high throughput human whole-genome sequencing) [Korbel, Urban,<br />

Affourtit et al., Science 2007]. We found hundreds <strong>of</strong> small to<br />

medium size sequence variations in the normal human genome. Many<br />

<strong>of</strong> those are in functionally relevant regions genome wide – and also,<br />

additional smaller sequence variants are enriched in 22q11.<br />

Methodology: These findings point to several genomic constellations<br />

as possible modifiers <strong>of</strong> the main 3 Mbp deletion on chromosome<br />

22q11: difference in deletion endpoints, small sequence variants<br />

within the deletion region on the non-deleted chromosome, and<br />

sequence variation genome wide. Here we report on multi-level<br />

genomics analyses <strong>of</strong> VCFS using 2 nd -generation sequencing<br />

(Illumina HiSeq 2000). We have sequenced the genomes <strong>of</strong> a panel<br />

<strong>of</strong> cell lines from VCFS patients (paired-end, 4x or deeper genomic<br />

coverage) and catalogued their genomic sequence variants from 1 bp<br />

to several hundreds <strong>of</strong> kbp in size. For the same cell lines as well as<br />

for controls we carried out very deep RNA-Seq analysis (up to 100<br />

million mapped 2x100bp paired-end constructs per cell line<br />

transcriptome). These data allow us to correlate with very high<br />

accuracy the effects on gene expression patterns <strong>of</strong> both the copy<br />

number changes caused by the main deletion and the additional,<br />

patient specific, genomic sequence variants. Results: We observe<br />

multiple small sequence variants, in the non-deleted 22q11 regions as<br />

well as genome-wide, with some variants potentially affecting loci <strong>of</strong><br />

functional significance. The gene expression patterns show evidence<br />

<strong>of</strong> gene dosage compensation for some genes as well as under- or<br />

over-compensation for others. The mechanisms controlling such<br />

effects will have to be studied in detail in follow-up work.<br />

Conclusions: 2 nd - generation sequencing technologies allow the<br />

analysis <strong>of</strong> the genetic and molecular etiology <strong>of</strong> complex<br />

neurodevelopmental disorders with a strong genomic contribution, at<br />

the level <strong>of</strong> resolution and the degree <strong>of</strong> comprehensiveness that will<br />

most likely be necessary to unravel the network <strong>of</strong> molecular<br />

interactions leading to the phenotype.


PP71 MULTIPLE NOVEL SMALL RNAS ASSOCIATED<br />

WITH SCHIZOPHRENIA FROM MASSIVELY PARALLEL<br />

SEQUENCING FROM PREFRONTAL CORTEX<br />

C. Jeffries*(1), D. Perkins(2)<br />

1. UNC Eshelman School <strong>of</strong> Pharmacy, Renaissance Computing<br />

Institute 2. Department <strong>of</strong> Psychiatry<br />

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

Introduction: Altered regulation <strong>of</strong> gene expression is emerging as<br />

pivotal to schizophrenia pathology. Discovery <strong>of</strong> small, noncoding,<br />

regulatory RNAs has ushered in a new era <strong>of</strong> genomics, with<br />

microRNAs (miRNAs) the first major class; several groups have<br />

reported altered miRNA expression in schizophrenia. Numerous<br />

other small RNA classes have been recently discovered, some<br />

seemingly arising from folded RNA hairpins and processed in a<br />

miRNA-like manner<br />

Methodology: In order to discover novel potentially regulatory<br />

RNAs involved in schizophrenia we used Illumina small RNA<br />

sequencing to interrogate total RNA from the post-mortem prefrontal<br />

cortex <strong>of</strong> ten schizophrenia and ten unaffected individuals from the<br />

Stanley RNA collection. After sample processing we compared<br />

unique reads against a library <strong>of</strong> 1222 human miRNAs, 182 viral<br />

miRNAs, and 579 other noncoding small RNAs<br />

Results: We compared the expression <strong>of</strong> the 100 most highly<br />

detected probes (in average >80 reads per sample) between groups.<br />

With a false discovery rate <strong>of</strong> .05, we found 14 small RNAs<br />

differentially expressed in schizophrenia compared to unaffected<br />

subjects, with an expected value <strong>of</strong> 5. Of the 14, 10 were miRNAs<br />

and 4 other small RNAs<br />

Conclusions: About a third <strong>of</strong> the most differentially expressed<br />

probes represented miRNAs, some already identified as associated<br />

with schizophrenia (e.g. miR-132). We report for the first time small<br />

RNAs other than miRNAs that also distinguish schizophrenia,<br />

anomalous levels <strong>of</strong> which might be manifestations <strong>of</strong> the disorder<br />

103<br />

PP72 ALTERED GENE EXPRESSION IN BIPOLAR I<br />

PATIENTS (WITH AND WITHOUT PSYCHOSIS)<br />

J. Neary*(1), D. Glahn(2), M. Zlojutro(1), T. Dyer(1), J.<br />

Blangero(1), M. Carless(1)<br />

1. Texas Biomedical Research Institute 2. Yale University School <strong>of</strong><br />

Medicine<br />

*jneary@sfbrgenetics.org<br />

Introduction: Bipolar disorder, a mood disorder delineated by<br />

drastic swings between high and low mood states, is estimated to<br />

manifest in 1-3% <strong>of</strong> the general population. However, close relatives<br />

<strong>of</strong> bipolar individuals are 10-20x times more likely to develop either<br />

depression or bipolar disorder, indicating significant heritability. Full<br />

understanding <strong>of</strong> the developmental mechanisms involved has thus<br />

far been hindered by the variability <strong>of</strong> clinical symptoms as well as<br />

associated genetic complexity. Here we focus on gene expression<br />

changes associated with bipolar I disorder (BPI) which is defined by<br />

at least one manic episode, with or without depression.<br />

Methodology: In our <strong>Genetics</strong> <strong>of</strong> Bipolar Disorder Study, we are<br />

presently collecting and associating neuroanatomic (MRI) data,<br />

neurocognitive data, and lymphocyte-derived RNA for 325<br />

individuals: 130 affected with BPI (65 with history <strong>of</strong> psychosis),130<br />

unaffected same-sex siblings and 65 unrelated controls. Here we<br />

report the results <strong>of</strong> genome-wide transcriptional pr<strong>of</strong>iles for the first<br />

30 BPI patients (15 with psychosis, 15 without), 25 unaffected<br />

siblings and 15 control subjects. For genes demonstrating a >5-fold<br />

difference in expression, we utilized Ingenuity Pathway Analysis.<br />

Results: In BPI patients versus controls, we found significant overrepresentation<br />

<strong>of</strong> genes involved in microglia cell binding<br />

(p=5.43x10 -3 ), including APP, IFNG, TNF and TNR. These results<br />

augment other reports linking microglial dysfunction to the<br />

manifestation <strong>of</strong> psychiatric disorders. Further analysis <strong>of</strong> BPI in<br />

psychotic versus non-psychotic BPI patients also indicated overrepresentation<br />

<strong>of</strong> genes involved with excretion <strong>of</strong> nitrates, release <strong>of</strong><br />

norepinephrine (NE), and release <strong>of</strong> reactive oxygen species (AGT,<br />

CCL24, HTR2A, IFNG, SST and TNF, p=7.64x10 -3 ). While<br />

pathways involving NE have been previously linked to psychosis and<br />

cognitive function, pathways involving reactive oxygen species may<br />

represent a potentially novel target for further study and therapeutic<br />

intervention.<br />

Conclusions: Here we present analyses <strong>of</strong> initial data generated by<br />

the <strong>Genetics</strong> <strong>of</strong> Bipolar Disorder study, which is still in progress. We<br />

are presently analyzing collected neuroanatomic and neurocognitive<br />

data, as well as miRNA transcripts and epigenetic markers for 325<br />

people in order to identify genes implicated in the development <strong>of</strong><br />

BPI-related endophenotypes.


PP73 SYNAPTOMEDB: AN ONTOLOGY-BASED<br />

KNOWLEDGEBASE FOR SYNAPTIC GENES<br />

M. Pirooznia*(1), T. Wang(2), D. Avramopoulos (2), D. Valle(2), G.<br />

Thomas(3), R. Huganir(3), P. Zandi(4), J. Potash(1)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Sciences, Johns Hopkins<br />

University 2. McKusick-Nathans Institute <strong>of</strong> Genetic Medicine,<br />

Johns Hopkins School <strong>of</strong> Medicine, Johns Hopkins University 3.<br />

Solomon H. Snyder Department <strong>of</strong> Neurocience, Johns Hopkins<br />

University 4. Department <strong>of</strong> Mental Health, Johns Hopkins<br />

Bloomberg School <strong>of</strong> Public Health, Johns Hopkins University<br />

*mpirooz1@jhmi.edu<br />

Introduction: The synapse is the fundamental structure for brain<br />

function through its role in connecting neurons into circuits. Over the<br />

past decade, the number <strong>of</strong> identified synaptic proteins has increased<br />

by ten fold, and the synapse has been proposed as an excellent<br />

candidate for large-scale studies such as second-generation<br />

sequencing. As a result, there is a need for a comprehensive resource<br />

to integrate information about synaptic proteins and the<br />

corresponding genes (the “synaptome”) from multiple heterogeneous<br />

sources. Here, we present an integrated database to retrieve, compile,<br />

and annotate genes encoding components <strong>of</strong> the synapse including<br />

neurotransmitters and their receptors, adhesion/cytoskeletal proteins,<br />

scaffold proteins, transporters, and others.<br />

Methodology: We compiled the human synaptome protein list from<br />

all published proteomics studies, including literature reviews, from<br />

2004 to 2010, as well as publicly available databases, for proteins in:<br />

postsynaptic, presynaptic, presynaptic active zone, and synaptic<br />

vesicle sites. We annotated the synaptome genes based on RefSeq<br />

(GRCh37/hg19), supplemented by data from UCSC (hg19) to<br />

identify human orthologs, their descriptions, and mapping<br />

data. Redundant genes were removed from the list to generate a<br />

unique gene set for the human synaptome. We then further annotated<br />

these genes by querying 42 databases covering all aspects <strong>of</strong> biology,<br />

including genes, proteins, pathways and other biological concepts.<br />

Results: We assembled a list <strong>of</strong> genes (n=1885) that encode all<br />

known proteins <strong>of</strong> the synapse. This is comprised <strong>of</strong> 575 genes<br />

encoding proteins in the presynaptic nerve terminal and active zone,<br />

107 from the synaptic vesicles, and 1755 from the postsynaptic<br />

density (there is some overlap between categories). The list includes<br />

strong candidates for bipolar disorder, such as ANK3, for major<br />

depression, such as GRM7, for schizophrenia, such as PDE4B, and<br />

for autism, such as SHANK3. We have developed a database with a<br />

web front application resource, called SynaptomeDB, to integrate the<br />

various and complex data sources for these synaptic genes. Current<br />

developments in the project are freely available at:<br />

http://psychiatry.igm.jhmi.edu/SynaptomeDB/<br />

Conclusions: A synaptome-based strategy for psychiatric research is<br />

valuable because there is evidence for synaptic proteins playing a role<br />

in psychiatric disorders, and because these proteins represent the<br />

most “druggable” targets for pursuit <strong>of</strong> novel therapies. Our<br />

application will further research in this area both in its current form<br />

and with additional modifications that will include incorporating<br />

variation data from the 1000 Genome Project and adding links to<br />

function based on GO and pathways.<br />

104<br />

PP74 MUTATION Y317X OF SLC6A8 GENE IS A POSSIBLE<br />

POLYMORPHISM PRESENT IN PATIENTS WITH MENTAL<br />

RETARDATION OF PSYCHIATRIC HOSPITAL "DR.<br />

SAMUEL RAMIREZ MORENO”<br />

C. Jiménez Z*(1), M. López N(1,2), R. Castro R(2), M. Martínez<br />

G(3), A. Miliar G(3)<br />

1. Secretaria de Salud 2. Instituto de Salud del Estado de Mexico 3.<br />

Instituto Politecnico Nacional<br />

*carlosajz@msn.com<br />

Introduction: Mental disorders account for 12% <strong>of</strong> the global burden<br />

<strong>of</strong> all diseases. Mental retardation (MR) is one <strong>of</strong> them and is defined<br />

as a disorder characterized by impairment <strong>of</strong> intellectual capacity<br />

which is below the average (IQ 70 or lower), with an age <strong>of</strong> onset<br />

before or equal to 18 years and concurrent deficits or inadequacies in<br />

adaptive functioning. Among the causes <strong>of</strong> MR, 30% seems to be<br />

genetic (chromosomal, monogenic and multifactorial) and 15% has<br />

been attributed to environmental factors (such as birth asphyxia and<br />

infections). The rest <strong>of</strong> cases (over 50%) have an unknown etiology,<br />

although the Human Genome Project recently completed provides<br />

information on new mutations in genes responsible for the<br />

appearance <strong>of</strong> this disability, is necessary the studies about this<br />

disease. In the same types syndromes, have been described cerebral<br />

creatine deficiency, due to inborn errors <strong>of</strong> metabolism that include<br />

two autosomal recessive disorders that affect the biosynthesis <strong>of</strong><br />

creatine, this has been linked to various mutations from SCL6A8<br />

gene, including the Y317X.<br />

Methodology: Genomic DNA was obtained from 110 patients by<br />

establishing a database. Although HybeProbe type probes were<br />

genotyped each sample and sequenced to corroborate some <strong>of</strong> the<br />

genotyping results. Was performed the statistical analysis <strong>of</strong> data<br />

derived from medical, psychiatric and nutritional diagnosis, as well<br />

as serum glucose, cholesterol, triglycerides, sodium, between others.<br />

Results: After performing the genotyping <strong>of</strong> the Y317X mutation in<br />

the DNA <strong>of</strong> 110 patients was not observed allelic variant but we<br />

could identify a different variation to the sequence reported in<br />

GeneBank confirmed by sequencing. On the other hand could not be<br />

correlated with other parameters obtained from the patients, but the<br />

study population showed no alterations in metabolic rate over 50%<br />

were in normal weight ranges. It was also observed different degrees<br />

<strong>of</strong> mental retardation.<br />

Conclusions: The Y317X mutation in the SLC6A8 gene reported in<br />

other populations and associated with mental retardation is not<br />

present in the patients <strong>of</strong> the psychiatric hospital "DR. SAMUEL<br />

RAMIREZ MORENO. The same patient population allele has a<br />

change <strong>of</strong> C/T so far not reported in other populations. There are<br />

different degrees <strong>of</strong> mental retardation in the study subjects but does<br />

not correlate with metabolic and anthropometric variables.


PP75 REDUCED FUNCTIONAL VARIATION AMONG<br />

NERVOUS SYSTEM GENES: IMPLICATIONS FOR<br />

GENETIC DISEASE ARCHITECTURE<br />

J. Freudenberg*(1), P. Gregersen(1), Y. Freudenberg-Hua(2)<br />

1. Center for Human <strong>Genetics</strong> and Genomics Feinstein Institute for<br />

Medical Research 2. Center for Human <strong>Genetics</strong> and Genomics<br />

Feinstein Institute for Medical Research 3. Department <strong>of</strong> Psychiatry,<br />

Northshore LIJ Healthsystem<br />

*jan.freudenberg@nslij-genetics.org<br />

Introduction: We notice that human genetic studies have identified<br />

more loci that cause monogenic nervous system than monogenic<br />

immune system diseases. On the other hand, relatively few<br />

susceptibility genes for complex neuropsychiatric disorders are<br />

unambiguously established, whereas far more solidly associated loci<br />

exist for immunological disorders. Understanding the genetic<br />

architecture causing these differences can improve the design <strong>of</strong><br />

future studies.<br />

Methodology: Therefore, we evaluate the patterns <strong>of</strong> coding single<br />

nucleotide variants (SNVs) among candidate genes with roles in the<br />

immune system and the nervous system, using a published diploid<br />

genome sequence generated by traditional Sanger technology and 200<br />

published exomes <strong>of</strong> Danish individuals generated by next generation<br />

sequencing.<br />

Results: In both datasets, we find that nonsynonymous variants<br />

(nsSNVs) are significantly less frequent in nervous system genes,<br />

despite their longer sequences. This applies both to candidate genes<br />

defined by their specific expression pattern and defined by their<br />

functional annotations. Based on the observed ratio between nsSNVs<br />

per nonsynonymous site (nsSite) and synonymous SNVs per<br />

synonymous site, we subsequently estimate that nervous system<br />

genes are composed <strong>of</strong> more strongly deleterious nsSites, which may<br />

contribute to more monogenic disease manifestations. In addition, we<br />

estimate that they contain equal or more mildly deleterious nsSites, as<br />

compared with immune system or random genes. Accordingly, a<br />

wider spread <strong>of</strong> susceptibility alleles over larger-sized gene regions,<br />

potentially combined with lower allele frequencies, could contribute<br />

to a reduced yield <strong>of</strong> association studies.<br />

Conclusions: One consequence could be a greater required effort for<br />

imputing susceptibility alleles.<br />

105


POSTER SESSION II<br />

ABSTRACTS<br />

106


ELSI, COUNSELING, AND GENETIC TESTING<br />

PP76 DO PARTICIPANTS IN SEQUENCING STUDIES WISH<br />

TO BE INFORMED OF THEIR RESULTS?<br />

E. Bui, L. Kassem*, F. McMahon<br />

Human <strong>Genetics</strong> Branch, Intramural Research Program, National<br />

Institute <strong>of</strong> Mental Health/National Institutes <strong>of</strong> Health<br />

*kasseml@mail.nih.gov<br />

Introduction: As whole-genome sequencing technology rapidly<br />

advances, the debate surrounding the reporting <strong>of</strong> individual results to<br />

participants increases, introducing many ethical and logistical<br />

issues. While these issues are being debated by bioethicists and<br />

researchers, little is known about actual participants’ opinions.<br />

Methodology: 59 volunteers were identified through an ongoing<br />

bipolar disorder genetics study and surveyed using a questionnaire<br />

we developed, the "Genome Sequencing Attitudes Survey<br />

(GSAS).” Of study participants contacted, 70% agreed to participate<br />

in the Genome Sequencing Attitudes Survey. The GSAS covers<br />

three main areas: what information participants would like to receive<br />

should their DNA be sequenced, what they would do with that<br />

information, and any concerns about privacy they may have. All<br />

interviews were conducted over the telephone by a single interviewer.<br />

Results: The findings suggest a large majority <strong>of</strong> study volunteers<br />

(97%) are interested in participating in sequencing studies. A large<br />

proportion (86%) would like to be fully informed about their genetic<br />

risks for various diseases, including those that are not actionable.<br />

96% <strong>of</strong> respondents would consent to their anonymized personal data<br />

being shared with other researchers. 41% <strong>of</strong> respondents also<br />

expressed concerns about their insurance companies and other<br />

entities accessing their personal genomic data, but would still be<br />

interested in participating nonetheless.<br />

Conclusions: Participants in genetics studies are very interested in<br />

participating in detailed genome sequencing experiments and most<br />

would appreciate or even expect to be <strong>of</strong>fered individual results. The<br />

public's knowledge about genetic risks, however, is quite lacking.<br />

Researchers need to take these limitations into account when<br />

providing individual results, and also should prepare to better educate<br />

potential research participants about the implications <strong>of</strong> having this<br />

information.<br />

107<br />

PP77 IDENTIFICATION OF CHROMOSOMAL<br />

ALTERATIONS IN AUTISM SPECTRUM DISORDER’S<br />

PATIENTS, TAMILNADU POPULATION, INDIA<br />

M. Arun*, V. Balachandar, K. Sasikala<br />

Human Molecular <strong>Genetics</strong> Laboratory, School <strong>of</strong> Life Sciences,<br />

Bharathiar University<br />

*arun47biotech@gmail.com<br />

Introduction: Autism is a complex neurobiological disorder that<br />

typically lasts throughout a person's lifetime. It is part <strong>of</strong> a group <strong>of</strong><br />

disorders known as autism spectrum disorders (ASD). It is <strong>of</strong>ten<br />

associated with other conditions, such as disorders <strong>of</strong> the CNS<br />

(tuberous sclerosis), developmental delay, attention deficit, epilepsy,<br />

and anxiety and mood disorders. A reasonable extraction <strong>of</strong> the<br />

overall data when applied to the male population finds 1 in 93 males<br />

(54 per 10,000) are likely affected with autism. At the current time,<br />

the exact causes <strong>of</strong> autism remain elusive, but researchers<br />

increasingly believe that both genetics and environment play a role.<br />

The prime aim <strong>of</strong> the present study was to identify the chromosomal<br />

aberrations with autism patients in Coimbatore region.<br />

Methodology: In order to investigate the possible cytogenetic<br />

damage in autism patients, peripheral blood lymphocyte culture<br />

(PBLC) method was carried out on the lymphocytes <strong>of</strong> 39 autism<br />

patient samples and equal number <strong>of</strong> controls was selected, based on<br />

the detailed questionnaire.An autism child has been followed up<br />

regularly at frequent intervals <strong>of</strong> 4-6 weeks both by the paediatrician<br />

and child psychologist. Parameters during follow up included enquiry<br />

into the parents' assessment <strong>of</strong> the child as a whole, level <strong>of</strong><br />

hyperactivity after behavioural and pharmacological intervention.<br />

Moreover, the present investigation has been conducted in the year <strong>of</strong><br />

2008 to 2011.In the present study volunteers provided blood samples<br />

(5 ml) to establish cell cultures at 72 h. For karyotyping, 100<br />

complete metaphase cells from each subject were evaluated using<br />

Trypsin – Giemsa Banding method.<br />

Results: In our study chromosomal alterations were frequently<br />

observed in chromosomes 2, 3, 7, 16 and X (2q32, 3q25-q27, 7q31q35,<br />

15q11-q13, 16p13, Xp22, and Xq13). The chromosomal<br />

aberration frequency among 39 subjects was: 2/39 supernumerary<br />

markers 7/39 deletions 1/39 duplication 1/39 inversions 2/39<br />

translocations. In the present study, chromosomal aberrations showed<br />

higher degree in experimentals compared to controls (P


PP78 BIOMARKERS IN PSYCHIATRY: FROM GENETICS<br />

TO BIOLOGY TO PREDICTIVE MEDICINE<br />

A. Niculescu*<br />

Indiana University School <strong>of</strong> Medicine<br />

*anicules@iupui.edu<br />

Introduction: Biomarkers are at the interface <strong>of</strong> genetic research and<br />

practical applications.<br />

Methodology: Functional genomics has emerged as a useful partner<br />

to classic genetic approaches, and combined approaches may provide<br />

shortcuts to discovery <strong>of</strong> genes and overall understanding <strong>of</strong> the<br />

neurobiology involved.<br />

Results: I will present multi-modal data from our group showing<br />

how panels <strong>of</strong> markers identified through Convergent Functional<br />

Genomics have predictive ability in independent cohorts, the key<br />

litmus test for any genetic or biomarker study. Specifically, I will<br />

present data on trait genetic testing for bipolar disorder, and state<br />

blood biomarker testing for mood, hallucinations, delusions and<br />

anxiety.<br />

Conclusions: Lastly, I will describe prototypes <strong>of</strong> how such testing<br />

could be used to categorize disease risk in individuals and aid<br />

personalized medicine approaches in psychiatry.<br />

108<br />

PP79 ETHNIC DISPARITIES IN THE PERCEPTION OF<br />

ETHICAL RISKS FROM PSYCHIATRIC GENETIC STUDIES<br />

E. Nwulia*(1), M. Hipolito(1), W. Lawson(1), J. Nurnberger Jr(2)<br />

1. Howard University 2. Howard University 3. Howard University 4.<br />

Indiana University<br />

*enwulia@howard.edu<br />

Introduction: Participation <strong>of</strong> ethnic minorities, particularly Blacks<br />

in psychiatric genetic studies have been poor. Existing literature<br />

from studies <strong>of</strong> convenient community samples suggest that mistrust<br />

<strong>of</strong> biomedical researchers, based on the historical legacy <strong>of</strong> the<br />

Tuskegee study (Schultz et al., 2003), fear <strong>of</strong> harm (Murphy et al.,<br />

2009), wariness <strong>of</strong> ultimate uses <strong>of</strong> information, e.g. misapplication<br />

<strong>of</strong> genetic data to explain racial differences in intelligence (Hernstein<br />

and Murray 1994), mental illness stigma and associated<br />

misperception <strong>of</strong> etiology <strong>of</strong> mental illness (Furr et al., 2002) are<br />

some <strong>of</strong> the potential impediments to representation <strong>of</strong> Blacks in<br />

research. However, it is unknown if these concerns persist among<br />

participants in an actual psychiatric genetic study.<br />

Methodology: Research participants in the Bipolar Genome Study<br />

(BIGS) were examined on six items <strong>of</strong> concerns in the Questionnaire<br />

on Genetic Risk (QGR). Responses from non-Hispanic Black<br />

(N=188) and White participants (N=1065) formed the base for this<br />

analysis. Polytomous multinomial models were specified for analysis<br />

<strong>of</strong> the likert-response format <strong>of</strong> participants concerns. Adjusting for<br />

demographic variables, the independent association <strong>of</strong> ethnicity to<br />

each <strong>of</strong> the six perceived concern items were examined using<br />

multivariate multinomial logistic regression.<br />

Results: Concerns about unfavorable consequences <strong>of</strong> conducting<br />

psychiatric genetic study were prevalent in the whole sample. Blacks<br />

expressed stronger concerns for all consequences except for medical<br />

insurance (P


EPIGENOMICS<br />

PP80 DISRUPTION OF EARLY MATERNAL CARE IN<br />

RHESUS MACAQUES RESULTS IN ALTERED EPIGENETIC<br />

REGULATION OF THE OXYTOCIN RECEPTOR (OXTR)<br />

GENE<br />

M. Baker*(1), S. Lindell(1), Q. Yuan(2), Z. Zhou(2), S. Suomi(3),<br />

C. Barr(1)<br />

1. Section <strong>of</strong> Comparative Behavioral Genomics, NIH/NIAAA/LNG<br />

2. Section <strong>of</strong> Human Neurogenetics, NIH/NIAAA, LNG 3.<br />

Laboratory <strong>of</strong> Comparative Ethology, NIH/NICHD<br />

*bakermb@mail.nih.gov<br />

Introduction: Oxytocin is a neuropeptide that produces affiliative,<br />

amnesic and anxiolytic affects. Given its roles in these processes,<br />

regulation <strong>of</strong> this system may moderate risk for stress-related<br />

disorders. We wanted to examine effects <strong>of</strong> disruption <strong>of</strong> early<br />

maternal care in rhesus macaque infants on binding <strong>of</strong> an activating<br />

histone (H3K4me3) and mRNA expression levels in adult brain.<br />

Methodology: Brains (N=14) were archived from male macaques<br />

(M. mulatta) that were raised by their mothers (MR) or in peer-only<br />

groups (PR), an established model <strong>of</strong> early adversity. Hippocampal<br />

samples were dissected for DNA and RNA isolation. Chromatin<br />

Immunoprecipitation was performed with anti- H3K4me3 antibody,<br />

and ChIP- and RNA-seq were performed on an Illumina GAII. Data<br />

previously generated by microarray was used to verify RNA-seq<br />

results.<br />

Results: We observed H3K4me3 binding that spanned the first exon<br />

<strong>of</strong> the OXTR gene (Chr2: 52,233,200 – 52,234,100), with peak<br />

binding observed in a central 300 nucleotide region. Both overall<br />

H3K4me3 binding and within-peak binding were decreased in PR<br />

monkeys (P = 0.01 and P = 0.005). OXTR mRNA expression levels<br />

were also decreased (P = 0.008).<br />

Conclusions: Variation in maternal care as it relates to oxytocin<br />

system functioning has been demonstrated in rodents. Our results<br />

show that disrupted maternal care produces decreased binding <strong>of</strong> an<br />

activating histone and lower OXTR mRNA expression levels in adult<br />

macaque brain. Early environment-induced epigenetic regulation <strong>of</strong><br />

OXTR may increase vulnerability to stress-related disorders, either<br />

alone or interactively with functional genetic variation. It could also<br />

produce long lasting effects on social cognition and affiliative<br />

behavior.<br />

109<br />

PP81 DNA HYPOMETHYLATION OF MB-COMT<br />

PROMOTER IN THE DNA DERIVED FROM SALIVA IN<br />

SCHIZOPHRENIA AND BIPOLAR DISORDER<br />

S. Nohesara(1), M. Ghadiri(1), H. Ahmadkhaniha(1), S.<br />

Mostafavi(2), S. Thiagalingam(2), H. Mostafavi Abdolmalek*(1,2)<br />

1. Tehran University <strong>of</strong> Medical Sciences 2. Boston University<br />

*hamostafavi@yahoo.com<br />

Introduction: The molecular basis <strong>of</strong> pathogenesis remains<br />

unrecognized in major mental diseases despite extensive genetic<br />

studies. This failure in the discovery <strong>of</strong> etiology <strong>of</strong> psychiatric<br />

diseases directed the attention <strong>of</strong> the scientific community to the<br />

possible contributions <strong>of</strong> epigenetic modulations such as DNA<br />

methylation and histone modifications, which play important roles in<br />

the regulation <strong>of</strong> gene expression in response to environmental<br />

factors. However, most epigenetic modifications are tissue specific<br />

and the availability <strong>of</strong> brain tissue to identify epigenetic aberrations<br />

in living condition is limited. The detection <strong>of</strong> epigenetic<br />

abnormalities in other tissues that represent the brain epigenetic<br />

marks is one <strong>of</strong> the critical steps to develop diagnostic and<br />

therapeutic biomarkers for mental diseases. Formerly, we analyzed<br />

115 human post-mortem brain samples from the frontal lobe and<br />

reported an increased expression <strong>of</strong> membrane-bound catechol-Omethyltransferase<br />

(MB-COMT) due to DNA hypo-methylation <strong>of</strong> the<br />

gene promoter as a risk factor for schizophrenia (SCZ) and bipolar<br />

disorder (BD). Here, hypothesizing that those factors that lead to the<br />

brain MB-COMT promoter DNA hypo-methylation may also cause<br />

concurrent epigenetic aberrations in peripheral tissues, we analyzed<br />

MB-COMT promoter methylation in DNA derived from the saliva in<br />

SCZ, BD and control subjects<br />

Methodology: DNA was extracted from saliva using oragene DNA<br />

kit. Bisulfate DNA sequencing and quantitative methylation specific<br />

PCR was used to measure the degree <strong>of</strong> MB-COMT<br />

promoter DNA methylation in SCZ, BD, non-affected family<br />

members <strong>of</strong> patients and control subjects (each group 20 cases).<br />

Results: We found that similar to the brain, MB-COMT promoter<br />

DNA is hypo-methylated (~%50) in DNA derived from the saliva in<br />

SCZ and BD compared to the controls ( p=0.02 and 0.037,<br />

respectively). DNA Methylation <strong>of</strong> MB-COMT promoter in<br />

unaffected family members <strong>of</strong> patients was not different compared to<br />

the control subjects.<br />

Conclusions: These studies suggest that DNA methylation analysis<br />

<strong>of</strong> MB-COMT promoter in saliva can be used as an available<br />

epigenetic bio-marker for disease state in SCZ and BD.


PP82 EPIGENETIC MODELING OF SEROTONIN<br />

TRANSPORTER IN NURSES FROM HIGH/LOW STRESS<br />

WORKING ENVIRONMENT<br />

J. Alasaari(1,2), M. Lagus(1,2), M. Härmä(3), S. Puttonen(3),<br />

T. Paunio*(1,2)<br />

1. National Institute for Health and Welfare 2. Helsinki University<br />

Central Hospital 3. 3. Finnish Institute <strong>of</strong> Occupational Health<br />

*tiina.paunio@thl.fi<br />

Introduction: Methylation <strong>of</strong> genomic DNA is part <strong>of</strong> an epigenetic<br />

regulatory system that has been linked to a number <strong>of</strong> adaptive<br />

processes such as stress regulation and learning. Methylation states<br />

<strong>of</strong> the CpG dinucleotides are dynamic and semi stable by nature and<br />

when this occurs within a gene promoter region, the result is<br />

silencing <strong>of</strong> promoter activity, ultimately leading to silencing <strong>of</strong> the<br />

gene itself. Methylation has been suggested to play a significant role<br />

in the interplay between gene and environment. The serotonin<br />

receptor gene (SLC6A4), a key regulator <strong>of</strong> serotonergic<br />

neurotransmission, is <strong>of</strong> particular interest in the context <strong>of</strong><br />

stress.Changes in serotonergic neurotransmission are implicated in<br />

disorders such as alcoholism, autism, migraine headaches and major<br />

depression.<br />

Methodology: A cohort <strong>of</strong> 99 Finnish female shift working nurses<br />

was recruited from five hospitals and four towns belonging to an<br />

ongoing Finnish Public Sector Study. Participants were selected from<br />

wards that were placed among highest or lowest quartiles on job<br />

strain based on Karasek's Job Content Questionnaire. The division<br />

into high and low job strain groups was based on the average <strong>of</strong> all<br />

the answers <strong>of</strong> a ward. The high job strain study group (N=31)<br />

consisted <strong>of</strong> nurses who had self-evaluated their own job strain at<br />

least as high as their ward on average in the highest quartile on job<br />

strain. The low job strain study group (N=46) consisted <strong>of</strong> nurses<br />

who had self-evaluated their own job strain at least as low as their<br />

ward on average in the lowest quartile on job strain. Individual<br />

experiences from work-related stress and depression were assessed<br />

by the Maslach Burnout Inventory - General Survey (MBI-GS) and<br />

Beck Depression Inventory (BDI). The inclusion criteria was age<br />

between 30-58 years, mother tongue Finnish, BMI under 35, at least<br />

3 years work experience in the same ward and absenteeism from<br />

work <strong>of</strong> no longer than 6 months during the past 3 years. Nurses with<br />

high alcohol consumption, frequent smoking, hormonal medication<br />

and medication affecting cognitive functions were excluded. We<br />

genotyped the high and low job strain study groups by bisulfate<br />

sequencing <strong>of</strong> CpG sites <strong>of</strong> a 200bp region upstream <strong>of</strong> SLC6A4 and<br />

calculated methylation percentages for each CpG residue using<br />

Sequence Scanner v1.0. The differences in the two groups were<br />

evaluated using two-tailed student's T-test.<br />

Results: None <strong>of</strong> the individuals in the study cohort scored 3.5 or<br />

higher (severe burnout) in the MBI-GS. The percentage <strong>of</strong><br />

individuals scoring between 1.5-3.49 (moderate burnout) were 42%<br />

and 11% in the high and low work strain groups respectively. The<br />

percentage <strong>of</strong> individuals having mild depression (scores between 10-<br />

18) was 39% and 11% in the high and low job strain groups<br />

respectively. Bisulfite sequencing <strong>of</strong> the promoter region <strong>of</strong> SLC6A4<br />

revealed a robust and consistent difference between methylation<br />

status <strong>of</strong> individuals from the two job strain groups at the five CpG<br />

sites examined. The CpG methylation in the high work strain group<br />

was lower as compared to the low work strain group at all five sites,<br />

ranging from 6 to 13% in the low strain and from 3 to 9% in the high<br />

strain group (P=0.001-0.02, respectively).<br />

Conclusions: The results evidence for hypomethylation <strong>of</strong> promoter<br />

region <strong>of</strong> serotonin transporter in adaptation to environmental stress.<br />

Environmental stress may have lasting effects in the epigenome and<br />

gene regulation.<br />

110<br />

PP83 COMPLEXITY ANALYSIS OF COMPLEX DISEASE:<br />

SCHIZOPHRENIA<br />

C. Smith*(1), K. Huang(2)<br />

1. Boston Univeristy 2. Federal Institute <strong>of</strong> Technology<br />

*clsmith@bu.edu<br />

Introduction: Schizophrenia is a complex disease linked to both<br />

genetic and environmental factors. Most recent research focuses on<br />

identifying the genetic components <strong>of</strong> disease. Large international<br />

genome wide studies identified rare and common alleles <strong>of</strong> over 100<br />

genes linked to schizophrenia, as well an increased level <strong>of</strong> copy<br />

number variants. Environmental components linked to disease occur<br />

early during development and include malnutrition, folate deficiency,<br />

viral infection, and elder fathers. Our focus has been on<br />

understanding how genetic and environmental components<br />

interaction.<br />

Methodology: Here, we present the results <strong>of</strong> preliminary studies<br />

using complexity modeling methods and the concept <strong>of</strong> emergence to<br />

ask what influences the level <strong>of</strong> schizophrenia in a population.<br />

Results: The results suggest that the level <strong>of</strong> schizophrenia in a<br />

population is correlated with the de novo mutation rate.<br />

Conclusions: These results demonstrate the usefulness <strong>of</strong> complexity<br />

approaches for understanding how the multiple factors linked to<br />

schizophrenia interactact. Future analysis will apply additional<br />

principles <strong>of</strong> complexity analysis to further understand the<br />

complexities <strong>of</strong> schizophrenia.


PP84 ALTERNATIVE STRATEGIES FOR CALCULATING<br />

ANTIPSYCHOTIC DOSAGE EQUIVALENTS:<br />

ASSOCIATION WITH DIFFERENT ETHNIC CLUSTERS<br />

A. Hassan*, V. De Luca, A. Ravindran, C. Teo, J. Kennedy<br />

University <strong>of</strong> Toronto<br />

*nabeel.hassan@utoronto.ca<br />

Introduction: BACKGROUND:Side effects <strong>of</strong> antipsychotic<br />

medication are among the first reasons for noncompliant with<br />

treatment for schizophrenia or schizoaffective disorder. It is very<br />

common but hard to predict. These side effects are usually seen<br />

above the recommended dosage <strong>of</strong> antipsychotic prescription, which<br />

falls between 300 and 1000 chlorpromazine equivalents (CPZe) per<br />

day. Several studies have indicated that demographic characteristics<br />

can influence the individual’s dosage <strong>of</strong> medication to fall above the<br />

recommended range. Some <strong>of</strong> the factors included to influence excess<br />

antipsychotic dosing were the hospital cultures. A number <strong>of</strong><br />

guidelines have been developed to calculate the antipsychotic dosage<br />

equivalents.<br />

AIM: This retrospective study determines if high dosing <strong>of</strong><br />

antipsychotic is more common for certain ethnicity considering both<br />

the self-reported ethnicity and the geographical ancestry calculated<br />

using 191 DNA markers.<br />

Methodology: We collected ethnicity data (self-reported) and DNA<br />

sample from 241 schizophrenia patients at CAMH. We calculated the<br />

CPZe at the time <strong>of</strong> the assessment as our main outcome.<br />

Results: We did not find any significant difference between White<br />

Europeans and Non White Europeans (self-reported) regarding the<br />

CPZe (p=0.23) furthermore when we considered the geographical<br />

ancestry determined by using the 191 SNPs we could not find any<br />

association between the White ancestry and CPZe.<br />

Conclusions: Our preliminary analysis shows that there is no<br />

evidence that different ethnic groups receive different dose <strong>of</strong><br />

antipsychotics. However, there are many ways to calculate CPZ<br />

equivalents doses and this makes it more difficult to perform<br />

replication studies.<br />

111<br />

PP85 CEREBRAL DOMINANCE IS EPIGENETICALLY<br />

TRANSMITTED<br />

T. Crow(1), S. Leask*(2)<br />

1. University <strong>of</strong> Oxford 2. University <strong>of</strong> Nottingham<br />

*stuart.leask@nottingham.ac.uk<br />

Introduction: Approximately 90% <strong>of</strong> individuals are right-handed, a<br />

population characteristic that distinguishes humans from the great<br />

apes. A single Mendelian gene with two alleles has been suggested. It<br />

has been proposed that cerebral asymmetry is the defininig feature <strong>of</strong><br />

the human brain, and the location <strong>of</strong> the variation underlying<br />

psychosis. Relative handskill is an index <strong>of</strong> cerebral asymmetry, and<br />

has been found to correlate with verbal and non-verbal ability in the<br />

general population.<br />

Methodology: Here we investigate handedness at age 11 years as a<br />

continuous variable in relation to parental age at the birth <strong>of</strong> the child.<br />

If determined by DNA sequence (Mendelian) variation the age <strong>of</strong> a<br />

parent would be without influence.<br />

Results: On the contrary we find that mothers below the age <strong>of</strong> 20<br />

are increasingly likely to have children who are more left-handed<br />

than expected; fathers below the age <strong>of</strong> 20 have children who are<br />

more right-handed. Thus variation in handedness is not Mendelian,<br />

but a function <strong>of</strong> developmental stage and sex <strong>of</strong> the parent.<br />

Conclusions: The analysis reveals an epigenetic trans-generational<br />

influence relating to the faculty that defines the species – language.<br />

Such transmission is relevant to human-specific heritability<br />

predisposing to neuropsychiatric disease.<br />

Crow, TJ, Crow, LR, Done, DJ, & Leask, S.J. 1998. Relative hand<br />

skill predicts academic ability: global deficits at the point <strong>of</strong><br />

hemispheric indecision. Neuropsychologia 36(12): 1275-1282.<br />

Peters, M, Reimers, S. & Manning, T.J. 2006. Hand preference for<br />

writing and associations with selected demographic and behavioral<br />

variables in 255,100 subjects: The BBC internet study. Brain &<br />

Cognition 62: 177-189.


PHENOTYPES & ENDOPHENOTYPES<br />

PP86 GENETIC PLASTICITY IN INDISCRIMINANT<br />

BEHAVIOR AS A CONSEQUENCE OF EARLY SEVERE<br />

SOCIAL DEPRIVATION AND INSTITUTIONAL CARE<br />

S. Drury*(1), C. Nelson(2), N. Fox(3), C. Zeanah(1), K. Theall(1)<br />

1. Tulane University 2. Harvard 3. University <strong>of</strong> Maryland<br />

*sdrury@tulane.edu<br />

Introduction: Recent theories challenge the vulnerability/resilience<br />

concept <strong>of</strong> gene-environment interactions suggesting instead<br />

that these interactions are better characterized in terms <strong>of</strong> biological<br />

sensitivity to context. In this model genetic variation results in<br />

greater sensitivity to the environment, for better or worse. To test this<br />

model we explored the genetic contribution to indiscriminant social<br />

behavior, an established lasting behavioral construct prevalent in<br />

children exposed to early severe social deprivation. As symptoms <strong>of</strong><br />

indiscriminant behavior develop specifically in the environmental<br />

setting <strong>of</strong> pathogenic caregiving and only a subset <strong>of</strong> children recover<br />

when placed in an enhanced environment genetic plasticity may<br />

explain this differential response to the caregiving environment.<br />

Methodology: 98 children enrolled in the Bucharest Early<br />

Intervention Project (BEIP) were randomized, before 30 months <strong>of</strong><br />

age, to either continued care as usual (CAUG) or high quality foster<br />

care (FCG) and followed longitudinally. Indiscriminant behavior was<br />

assessed at four time points by caregiver report using the same<br />

validated semi-structured interview, the Disturbances <strong>of</strong> Attachment<br />

Interview (DAI). (2)We examined the association <strong>of</strong> indiscriminant<br />

behavior at each time point with functional polymorphisms in two<br />

genes previously associated with differential susceptibility to early<br />

adversity, Brain Derived Neurotrophic Factor (BDNF) val 66met and<br />

the serotonin transporter (5HTT) 5httlpr. We additionally explored<br />

the hypothesis that the met allele <strong>of</strong> BDNF and the short allele <strong>of</strong> the<br />

5httlpr would reflect elevated sensitivity to change in the caregiving<br />

environment such that they would be associated with the highest level<br />

<strong>of</strong> symptoms in the CAUG but the lowest level <strong>of</strong> symptoms when<br />

children were placed in foster care. Finally as these two<br />

polymorphisms have previously been found to have an additive<br />

influence on outcomes (3)we explored the association between<br />

indiscriminant symptoms and a combined plasticity genotype (met<br />

allele carriers and s/s homozygotes).<br />

Results: An interaction between group status (FCG or CAUG) and<br />

genotype was detected at multiple time points. Children with the s/s<br />

5httlpr genotype or carriers <strong>of</strong> the met 66 allele in BDNF<br />

demonstrated both the greatest decrease in DAI scores in the FCG<br />

and the highest persistent rate <strong>of</strong> indiscriminate social behavior in the<br />

CAUG. Regression analysis revealed that 5httlpr genotype had the<br />

greatest impact on change in DAI score between baseline and 42<br />

months(p=.02, r2=.17) while the greatest impact <strong>of</strong> BDNF genotype<br />

occurred between 30 and 54 months (p=003 r2=.11). Children with<br />

both s/s 5htt genotype and met/ allele carriers <strong>of</strong> BDNF in the CAUG<br />

had the highest number <strong>of</strong> indiscriminate symptoms at 54 months<br />

while those with the same genotype in the FCG had the lowest<br />

number <strong>of</strong> symptoms at 54 months.<br />

Conclusions: These findings represent the first known genetic<br />

associations with indiscriminant social behavior. This is the first<br />

study, to our knowledge, to demonstrate genetic biological sensitivity<br />

to context in the same children exposed to well-defined changes in<br />

the caregiving environment associated with institutional rearing.<br />

These findings add to the growing literature demonstrating biological<br />

sensitivity to context and highlight the need for careful consideration<br />

<strong>of</strong> this model in future gene-environment interaction studies.<br />

112<br />

PP87 PATTERNS OF DEFICITS IN BRAIN FUNCTION IN<br />

BIPOLAR DISORDER AND SCHIZOPHRENIA: A CLUSTER<br />

ANALYTIC STUDY<br />

M. Hall*(1,2,3), J. Smoller(3), K. Schulze(4), F. Rijsdijk(4), P.<br />

Lee(3), G. Taylor(2), E. Bramon(4), M. Coleman(1), R. Murray(4),<br />

D. Salisbury(2), D. Levy(1)<br />

1. Psychology Research Laboratory, McLean Hospital, Harvard<br />

Medical School 2. Cognitive Neuroscience Laboratory, McLean<br />

Hospital, Harvard Medical School 3. <strong>Psychiatric</strong> <strong>Genetics</strong> Program in<br />

Mood and Anxiety Disorders, Massachusetts General Hospital,<br />

Harvard Medical School 4. Division <strong>of</strong> Psychological Medicine,<br />

Institute <strong>of</strong> Psychiatry, King's College London<br />

*mhall@mclean.harvard.edu<br />

Introduction: Historically, bipolar disorder (BPD) and schizophrenia<br />

(SCZ) have been considered distinct disorders with differing<br />

etiologies and courses <strong>of</strong> illness. Growing evidence suggests that<br />

overlapping genetic influences contribute to risk for these disorders<br />

and that each disease is genetically heterogeneous. In this study, we<br />

explored the use <strong>of</strong> cluster analytical approach to extract<br />

neurophysiological pr<strong>of</strong>iles in patients with diagnoses <strong>of</strong> SCZ or<br />

BPD, their unaffected relatives, and control subjects. The main goals<br />

<strong>of</strong> this study were to: 1) identify and characterize distinct<br />

neurophysiological functional pr<strong>of</strong>iles in independent samples <strong>of</strong><br />

psychotic patients, their unaffected first-degree relatives and controls<br />

2) assess the magnitude and type <strong>of</strong> brain functional abnormalities in<br />

relation to DSM diagnoses and 3) compare these empirically derived<br />

clusters <strong>of</strong> psychotic patients on demographic features and illness<br />

characteristics.<br />

Methodology: Seven auditory neurophysiologic phenotypes,<br />

including P50 sensory gating, evoked gamma band response, P3<br />

amplitude, P3 latency, P2 amplitude, N1 amplitude, and MMN<br />

amplitude, were collected from two independent cohorts <strong>of</strong> SCZ and<br />

BPD families at two research institutions, including probands<br />

(n=157), relatives (n=82) and well controls (n=230). The individuals<br />

were clustered using the K-means algorithm.<br />

Results: Three neurophysiologically distinct pr<strong>of</strong>iles emerged from<br />

the cluster analysis (Figure 1). Individuals in cluster one, the global<br />

impaired group, exhibited severe functional abnormalities and had<br />

the worst scores on all measures. Individuals in cluster two, the high<br />

sensory processing group, performed the best on tasks that tap the<br />

early stages <strong>of</strong> sensory processing. Individuals in cluster three, the<br />

high cognitive group, had the best scores on tasks that probe<br />

inhibitory function and higher cognitive processes. These overall<br />

patterns were observed in both independent samples and thus seem to<br />

robustly reflect three distinct neurocognitive pr<strong>of</strong>iles. Significant<br />

heritability was found for these three empirically derived behavioral<br />

pr<strong>of</strong>iles. Notably, these pr<strong>of</strong>iles are not specific to either SCZ or<br />

BPD.<br />

Conclusions: The results suggest that three distinct clusters <strong>of</strong><br />

individuals can be identified who may differ in underlying<br />

neurobiology and genetic risk factors. These results raise the<br />

intriguing possibility that neurocognitive pr<strong>of</strong>iles may be more<br />

powerful discriminators than diagnoses in genetic analyses.


PP88 ROLE OF THE GRIN2B GENE IN THE PRESENCE OF<br />

COMORBID PSYCHIATRIC DIAGNOSES IN WOMEN WITH<br />

BULIMIA NERVOSA<br />

Z. Yilmaz*(1,2), A. Kaplan(1,2), C. Zai(1,2), R. Levitan(1,2), J.<br />

Kennedy(1,2)<br />

1. University <strong>of</strong> Toronto 2. Centre for Addiction and Mental Health<br />

*zeynep.yilmaz@utoronto.ca<br />

Introduction: GRIN2B gene regulates the activity <strong>of</strong> NMDA<br />

receptor NR2 subunit, which acts as the agonist binding site for<br />

glutamate and has been associated with anxiety and impulse controlrelated<br />

disorders. Majority <strong>of</strong> patients with bulimia nervosa (BN) also<br />

report a history <strong>of</strong> anxiety disorders and a significant proportion have<br />

impulse control problems, suggesting that BN may also be associated<br />

with glutamatergic abnormalities. The purpose <strong>of</strong> this study is to (1)<br />

examine the frequency <strong>of</strong> GRIN2B genetic variants in BN and<br />

healthy controls and (2) explore the role <strong>of</strong> the GRIN2B gene in<br />

comorbid psychiatric disorders among bulimic women.<br />

Methodology: For the first part <strong>of</strong> the study, we genotyped 243<br />

women with BN and equal number <strong>of</strong> ethnicity-matched female<br />

controls for GRIN2B rs2284411, rs1806201, rs1019385 and rs890,<br />

markers previously associated with psychiatric disorders. We then<br />

performed genetic analyses on the BN probands to investigate if the<br />

GRIN2B variants and haplotypes were associated with comorbid<br />

psychiatric diagnoses and severity <strong>of</strong> eating disorder symptoms.<br />

Results: The data analysis for the first part <strong>of</strong> the study is currently<br />

underway. Within the BN group, we found a significant association<br />

<strong>of</strong> GRIN2B markers and haplotypes with a lifetime history <strong>of</strong> anxiety<br />

disorders (p = .015). GRIN2B genotypes or haplotypes were not<br />

associated with childhood ADHD or history <strong>of</strong> substance use in BN<br />

probands.<br />

Conclusions: To our knowledge, this is the first study to look at the<br />

role <strong>of</strong> GRIN2B gene in BN. The pathophysiology <strong>of</strong> BN with<br />

comorbid anxiety disorders may be a distinct subphenotype related to<br />

underlying glutamatergic abnormalities, and these finding may have<br />

implications for treatment for BN patients with comorbid anxiety<br />

disorders.<br />

113<br />

PP89 INVESTIGATION OF THE ASSOCIATION OF<br />

POLYGENIC SIGNALS FOR ADHD TO PREDICT<br />

COGNITIVE MEASURES THAT SHARE GENETIC<br />

INFLUENCES WITH ADHD<br />

G. Mould*(1), T. Price(1), T. Banaschewski(4), M. Gill(2), I.<br />

Manor(5), A. Miranda(6), F. Mulas(7), R. Oades(8), A.<br />

Rothenberger(3), H. Royers(12), H. Steinhausen(9,10,11), J. van der<br />

Meere(13), S. Faraone(14), P. Asherson(1), J. Kuntsi(1)<br />

1. King's College London 2. Trinity Centre for Health Sciences 3.<br />

University <strong>of</strong> Göttingen 4. University <strong>of</strong> Heidelberg 5. S. Herzog<br />

Memorial Hospital 6. University <strong>of</strong> Valencia 7. La Fe University<br />

Hospital 8. University <strong>of</strong> Duisburg-Essen 9. University <strong>of</strong> Zurich 10.<br />

University <strong>of</strong> Basel 11. University Hospital Aarhus 12. Ghent<br />

University 13. University <strong>of</strong> Groningen 14. State University <strong>of</strong> New<br />

York Upstate Medical University<br />

*glenn.mould@kcl.ac.uk<br />

Introduction: ADHD is associated with multiple cognitive<br />

performance deficits. Among these Kuntsi et al (2010, Archives <strong>of</strong><br />

General Psychiatry) selected the most promising indicators for a<br />

multivariate familial factor analysis. Two familial cognitive<br />

impairment factors were identified, which accounted for 85% and<br />

13% <strong>of</strong> the familial variance on ADHD. The first, large factor<br />

captured speed and variability <strong>of</strong> reaction times (RT) and the second<br />

factor captured commission (Ce) and omission (Oe) errors on a<br />

go/no-go task. Using the same sample we now evaluate whether<br />

polygenic signals for ADHD generated from genome wide SNP data<br />

are associated with the most promising cognitive variables.<br />

Methodology: Analysis used data from a collaboration <strong>of</strong> 8 teams<br />

from the International Multi-Centre ADHD Gene (IMAGE) project.<br />

Probands were all combined type ADHD, <strong>of</strong> white European decent<br />

and aged 6-18. DNA was available from probands and both parents.<br />

The transmission disequilibrium test (TDT) was conducted and the<br />

data used to generate polygenic scores at different significance<br />

thresholds. Polygenic scores from 352 parent-proband trios were used<br />

to predict cognitive performance using regression analysis, with RT<br />

variability, Oe, Ce and IQ using a 4-fold cross validation procedure.<br />

SNPs included in the analysis were in linkage equilibrium, to<br />

minimise the use <strong>of</strong> redundant genetic variation and potentially<br />

enhance statistical power.<br />

Results: Analyses tested ADHD associated genotype scores at<br />

different p-value thresholds. Significant associations were detected<br />

for Oe and RT variability at thresholds <strong>of</strong> P


PP90 THE INFLUENCE OF SCHIZOPHRENIA-RELATED<br />

DISC1 POLYMORPHISMS ON SENSORIMOTOR GATING,<br />

COGNITION AND PERSONALITY IN HEALTHY MALES<br />

P. Bitsios*(1), P. Roussos(1,2), S. Giakoumaki(1)<br />

1. Department <strong>of</strong> Psychiatry and behavioral Sciences, University <strong>of</strong><br />

Crete 2. Department <strong>of</strong> Psychiatry, Mount Sinai School <strong>of</strong> Medicine<br />

*pbitsios@med.uoc.gr<br />

Introduction: There is evidence supporting a role for the DISC1<br />

locus in schizophrenia, however the impact <strong>of</strong> DISC1 risk<br />

polymorphisms on schizophrenia endophenotypes has not been<br />

studied extensively.<br />

Methodology: We selected DISC1 polymorphisms that are<br />

functional or have shown significant association with SZ or SZrelated<br />

endophenotypes (rs3738401, rs1538979, rs2492367,<br />

rs6675281, rs1984895, rs1000731, rs999710, rs821577, rs821616,<br />

rs821633, rs3737597). These polymorphisms were analyzed in 530<br />

healthy males, phenotyped for acoustic startle and prepulse inhibition<br />

(PPI), performance in verbal and working memory, executive<br />

function, schizotypy, trait anxiety and affective personality traits.<br />

QTPHASE from the UNPHASED package was used for the<br />

association analysis <strong>of</strong> allelic data, withp values corrected for<br />

multiple testing by running 10000 permutations <strong>of</strong> the data.<br />

Results: The rs999710 T allele was most strongly associated with<br />

reduced PPI (p


the worse mean scores when compared to mixed episode and<br />

depression. In manic patients rs4680 allele G+ showed better<br />

performance in an executive function test. Cognitive performance<br />

was better for manic episode patients with G at rs165599 as shown<br />

through executive function tests: SCWT-1 SCWT-2 and SCWT-2<br />

errors. Mixed episode rs4680 G+ patients performed better in verbal<br />

fluency, non-verbal memory and executive function tests. Mixed<br />

episode patients with rs165599 G displayed higher mean scores in<br />

intelligence and executive function. Depressive patients were not<br />

significantly different from controls in all tests.<br />

Conclusions: Our data shows that rs4680 and rs165599 G+<br />

individuals had better cognitive function than G- during manic and<br />

mixed episodes. During manic episodes G+ had better performance<br />

than G- on executive function tests, and those with mixed episodes<br />

G+ patients performed better in executive, memory, verbal fluency<br />

and intelligence tests. We hypothesize that COMT genotype may<br />

partly determine the cognitive disorganization in some patients<br />

during mania and mixed episodes.<br />

115<br />

PP92 PCLO GENOTYPE EFFECT ON NEURAL<br />

CORRELATES OF EMOTIONAL MEMORY AND THE<br />

INFLUENCE ON THE HPA AXIS<br />

S. Woudstra*(1,2,3,7), W. Hoogendijk(1,7), Z. Bochdanovits(2,8),<br />

M. van Tol(1,3,4,6), L. Demenescu(5), S. Vreeburg(1), N. van der<br />

Wee(3,4), F. Zitman(3), E. Opmeer(6), A. Aleman(6), B.<br />

Penninx(1,2,3), D. Veltman(1)<br />

1. Department <strong>of</strong> Psychiatry, VU university Medical Centre 2.<br />

Department <strong>of</strong> Medical Genomics, VU university Medical Centre 3.<br />

Department <strong>of</strong> Psychiatry, Leiden University Medical Centre 4.<br />

Leiden Institute for Brain and Cognition, Leiden University 5.<br />

Department <strong>of</strong> Psychiatry and Psychotherapy, RWTH Aachen<br />

University 6. BCN Neuroimaging Center, University Medical Center<br />

Groningen 7. Neuroscience Campus Amsterdam 8. TI Pharma<br />

*s.woudstra@vumc.nl<br />

Introduction: Emotional memory information processing is thought<br />

to be related to anhedonia, one <strong>of</strong> the core symptoms in major<br />

depressive disorder (MDD). It was found that glucocorticoids -<br />

released by the hypothalamic-pituitary-adrenal (HPA) axis -<br />

influence emotional memory information processing and may<br />

contribute to the neuroendocrine abnormalities found in MDD<br />

(Wingenfeld & Wolf, 2011). Emotional memory information<br />

processing is also thought to be associated with monoamines (Merens<br />

et al., 2007). A GWA study for MDD implicated piccolo (PCLO),<br />

which protein is involved in monoaminergic neurotransmission<br />

(Sullivan et al., 2009), and we hypothesize that the piccolo risk allele<br />

(PLCO+) is associated with altered brain activity during an emotional<br />

word memory task. In addition, we hypothesize that the PCLO<br />

genotype moderates the association between emotional memory<br />

information processing and the HPA axis. Therefore, we will<br />

investigate whether PCLO+ is associated with dysregulation <strong>of</strong> the<br />

HPA-axis.<br />

Methodology: Using both genotype and functional MRI data from<br />

118 subjects (average age 38.4 yrs 62% female) from the NESDA<br />

neuroimaging study, we conducted an ANOVA with genotype as<br />

independent factor (p


PP93 A MULTI-TIERED TRANSLATIONAL FRAMEWORK<br />

FOR LONGITUDINAL PSYCHOSIS RESEARCH: THE<br />

PSYCHOSIS CENTER LOWER SAXONY IN GÖTTINGEN,<br />

GERMANY<br />

M. Budde*(1), D. Reich-Erkelenz(1), H. Bickeböller(2), J.<br />

Brockmöller(3), M. Tzvetkov(3), O. Gruber(1), P. Falkai(1), T.<br />

Schulze(1)<br />

1. Georg-August-University, University Medical Center, Department<br />

<strong>of</strong> Psychiatry and Psychotherapy 2. Georg-August-University,<br />

University Medical Center, Department <strong>of</strong> Genetic Epidemiology 3.<br />

Georg-August-University, University Medical Center, Department <strong>of</strong><br />

Clinical Pharmacology<br />

*monika.budde@med.uni-goettingen.de<br />

For a clinician, the longitudinal course <strong>of</strong> a patient suffering from<br />

schizophrenia (SZ) or bipolar disorder (BD) is an important<br />

phenotypic feature that is used to guide treatment options and<br />

prognostic considerations. Moreover, it is the course (e.g. cognitive<br />

functioning, social withdrawal) that determines the economic burden<br />

<strong>of</strong> psychosis, both for the individual patient and society.<br />

Despite this, hardly any systematic analyses <strong>of</strong> complex genotypephenotype<br />

relationships in the longitudinal course <strong>of</strong> SZ and BD can<br />

be found. Therefore the Psychosis Center Lower Saxony has been<br />

established in Göttingen, Germany, aiming at analyses based on a<br />

large sample comprising 3000 prospectively-followed patients and<br />

prospectively-followed 3000 controls. To allow for the recruitment <strong>of</strong><br />

sufficiently large cohorts and their continued longitudinal follow-up,<br />

the Psychosis Center is tightly integrated with the inpatient and<br />

outpatient sections <strong>of</strong> the Department <strong>of</strong> Psychiatry and<br />

Psychotherapy <strong>of</strong> the University Medical Center <strong>of</strong> the Georg-<br />

August-University and will have associated recruitment centers at 13<br />

collaborating hospitals in Lower Saxony, Northern Hesse and<br />

Bremen.<br />

Each patient <strong>of</strong> 18 years <strong>of</strong> age or older with a DSM-IV diagnosis <strong>of</strong><br />

SZ or BD can take part in the project. Over a period <strong>of</strong> at least 1.5<br />

years each patient and control will be seen in regular visits every 6<br />

months or earlier, if needed (e.g. in case <strong>of</strong> relapse). At each visit,<br />

phenotypic features like differentiated psychopathology, response to<br />

treatment including adverse events, global functioning,<br />

neuropsychological performance, and life events will be assessed.<br />

These assessments will we paralleled with structural and functional<br />

imaging.<br />

In the long run, the major aim <strong>of</strong> the Psychosis Center Lower Saxony<br />

is on the one hand the creation <strong>of</strong> a phenotypic database that can<br />

easily be linked to already existing phenotypic databases and on the<br />

other hand the creation <strong>of</strong> a biobank (PsyCourse Biobank Göttingen)<br />

for genomics, epigenomics, transcriptomics, and proteomics.<br />

116<br />

PP94 WHITE MATTER ABNORMALITIES IN PSYCHOSIS<br />

ARE RELATED TO INCREASED GENETIC RISK<br />

E. Sprooten*, A. McIntosh, S. Lawrie, J. Sussmann<br />

University <strong>of</strong> Edinburgh<br />

*andrew.mcintosh@ed.ac.uk<br />

Introduction: Several lines <strong>of</strong> clinical, epidemiological and<br />

molecular evidence suggest that abnormalities <strong>of</strong> white matter may<br />

convey risk to major psychiatric disorder. Using diffusion tensor<br />

imaging (DTI), we sought to address whether white matter<br />

abnormalities are present in 1) individuals at high familial risk and 2)<br />

carriers <strong>of</strong> specific risk variants in NRG1-ErbB4 or DISC1<br />

Methodology: We used DTI in two large samples <strong>of</strong> people at highrisk<br />

<strong>of</strong> either schizophrenia or bipolar disorder to assess the<br />

relationship <strong>of</strong> fractional anisotropy to familial disease risk.<br />

Secondly, using similar methods, we assessed whether reduced<br />

fractional anisotropy was associated with functional risk variants in<br />

NRG1 (rs6994992, type IV promoter variant), ErbB4 (rs4673628,<br />

splice site) and DISC1 (rs821616, mis-sense)<br />

Results: Reduced white matter integrity was found in subjects at high<br />

risk <strong>of</strong> schizophrenia and in subjects at high risk <strong>of</strong> bipolar disorder<br />

for genetic reasons. Reduced anisotropy was also found in individuals<br />

with previously identified functional risk variants in the NRG1-<br />

ErbB4 signalling pathway and in DISC1.<br />

Conclusions: Emerging evidence from DTI suggests that risk<br />

variants for major mental illness have effects on white matter<br />

integrity that fail to respect diagnostic boundaries. This suggests that<br />

white matter integrity may be a useful quantitative trait for the<br />

identification <strong>of</strong> novel risk variants an that understanding its cellular<br />

basis may yield information about the aetiology <strong>of</strong> major mental<br />

illness.


PP95 THE GENETICS OF LANGUAGE LATERALIZATION<br />

AND LEFT-HANDEDNESS: LINKAGE ANALYSIS IN A<br />

DUTCH POPULATION ISOLATE<br />

M. Somers*(1), R. Oph<strong>of</strong>f(2,4), R. Cantor(3), M. Aukes(1,5), K. van<br />

Eijk(4), M. Dauwan(1), R. van 't Slot(4), E. Janson(4), M. Boks(1,5),<br />

R. Kahn(1), I. Sommer(1)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Psychiatry, University Medical Center Utrecht 2. Center for<br />

Neurobehavioral <strong>Genetics</strong>, Semel Institute for Neuroscience and<br />

Human Behavior, UCLA 3. ) Department <strong>of</strong> Human <strong>Genetics</strong>, David<br />

Geffen School <strong>of</strong> Medicine, UCLA 4. Department <strong>of</strong> Medical<br />

<strong>Genetics</strong>, University Medical Center Utrecht 5. Julius Center for<br />

Health Sciences and Primary Care, University Medical Center<br />

Utrecht<br />

*m.somers@umcutrecht.nl<br />

Introduction: Investigating endophenotypes can be an additional<br />

strategy to identify the genetic underpinnings <strong>of</strong> schizophrenia.<br />

Extensive evidence shows that decreased language lateralization is a<br />

useful endophenotype. Language lateralization is related to handpreference<br />

as almost 100% <strong>of</strong> right-handers have left-hemispheric<br />

lateralization, while left-handers show larger variability with 30%<br />

having right-hemispheric or bilateral language lateralization. As such,<br />

decreased language lateralization can be efficiently studied in nonschizophrenic<br />

left-handed subjects. This study applied an<br />

endophenotype approach to find genetic regions involved in<br />

schizophrenia through linkage analysis <strong>of</strong> decreased language<br />

lateralization and hand-preference, measured in large families with<br />

multiple left-handers from a unique Dutch population isolate: the<br />

former isle <strong>of</strong> Urk.<br />

Methodology: DNA was collected from 369 subjects <strong>of</strong> 37 large<br />

multi-generational families. Hand-preference was measured with the<br />

Edinburgh Handedness Inventory (EHI) and language lateralization<br />

was measured with functional transcranial Doppler (fTCD).<br />

Genotyping with the Illumina HumanLinkage-24 panel (5913 SNPs)<br />

was successful in 361 subjects. After quality control and<br />

checking for mendelian inconsistencies using PLINK and MERLIN,<br />

155 SNPs and 6 subjects were excluded, leaving a total <strong>of</strong> 355<br />

subjects with 5420 autosomal SNPs for linkage analysis with<br />

MERLIN. A parametric rare-dominant single-gene fluctuating<br />

asymmetry model (McManus 1985, van Agtmael et al, 2001) was<br />

tested for hand-preference as dichotomous trait. Non-parametric<br />

models were tested for hand-preference and language lateralization,<br />

both as dichotomous and quantitative traits.<br />

Results: The parametric analysis showed suggestive linkage in the<br />

22q13 region (HLOD Max=2.24) for hand-preference. Non-parametric<br />

multipoint analysis showed overlapping suggestive linkage in the<br />

same region (LODMax=2.69). There was no linkage for handpreference<br />

as a quantitative trait. Non-parametric multipoint analysis<br />

showed suggestive linkage for language lateralization as a<br />

dichotomous trait in the 7q34 region (LODMax=2.65) and as a<br />

quantitative trait in the 6p22 region (LODMax=2.56).<br />

Conclusions: Our results indicate that left-handedness, non-left<br />

hemispheric language lateralization and degree <strong>of</strong> language<br />

lateralization show genetic linkage in this population isolate, which is<br />

consistent with the notion that these traits have a genetic basis. The<br />

absence <strong>of</strong> overlap between the linked regions can point towards at<br />

least partly separate genetic underpinnings for each trait. Findings<br />

from our parametric analysis are consistent with a major gene model<br />

for hand-preference. The markers in the three regions will be tested<br />

for association with the traits the regions are linked to, in order to<br />

further investigate the validity <strong>of</strong> the current findings.<br />

117<br />

PP96 THE KCNH2 GENE IS ASSOCIATED WITH<br />

NEUROCOGNITION AND THE RISK OF SCHIZOPHRENIA<br />

K. Ohi*(1,3), R. Hashimoto(1,2,3), Y. Yasuda(1,3), M.<br />

Fukumoto(1,3), H. Yamamori(1,3,4), K. Kamino(1,5), T.<br />

Morihara(1), M. Iwase(1), H. Kazui(1), M. Takeda(1,2)<br />

1. Department <strong>of</strong> Psychiatry, Osaka University Graduate School <strong>of</strong><br />

Medicine 2. Molecular Research Center for Children's Mental<br />

Development, United Graduate School <strong>of</strong> Child Development, Osaka<br />

University, Kanazawa University and Hamamatsu University School<br />

<strong>of</strong> Medicine 3. CREST (Core Research for Evolutionary Science and<br />

Technology <strong>of</strong> JST (Japan Science and Technology Agency) 4.<br />

Department <strong>of</strong> Molecular Neuropsychiatry, Osaka University<br />

Graduate School <strong>of</strong> Medicine 5. Shoraiso National Hospital<br />

*ohi@psy.med.osaka-u.ac.jp<br />

Introduction: A genetic variant (rs3800779 M30) in the KCNH2<br />

gene has been associated with schizophrenia, a lower intelligence<br />

quotient (IQ) and processing speed scores, altered brain functions and<br />

increased KCNH3-3.1 mRNA levels in the hippocampus. The aims <strong>of</strong><br />

this study were to investigate whether the KCNH2 polymorphismis<br />

associated with schizophrenia-related neurocognitive deficits<br />

andtoconfirm the association between the variant and schizophrenia.<br />

Methodology: The effects <strong>of</strong> the riskgenotype on IQ and seven<br />

neurocognitive batteries were examined by the analysis <strong>of</strong> covariance<br />

in 191 healthy subjects. We performed a meta-analysis <strong>of</strong> the<br />

association between M30 and schizophrenia using five independent<br />

ethnic groups (1,720 cases 2,418 controls).<br />

Results: Consistent with the previous study, we provided evidence<br />

that subjects with the risk T carriers had significantly lower IQ scores<br />

than those with the G/G genotype(p=0.048, Figure1A). Of the seven<br />

neurocognitive batteries, subjects with the risk genotype<br />

demonstrated lower performances on attention/vigilance (p=0.0079,<br />

Figure1B) and working memory (p=0.0066, Figure1C) relative to<br />

subjects with the G/G genotype. Meta-analysis demonstrated<br />

evidence for an association between M30 and schizophrenia without<br />

showing heterogeneity across studies (odds ratio=1.18, p=0.0017).<br />

Conclusions: These data suggest that the KCNH2 polymorphism<br />

could be associated with schizophrenia-related neuropsychological<br />

deficits and the risk <strong>of</strong> developing schizophrenia.


PP97 VARIANTS OF THE RELA GENE ARE ASSOCIATED<br />

WITH SCHIZOPHRENIA AND THEIR STARTLE<br />

RESPONSES<br />

R. Hashimoto(1,2,4,7), K. Ohi(2,4), Y. Yasuda(2,4), M.<br />

Fukumoto(2,4), H. Yamamori(2,3,4), H. Takahashi(2,4), M.<br />

Iwase(2), T. Okochi(4,5), H. Kazui(2), O. Saitoh(6), M. Tatsumi(8),<br />

N. Iwata(4,5), N. Ozaki(4,9), K. Kamijima(10), H. Kunugi(7), M.<br />

Takeda(1)<br />

1. Molecular Research Center for Children's Mental Development,<br />

United Graduate School <strong>of</strong> Child Development, Osaka University,<br />

Kanazawa University and Hamamatsu University School <strong>of</strong> Medicine<br />

2. Department <strong>of</strong> Psychiatry, Osaka University Graduate School <strong>of</strong><br />

Medicine 3. Department <strong>of</strong> Molecular Neuropsychiatry, Osaka<br />

University Graduate School <strong>of</strong> Medicine 4. CREST (Core Research<br />

for Evolutionary Science and Technology), JST (Japan Science and<br />

Technology Agency) 5. Department <strong>of</strong> Psychiatry, Fujita Health<br />

University School <strong>of</strong> Medicine 6. Department <strong>of</strong> Psychiatry, National<br />

Center Hospital 7. Department <strong>of</strong> Mental Disorder Research,<br />

National Institute <strong>of</strong> Neuroscience, National Center <strong>of</strong> Neurology and<br />

Psychiatry 8. Yokohama Shinryo Clinic 9. Department <strong>of</strong> Psychiatry,<br />

Nagoya University Graduate School <strong>of</strong> Medicine 10. Department <strong>of</strong><br />

Psychiatry, Showa University School <strong>of</strong> Medicine<br />

Introduction: The pathogenesis <strong>of</strong> schizophrenia is thought to<br />

involve aberrant immune and inflammatory responses. Nuclear factor<br />

kappa B (NF-kappaB) plays important roles in the immune and<br />

inflammatory responses. The v-rel avian reticuloendotheliosis viral<br />

oncogene homolog A (RELA) gene encodes the major component <strong>of</strong><br />

the NF-kappaB complex.<br />

Methodology: We genotyped four single nucleotide polymorphisms<br />

(SNPs) in the RELA gene and performed a gene-based association<br />

analysisusing 1,224 patients with schizophrenia and 1,663 controls.<br />

In silico genotype-gene expression analysis was performed using web<br />

database and the WGAViewer s<strong>of</strong>tware. The impact <strong>of</strong> four RELA<br />

polymorphisms on prepulse inhibition (PPI) was investigated in 53<br />

patients with schizophrenia.<br />

Results: We found significant associations <strong>of</strong> three SNPs [rs7119750<br />

(SNP1): p=0.0080, rs2306365 (SNP3): p=0.0031 and rs11820062<br />

(SNP4): p=0.00011] with schizophrenia and stronger evidence for<br />

association in a multi-marker sliding window haplotype analysis (the<br />

lowest p=0.00006). The association between this gene and<br />

schizophrenia was evident in male subjects but not in female<br />

subjects, when separately analyzed by gender. In silico<br />

analysis revealed that these three schizophrenia-associated SNPs<br />

might be related to RELA mRNA expression in immortalized Blymphocytes.<br />

In silico analysis also suggested the putative promoter<br />

SNP4 might disrupts the consensus transcription factor binding<br />

sequence <strong>of</strong> the androgen receptor. In addition, we provided evidence<br />

that at risk genotypes <strong>of</strong> three SNPs (SNP1, 3 and 4) were associated<br />

with deficits in PPI, however, there was no effect <strong>of</strong> the one non-risk<br />

SNP2 on PPI (Figure1).<br />

Conclusions: These findings suggest that variants <strong>of</strong> the RELA gene<br />

are associated with risk for schizophrenia and PPI deficits in a<br />

Japanese population.<br />

118<br />

PP98 GENETIC DETERMINANTS OF IMPAIRED BRAIN<br />

FUNCTION IN SCHIZOPHRENIA<br />

S. Bergen(1,2,3), S. Purcell(1,2,3), J. Goldstein(1,4), R.<br />

McCarley(1,5), L. Seidman(1,6), M. Shenton(1,4,5), T. Woo(1,7), T.<br />

Petryshen*(1,2,3)<br />

1. Dept <strong>of</strong> Psychiatry, Harvard Medical School 2. <strong>Psychiatric</strong> and<br />

Neurodevelopmental <strong>Genetics</strong> Unit, Center for Human Genetic<br />

Research, Massachusetts General Hospital 3. Stanley Center for<br />

<strong>Psychiatric</strong> Research, Broad Institute 4. Dept <strong>of</strong> Psychiatry, Brigham<br />

and Women's Hospital 5. Dept <strong>of</strong> Psychiatry, VA Boston Healthcare<br />

System 6. Dept <strong>of</strong> Psychiatry, Beth Israel Deaconess Medical Center<br />

7. McLean Hospital<br />

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

Introduction: Schizophrenia (SCZ) is characterized by severe<br />

impairments in cognition and brain function for which the underlying<br />

neural abnormalities are essentially unknown. Recently, the SCZ<br />

genetics field has been ignited by the identification <strong>of</strong> genetic<br />

variants contributing to SCZ susceptibility from the consensus <strong>of</strong><br />

large-scale genome-wide association studies (GWAS). The next<br />

logical and necessary step is to translate these findings into a better<br />

understanding <strong>of</strong> cognitive and brain function impairments in SCZ to<br />

elucidate the molecular mechanisms <strong>of</strong> neural circuit dysfunction in<br />

this disease.<br />

Methodology: The genetic contribution to impaired brain function in<br />

SCZ is being evaluated using a unique case/control sample (total N =<br />

347) spanning prodrome, first episode, and chronic SCZ stages with<br />

longitudinal data from clinical, cognitive, structural and functional<br />

imaging, electrophysiological, and hormonal phenotypic<br />

domains. Candidate genes are selected that have a priori evidence<br />

for involvement in SCZ or brain function from genome-wide<br />

association studies (GWAS) or cell-specific postmortem expression<br />

studies. Regression analysis is performed using PLINK for each<br />

phenotypic measure, as well as multivariate analysis <strong>of</strong> principle<br />

components across phenotypic domains. Empirical P values are<br />

calculated by point-wise permutation for each SNP test.<br />

Results: Preliminary regression analysis was performed using a<br />

subset <strong>of</strong> 115 subjects (51 control, 64 SCZ across prodrome, first<br />

episode, and chronic stages) and 6 cognitive measures assessing<br />

attention/processing speed, language function, visuospatial ability,<br />

verbal learning & memory, and working memory. 61 SNPs tested<br />

were located in 23 genes/loci with near or above genome-wide<br />

significance in published GWAS <strong>of</strong> SCZ or cognition, or in the<br />

<strong>Psychiatric</strong> GWAS Consortium SCZ mega-analysis, or with<br />

significantly different expression in SZ cortical neurons. In combined<br />

analysis <strong>of</strong> cases and controls, the SCZ risk gene neurogranin<br />

(NRGN) was associated with language function and verbal learning<br />

(rs548181; WASI vocabulary, P = 0.003; WMS-3 logical memory, P<br />

= 0.007), in support <strong>of</strong> mouse studies reporting a role for NRGN in<br />

cognition. The KIBRA gene, originally identified in a memory<br />

GWAS, was associated with attention/processing speed (rs17070145;<br />

Trial-making Part A, P = 0.01) in this sample. Expanded analysis <strong>of</strong><br />

additional subjects and other phenotypes (gray matter MRI and white<br />

matter DTI measures, ERP components, hormone levels, working<br />

memory brain activity) is ongoing.<br />

Conclusions: This deeply-phenotyped patient sample provides an<br />

exceptional opportunity to elucidate the functions <strong>of</strong> existing SCZ<br />

risk variants in regulating specific neural processes underlying<br />

pathophysiology. Initial analyses indicate associations between<br />

cognitive traits and the NRGN and KIBRA genes which provide<br />

insights into their disease-relevant functions. Analysis <strong>of</strong> other<br />

phenotypes within and across neuroanatomical, functional, and<br />

physiological domains may highlight genetic relationships suggesting<br />

common neural mechanisms underlying brain function deficits in<br />

SCZ.


PP99 BDNF VAL66MET INTERACTS WITH STRESS AT<br />

AGE 13 TO PREDICT DEPRESSION AT AGE 15<br />

R. Salk*, L. Abramson, J. Hyde<br />

University <strong>of</strong> Wisconsin-Madison<br />

*rsalk@wisc.edu<br />

Introduction: The brain derived neurotrophic factor (BDNF) gene is<br />

an identified and theoretically relevant gene for investigating geneenvironment<br />

interactions across development that predict depression<br />

(Casey et al., 2009). The BDNF gene codes for a neurotrophin that is<br />

highly expressed in the central nervous system, heavily involved in<br />

synaptic plasticity, and strongly influenced by stress. This study<br />

explored BDNF-stress interactions across development to predict<br />

depression symptoms in adolescence.<br />

Methodology: Participants were 228 Caucasian youth (50% female)<br />

who have participated in a longitudinal study <strong>of</strong> child development<br />

since birth (the Wisconsin Study <strong>of</strong> Families and Work, Hyde et al.,<br />

1995). Data were collected during the summers following Grades 5<br />

(mean age = 11.49, SD = .29, 2002 and 2003), 7 (M = 13.49, SD =<br />

.31, 2004 and 2005), and 9 (M = 15.48, SD = .31, 2006 and 2007). At<br />

all time-points participants completed a measure <strong>of</strong> depression<br />

symptoms (CDI), negative life events (APES), and peer sexual<br />

harassment victimization (PSHV AAUW items). DNA was collected<br />

with buccal swabs at age 15. The recessive homozygous group was<br />

combined with the heterozygotes: Met carriers (both Val/ Met and<br />

Met/Met n = 60) and Val/Val homozygotes (n = 168).<br />

Results: Multiple regression was used to test whether the BDNF<br />

Val66Met polymorphism would interact with negative life events or<br />

peer sexual harassment victimization to predict depression symptoms<br />

at age 15 (controlling for depression symptoms at age 11). Maximum<br />

likelihood parameter estimates with standard errors were used to<br />

account for the skewed CDI scores. Sex and age 11 CDI were<br />

included in the model to increase power to test substantive questions<br />

about BDNF, stress measures, and their interaction on depression.<br />

We first regressed age 15 CDI on gender, age 11 CDI, BDNF, age 13<br />

APES, and the BDNF-APES interaction. The overall model<br />

accounted for a significant amount <strong>of</strong> variance in depression, R 2 =<br />

0.24, F(5, 222) = 3.72, p < .001. A significant main effect <strong>of</strong> BDNF<br />

was observed, b = 3.58, t(222) = 2.47, p = .013, such that Met carriers<br />

had more depressive symptoms than Val/Val individuals. A<br />

significant main effect <strong>of</strong> age 13 stress was observed, b = 0.38, t(222)<br />

= 3.24, p = .001, such that depression symptoms at age 15 increased<br />

with increasing stress at age 13. BDNF moderated the stress effect on<br />

depression, b = -0.41, t(222) = -2.54, p = 0.01. For Val/Val<br />

individuals, more stress predicted increased depression, b = 0.38, t<br />

(222) = 3.24, p = 0.001. Stress did not predict depression for Met<br />

carriers, b= 0.02, t (222) = 0.14, p = 0.89. This genetic-vulnerability<br />

by stress model was then repeated using age 13-peer sexual<br />

harassment victimization as the stressor. A significant effect <strong>of</strong> the<br />

BDNF-PSHV interaction was also observed, b = -0.58, t (222) = -<br />

2.23, p = 0.026, with increasing levels <strong>of</strong> PSHV only increasing<br />

depression symptoms in Val/Val individuals (b = 0.50, t(222) = 2.42,<br />

p = 0.02).<br />

Conclusions: Using both a general and specific measure <strong>of</strong> stress, the<br />

current study demonstrates a genetic vulnerability-stress interaction<br />

that predicts depression symptoms across development. BDNF<br />

Val66Met significantly moderated the relationship between stress at<br />

age 13 and depression symptoms at age 15 (averaging across gender<br />

and controlling for depression symptoms at age 11). With increasing<br />

stress at age 13, Val/Val youth were at increased risk for depression<br />

symptoms at age 15 however, there was no association between age<br />

13 stressors and age 15 depression for Met-carriers. Interestingly,<br />

Met carriers had elevated depression symptoms regardless <strong>of</strong> stress,<br />

whereas Val/Val individuals were stress reactive. Consistent with the<br />

neurotrophic hypothesis <strong>of</strong> depression, the Met allele was the risky<br />

allele for depressive symptoms under low to medium levels <strong>of</strong> stress<br />

(likely because <strong>of</strong> its lower BDNF secretion and decreased synaptic<br />

plasticity). However, under higher levels <strong>of</strong> stress, Val/Val<br />

individuals were at increased risk for depression symptoms. These<br />

findings support a nuanced depiction <strong>of</strong> BDNF Val66Met by stress<br />

interactions that predict depression symptoms in adolescence.<br />

119<br />

PP100<br />

WITHDRAWN


PP101 SCN1A ALTERS BRAIN STRUCTURE AND<br />

MODULATES AGE-DEPENDENT ALTERATIONS IN<br />

POSTERIOR CINGULATE ACTIVATION DURING A<br />

WORKING MEMORY TASK<br />

S. Meier*(1), T. Demirakca(2), W. Brusniak(3), I. Wolf(2), K.<br />

Liebsch(3), N. Tunc-Skarka(2), V. Nieratschker(1), S. Witt(1), F.<br />

Matthäus(4), G. Ende(2), H. Flor(3), M. Rietschel(1), C. Diener(3),<br />

T. Schulze(5)<br />

1. Department <strong>of</strong> Genetic Epidemiology in Psychiatry, Central<br />

Institute <strong>of</strong> Mental Health, University <strong>of</strong> Heidelberg 2. Department <strong>of</strong><br />

Neuroimaging, Central Institute <strong>of</strong> Mental Health, University <strong>of</strong> 3.<br />

Department <strong>of</strong> Cognitive and Clinical Neuroscience, Central Institute<br />

<strong>of</strong> Mental 4. Institute <strong>of</strong> Applied Mathematics, University <strong>of</strong><br />

Heidelberg 5. Department <strong>of</strong> Psychiatry and Psychotherapy,<br />

University Medical Center<br />

*sandra.meier@zi-mannheim.de<br />

Introduction: SCN1A encodesthe sodium channel alpha subunit and<br />

is a susceptibility gene for neurological disorders. The C allele <strong>of</strong> the<br />

SCN1A variant rs10930201 has recently been found associated with<br />

poor short-term memory performance in a genome wide association<br />

study. Further rs10930201 was observed to be related to brain<br />

activity during a working memory task.<br />

Methodology: As age strongly affects cognitive functioning and agerelated<br />

changes in brain structure as well as in task-induced brain<br />

activity are well documented, we investigated the effects <strong>of</strong> age<br />

andrs10930201 on brain structure and cortical function during a<br />

working memory.<br />

Results: We observed a main effect <strong>of</strong> the genotype on grey matter<br />

density, carriers <strong>of</strong> the C allele showed reduced gray matter densities<br />

in frontal and insular regions. FMRI analysis revealed no main<br />

effects for the genotype, however C allele carrriers displayed<br />

increased activity in the posterior cingulate cortex.<br />

Conclusions: Activation <strong>of</strong> PCC has been reported to be agedependent<br />

and enhanced activity is related to poorer performance.<br />

Our results are in line with this suggestion, since only subjects with<br />

the vulnerability allele <strong>of</strong> rs10930201 displayed a significantly<br />

increased activity <strong>of</strong> the PCC with increasing age. In summary, our<br />

results illustrate a modulatory effect <strong>of</strong> the SCN1A polymorphism on<br />

brain morphology and age-related changes in the brain activation in<br />

regions subserving working memory.<br />

120<br />

PP102 ASSOCIATION ANALYSIS OF ANK3 GENE<br />

VARIANTS IN NORDIC BIPOLAR DISORDER AND<br />

SCHIZOPHRENIA CASE-CONTROL SAMPLES<br />

M. Tesli*(1,2), P. Koefoed(3,4), L. Athanasiu(1,2,5), M.<br />

Mattingsdal(5), O. Gustafsson(2,6), I. Agartz(1,7,8), L. Rimol(8), A.<br />

Brown(1,9,10), K. Wirgenes(1,2), E. Jönsson(7), A. Kähler(2,11,12),<br />

T. Werge(13), O. Mors(14), I. Melle(1,2), S. Djurovic(1,2,5), O.<br />

Andreassen(1,2)<br />

1. Institute <strong>of</strong> Clinical Medicine, University <strong>of</strong> Oslo 2. Division <strong>of</strong><br />

Mental Health and Addiction, Oslo University Hospital 3.<br />

Department <strong>of</strong> Neuroscience and Pharmacology, University <strong>of</strong><br />

Copenhagen 4. Department O, Center <strong>of</strong> Psychiatry 5. Department <strong>of</strong><br />

Medical <strong>Genetics</strong>, Oslo University Hospital 6. deCODE <strong>Genetics</strong> 7.<br />

Department <strong>of</strong> Clinical Neuroscience, Karolinska Institutet and<br />

Hospital 8. Department <strong>of</strong> <strong>Psychiatric</strong> Research, Diakonhjemmet<br />

Hospital 9. Department <strong>of</strong> Biostatistics, University <strong>of</strong> Oslo 10.<br />

Department <strong>of</strong> Mathematics, University <strong>of</strong> Oslo 11. Department <strong>of</strong><br />

<strong>Genetics</strong>, University <strong>of</strong> North Carolina at Chapel Hill 12. Department<br />

<strong>of</strong> Medical Epidemiology and Biostatistics, Karolinska Institutet 13.<br />

Research Institute <strong>of</strong> Biological Psychiatry, Copenhagen 14. Center<br />

for <strong>Psychiatric</strong> Research, Aarhus University Hospital<br />

*m.s.tesli@medisin.uio.no<br />

Introduction: Several studies have found genetic variants in Ankyrin<br />

3 (ANK3) to be associated with bipolar disorder (BD). We wanted to<br />

investigate the potential association between two previously<br />

identified ANK3 SNPs and BD in a Nordic case-control sample. Due<br />

to evidence <strong>of</strong> genetic overlap with schizophrenia (SZ), we also<br />

tested for association between these two SNPs and SZ.<br />

Methodology: The two ANK3 SNPs rs10994336 and rs1938526 were<br />

genotyped in a Scandinavian case-control sample (N = 854 BD cases,<br />

1073 SZ cases and 2919 healthy control subjects). Statistical analyses<br />

in the Scandinavian sample were performed with the s<strong>of</strong>tware<br />

PLINK, using the Cochrane-Mantel-Haenszel test to correct for<br />

population stratification.<br />

Results: When we combined the results from our Scandinavian<br />

sample with the results from an Icelandic sample (N = 435 BD cases,<br />

651 SZ cases and 11491 healthy controls), we found ANK3 SNP<br />

rs10994336 to be nominally significantly associated with BD in this<br />

combined Nordic BD sample (N = 1289 /14105) (P=0.024 using<br />

Fisher`s combined probability test). None <strong>of</strong> the SNPs were<br />

significantly associated with SZ in the combined Nordic SZ sample<br />

(N = 1724/14410).<br />

Conclusions: These results give further support to the hypothesis that<br />

ANK3 is a BD susceptibility gene and that ion channel dysfunction<br />

may be involved in the underlying pathophysiological mechanisms <strong>of</strong><br />

the BD clinical phenotype.


PP103 SIMILARITIES AND DIFFERENCES IN<br />

PERIPHERAL BLOOD GENE EXPRESSION SIGNATURES<br />

OF INDIVIDUALS WITH SCHIZOPHRENIA AND THEIR<br />

FIRST-DEGREE BIOLOGICAL RELATIVES<br />

S. Glatt*(1), W. Stone(2), N. Nossova(3), C. Liew(3), L.<br />

Seidman(2), M. Tsuang(4)<br />

1. SUNY Upstate Medical University 2. Harvard Medical School 3.<br />

GeneNews, Ltd. 4. University <strong>of</strong> California, San Diego<br />

*glatts@upstate.edu<br />

Introduction: Several studies have evaluated the potential utility <strong>of</strong><br />

blood-based whole-transcriptome signatures as a source <strong>of</strong><br />

biomarkers for schizophrenia. This endeavor has been complicated<br />

by the fact that individuals with schizophrenia <strong>of</strong>ten differ from<br />

appropriate comparison subjects on more than just the presence <strong>of</strong> the<br />

disorder for example, individuals with schizophrenia typically receive<br />

antipsychotic medications, and have been dealing with the sequelae<br />

<strong>of</strong> this chronic illness for some time. The inability to control such<br />

factors introduces a considerable degree <strong>of</strong> uncertainty in the results<br />

to date.<br />

Methodology: To overcome this, we performed a blood-based geneexpression<br />

pr<strong>of</strong>iling study <strong>of</strong> schizophrenia patients (n=9) and<br />

unaffected comparison subjects (n=12), while also including a sample<br />

<strong>of</strong> nonpsychotic biological siblings <strong>of</strong> the individuals with<br />

schizophrenia (n=9).<br />

Results: These unaffected biological siblings, who may harbor some<br />

<strong>of</strong> the genetic predisposition to schizophrenia, exhibited a host <strong>of</strong><br />

differences from unaffected comparison subjects, many <strong>of</strong> which<br />

were shared by their schizophrenic siblings, perhaps indicative <strong>of</strong><br />

underlying risk factors for the disorder. Several genes that were<br />

dysregulated in both individuals with schizophrenia and their siblings<br />

related to nucleosome and histone structure and function, suggesting<br />

a potential epigenetic mechanism underlying the risk state for the<br />

disorder. Nonpsychotic siblings also displayed some differences<br />

from comparison subjects that were not found in their affected<br />

siblings, suggesting that the dysregulation <strong>of</strong> some genes in<br />

peripheral blood may be indicative <strong>of</strong> underlying protective factors.<br />

Conclusions: This study illustrated the potential utility and increased<br />

informativeness <strong>of</strong> including unaffected first-degree relatives in<br />

research in pursuit <strong>of</strong> peripheral biomarkers for schizophrenia.<br />

121<br />

PP104 BLOOD-BASED GENE EXPRESSION SIGNATURES<br />

OF EARLY AUTISM<br />

S. Glatt*(1), M. Tsuang(2), M. Winn(3), L. Lopez(2), K. Pierce(2),<br />

M. Weinfeld(2), C. Carter(2), N. Schork(3), E. Courchesne(2)<br />

1. SUNY Upstate Medical University 2. University <strong>of</strong> California, San<br />

Diego 3. The Scripps Research Institute<br />

*glatts@upstate.edu<br />

Introduction: One way to combat autistic spectrum disorders<br />

(ASDs) would be to discover their biomarkers, which potentially<br />

could revolutionize diagnosis and management <strong>of</strong> these<br />

disorders. Here we report the first cross-sectional results <strong>of</strong> wholetranscriptome<br />

expression pr<strong>of</strong>iling in freshly drawn peripheral blood<br />

mononuclear cells (PBMCs) from a longitudinal study <strong>of</strong> young<br />

children with a variety <strong>of</strong> neurodevelopmental disorders including<br />

ASDs. Our main objective was to determine the potential utility <strong>of</strong> a<br />

transcriptomic signature as a biomarker <strong>of</strong> early autism by<br />

discriminating toddlers with an ASD from those with other<br />

developmental disorders, such as language delay and global<br />

developmental delay, as well as typically developing (TD) toddlers.<br />

Methodology: Enrolled subjects were first diagnosed with (or<br />

identified as being at-risk for) an ASD, language delay, other<br />

developmental delay, or TD between the ages <strong>of</strong> 12 and 46<br />

months. Each subject’s PBMC mRNA expression pr<strong>of</strong>ile was<br />

determined by microarray.<br />

Results: Numerous genes with low false-discovery rates<br />

distinguished developmental disorders from each other and from TD<br />

subjects. The 313 genes most reliably changed in AD subjects<br />

relative to two independent TD groups were most strongly associated<br />

with mitotic cell cycle regulation. Other over-represented biological<br />

processes included endopeptidase activity, cerebral cortex<br />

development, microtubule processes, negative regulation <strong>of</strong> neuron<br />

apoptosis, and neuron migration.<br />

Conclusions: PBMCs may provide a useful source <strong>of</strong> biomarkers<br />

that are highly specific to the early stages <strong>of</strong> particular<br />

neurodevelopmental disorders including ASDs. Ongoing<br />

longitudinal analyses will determine if these blood-based biomarker<br />

pr<strong>of</strong>iles fluctuate as symptom pr<strong>of</strong>iles change over time with<br />

intensive behavioral treatment.


PP105 THE PSYCHIATRIC SUSCEPTIBILITY GENE<br />

CACNA1C AND ITS RELATIONSHIP WITH PERSONALITY<br />

TRAITS, DEPRESSIVE SYMPTOMS, AND COGNITIVE<br />

FUNCTION IN THE GENERAL POPULATION<br />

ECIP<br />

J. Strohmaier*(1), S. Wüst(1), S. Witt(1), V. Nieratschker(1), S.<br />

Meier(1), A. Loerbroks(2), J. Frank(1), T. Stürmer(3,4,5), M.<br />

Amelang(6), M. Rietschel(1), T. Schulze(7)<br />

1. Central Institute <strong>of</strong> Mental Health, Division <strong>of</strong> Genetic<br />

Epidemiology in Psychiatry 2. Mannheim Institute <strong>of</strong> Public Health,<br />

Social and Preventive Medicine, Universitätsmedizin Mannheim,<br />

Medical Faculty Mannheim, Heidelberg University 3. Division <strong>of</strong><br />

Pharmacoepidemiology and Pharmacoeconomics and Division <strong>of</strong><br />

Preventive Medicine, Brigham and Women's Hospital, Harvard<br />

Medical School 4. Department <strong>of</strong> Epidemiology, University <strong>of</strong> North<br />

Carolina, Gillings School <strong>of</strong> Global Public Health 5. Department <strong>of</strong><br />

Internal Medicine VI, University <strong>of</strong> Heidelberg 6. Institute <strong>of</strong><br />

Psychology, University <strong>of</strong> Heidelberg 7. Department <strong>of</strong> Psychiatry<br />

and Psychotherapy, University Medical Center, Georg-August-<br />

University<br />

*jana.strohmaier@zi-mannheim.de<br />

Introduction: Recent genome-wide association studies report strong<br />

evidence for an association between the A-allele <strong>of</strong> rs1006737 in<br />

CACNA1C and bipolar disorder, schizophrenia, and major<br />

depression. We assessed the relationship <strong>of</strong> rs1006737 with<br />

endophenotypes <strong>of</strong> these disorders (neuroticism, extraversion, anger<br />

suppression, sense <strong>of</strong> coherence, dispositional optimism, depression,<br />

perceived social support, psychoticism, working memory, and<br />

declarative memory) within a large longitudinal cohort study. We<br />

further assessed a possible recently described sex-specific effect <strong>of</strong><br />

rs1006737 (Dao et al. 2010).<br />

Methodology: A population-based sample <strong>of</strong> 3793 subjects aged 40-<br />

65 was screened for personality with the Eysenck-Personality-<br />

Inventory (EPI),the State-Trait Anger Expression Inventory (STAXI),<br />

the Sense <strong>of</strong> Coherence Scale (SOC), the Life Orientation Test<br />

(LOT), for perceived social support questionnaire (SozU), depression<br />

and psychoticism. At 10-years follow-up a subsample <strong>of</strong> all subjects<br />

aged 70+ was screened for depression and cognition with the<br />

Geriatric Depression Scale (GDS), the Telephone Interview <strong>of</strong><br />

Cognitive Status (TICS), the East Boston Memory Test (EBMT), a<br />

Verbal Fluency Test und the Digit- Span Backwards Test. All<br />

subjects were genotyped for rs1006737.<br />

Results: In women, rs1006737 was significantly associated with<br />

depression, neuroticism, sense <strong>of</strong> coherence, perceived social<br />

support, and psychoticism. G-allele carriers scored higher on<br />

depression, neuroticism, psychoticism, and lower on sense <strong>of</strong><br />

coherence and perceived social support than AA-homozygotes. The<br />

strongest association was observed for the resilience factor sense <strong>of</strong><br />

coherence (p=0.001 Bonferroni corrected for multiple testing). In<br />

women, rs1006737 was further associated with cognitive function on<br />

a nominal level. Again, we observed an association with the opposite<br />

allele than the previously described risk-allele.<br />

Conclusions: The risk variant rs1006737 is associated with<br />

personality traits and cognitive function in women <strong>of</strong> the general<br />

population. Two previous findings also reported associations <strong>of</strong> the<br />

A-allele with low neuroticism (Kühner et al.2010 small healthy<br />

sample), and less baseline depression severity (Casamassima et al.<br />

large clinically depressed sample). The sex-specific effects <strong>of</strong><br />

rs1006737 need to be disentangled in larger samples. Our results<br />

suggest that phenotype characterization for psychiatric genetic<br />

association studies need to include the whole continuum from<br />

maladaptive to adaptive/competent functioning.<br />

122<br />

PP106 MICRODELETION IN 10Q23.1 CORRELATED WITH<br />

A SUBTYPE OF SCHIZOPHRENIA WITH SEVERE<br />

DELUSIONS<br />

M. Zeledon*(1,2,3), M. Taub(5), P. Chen(6), A. Pulver(3,4), D.<br />

Avramopoulos(1,3), A. Sawa(3), D. Valle(1)<br />

1. McKusick-Nathans Institute <strong>of</strong> Genetic Medicine, Johns Hopkins<br />

University School <strong>of</strong> Medicine 2. Predoctoral Training Program in<br />

Human <strong>Genetics</strong>, Johns Hopkins University School <strong>of</strong> Medicine 3.<br />

Department <strong>of</strong> Psychiatry and Behavioral Sciences, Johns Hopkins<br />

University School <strong>of</strong> Medicine 4. Department <strong>of</strong> Epidemiology,<br />

Bloomberg School <strong>of</strong> Public Health 5. Biostatistics, Bloomberg<br />

School <strong>of</strong> Public Health 6. Department <strong>of</strong> Medical <strong>Genetics</strong>, National<br />

Taiwan University Hospital<br />

*mzeledo3@jhmi.edu<br />

Introduction: Schizophrenia (SZ) is a severely disabling psychiatric<br />

disease that affects 1% <strong>of</strong> the world's population. Patient symptoms<br />

are <strong>of</strong>ten categorized as either cognitive, positive (hallucinations,<br />

delusions, bizarre thoughts) or negative (social withdrawal, loss <strong>of</strong><br />

motivation, affect flattening, and apathy). In 2003, Fallin et al.<br />

reported a linkage peak (NPL <strong>of</strong> 4.7) at 10q22 in the Ashkenazi<br />

Jewish (AJ) population. In 2009, Chen et al. followed up with fine<br />

mapping association studies in the AJ and found strong evidence <strong>of</strong><br />

association between the quantitative trait "delusion” and three SNPs<br />

in the first intron <strong>of</strong> NRG3. Two other independent groups have since<br />

replicated these findings, making NRG3 a strong candidate gene for a<br />

subtype <strong>of</strong> schizophrenia that presents with florid delusions.<br />

Methodology: In this study, we set out to identify causative variants<br />

in the 162 kb LD block covering the 5’ end <strong>of</strong> NRG3 and containing<br />

the 3 SNPs.<br />

Results: Next-generation Illumina sequencing <strong>of</strong> 47 AJ SZ patients<br />

at either extreme <strong>of</strong> the delusion quantitative trait revealed a novel<br />

1.8kb inherited microdeletion 21.5 kb 5’ <strong>of</strong> NRG3 and located<br />

between two clusters <strong>of</strong> predicted transcription factor binding sites.<br />

The deletion was found with similar frequencies in the general AJ SZ<br />

(2.7%), AJ Bipolar I (2.6%), and AJ Control (2.8%) populations and<br />

was not found in 346 Outbred SZ cases, leading us to believe it may<br />

be specific to the Ashkenazim. The deletion frequency among the AJ<br />

SZ patients with the highest delusion scores (MAF=10.1% n=69, 7<br />

carrying the deletion) was higher than in patients with the lowest<br />

delusion scores (MAF=0%, n=67) whose deletion frequency was<br />

lower than the frequency in the general AJ SZ population. The<br />

frequency <strong>of</strong> the deletion between the high and low delusion groups<br />

was also significantly different (p=0.013).Given the difficulties <strong>of</strong><br />

classifying patients with intermediate delusion scores into high or<br />

low delusion categories, we tested the correlation between delusion<br />

scores and deletion frequency for 658 AJ SZ patients across the range<br />

<strong>of</strong> delusion scores, and found this deletion to be significantly<br />

correlated with higher delusion factor scores (p genotype<br />

effects=0.0069).<br />

Conclusions: Due to its location 21.5kb 5’ <strong>of</strong> the annotated<br />

transcription start site and between annotated transcription factor<br />

binding sites, this deletion may be causative by perturbing regulation<br />

<strong>of</strong> NRG3 expression. Alternatively, it may have no effect itself but<br />

tag a haplotype containing the causative variant or variants.<br />

Understanding the ways in which variation in this region affect NRG3<br />

expression would allow us to move from these genetic observations<br />

to deciphering the mechanistic pathways that may lead to risk for SZ<br />

or modifications <strong>of</strong> the SZ phenotype. We are currently testing this<br />

hypothesis in vitro through Dual-Luciferase Assays in HEK239,<br />

HT22 and cultured mouse primary cortical neurons, as well as in vivo<br />

using a LacZ reporter in zebrafish.


PP107 INDEPENDENT EFFECTS OF SEROTONERGIC AND<br />

DOPAMINERGIC POLYMORPHISMS ON TRAIT<br />

IMPULSIVITY<br />

G. Varga*(1), A. Szekely(1), P. Antal(2), P. Sarkozy(2), Z.<br />

Demetrovics(1), Z. Nemoda(3), M. Sasvari-Szekely(3)<br />

1. Institute <strong>of</strong> Psychology, Eötvös Loránd University 2. Department<br />

<strong>of</strong> Measurement and Information Systems, Budapest University <strong>of</strong><br />

Technology and Economics 3. Institute <strong>of</strong> Medical Chemistry,<br />

Molecular Biology and Pathobiochemistry, Semmelweis University<br />

*triptamin@gmail.com<br />

Introduction: According to twin studies heritability <strong>of</strong> trait<br />

impulsivity is approximately 45%. Results from candidate gene<br />

studies are contradictory impulsivity phenotypes measured with<br />

different self report methods demonstrate either the role <strong>of</strong> the<br />

dopaminergic or the serotonergic neurotransmitter system. Our<br />

present study comprises polymorphisms from both neurotransmitter<br />

systems and investigates association with trait impulsivity.<br />

Methodology: The Barratt Impulsiveness Scale (BIS-11) was used to<br />

assess trait impulsivity in a sample <strong>of</strong> 687 Caucasian young adults<br />

within the age range <strong>of</strong> 18-33 years. Analyses <strong>of</strong> 6 genetic<br />

polymorphisms <strong>of</strong> the dopaminergic (DRD2, DRD4, COMT) and<br />

serotonergic (SLC6A4, HTR1A, HTR1B) system were carried out<br />

using ANCOVAs with sex as a covariate.<br />

Results: Significant association <strong>of</strong> a dopaminergic and a serotonergic<br />

polymorphism is reported: lower impulsivity scores characterized the<br />

group carrying the DRD4 7–repeat (p=.006) and the HTR1B 1997 G<br />

alleles (p=.003). Findings endured after Bonferroni correction.<br />

Conclusions: Our study examined the effect <strong>of</strong> three serotonergic<br />

and three dopaminergic candidate polymorphisms on trait<br />

impulsivity, using a large, ethnically homogeneous sample <strong>of</strong><br />

Caucasian young adults from a narrow age range. The presented<br />

results indicate significant and additive effect <strong>of</strong> two <strong>of</strong> the studied<br />

polymorphisms. These results support the combined role <strong>of</strong><br />

dopaminergic and serotonergic transmitter systems on self-report<br />

impulsivity.<br />

123<br />

PP108 PERIPHERAL BLOOD-EXPRESSION PROFILES AS<br />

BIOMARKERS FOR POSTPARTUM DEPRESSION IN A<br />

HIGH-RISK POPULATION<br />

D. Mehta*(1), L. Kraus(1), M. Rex-Haffner(1), J. Newport(2), Z.<br />

Stowe(2,3), E. Binder(1,2)<br />

1. Max Planck Institute <strong>of</strong> Psychiatry 2. Emory University School <strong>of</strong><br />

Medicine, Department <strong>of</strong> Psychiatry and Behavioral Sciences 3.<br />

Emory University School <strong>of</strong> Medicine, Department <strong>of</strong> Obstetrics and<br />

Gynecology<br />

*divs5@mpipsykl.mpg.de<br />

Introduction: Postpartum depression affects as many as 13% <strong>of</strong><br />

women and has a strong negative impact not only on the mother, but<br />

also on the development <strong>of</strong> the infant. Early detection, possibly by<br />

using biomarkers and timely treatment <strong>of</strong> the disorder is therefore<br />

very important for both mother and child. The aim <strong>of</strong> this study was<br />

the identification <strong>of</strong> biomarkers for postpartum depression in a high<br />

risk population by global assessment <strong>of</strong> expression changes in<br />

peripheral blood over pregnancy and the early postpartum period and<br />

the characterization <strong>of</strong> effects <strong>of</strong> post-partum onset depression on<br />

transcriptional pr<strong>of</strong>iles.<br />

Methodology: Multiple measurements from 46 women at high risk<br />

for depressive symptoms during pregnancy and the early postpartum<br />

period were collected in a longitudinal study <strong>of</strong> peripartum<br />

depression. All women had a history <strong>of</strong> a mood or anxiety disorder<br />

and included 3 groups. 15 women had postpartum onset depression,<br />

21 showed no depressive symptoms during the time <strong>of</strong> observation<br />

and 10 women were continuously depressed. Current depressive<br />

symptoms were assessed using the SCID mood module and the Beck<br />

Depression Inventory (BDI). RNA was extracted from whole blood<br />

in Tempus tubes, amplified and hybridized on the Illumina Human<br />

HT12 v4 microarray containing 47,231 probes. Probes were filtered<br />

for background correction and normalized using the VSN<br />

normalization, resulting in 13488 probes for further analysis.<br />

Statistical analysis was performed in R.<br />

Results: Expression changes across pregnancy and the postpartum<br />

period were assessed using mixed model analysis. Transcriptional<br />

pr<strong>of</strong>iles <strong>of</strong> >3000 transcripts were significantly changed across<br />

pregnancy and the early postpartum period after correcting for<br />

multiple testing using the Bonferroni method <strong>of</strong> correction (p < 3.7 x<br />

10 -6 ). A significant overrepresentation <strong>of</strong> steroid hormone<br />

transcription factor binding sites within these transcripts was<br />

observed. Comparison <strong>of</strong> expression pr<strong>of</strong>iles in the third trimester <strong>of</strong><br />

pregnancy between women with and without postpartum onset<br />

depression using t-tests revealed 496 significant transcripts. Using<br />

expression pr<strong>of</strong>iles from these 496 transcripts, women with and<br />

without postpartum depression could be classified with an overall<br />

prediction <strong>of</strong> 88%, sensitivity <strong>of</strong> 82.4% and specificity <strong>of</strong> 93.3%<br />

using the PAMR package.<br />

Conclusions: Our preliminary results indicate that transcriptional<br />

pr<strong>of</strong>iles <strong>of</strong> a large number <strong>of</strong> transcripts are significantly regulated<br />

across pregnancy, most likely due to the large changes in steroid<br />

hormone levels. Furthermore, there is a subset <strong>of</strong> transcripts whose<br />

expression in the third trimester <strong>of</strong> pregnancy is significantly<br />

different between women who will or will not experience postpartum<br />

depression. These transcripts showed a high classification accuracy<br />

and expression levels <strong>of</strong> these transcripts may serve as potential<br />

biomarkers for peripartum depression. Validation <strong>of</strong> these results<br />

using real-time PCR and addition <strong>of</strong> more samples to the current<br />

dataset to add power to the study are currently underway. Our data<br />

suggest that a risk for postpartum depression might be detected as<br />

early as the last trimester <strong>of</strong> pregnancy allowing timely prevention<br />

and treatment strategies.


PP109 MITOCHONDRIAL DYSFUNCTION IN AUTISM<br />

A. Ayyappan*(1), K. Nakamura(2), T. Ismail(2), S. Suda(3), K.<br />

Yamada(4), Y. Iwayama(4), T. Yoshikawa(4), N. Takei(1), N.<br />

Mori(1,2)<br />

1. Research Center for Child Mental Development, Hamamatsu Univ<br />

Sch Med 2. Dept <strong>of</strong> Psychiatry & Neurology, Hamamatsu Univ Sch<br />

Med 3. Dept <strong>of</strong> Psychiatry, Jichi Med Univ 4. Laboratory for<br />

Molecular Psychiatry, RIKEN Brain Sci Inst<br />

*anitha@hama-med.ac.jp<br />

Introduction: Mitochondria serve as the energy powerhouses <strong>of</strong><br />

eukaryotic cells since they generate most <strong>of</strong> the adenosine<br />

triphosphate (ATP), the source <strong>of</strong> chemical energy in cells. Defects<br />

in brain energy metabolism resulting from mitochondrial dysfunction<br />

(MtD) have been implicated in autism, as evidenced by diminished<br />

levels <strong>of</strong> ATP in autistic brains, and mitochondrial abnormalities that<br />

have been observed in the animal models <strong>of</strong> this disorder. Abnormal<br />

energy metabolism may lead to damage to brain cells, resulting in<br />

cognitive-, language- and behavioral- abnormalities. Previous studies<br />

<strong>of</strong> MtD in autism had been restricted to common enzymes involved<br />

in energy metabolism. Most <strong>of</strong> the genetic studies were based on<br />

mutations in the mitochondrial DNA (mtDNA). However, despite the<br />

mtDNA, the genomic DNA encodes several essential proteins<br />

involved in mitochondrial activities. We carried out a comprehensive<br />

study <strong>of</strong> the role <strong>of</strong> MtD in the pathogenesis <strong>of</strong> autism. Expressions<br />

<strong>of</strong> genes involved in diverse mitochondrial functions (e.g.<br />

mitochondrial biogenesis, transport and energy production) were<br />

compared between the postmortem brains <strong>of</strong> autism patients and<br />

healthy controls. The genes that showed altered expressions in autism<br />

were further checked for association with this disorder, in a familybased<br />

study.<br />

Methodology: Frozensamples from autism patients (n=8) and<br />

controls (n=10), belonging to the following brain regions, were<br />

obtained from the Autism Tissue Program, U.S.A., (i) anterior<br />

cingulate gyrus (ACG 8 autism, 13 controls), (ii) motor cortex (MC 7<br />

autism, 8 controls), and (iii) thalamus (TH 8 autism, 9 controls). The<br />

Human Mitochondria PCR Array (SABiosciences, MD, USA), which<br />

pr<strong>of</strong>iles the expressions <strong>of</strong> 84 genes involved in diverse functions <strong>of</strong><br />

mitochondria, was used to quantify gene expression based on SYBR<br />

Green real-time qRT-PCR method. For the association study, we<br />

analyzed DNA samples from 841 Caucasian- and 188 Japanese-<br />

families. Single nucleotide polymorphisms (SNPs) for the study were<br />

selected from the data <strong>of</strong> International HapMap Project. TaqMan<br />

method <strong>of</strong> SNP genotyping was used to score the SNPs. Family<br />

Based Association Test (FBAT) was used to examine the association<br />

<strong>of</strong> SNPs with autism.<br />

Results: Several genes showed altered expressions in the brains <strong>of</strong><br />

autism patients compared to controls. Among these, metaxin 2<br />

(MTX2 2q31.1), neur<strong>of</strong>ilament, light polypeptide (NEFL 8p21.2) and<br />

solute carrier family 25, member 27 [SLC25A27, also known as<br />

uncoupling protein 4 (UCP4) 6p12.3] showed consistently reduced<br />

expressions in the ACG, MC and TH <strong>of</strong> autism patients compared to<br />

controls. Further, we checked the associations <strong>of</strong> MTX2, NEFL and<br />

SLC25A27 with autism in Caucasian- and Japanese- family samples.<br />

NEFL (rs2979704 in Exon 4 UTR) showed a significant association<br />

(p=0.038) with autism in the Caucasian samples.<br />

Conclusions: This is the first comprehensive study <strong>of</strong> MtD in autism.<br />

Considering our results, NEFL is a promising candidate gene for MtD<br />

in this disorder. NEFL is one <strong>of</strong> the most abundant cytoskeletal<br />

components <strong>of</strong> neurons. It plays a pivotal function in the assembly<br />

and maintenance <strong>of</strong> axonal cytoskeleton, playing a role in regulating<br />

neuronal development, specifically neurite outgrowth. NEFL is<br />

located in 8p21.2, which has been identified as a potential<br />

susceptibility locus in a previous genome-wide association study.<br />

Furthermore, chromosome 8p has been identified as a potential hub<br />

for developmental neuropsychiatric disorders.<br />

124<br />

PP110 INVESTIGATING THE RELATIONSHIP<br />

BETWEEN IRON AND IQ OR DEPRESSION<br />

IN ADOLESCENTS<br />

N. Mills*, N. Hansell, M. Wright, J. Whitfield, N. Martin, N. Wray<br />

Queensland Institute <strong>of</strong> Medical Research<br />

*natalie.mills@qimr.edu.au<br />

Introduction: Inflammatory markers, including ferritin, C-reactive<br />

protein, and pro-inflammatory cytokines have been implicated in the<br />

etiology <strong>of</strong> psychiatric disorders such as major depressive disorder<br />

they have also been related to measures <strong>of</strong> cognition. Only a few<br />

studies have investigated this relationship and these studies have<br />

relatively small sample size and have rarely focussed on adolescents.<br />

Our objective is to investigate the phenotypic and genetic relationship<br />

between measures <strong>of</strong> circulating levels <strong>of</strong> iron (ferritin, transferrin,<br />

and saturation <strong>of</strong> transferrin) and <strong>of</strong> IQ and depressive symptoms in<br />

adolescents. (Transferrin saturation is a measure <strong>of</strong> the degree to<br />

which transferrin (iron transport protein) receptors are occupied by<br />

iron).<br />

Methodology: Data was collected from the ongoing Brisbane<br />

Adolescent Twin Study, based at Queensland Institute <strong>of</strong> Medical<br />

Research. Here, we report data from twin adolescents, all<br />

aged greater than 15.0 years but less than 17.0 years. The sample<br />

size comprised between 1,434 and 2,463 twin adolescents, depending<br />

on the measure. Full-scale IQ (FIQ) was assessed by the<br />

Multidimensional Aptitude Battery (MAB) and can be expressed on<br />

the sub-scales <strong>of</strong> verbal IQ (VIQ), and performance IQ<br />

(PIQ). Depressive symptoms were extracted from the Somatic and<br />

Psychological Health Report (SPHERE) and a sum score <strong>of</strong> item<br />

response was created for each individual. A log transformation was<br />

applied to the ferritin measures. Data was analysed using the<br />

statistical program Mx and the sib-pair program.<br />

Results: The measures <strong>of</strong> iron, transferrin, transferrin saturation and<br />

log10 <strong>of</strong> ferritin are all highly heritable, with heritabilities <strong>of</strong> 0.67 for<br />

transferrin, 0.77 for transferrin saturation, 0.73 for iron, and 0.75 for<br />

log10 <strong>of</strong> ferritin. Heritabilities <strong>of</strong> FIQ, VIQ and PIQ were found to<br />

be 0.66, 0.59, and 0.68 respectively. The heritability <strong>of</strong> total<br />

SPHERE score was 0.34. Analyses about the phenotypic and genetic<br />

relationship between these measures and depression and cognition<br />

measures are ongoing.<br />

Conclusions: We aim to provide insight into the relationship<br />

between inflammatory markers and depression and cognition.


PP111 THE ASSOCIATION OF THE NICOTINIC<br />

ACETYLCHOLINE RECEPTOR (CHRNα5) GENE AND THE<br />

BRAIN-DERIVED NEUROTROPHIC FACTOR (BDNF) GENE<br />

WITH DIFFERENT ASPECTS OF SMOKING BEHAVIOR<br />

E. Breetvelt*(1,2), M. Numans(2), W. Hoeben(1), E. Strengman(3),<br />

J. Luyxk(1), M. Aukes(1,2), S. Bakker(1), R. Kahn(1), R.<br />

Oph<strong>of</strong>f(1,3,4), M. Boks(1,2)<br />

1. a) Rudolf Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Psychiatry, University Medical Center Utrecht 2. b) Julius Center for<br />

Health Sciences and Primary Care, University Medical Center<br />

Utrecht 3. c) Department <strong>of</strong> Medical <strong>Genetics</strong>, University Medical<br />

Center Utrecht, Utrecht 4. d) Center for Neurobehavioral <strong>Genetics</strong>,<br />

University <strong>of</strong> California Los Angeles<br />

*e.j.breetvelt@umcutrecht.nl<br />

Introduction: Background: Recent studies show that different<br />

aspects <strong>of</strong> smoking behavior are associated with the alpha-5 subunit<br />

<strong>of</strong> the nicotinic acetylcholine receptor (CHRNA5) gene and the gene<br />

coding for brain-derived neurotrophic factor (BDNF). This raises the<br />

question whether the amount <strong>of</strong> cigarettes used per day has a<br />

different genetic background than smoking initiation and what other<br />

smoking phenotypes may be relevant. Aim: Replicate these recent<br />

findings in a large population based sample.<br />

Methodology: We investigated the association with smoking<br />

initiation and the number <strong>of</strong> cigarettes used per day as well as<br />

additional smoking phenotypes in a population based sample <strong>of</strong> 2.166<br />

participants <strong>of</strong> Dutch origin.<br />

Results: rs16969968 in the CHRNA5 gene was associated with the<br />

amount <strong>of</strong> nicotine use and in particular smoking 25 cigarettes or<br />

more per day. rs6265 in the BDNF gene was not associated with<br />

smoking initiation. Instead, in this sample this SNP was associated<br />

with smoking cessation.<br />

Lifetime Current<br />

smoking smoking<br />

Cigarettes<br />

Smoking<br />

smoked per<br />

cessation<br />

day<br />

AA 52.8% 21.4% 59.4% 16.2 9.6 20.0%<br />

Smoking 25<br />

(SD) cigarettes or more<br />

per day<br />

p-value 0.43 0.80 0.83 0.02 0.01<br />

AG 54.0% 23.7% 56.3% 13.8 8.6 13.5%<br />

p-value 0.51 0.42 0.17 0.36 0.05<br />

GG 55.5% 22.2% 60.4% 13.1 8.5 7.7%<br />

reference reference reference reference reference<br />

Total 54.4% 22.8% 41.5% 13.7 8.7 11.6%<br />

table 1<br />

Results for CHRNA5 rs16969968. P-values were calculated using<br />

homozygotic wild-type as reference group. P-values below 0.05 are<br />

highlighted in bold.<br />

Ever<br />

smoking<br />

Current Smoking<br />

smoking cessation<br />

Cigarettes<br />

smoked per<br />

day<br />

Smoking 25<br />

(SD) cigarettes or more<br />

per day<br />

CC 62.5% 13.6% 77.8% 9.5 5.8 0.0%<br />

p-value 0.21 0.10 0.02 0.14 n.a.<br />

CT 53.1% 25.0% 53.2% 14.2 8.9 12.0%<br />

p-value 0.42 0.14 0.03 0.51 0.99<br />

TT 54.9% 22.8% 60.0% 13.5 8.6 11.7%<br />

reference reference reference reference reference<br />

Total 54.4% 22.8% 41.5% 13.7 8.7 11.6%<br />

table 2<br />

Results for BDNF rs6265. p-values were calculated using<br />

homozygotic wild-type as reference group. P-values below 0.05 are<br />

high-lighted in bold.<br />

Conclusions: Overall the results confirm the involvement <strong>of</strong> the<br />

nicotinic acetylcholine receptor gene in modifying the amount <strong>of</strong><br />

nicotine use and further suggest involvement <strong>of</strong> the brain-derived<br />

neurotrophic factor gene in smoking behavior.<br />

125<br />

PP112 ASSOCIATION STUDY AMONG THREE BDNF<br />

VARIANTS IN MEXICAN PATIENTS WITH OCD: CASE-<br />

CONTROL AND FAMILY-BASED STUDIES<br />

L. Márquez*(1), B. Camarena(2), C. Lóyzaga(1), S. Hernández(2),<br />

L. Vargas(1), I. Vargas(2), A. Aguilar(2), H. Nicolini(3)<br />

1. Clinic <strong>of</strong> Obsessive-Compulsive Disorder and Spectrum Disorders,<br />

National Institute <strong>of</strong> Psychiatry Ramón de la Fuente Muñiz 2.<br />

Department <strong>of</strong> <strong>Psychiatric</strong> <strong>Genetics</strong>, National Institute <strong>of</strong> Psychiatry<br />

Ramón de la Fuente Muñiz 3. Autonomous University <strong>of</strong> Mexico<br />

City<br />

*pita.rocker@gmail.com<br />

Introduction: Obsessive-compulsive disorder (OCD) is a psychiatric<br />

disorder whose etiology is not yet known, even though it is clear that<br />

genetic factors play an important role. In 2003, an association<br />

between the Brain-Derived Neurotrophic Factor (BDNF) and OCD<br />

was described in a family-based association study, but those results<br />

has not been replicated to date, although some findings support the<br />

theory that BDNF may be implicated in the molecular etiology <strong>of</strong><br />

OCD. In this study, we investigate the role <strong>of</strong> three variants <strong>of</strong> the<br />

BDNF gene (rs6265, rs1519480 and rs7124442) by single SNP and<br />

haplotypes analysis in OCD Mexican patients using a case-control<br />

and family-based association design.<br />

Methodology: Three polymorphic variants <strong>of</strong> BDNF gene were<br />

genotyped in a control group (n=283), OCD subjects (n=260) and<br />

first degree relatives <strong>of</strong> 109 OCD subjects (n=185). All samples were<br />

genotyped using TaqMan allelic discrimination assays. Single SNP<br />

and haplotype analyses were conducted to determine association<br />

between BDNF variants and OCD. Finally, clinical information (age<br />

at onset, Yale-Brown Obsessive-Compulsive Severity Scale and<br />

Symptom Checklist, comorbidity, family history <strong>of</strong> psychiatric<br />

disorders) were obtained in 143 OCD patients for which statistical<br />

analysis was conducted in order to investigate whether any genetic<br />

variant was associated with a specific clinical phenotypes in OCD.<br />

The diagnosis <strong>of</strong> OCD and the clinical assessment were evaluated by<br />

psychiatrists experts in OCD.<br />

Results: Single SNP analysis in case-control study showed a<br />

statistically significant association between rs6265 and OCD. In<br />

particular, we observed a high frequency <strong>of</strong> Val/Val genotype and<br />

Val allele in OCD patients compared with control group ( χ 2 =26.6,<br />

p=0.0000 χ 2 =28.5, p=0.0000, respectively). Also, analysis <strong>of</strong><br />

rs1519480 showed a high frequency <strong>of</strong> G allele in OCD patients<br />

compared with the control group ( χ 2 =25.8, p=0.0000). There was no<br />

statistically significant association between the rs7124442 and OCD.<br />

Haplotype analysis showed that A-A-T (rs6265-rs1519480rs7124442)<br />

was found statistically more frequent in OCD group than<br />

control group (p=0.0041), showing a 2.8-fold increased risk <strong>of</strong> OCD.<br />

Also, a low frequency <strong>of</strong> A-G-T haplotype was observed in OCD<br />

patients (p=0.0024).<br />

When age at onset <strong>of</strong> OCD, symptom dimensions, symptom severity<br />

and gender was analyzed in single locus and haplotype-based<br />

analysis, no significant association between any variant or haplotype<br />

was observed, but an increased risk <strong>of</strong> the contamination/cleaning<br />

dimension for males carriers <strong>of</strong> the Val allele <strong>of</strong> rs6265 (p=0.0096).<br />

Finally, the family-based association study showed no significant<br />

differences in the transmission <strong>of</strong> any variant.<br />

Conclusions: We replicated the association between Val allele <strong>of</strong><br />

rs6265 BDNF gene polymorphism and OCD. We found significant<br />

association <strong>of</strong> rs1519480 in OCD patients compared with a control<br />

group. Finally, we observed a high risk haplotype (A-A-T) in OCD<br />

patients and a protective A-G-T haplotype for OCD. Interestingly, the<br />

risk to develop OCD could be dependent <strong>of</strong> being a carrier <strong>of</strong> A<br />

variant <strong>of</strong> rs1519480, region that has never been analyzed in OCD.<br />

Therefore, our findings suggest that BDNF gene could be related to<br />

the development <strong>of</strong> OCD.


PP113 ASSOCIATION OF EVENT-RELATED DELTA BAND<br />

OSCILLATIONS AND SINGLE NUCLEOTIDE<br />

POLYMORPHISMS IN THE CHOLINERGIC NICOTINIC<br />

RECEPTOR GENE LOCUS (CHRNA6-CHRNB3) ON<br />

CHROMOSOME 8<br />

N. Manz*(1), M. Rangaswamy(1), S. Kang(1), A. Goate(2), K.<br />

Bucholz(2), J. Wang(2), J. Rice(2), L. Almasy(3), J. Tischfield(4), M.<br />

Schuckit(5), J. Kramer(6), H. Edenberg(7), L. Bierut(2), B.<br />

Porjesz(1)<br />

1. SUNY Downstate Medical Center 2. Washington University<br />

School <strong>of</strong> Medicine 3. Texas Biomedical Research Institute 4.<br />

Rutgers University 5. University <strong>of</strong> California San Diego 6.<br />

University <strong>of</strong> Iowa 7. Indiana University School <strong>of</strong> Medicine<br />

*nmanz@hbnl.downstate.edu<br />

Introduction: Brain electrical activity recorded using non-invasive<br />

electroencephalography (EEG) provide sensitive measures <strong>of</strong> brain<br />

function with exquisite temporal resolution during cognitive tasks.<br />

Extensive existing literature indicates that the P3 component <strong>of</strong> the<br />

event-related potential (ERP) is reduced in abstinent alcoholics and in<br />

<strong>of</strong>fspring at risk. More recently it has been reported that the theta and<br />

delta event-related oscillations (EROs) underlying P3 are also<br />

reduced in alcoholics and their <strong>of</strong>fspring; theta has a frontal<br />

topography and is related to attention, while delta has a posterior<br />

topography and is related to decision making processes. These ERO<br />

endophenotypes are highly heritableand more proximal to gene<br />

function than diagnosis, providing a powerful strategy in searching<br />

for genes in psychiatric disorders, such as alcoholism. In the<br />

Collaborative Study on the <strong>Genetics</strong> <strong>of</strong> Alcoholism (COGA) we have<br />

found that these ERO phenotypes have been useful in gene<br />

identification associated with a predisposition to develop alcohol<br />

dependence and related disorders. There is evidence in the literature<br />

that nicotinic receptor genes in the CHRNA6-CHRNB3 locus are<br />

associated with smoking and alcohol behaviors.<br />

Methodology: EROs in the delta band (1-3 Hz) were calculated from<br />

EEG recordings during a visual oddball task in 988 adolescents and<br />

young adults (12-25 years) from COGA alcoholic and control<br />

families (643 European-American, 247 African-American, 98 Others)<br />

along with drinking and smoking variables. Analyses <strong>of</strong> delta EROs<br />

at the midline posterior lead where they are maximum and single<br />

nucleotide polymorphisms (SNPs) in the cholinergic nicotinic<br />

receptor gene cluster CHRNA6-CHRNB3 was performed using a<br />

linear regression model for a case-control study with 263 subjects<br />

from families affected with alcoholism and 725 subjects from control<br />

families.<br />

Results: We find evidence for several SNPs in the CHRNA6-<br />

CHRNB3 locus on chromosome 8 to be significantly associated (FDR<br />

corrected p=0.02-0.004) with delta EROs at the posterior midline<br />

electrode (Pz) in response to the target stimulus.<br />

Conclusions: These findings support a role for the cholinergic<br />

nicotinic receptor genes on chromosome 8 (CHRNA6 and CHRNB3)<br />

in the genetic predisposition towards alcoholism, and underscore the<br />

utility <strong>of</strong> brain oscillations and the endophenotype approach in<br />

understanding complex psychiatric disorders.<br />

126<br />

PP114 CORTICOTROPIN-RELEASING HORMONE<br />

RECEPTOR1 GENE SNPS INTERACT WITH STRESS TO<br />

PREDICT DEPRESSION LONGITUDINALLY<br />

M. Moore*(1), C. Van Hulle(1), E. Shirtcliff(2), M. Essex(1), K.<br />

Lemery-Chalfant(3), H. Goldsmith(1)<br />

1. University <strong>of</strong> Wisconsin-Madison 2. University <strong>of</strong> New Orleans 3.<br />

Arizona State University<br />

*mnmoore@wisc.edu<br />

Introduction: The genetic diathesis-environmental stress model<br />

posits that studying either environmental or genetic factors in<br />

isolation is likely to prove less fruitful than studying the interplay<br />

between the two (Rutter, 2005). The importance <strong>of</strong> stress in<br />

precipitating depressive symptoms has led to a focus on the<br />

hypothalamic- pituitary-adrenal (HPA) axis in depression’s<br />

etiology. Corticotropin-releasing hormone (CRF) is a neuropeptide<br />

that regulates activity <strong>of</strong> the HPA axis. Polymorphisms <strong>of</strong><br />

corticotropin-releasing hormone receptor1 (CRHR1), the gene that<br />

codes for the CRF 1 receptor, are thus likely to have an impact on<br />

HPA axis activity (Binder & Nemer<strong>of</strong>f, 2010). Although CRH<br />

receptors have received only modest research attention, CRHR1<br />

polymorphisms appear to be associated with the risk for depression in<br />

adult samples (Liu et al., 2006; Bradley et al, 2008). Patterns <strong>of</strong><br />

family resemblance for afternoon cortisol levels are strongly<br />

influenced by environmental (not genetic) factors and thus may act as<br />

a proxy <strong>of</strong> family stress (Schreiber et al., 2006; Ellenboge et al.,<br />

2004; Burke et al., 2005). We examined two allelic variants in the<br />

CRHR1 gene and their interaction with afternoon cortisol to predict<br />

depressive symptoms longitudinally from middle childhood to early<br />

adolescence.<br />

Methodology: Participants were from a longitudinal, birth recordbased<br />

twin panel; relevant data were available for 493 Caucasian<br />

individuals (48.9% female). At mean age 8.02 years (SD=.80),<br />

participants and their parents completed assessments that included<br />

diagnostic interviews and DNA collection via<br />

buccal swabs. Participants also provided saliva samples, collected<br />

between 1500h and 1900h on three consecutive days, which were<br />

used for cortisol assays. At mean age 13.40 years (SD=1.30)<br />

participants again completed diagnostic interviews to assess<br />

depressive symptoms.<br />

Results: For both rs242938 and rs242939 (which were in strong<br />

linkage disequilibrium), the homozygous minor allele group was<br />

combined with the heterozygous group and compared to the<br />

homozygous major allele group (for rs242938, T carriers [n=81] were<br />

compared to CC homozygotes [n=409]; for rs242939, G carriers<br />

[n=59] were compared to AA homozygotes [n=399]). We tested<br />

main and interactive effects <strong>of</strong> the polymorphisms and afternoon<br />

cortisol level on depressive symptoms using generalized estimating<br />

equations to account for the paired nature <strong>of</strong> twin data, and included<br />

gender as a covariate. We observed main effects for both<br />

polymorphisms, such that minor allele carriers reported higher<br />

depressive symptoms in adolescence (for rs242938, b=.36, z=2.03,<br />

p=.04; for rs242939, b=.46, z=2.14, p=.03). In interaction with<br />

afternoon cortisol, rs242938 significantly predicted later depression,<br />

b=6.83, z=3.05, p=.002, such that T carriers with higher afternoon<br />

cortisol levels at age 8 reported higher depressive symptoms in<br />

adolescence. A similar interaction was found for G carriers <strong>of</strong><br />

rs242939, b=7.72, z=2.66, p=.008. For both polymorphisms, family<br />

stress was a significant predictor <strong>of</strong> later depression for minor allele<br />

carriers (for rs242938, p


PP115 A GENOME-WIDE ASSOCIATION STUDY OF<br />

BIPOLAR DISORDER AND CO-MORBID MIGRAINE:<br />

REPORTING ASSOCIATION WITH THE GENE<br />

NEUROBEACHIN (13Q13.2)<br />

K. Oedegaard*(1,2,3), S. Johansson(4,5), T. Greenwood(3), O.<br />

Fasmer(1,2), H. Akiskal(3), B. Genome Study(6), J. Haavik(4), J.<br />

Kelsoe(3)<br />

1. MoodNet, Haukeland University Hospital, <strong>Psychiatric</strong> Division 2.<br />

University <strong>of</strong> Bergen, Department <strong>of</strong> Clinical Medicine, Section <strong>of</strong><br />

Psychiatry 3. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California 4.<br />

Department <strong>of</strong> Biomedicine, University <strong>of</strong> Bergen 5. Center for<br />

Medical <strong>Genetics</strong> and Molecular Medicine, Haukeland University<br />

Hospital 6. Bipolar Genome Study<br />

*keti@haukeland.no<br />

Introduction: Both bipolar disorder (BPAD) and migraine are highly<br />

heritable disorders with considerable non-genetic components. The<br />

co-morbidity between migraine and BPAD has been demonstrated in<br />

a large amount <strong>of</strong> epidemiological and clinical studies, and there is<br />

also some evidence for shared genetic vulnerability.<br />

Methodology: To identify susceptibility factors for the<br />

BPAD/migraine phenotype, we conducted a GWAS in 1157 cases<br />

with bipolar disorder collected through the NIMH <strong>Genetics</strong> Initiative<br />

for Bipolar Disorder and genotyped at 1M SNPs by the Translational<br />

Genomics Institute (TGEN) in Phoenix (TGEN1). We compared<br />

BPAD patients without migraine (n= 765) to BPAD patients with<br />

migraine (n=392).<br />

Results: The strongest evidence for association was found for several<br />

SNPs on chromosome 13 in a region encompassing the gene<br />

Neurobeachin with the strongest signal at rs1160720 (P= 5.96x 10-8).<br />

Conclusions: We hypothesised that the subgroup <strong>of</strong> patients<br />

presenting combined symptoms from two different brain disorders<br />

associated with disturbances in neuronal membrane passage (i.e<br />

migraine and BPAD) might reflect a complex neurodegenerative<br />

phenotype that could share an underlying genetic susceptibility. The<br />

findings <strong>of</strong> this study suggest that NBEA may predispose to a<br />

combined phenotype <strong>of</strong> bipolar disorder and migraine headache,<br />

which comprises a subset <strong>of</strong> bipolar patients.<br />

127<br />

PP116 THE GENETICS OF DEPRESSION SUBGROUPS AND<br />

DIMENSIONS<br />

R. Power*, R. Uher, P. McGuffin, C. Lewis<br />

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

*robert.r.power@kcl.ac.uk<br />

Introduction: Depression has a lifetime prevalence <strong>of</strong> 16% yet as a<br />

phenotype it shows variation in symptoms and severity and differing<br />

response to treatment. The lack <strong>of</strong> replicable genetic findings in<br />

depression further suggests that what is defined as one group<br />

clinically may be heterogeneous. Using the combined data from three<br />

depression studies (DeCC, DeNT, and GENDEP) totalling 3300<br />

cases, we sought to create a more homogenous grouping <strong>of</strong> classes<br />

for genome wide analysis.<br />

Methodology: Three methods were used to explore the symptom<br />

pr<strong>of</strong>iles <strong>of</strong> the individuals, based on 16 binary symptom scores.<br />

Firstly, latent class analysis (LCA) was used to explore a categorical<br />

view <strong>of</strong> depression, aiming to empirically replicate recognized<br />

subtypes <strong>of</strong> depression (melancholic, atypical, etc.). Secondly, factor<br />

analysis (FA) was used to construct quantitative traits, aiming to<br />

provide a more dimensional view <strong>of</strong> depression. Lastly, factor<br />

mixture analysis (FMA) was used to create latent classes within<br />

which individual variation could be accounted for by factors. As even<br />

if depression is made up <strong>of</strong> one or more quantitative traits, the<br />

selection <strong>of</strong> individuals into case control studies would likely result in<br />

class-like distributions.<br />

Results: All three methods produced similar results. LCA resulted in<br />

a 4 class model best fitting the data. The classes could be defined as a<br />

moderate depression class, a severe class, and two classes<br />

intermediate in severity, one with atypical rather than typical features<br />

(increased appetite and sleep, rather than a decrease). FA provided<br />

the best fitted model with 2 factors, and parallel analysis showed<br />

most information was held in these first two factors. The first factor<br />

was loaded on all symptoms, particularly those crucial to diagnosis <strong>of</strong><br />

depression (anhedonia, low mood, etc.), suggesting it represented a<br />

score <strong>of</strong> overall severity. The second factor's loadings reflected the<br />

link between eating and sleeping symptoms, i.e. that overeating<br />

correlated with oversleeping, and the reverse. FMA showed the best<br />

fit model contained 4 classes and 2 factors, and gave similar results to<br />

the LCA model. GWAS analysis <strong>of</strong> the individuals in each <strong>of</strong> the 4<br />

LCA classes against 1600 controls identified several novel<br />

associations at suggestive significance, but none reaching genomewide<br />

significance. The severe class (n=620) had 5 SNPs reaching<br />

suggestive significance. This included one SNP in the q14 region <strong>of</strong><br />

chromosome 2, one in the in the q26 region <strong>of</strong> chromosome 3, and<br />

three on chromosome 5. The class with atypical features <strong>of</strong><br />

depression (n=710) showed association with one SNP in the p25<br />

region <strong>of</strong> chromosome 3. No SNPs were found to be associated with<br />

the other two classes.<br />

Conclusions: The LCA and FA gave empirical evidence for two<br />

distinctions in the diagnosis <strong>of</strong> depression: overall severity and the<br />

correlation <strong>of</strong> eating and sleeping symptoms. Sadly the reduction in<br />

heterogeneity failed to lead to GWAS findings <strong>of</strong> genome wise<br />

significance. The next stage <strong>of</strong> analysis will be to re-analyse with<br />

weighting <strong>of</strong> individuals by their probability <strong>of</strong> belonging to each<br />

class. This should provide a better integration <strong>of</strong> the FMA results and<br />

GWAS data, and hopefully strengthen the suggestive association<br />

findings. We also plan to evaluate the heritability <strong>of</strong> these classes.<br />

We will use confirmatory analysis <strong>of</strong> class allocation in 1600 siblings<br />

<strong>of</strong> the DENT sample that were not included in class construction.<br />

Identification <strong>of</strong> heritable symptom patterns in depression should<br />

produce more heterogeneous populations and diagnoses, which in<br />

turn can be used to improve power in GWAS.


PP117 THE NUMBERS SENSE AND ITS RELATIONSHIP<br />

WITH MATHEMATICAL ABILITY AND DISABILITY<br />

M. Tosto(1), Y. Kovas(1,2), R. Plomin*(2)<br />

1. Psychology Department, Goldsmiths College 2. Social, Genetic<br />

and Developmental Psychiatry Centre, Institute <strong>of</strong> Psychiatry<br />

*robert.plomin@kcl.ac.uk<br />

Introduction: Number Sense refers to the untaught, non-verbal<br />

abilities to estimate and approximate numerosities. These abilities<br />

seem to be specific to the mathematical domain. Studies have shown<br />

that individual differences in Number Sense abilities positively relate<br />

to individual variation in mathematical tests scores and achievement.<br />

To date, the nature <strong>of</strong> these relationships is largely unknown. As part<br />

<strong>of</strong> the Twins Early Development Study (TEDS), we present the first<br />

genetically sensitive investigation looking into the aetiology <strong>of</strong> the<br />

Number Sense and the relationship between Number Sense and<br />

mathematical ability and mathematical disability.<br />

Methodology: A pilot study was conducted to assemble and validate<br />

an on-line battery comprising seven tests designed to assess the<br />

specific ability to approximate numerosity, numerical magnitude,<br />

non-verbal spatial memory, speed <strong>of</strong> processing, and a range <strong>of</strong><br />

mathematical skills. All tests were age appropriate for 16 year olds.<br />

The battery is currently being administered to the TEDS as part <strong>of</strong> the<br />

assessment at 16-years <strong>of</strong> age. 2,274 twins belonging to the first<br />

cohort have already completed the assessment. More data will be<br />

available when the second cohort will start testing at the end <strong>of</strong><br />

August 2011. The twins also completed the PISA (Programme for<br />

International Student Assessment) questionnaires measuring attitude,<br />

interest, self efficacy and time spent on mathematics.<br />

Results: Preliminary analysis on the data from the first cohort shows<br />

that Mathematical performance is significantly predicted by Number<br />

Sense and general cognitive abilities measures. The genetic analysis<br />

<strong>of</strong> mathematical performance at 16 showed the pattern similar to<br />

previous ages, with a moderate (.6 - .5) genetic component, almost<br />

nonexistent shared environment and high (.5 - .7) non-shared<br />

environment. The first genetic analysis on the Number Sense<br />

measures showed a surprisingly low heritability, and a large nonshared<br />

environmental component. Among the PISA questionnaire,<br />

mathematical ability was related to mathematics self-efficacy and the<br />

time spent doing mathematical activities. More detailed behavioural<br />

and quantitative genetic analyses are currently under way.<br />

Conclusions: To date the Number Sense was thought to have<br />

evolutionary origin, however the study presented in this paper is the<br />

first to show that these abilities have, in fact, low familiality. The<br />

results <strong>of</strong> this study show that mathematics remains a heritable trait at<br />

16 years <strong>of</strong> age. Multivariate genetic analyses will uncover the<br />

aetiology <strong>of</strong> the co-variance among the traits. The results <strong>of</strong> this study<br />

will help to understand better the mechanisms underlying<br />

mathematical variation and will help explaining the development <strong>of</strong><br />

mathematical disability.<br />

128<br />

PP118 AUTISM AND SCHIZOPHRENIA IN 22Q11DS, TRUE<br />

PLEIOTROPY OR DIFFERENT WAYS OF LOOKING AT<br />

THE SAME PHENOTYPE<br />

J. Vorstman*(1), E. Breetvelt(1), E. Chow(2,3), A. Bassett(2,3)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Psychiatry, University Medical Center Utrecht 2. Clinical <strong>Genetics</strong><br />

Research Program, Centre for Addiction and Mental Health 3.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Toronto<br />

*j.a.s.vorstman@umcutrecht.nl<br />

Introduction: An increasing number <strong>of</strong> structural genomic variants,<br />

in particular copy number variants (CNVs), is identified as large<br />

effect risk factors for neuropsychiatric diseases. Notably, while at<br />

first specific CNVs were thought to be associated with either<br />

schizophrenia or autism spectrum disorders (ASDs), subsequent<br />

studies reported the occurrence <strong>of</strong> many <strong>of</strong> these CNVs in both<br />

disorders. When one genetic variant can lead to two or more distinct<br />

phenotypes, the concept <strong>of</strong> pleiotropy can be applied. For instance,<br />

deletions at 15q13.3, 17q12, 16p11.2, and 22q11.2, as well as<br />

deletions <strong>of</strong> NRXN1 and duplications <strong>of</strong> VIPR2 are all associated<br />

with both schizophrenia and ASDs. How can we better understand<br />

this pleiotropy? Here, the question is whether the observation <strong>of</strong> both<br />

ASDs and schizophrenia in carriers <strong>of</strong> a specific CNV is to be<br />

considered true pleiotropy or rather as the same phenotype or<br />

expressions <strong>of</strong> the same pathological process, i.e. schizophrenia,<br />

observed at different stages. The 22q11.2 deletion syndrome<br />

(22q11DS) occurs relatively frequently allowing for sufficiently<br />

powered studies to examine the neuropsychiatric pleiotropy observed<br />

in relation to many CNVs. We hypothesized that in 22q11DS, autistic<br />

features during childhood are a precursor manifestation <strong>of</strong><br />

schizophrenia. Therefore ASD in patients with 22q11DS would be<br />

associated with higher rates <strong>of</strong> subsequent schizophrenia in<br />

adulthood.<br />

Methodology: We have tested this hypothesis by studying a cohort<br />

<strong>of</strong> 78 adult 22q11DS patients who underwent thorough and<br />

standardized psychiatric evaluation (as described previously)<br />

allowing a lifetime diagnosis <strong>of</strong> schizophrenia to be made or ruled<br />

out. In addition, caregivers <strong>of</strong> these patients were asked to<br />

retrospectively report ASD symptoms <strong>of</strong> their child during childhood<br />

using the social responsiveness Scale (SRS) and the Social<br />

Communication Questionnaire lifetime (SCQ). We compared mean<br />

SRS T-scores in 22q11DS patients with and without schizophrenia,<br />

and generated 95% Confidence Intervals (CIs) through a<br />

bootstrapping procedure (n = 1000). A Mann-Withney-Wilcoxon test<br />

was used to test differences between the two groups. Using the<br />

questionnaire data in a dichotomous fashion, we applied a binary<br />

logistic model with cut-<strong>of</strong>f scores <strong>of</strong> 12 and 15 for the SCQ and 60<br />

for the SRS T-score as predictors. We calculated Odds Ratio (ORs)<br />

and their 95%CIs, entering age, IQ and sex as covariates.<br />

Results: Mean SRS scores and 95%CIs were virtually identical in<br />

22q11DS patients with and those without schizophrenia (p=0.36).<br />

Results from the logistic regression demonstrated no significant<br />

predictive effect <strong>of</strong> ASD as a dichotomous variable measured by the<br />

SRS or the SCQ.<br />

Conclusions: The results demonstrated that ASD during childhood is<br />

not associated with an increased risk for schizophrenia later in life in<br />

22q11DS patients. The average SRS scores were highly similar<br />

between those with and those without schizophrenia suggesting that<br />

even small effect sizes are unlikely. The previously reported<br />

increased rates <strong>of</strong> both schizophrenia and ASD in 22q11DS patients,<br />

together with the absence <strong>of</strong> any association between these two<br />

disorders in the current study, lend evidence to the possibility that<br />

ASD and schizophrenia should be considered as two distinct,<br />

pleiotropic neuropsychiatric consequences <strong>of</strong> the 22q11.2 deletion.


PP119 ANTIPSYCHOTIC TREATMENT AND COPY<br />

NUMBER VARIATIONS - A PILOT STUDY<br />

C. Skjødt*, A. Ingason, K. Jakobsen, T. Hansen, J. Thygesen, L.<br />

Duong, T. Werge<br />

Research Institute <strong>of</strong> Biological Psychiatry, Mental Health Center Sct<br />

Hans, Copenhagen University Hospital<br />

*celina@regionh.dk<br />

Introduction: The identification <strong>of</strong> multiple genetic high risk factors<br />

<strong>of</strong> schizophrenia seems to suggest the existence <strong>of</strong> a corresponding<br />

number <strong>of</strong> etiological subtypes <strong>of</strong> the disorder subtypes that might<br />

well be characterized by correspondingly distinct pr<strong>of</strong>iles <strong>of</strong> response<br />

to medical therapy. To test this hypothesis, we conducted a blinded,<br />

matched case-control pilot study to examine the correlation between<br />

effect <strong>of</strong> antipsychotic treatment and CNV status.<br />

Methodology: 12 schizophrenia patients carrying CNVs associated<br />

with schizophrenia were matched one-by-one on age and gender to<br />

12 schizophrenia patients without CNVs all 24 patients were<br />

originally ascertained together to the same sample. Clinical data from<br />

ten years <strong>of</strong> medical records (2000-2010) and data from the<br />

comprehensive, national health registers were collected for all 24<br />

patients. Medical records were rated without knowledge <strong>of</strong> CNV<br />

status.<br />

Results: Paired analysis showed that body mass index, smoking,<br />

CYP2D6 phenotype, substance abuse, suicidal behavior and<br />

habitation were similar between the two groups. The CNV carriers<br />

had experienced fewer positive antipsychotic drug trials than the noncarriers<br />

(1.42±1 vs. 2.08±1) and more antipsychotic drug trials<br />

altogether (3.42±2.31 vs. 2.72±1.60), corresponding to a significant<br />

lower success rate per drug trial (p=0.019). Also, we found an<br />

indication that CNV carriers might have an earlier age <strong>of</strong> onset (p =<br />

0.086) and were more hospitalized during their illness, measured as<br />

percentage <strong>of</strong> time spent in hospital during their illness (p = 0.062). A<br />

sample size <strong>of</strong> 48 patients with the same outcome would have yielded<br />

significant results for these last two trends.<br />

Conclusions: These observations seem to support general<br />

expectations that CNV-carrying patients may have an earlier onset <strong>of</strong><br />

illness which also could explain their greater deal <strong>of</strong> hospitalization<br />

and poorer effect <strong>of</strong> antipsychotic treatment. A larger study is needed<br />

to investigate these findings further importantly a sample only twice<br />

as large would have sufficient power to capture significant<br />

differences in these variables.<br />

129<br />

PP120 RELEVANCE OF RECENTLY-IDENTIFIED<br />

SCHIZOPHRENIA RISK VARIANTS TO A CHILD AND<br />

ADOLESCENT CLINICAL POPULATION<br />

A. Doyle*(1,2,3,4), E. Braaten(2,3), C. O'Dushlaine(4), D.<br />

Ruderfer(1,4), D. Toner(1), N. Doty(2,3), K. Chambert(4), J.<br />

Moran(4), E. Hill(1), B. Neale(1,3,4), S. Purcell(1,2,3,4), J.<br />

Smoller(1,2,3,4), P. Sklar(5)<br />

1. Center for Human <strong>Genetics</strong> Research, Massachusetts General<br />

Hospital 2. Department <strong>of</strong> Psychiatry, Massachusetts General<br />

Hospital 3. Harvard Medical School 4. Stanley Center for <strong>Psychiatric</strong><br />

Research at the Broad Institute <strong>of</strong> Harvard & MIT 5. Mount Sinai<br />

School <strong>of</strong> Medicine<br />

*aedoyle@partners.org<br />

Introduction: It is well- documented that the risk mechanisms<br />

leading to severe adult psychopathology begin to unfold in childhood<br />

and adolescence and that treatment prior to the onset <strong>of</strong> disorder<br />

reduces morbidity and mortality. Yet, there are multiple hurdles to<br />

effective early identification and intervention in the field <strong>of</strong> child<br />

psychiatry. Given that the initial clinical manifestations <strong>of</strong> conditions<br />

such as schizophrenia and bipolar disorder are far more common than<br />

conversion to illness, enumerating signs and symptoms has limited<br />

potential to distinguish youth at greatest risk for poor outcome from<br />

those on a more benign course. The search for objective criteria by<br />

which to determine risk is further hindered by imprecise<br />

measurement tools and uncertain diagnostic boundaries.<br />

Unprecedented progress in the past two years in the identification <strong>of</strong><br />

genetic risk variants for schizophrenia and bipolar disorder creates<br />

the potential for improved prognostic algorithms and early and<br />

personalized intervention by incorporating genetic data into clinical<br />

practice. Yet, the probabilistic nature <strong>of</strong> genetic risk in multifactorial<br />

phenotypes and the shared etiology among disorders suggests that the<br />

pathways from genes to behavior will be complex, with risk variants<br />

likely to index traits that cut across what are currently considered<br />

different conditions. Thus, there is considerable work to be done<br />

before the translation <strong>of</strong> such findings to patients will rest on solid<br />

empirical ground.<br />

Methodology: We are merging state-<strong>of</strong>-the-art human genetics<br />

methods with high-quality, comprehensive assessment <strong>of</strong><br />

neurocognitive and psychiatric quantitative traits in child and<br />

adolescent outpatients being evaluated in a new pediatric<br />

neurpsychology clinic at Massachusetts General Hospital. Currently,<br />

we are aiming to describe the full phenotypic spectrum <strong>of</strong> replicated<br />

risk variants from the recent literature. Our eventual goal is to map<br />

trajectories to major adult psychiatric illness. As pro<strong>of</strong> <strong>of</strong> concept for<br />

this work, we have obtained genomewide genotyping and extensive<br />

phenotyping on a pilot sample <strong>of</strong> 74 outpatients from this clinic using<br />

an Illumina 2.5 v1 array. We explored genomewide CNV burden<br />

using PennCNV and extrapolated a polygenic schizophrenia risk<br />

score based on GWAS data from the International Schizophrenia<br />

Consortium.<br />

Results: We have detected both large (greater than 500kb) and small<br />

(between 100 and 30kb) structural variants in regions highlighted in<br />

the recent schizophrenia literature in clinic patients, some <strong>of</strong> whom<br />

already show signs <strong>of</strong> severe psychiatric illness. We have also<br />

documented an association between CNV burden and specific clinical<br />

phenotypes. Additionally, we have established the relevance <strong>of</strong> a<br />

polygenic schizophrenia risk score to the sample, which distinguishes<br />

patients as a whole from healthy adult controls.<br />

Conclusions: Results speak to the promise <strong>of</strong> studying variants from<br />

the recent psychiatric genetics literature in well-characterized child<br />

and adolescent clinical samples that can be followed longitudinally.


PP121 KYNURENE METABOLITE LEVEL (KYNA)<br />

ALTERATION IN BIPOLAR DISORDER PSYCHOSIS AND<br />

ASSOCIATED KMO GENETIC VARIATION<br />

C. Lavebratt*(1), S. Olsson(1), L. Backlund(1), U. Ösby(1), L.<br />

Frisén(1), M. Landén(1,2), S. Erhardt(1), G. Engberg(1), M.<br />

Schalling(1)<br />

1. Karolinska Institutet 2. The Sahlgrenska Academy at Gothenburg<br />

University<br />

*catharina.lavebratt@ki.se<br />

Introduction: Cognitive impairments <strong>of</strong> executive function, attention<br />

and memory are present in bipolar disorder.Kynurenic acid (KYNA)<br />

influences brain glutamatergic and cholinergic transmission by<br />

antagonizing the NMDA receptorand the a7 nicotinic acetylcholine<br />

receptor. In addition, elevated levels <strong>of</strong> endogenous KYNA increases<br />

brain dopamine firing. The concentration <strong>of</strong> KYNA in CSF was<br />

previously reported to be elevated in bipolar disorder. KYNA is one<br />

<strong>of</strong> three products <strong>of</strong> three different enzymatic modifications <strong>of</strong> Lkynurenine.<br />

The enzymes producing KYNA are kynurenine<br />

aminotransferases (KATs). Kynurenine 3-monooxygenase (KMO),<br />

which forms 3-hydroxykynurenine, is rate limiting in the metabolism<br />

<strong>of</strong> KYNA since it is the enzyme being most specific and with the<br />

highest affinity for kynurenine. Reduced KMO activity will thus<br />

shunt the synthesis towards KYNA. The purpose <strong>of</strong> this study was to<br />

elucidate if genetic variation in KMO contributes to the elevated<br />

KYNA level observed in CSF <strong>of</strong> bipolar disorder type 1 patients.<br />

Methodology: KYNA levels were measured in CSF from 99<br />

Swedish bipolar disorder patients. SNPs covering 79% <strong>of</strong> the<br />

variation in KMO were genotyped in these patients and in a second<br />

sample <strong>of</strong> 487 Swedish bipolar disorder type 1 patients and 1044<br />

Swedish anonymous blood donors.<br />

Results: We found that KYNA levels were elevated in CSF <strong>of</strong><br />

bipolar disorder patients with a history <strong>of</strong> psychotic episodes (n=48),<br />

compared to non-psychotic bipolar disorder patients (P=0.039).<br />

KYNA levels in CSF were higher in those psychotic bipolar patients<br />

homozygous for the major C-allele <strong>of</strong> the KMO SNP rs1053230<br />

(P=0.010, n=31). This link between psychotic bipolar patients and<br />

homozygosity for the rs1053230 C-allele was supported by findings<br />

in 487 bipolar type 1 patients where homozygosity <strong>of</strong> the C-allele<br />

was more common among bipolar disorder type 1 patients with<br />

history <strong>of</strong> psychosis (n= 344) compared to other bipolar disorder type<br />

1 patients (OR=1.6, P=0.0093). Haplotype analysis further indicated<br />

that haplotype carrying the rs1053230 C-allele was overrepresented<br />

in psychotic bipolar type 1 patients compared to non-psychotic<br />

bipolar patients (P=0.0096), and compared to healthy blood donors<br />

(n=1044, P=0.030).KMO is located in the outer membrane <strong>of</strong><br />

mitochondria, primarily inside the membrane. The SNP rs1053230<br />

lies at a putative site for substrate interaction outside <strong>of</strong> the<br />

membrane and defines a variation between hydrophilic Arg and<br />

hydrophobic Cys.<br />

Conclusions: Variation in KMO leads to variation in KYNA, that<br />

influences glutamatergic, cholinergic and dopaminergic transmission.<br />

Variation in KMO provides a genetic marker <strong>of</strong> cognitive impairment<br />

in bipolar disorder patients with psychosis.<br />

130<br />

PP122 COMMON POLYGENIC VARIATION<br />

CONTRIBUTING TO SCHIZOPHRENIA RISK ALSO<br />

EXPLAINS VARIATION IN TOTAL BRAIN VOLUME<br />

ECIP<br />

A. Terwisscha van Schel*(1), S. Bakker(1), N. van Haren(1), H.<br />

Boos(1), W. Cahn(1), H. Hulsh<strong>of</strong>f Pol(1), R. Oph<strong>of</strong>f(1,2), R. Kahn(1)<br />

1. University Medical Centre Utrecht 2. University <strong>of</strong> California, Los<br />

Angeles<br />

*aterwiss@umcutrecht.nl<br />

Introduction: Schizophrenia patients have, on average, 3% reduced<br />

brain volumes on magnetic resonance imaging (MRI) scans. Brain<br />

volume reductions in schizophrenia are associated with cognitive<br />

decline and poor outcome. The factors underlying these volume<br />

changes may therefore influence the core features <strong>of</strong> the disorder.<br />

Moreover, brain volume reductions are highly heritable. The aim <strong>of</strong><br />

our study was to investigate if a large combined set <strong>of</strong> single<br />

nucleotide polymorphisms (SNPs) associated with schizophrenia<br />

explains brain volume, both in patients and controls.<br />

Methodology: Subjects were genotyped using the Illumina<br />

HumanHap550 beadchip. After quality control 122,462 SNPs were<br />

used in the analysis. In the discovery sample (568 schizophrenia<br />

patients and 511 controls) we performed a GWAS. We then defined<br />

sets <strong>of</strong> SNPs with p-values below different thresholds (PT's). For each<br />

individual in de target sample <strong>of</strong> 159 schizophrenia patients and 145<br />

healthy controls we calculated the number <strong>of</strong> score alleles. Individual<br />

polygenic schizophrenia scores (PSS) were calculated for every PT-<br />

SNP set by weighting the number <strong>of</strong> score alleles by the logarithm <strong>of</strong><br />

the odds ratio from the GWAS (conform Purcell et al., 2009). We<br />

measured total brain, gray matter, white matter, lateral and third<br />

ventricle volume on a 1.5 Tesla MRI scan and corrected these for<br />

age, sex and intracranial volume. We then performed linear<br />

regressions using brain volumes as dependent variable and PSS as<br />

independent variable. The analyses were repeated including also<br />

disease status and disease*PSS interaction as independent variables.<br />

Results: A higher PSS was significantly associated with reduced<br />

total brain volume (TBV) at different PT's (e.g., for PT 0.3: R 2 =<br />

0.036, p = 0.009) in the total sample. Similar effects <strong>of</strong> PSS were<br />

seen on all other brain volume measures, with the largest effect on<br />

third ventricle volume (R 2 = 0.066, p = 0.002). There was no<br />

significant effect <strong>of</strong> PSS on disease status. The association is also<br />

significant in the patient group only, but not in the control group<br />

only.<br />

Conclusions: This is the first study to show the genetic overlap<br />

between schizophrenia and brain volume based on genotyping data,<br />

suggesting shared biological pathways. Studying the biological<br />

mechanisms <strong>of</strong> the aberrant brain structure in schizophrenia patients<br />

might therefore culminate in important insights in disease<br />

mechanisms. Finally, this study underlines the possible advantages <strong>of</strong><br />

using highly heritable, quantitative phenotypes, such as brain volume,<br />

in genetic studies <strong>of</strong> complex disorders.


PP123 PRELIMINARY EVIDENCE OF GENE X<br />

ENVIRONMENT INTERACTION BETWEEN 5HTTLPR<br />

POLYMORPHISM AND PUBERTAL STATUS IN<br />

ADOLESCENTS’ DEPRESSIVE SYMPTOMS: UNCOVERING<br />

THE ROLE OF GENES IN SENSITIVE PERIODS OF<br />

DEVELOPMENT<br />

G. Salum*(1,2,4,6), A. Bortoluzzi(6,7), P. Silveira(1,3), V.<br />

Bosa(1,3), I. Schuch(1,3), M. Goldani(3), C. Blaya(5,6), S. Leistner-<br />

Segal(1), G. Manfro(1,2,4,6)<br />

1. Federal University <strong>of</strong> Rio Grande do Sul 2. Post-Graduate Program<br />

in Medical Sicences: Psychiatry 3. Center for Child and Adolescent<br />

Health Studies (NESCA) 4. National Institute <strong>of</strong> Developmental<br />

Psychiatry for Children and Adolescents (CNPq, Brazil) 5. Health<br />

Sciences Federal University <strong>of</strong> Porto Alegre 6. Anxiety disorders<br />

program for Child and Adolescent Psychiatry (PROTAIA) 7. Postgraduate<br />

Program in Neuroscience. Health Basic Sciences Institute <strong>of</strong><br />

(ICBS)<br />

*gsalumjr@gmail.com<br />

Introduction: Puberty is a critical sensitive period associated with<br />

the onset <strong>of</strong> several clinical phenotypes <strong>of</strong> psychiatric disorders. The<br />

5HTTLPR polymorphism, with the embedded functional Single<br />

Nucleotide Polymorphism (SNP), is consistently implicated as a<br />

moderator <strong>of</strong> the effects <strong>of</strong> psychosocial stressors in psychiatric<br />

disorders. The aim <strong>of</strong> this study is to investigate if puberty can be<br />

experienced differently regarding depressive symptomatology<br />

between those with high and low functional copies <strong>of</strong> 5HTTLPR.<br />

Methodology: A total <strong>of</strong> 86 adolescents participated in a crosssectional<br />

study including pre-pubertal, pubertal and post-pubertal<br />

adolescents measured through Tanner stages. Depressive<br />

symptomatology was assessed using self-rated the Childhood<br />

Depressive Inventory (CDI). The 5HTTLPR was analyzed into three<br />

groups classified in accordance with expression: LaLa vs. (LgLa or<br />

LaS) vs. (LgLg or LgS or SS). We constructed a Generalized Linear<br />

Model using depressive scores as dependent variable, 5HTTLPR and<br />

Tanner stages as independent variables, as well as their interaction<br />

term using robust estimators. In addition we controlled the analysis<br />

for possible confounding factors as: age, gender, body fat, physical<br />

inactivity. Interaction terms <strong>of</strong> the model were interpreted using<br />

pairwise contrasts.<br />

Results: We found a significant gene x environment interaction<br />

between pubertal status and 5HTTLPR (p interaction=0.001). No<br />

main effects were found for 5HTTLPR or Tanner stages itself.<br />

Pairwise contrasts <strong>of</strong> CDI estimated marginal means between groups<br />

reveal a dose-response gene x environment interaction. In those with<br />

two copies <strong>of</strong> the high function allele (LaLa) there is an important<br />

decrease in depressive symptoms after puberty - pre-pubertal 8.09<br />

(SE=1.58), pubertal 9.51 (SE=2.06), post pubertal 1.84 (SE=0.91). In<br />

those with one low functional copy (LgLa, LgS) this decrease is<br />

attenuated - 8.80 (SE=2.31), 7.99 (SE=0.65), 6.10 (SE=0.95) and<br />

there is no decrease in those with two copies <strong>of</strong> the functional allele<br />

10.43 (SE=10.43), 8.71 (SE=2.05), 9.34 (SE=1.01).<br />

Conclusions: Our findings suggest that 5HTTLPR has a major role<br />

mediating persistence <strong>of</strong> depressive symptoms after puberty. Further<br />

prospective studies should investigate this preliminary hypothesis.<br />

131<br />

PP124 COMMON AND DIFFERENT GENETIC<br />

BACKGROUND WITHIN MAOA GENE FOR BOTH<br />

SCHIZOPHRENIA AND MAJOR DEPRESSION DISORDER:<br />

A CIS-PHASE INTERACTION STUDY<br />

Q. Xu*, J. Zhang, Y. Chen, Y. Shen<br />

Institute <strong>of</strong> Basic Medical Sciences, Chinese Academy <strong>of</strong> Medical<br />

Sciences<br />

*xuqi@pumc.edu.cn<br />

Introduction: The genetic basis <strong>of</strong> schizophrenia and major<br />

depression has been explored extensively, but a causal relationship<br />

has been difficult to identify. We aimed to examine the effect <strong>of</strong><br />

multiple variants within MAOA gene on the pathogenesis <strong>of</strong> these<br />

psychiatric disorders.<br />

Methodology: We genotypedMAOA VNTR polymorphism and 19<br />

SNPs within this gene in 512 unrelated major depression patients,<br />

555 patients with paranoid SCZ and 567 controls from a Chinese<br />

population. We explored the VNTR-SNP additive and cis-phase<br />

interaction effect on the development <strong>of</strong> these two diseases. The<br />

expression level <strong>of</strong> MAOA gene was also determined between<br />

patients and controls by RT-PCR assays.<br />

Results: The main findings <strong>of</strong> present study were that the MAOA<br />

VNTR and functional SNPs may confer disease susceptibility to both<br />

paranoid SCZ and MDD in the Chinese Han population through<br />

different genetic interaction mechanism. In paranoid SCZ, MAOA<br />

may act via multiple variant interactions (corrected P=0.021 in<br />

female) whereas in MDD VNTR-L may act through the cisregulation<br />

to inhibit gene expression (corrected P=0.017 in male).<br />

Moreover, genetic variants within this gene could modify disease<br />

susceptibility by altering gene expression (P=5.86E-07 in SCZ and<br />

P=5.69E-05 in MDD). Also, genders can modify the genetic effect <strong>of</strong><br />

MAOAvariant in both disease<br />

Conclusions: Our study provides useful information that multiple<br />

variants interaction within MAOA gene plays an important role in the<br />

pathogenesis <strong>of</strong> severe mental disorder and which may lead to better<br />

understand <strong>of</strong> complex disorders genetic entities and aid genetic<br />

analysis <strong>of</strong> most complex diseases at a deeper level.


PP125 ASSOCIATION STUDY OF GENETIC VARIATIONS<br />

IN THE HUMAN MELATONIN RECEPTOR (MTNR1A AND<br />

MTNR1B) GENES WITH MOOD DISORDERS AND<br />

CHRONOBIOLOGICAL RELATED PHENOTYPES<br />

V. Soria(1), M. Gratacòs *(2), V. Gálvez(1), G. Escaramís (2), J.<br />

Crespo (1,3), J. Valero (4), A. Gutiérrez-Zotes (4), L. Martorell (4),<br />

E. Vilella (4), J. Menchón(1,3),. Estivill (2,5), M. Urretavizcaya(1,3)<br />

1. Department <strong>of</strong> Psychiatry, Bellvitge University Hospital, IDIBELL<br />

2. Center for Genomic Regulation (CRG-UPF), CIBERESP (CIBER<br />

en Epidemiología y Salud Pública) 3. Department <strong>of</strong> Clinical<br />

Sciences, Bellvitge Campus, Barcelona University, CIBERSAM<br />

(CIBER en Salud Mental) 4. Hospital Psiquiàtric Universitari Institut<br />

Pere Mata, IISPV, Rovira i Virgili University 5. Experimental and<br />

Health Sciences Department, Pompeu Fabra University<br />

*monica.gratacos@crg.cat<br />

Introduction: A large body <strong>of</strong> literature suggests that circadian<br />

dysfunction plays an important role in the pathophysiology <strong>of</strong> mood<br />

disorders (MD). Among the clinical hallmarks <strong>of</strong> MD are<br />

disturbances in sleep/wake cycle, diurnal mood variation and a<br />

seasonal pattern <strong>of</strong> symptom recurrence and remission. In fact,<br />

disruptions <strong>of</strong> circadianrhythms, including abnormalities <strong>of</strong> circadian<br />

phase position and in circadian oscillations <strong>of</strong> melatonin levels have<br />

been described in both unipolar major depressive disorder (MDD)<br />

and bipolar disorder (BD). Melatonin is a pineal gland hormone<br />

involved in the synchronization <strong>of</strong> the circadian clock which signals<br />

day-night information to the endogenous circadian pacemaker located<br />

in the suprachiasmatic nucleus <strong>of</strong> the hypothalamus.<br />

Methodology: We hypothesized that genetic variation in the<br />

melatonin receptor genes could be associated to MD phenotypes. To<br />

test this hypothesis we performed a case-control study including 440<br />

screened control subjects and 445 unrelated patients with MD (256<br />

MDD and 189 BD) diagnosed according to DSM-IV criteria. Patients<br />

completed the Spanish versions <strong>of</strong> the Seasonal Pattern Assessment<br />

Questionnaire and the Horne-Östberg Morningness-Eveningness<br />

Questionnaire in order to evaluate chronobiological phenotypes<br />

which presumably have a strong inherited component, such as<br />

seasonality and diurnal preference or chronotype. First we selected a<br />

set <strong>of</strong> TagSNPs representative <strong>of</strong> the common variation patterns<br />

identified in the European population in the genomic region<br />

containing the human melatonin receptors' genes (MTNR1A,<br />

MTNR1B). We successfully genotyped 12 SNPs using the SNPlex<br />

Genotyping System. Genetic association under four different models<br />

<strong>of</strong> inheritance adjusted for covariates <strong>of</strong> interest were analyzed using<br />

the statistical s<strong>of</strong>tware environment R and the SNPassoc package.<br />

Results: We found significant differences in genotype distributions<br />

between MD cases and controls for 3 SNPs located in MTNR1B gene<br />

(OR=1.72, P unc= 0.003 OR=1.41, Punc= 0.036 OR=1.42, Punc= 0.034).<br />

In addition, a MTNR1B intronic variant was nominally associated<br />

with Global Seasonality Scores (P=0.01) and a SNP located in the 5'<br />

near region <strong>of</strong> MTNR1A gene was associated with diurnal preference<br />

in MD patients (P=0.003).<br />

Conclusions: These preliminary data suggest that the genetic<br />

variability in MTNR1B gene could be involved in the susceptibility to<br />

mood disorders. Further studies in larger samples are needed to<br />

confirm that genetic variants in the melatonin receptor genes might<br />

be a contributing factor to the susceptibility to MD and their<br />

association with chronobiological related phenotypes.<br />

132<br />

PP126 GENOME-WIDE SIGNIFICANT RISK VARIANTS<br />

FOR PSYCHOSIS AND NEURAL INTERMEDIATE<br />

PHENOTYPES<br />

A. Meyer-Lindenberg*<br />

Central Institute <strong>of</strong> Mental Health<br />

*a.meyer-lindenberg@zi-mannheim.de<br />

Introduction: While GWAS will probably not provide all answers<br />

about the genetics <strong>of</strong> schizophrenia, any common variant that does<br />

survive the extreme amount <strong>of</strong> statistical thresholding that this<br />

method requires certainly merits study using intermediate imaging<br />

phenotypes. Of those variants, the one with the strongest support is<br />

zinc finger protein 804A (ZNF804A) encoding a zinc-finger protein<br />

<strong>of</strong> unknown, but possibly regulatory function. Like many candidate<br />

gene variants, ZNF804A is pleiotropic on the level <strong>of</strong> psychiatric<br />

diagnoses, also being associated with bipolar disorder.<br />

Methodology: We use an imaging genetics approach in a sample <strong>of</strong><br />

n=110 Germans <strong>of</strong> German descent genotyped for ZNF804A risk<br />

SNPs and studied using functional neuroimaging,<br />

Results: In functional neuroimaging with an n-back working memory<br />

probe, healthy carriers <strong>of</strong> risk genotypes exhibit no changes in<br />

regional activity. However, they did exhibit pronounced gene dosagedependent<br />

alterations in functional connectivity, which was decreased<br />

from DLPFC across hemispheres and increased with<br />

hippocampus, similar to findings in patients. These connectivity<br />

abnormalities were specific to working memory. Subsequent<br />

workshowed an inability to downregulate key parts <strong>of</strong> the<br />

mentalizing system in conjunction with impaired connectivity <strong>of</strong> this<br />

system to DLPFC, suggesting possible downstream functional<br />

activation effects <strong>of</strong> impaired prefrontal connectivity that mirror<br />

findings in patients. Interestingly, abnormally increased coupling <strong>of</strong><br />

amygdala was also observed, a phenotype unlikely to be related to<br />

heritable risk for schizophrenia and therefore possibly related to risk<br />

for bipolar disorder, where similar findings in patients have been<br />

described.<br />

Conclusions: Neural intermediate phenotypes related to risk for<br />

schizophrenia and bipolar disorder through Z NF804A genetic risk<br />

variants can be defined and map onto systems previously implicated<br />

in patients.


PP127 RISK FACTORS FOR SUICIDE ATTEMPTS IN<br />

PATIENTS WITH BIPOLAR DISORDER<br />

Y. Ahn *(1,2,3), J. Song(1,2), H. Yu(2), Y. Kim (1,2,3)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Science, Seoul National<br />

University College <strong>of</strong> Medicine 2. Department <strong>of</strong> Neuropsychiatry,<br />

Seoul National University Hospital 3. Institute <strong>of</strong> Human Behavioral<br />

Medicine, Seoul National University College <strong>of</strong> Medicine<br />

*aym@snu.ac.kr<br />

Introduction: Suicide is one <strong>of</strong> the greatest burdens among patients<br />

with psychiatric disorders. According to many studies, bipolar<br />

disorder is one <strong>of</strong> the most consistent risk factors for suicide. This<br />

study compared the characteristics <strong>of</strong> bipolar disorder patients with<br />

and without a history <strong>of</strong> suicide attempts to identify the risk factors<br />

for suicide in this disorder.<br />

Methodology: Two hundred twelve patients with bipolar disorder<br />

were recruited from the Bipolar Disorder Clinic at the Department <strong>of</strong><br />

Neuropsychiatry, Seoul National University Hospital. The patients<br />

participated in the genetic study and were individually interviewed<br />

with the Diagnostic Interview for Genetic Studies (DIGS). We<br />

compared the demographic and clinical characteristics <strong>of</strong> those<br />

participants who did and did not attempt suicide.<br />

Results: Forty-four (21.2%) patients had histories <strong>of</strong> suicide<br />

attempts. Suicide attempters were younger, more likely to be<br />

diagnosed with bipolar II disorder. The variables that differentiated<br />

those who did from those who did not attempt suicide included age at<br />

first contact, lifetime history <strong>of</strong> antidepressant use, major depressive<br />

episode, mixed episode, auditory hallucinations, rapid cycling, the<br />

number <strong>of</strong> previous mood episodes, age <strong>of</strong> first depressive episode,<br />

and age <strong>of</strong> first psychotic symptoms. Strong predictors for suicide<br />

attempts were younger age at onset, lifetime history <strong>of</strong> auditory<br />

hallucinations, and history <strong>of</strong> antidepressant use. Discrete-time<br />

survival analysis revealed that antecedent depressive episodes and<br />

psychotic symptoms predicted the first suicide attempt in patients<br />

with bipolar disorder.<br />

Conclusions: Bipolar disorder is one <strong>of</strong> the major psychiatric<br />

conditions associated with the high lifetime risk for suicide. Thus, it<br />

is important that clinicians understand the major risk factors for<br />

suicidal behavior so that they can develop and implement improved<br />

strategies for dealing with this complex behavior.<br />

133<br />

PP128 EVIDENCE OF AN ASSOCIATION BETWEEN<br />

DYSBINDIN GENE AND AGE AT ONSET IN<br />

SCHIZOPHRENIA<br />

K. Lee*(1), E. Joo(1), Y. Kim(2)<br />

1. Department <strong>of</strong> Neuropsychiatry, Eulji University School <strong>of</strong><br />

Medicine, Eulji General Hospital 2. Department <strong>of</strong> Psychiatry &<br />

Behavioral Science and Institute <strong>of</strong> Human Behavioral Medicine,<br />

Seoul National University College <strong>of</strong> Medicine<br />

*lky@eulji.ac.kr<br />

Introduction: The dysbindin gene (DTNBP1) is located in<br />

chromosome 6p22.3, one <strong>of</strong> the regions <strong>of</strong> positive linkage for<br />

schizophrenia. In particular, dysbindin protein has been found to play<br />

a role in the glutatmate neural transmission in the brain. A strong<br />

genetic association between DTNBP1 and schizophrenia has been<br />

replicated through many recent studies. However, we have not<br />

replicated positive association between DTNBP1 and schizophrenia<br />

in our Korean sample. Because schizophrenia has been regarded as a<br />

disease with a quite heterogeneous origin and evolvement, it is useful<br />

to categorize patients with schizophrenia into relatively homogeneous<br />

subsets based on clinical characteristics including age at onset<br />

(AAO). Therefore, we investigated the association between DTNBP1<br />

and AAO<strong>of</strong> schizophrenia.<br />

Methodology: We assessed age at first occurrence <strong>of</strong> positive<br />

psychotic symptoms<strong>of</strong> 197 patients with schizophrenia. Three SNPs,<br />

SNPA, P1763, P1320 <strong>of</strong> DTNBP1were genotyped.<br />

Results: In SNPA, patients with AT (N=10) showed significant<br />

earlier AAO than those with AA (N=187) (p


PP129 A CANDIDATE GENE STUDY OF CHILD ANXIETY<br />

DISORDERS AND RESPONSE TO COGNITIVE BEHAVIOR<br />

THERAPY<br />

ECIP<br />

K. Lester*(1), J. Hudson(2), C. Creswell(3), M. Tropeano(1), P.<br />

Cooper(3), A. Farmer(1), C. Lewis(1), H. Lyneham(2), R. Rapee(2),<br />

D. Collier(1), T. Eley(1)<br />

1. King's College London, MRC Social, Genetic and Developmental<br />

Psychiatry Centre, Institute <strong>of</strong> Psychiatry 2. Centre for Emotional<br />

Health, Department <strong>of</strong> Psychology, Macquarie University 3.<br />

Winnicott Research Unit, School <strong>of</strong> Psychology and Clinical<br />

Languages Sciences, University <strong>of</strong> Reading<br />

*kathryn.lester@kcl.ac.uk<br />

Introduction: Anxiety disorders are highly prevalent and debilitating<br />

conditions that frequently emerge during childhood. Childhood<br />

anxiety is associated with a wide range <strong>of</strong> impairments and <strong>of</strong>ten<br />

precedes other major psychological and physical problems. Like most<br />

complex traits, anxiety disorders are under genetic influence, but<br />

little is known about specific genes involved. Given the pervasive<br />

negative effects <strong>of</strong> anxiety on children's development, it is important<br />

to not only understand the factors involved in the onset and<br />

maintenance <strong>of</strong> child anxiety but it is also <strong>of</strong> critical importance to<br />

treat child anxiety disorders successfully. Cognitive-behavior therapy<br />

(CBT) is the treatment <strong>of</strong> choice for child anxiety disorders, and is<br />

effective in around 60% <strong>of</strong> cases. Poor treatment prognosis is<br />

associated with increased symptom severity, parental<br />

psychopathology and comorbid mood disorders, all <strong>of</strong> which could<br />

reflect genetic influence. The role <strong>of</strong> specific genetic markers in<br />

predicting response to psychological therapy has received almost no<br />

attention to date. However, our group has recently shown that the<br />

5HTTLPR moderates CBT treatment response.<br />

Methodology: 467 anxiety-disordered children aged 6 to 13 years<br />

(with 4 grandparents <strong>of</strong> white European ancestry) undergoing<br />

manualised CBT were recruited from Reading, UK and Sydney,<br />

Australia. DNA was extracted from buccal-cells. A comparison<br />

control sample comprised 459 never psychiatrically-ill white<br />

Europeans. CBT response was defined using parent and child<br />

structured diagnostic interviews and clinician severity ratings for<br />

primary and all anxiety disorders. Genotyping was performed for<br />

markers in selected candidate genes previously associated with<br />

relevant phenotypes: Glucocorticoid Receptor (GR) Corticotrophin<br />

Releasing Hormone (CRH) Regulator <strong>of</strong> G Protein Signalling 2<br />

(RGS2) Glutamic Acid Decarboxylase (GAD) and Nerve Growth<br />

Factor (NGF).<br />

Results: Case-control differences were significant for GR bcl1 (X²(1)<br />

= 4.40, p = .04) RGS2 rs10801152 (X²(2) = 7.73, p = .02), RGS2<br />

rs6428136 X²(2) = 6.42, p = .04) and GAD rs2241165 (X²(2) =<br />

10.23, p = .006) but not CRH. An association was also observed<br />

between NGF rs6330 and treatment response at follow-up. Positive<br />

response at follow-up was seen in 17% and 13.2% more children with<br />

the TT than CT/TT genotype for primary (X²(1) = 5.81, p = .02) and<br />

all (X²(2) = 3.26, p = .07) anxiety disorder response respectively.<br />

Conclusions: These findings support the notion that variants <strong>of</strong> the<br />

GR, RGS2 and GAD gene may play a role in the etiology <strong>of</strong><br />

childhood anxiety disorders and that variants <strong>of</strong> the NGF gene may<br />

contribute to the variability <strong>of</strong> response to psychological therapy.<br />

Identifying factors that help detect children at risk for developing<br />

anxiety disorders could lead to prevention programmes targeting<br />

vulnerable children during periods <strong>of</strong> high stress. Knowledge <strong>of</strong><br />

genetic factors could also be used to inform treatment choices with<br />

respect to psychotherapeutic interventions.<br />

134<br />

PP130 ASSOCIATION BETWEEN COPY NUMBER<br />

VARIATION OF GLYCOGEN SYNTHASE KINASE 3 BETA<br />

AND MOOD DISORDER<br />

Z. Elek*, E. Szántai, R. Kovács-Nagy, G. Faludi, Z. Rónai, M.<br />

Sasvári-Székely<br />

SE<br />

*el.zsuzsa@gmail.com<br />

Introduction: Investigation <strong>of</strong> copy number variations (CNV) is a<br />

novel approach in candidate gene studies. CNVs are caused by the<br />

gain or loss <strong>of</strong> genetic material <strong>of</strong> 150–500 kb length, these<br />

polymorphisms have been shown to be the significant source <strong>of</strong><br />

genetic variation between humans. Recently, glycogen synthase<br />

kinase 3 beta (Gsk3beta) and the adjacent gene, Nr1i2 (pregnane X<br />

receptor is<strong>of</strong>orm) have been reported to associate with bipolar<br />

depression.<br />

Methodology: Here we present a case-control study <strong>of</strong> 221 patients<br />

with mood disorder and 180 controls, involving the chromosomal<br />

region <strong>of</strong> glycogen synthase kinase 3 beta (Gsk3beta) and the<br />

flanking genes, Nr1i2 and C3orf15. Gene dosages have been<br />

measured by real time PCR, analyses were confirmed by<br />

conventional PCR and subsequent capillary electrophoresis.<br />

Results: In accordance with previously published results, an<br />

accumulation <strong>of</strong> increased copy number has been found in the patient<br />

group (5/221 vs. 1/180). Simultaneous analysis <strong>of</strong> our results and<br />

those published by Lachman et al. (1) suggests that the amplification<br />

<strong>of</strong> this region is in association with mood disorder (p = 0.006). We<br />

extended our study to analyze the expansion <strong>of</strong> the region affected by<br />

the CNV, and demonstrated the altered copy number <strong>of</strong> exon 9 <strong>of</strong><br />

Nr1i2 and exons 5 as well as 9 <strong>of</strong> Gsk3beta regions, however neither<br />

the Gsk3beta, nor the Nr1i2 promoters were involved in the CNV<br />

region. Consequently, the increased gene dosage measured in the<br />

Gsk3beta coding region does not include necessarily the complete<br />

functional unit <strong>of</strong> the gene.<br />

Conclusions: In conclusion our study confirmed that CNVs are<br />

important components <strong>of</strong> the genetic background <strong>of</strong> complex<br />

psychiatric disorders, identification <strong>of</strong> their accurate molecular<br />

biological role requires however further analyses.


PP131 COGNITIVE SYMPTOMS IN MANIA ASSOCIATED<br />

WITH THE DAOA AND COMT GENES<br />

D. Hukic*, C. Lavebratt, L. Frisén, L. Backlund, M. Schalling, U.<br />

Ösby<br />

Karolinska Institutet<br />

*dzana8@hotmail.com<br />

Introduction: Manic symptoms are pathognomonic in bipolar<br />

disorder. Individual symptoms <strong>of</strong> mania can be difficult to distinguish<br />

but previous factor analyses have identified cognitive symptoms as a<br />

valid group <strong>of</strong> manic symptoms. Combinations <strong>of</strong> manic symptoms<br />

identified by factor analyses have until now not been used as<br />

phenotypes in genetic analyses. The aim <strong>of</strong> this study was to<br />

investigate genetic associations between cognitive symptoms in<br />

bipolar mania grouped by factor analyses, and SNPs in the candidate<br />

genes DAOA and COMT, in case-case and case-control analysis.<br />

Methodology: Bipolar type 1 patients (n=631) from Sweden were<br />

phenotyped focusing on the most severe episode <strong>of</strong> mania. A factor<br />

analysis showed that the symptoms <strong>of</strong> pressured speech - disturbed<br />

concentration - racing thoughts, formed a factor <strong>of</strong> cognitive<br />

dysfunction (n=291; 46%). DNA was extracted from venous blood,<br />

and 78 SNPs were selected. 1,044 anonymous blood donors served as<br />

controls (ABD controls). The TaqMan OpenArray system (Applied<br />

Biosystems) was used for genotyping and allelic discrimination was<br />

analyzed with the TaqMan Genotyper S<strong>of</strong>tware (Applied<br />

Biosystems).<br />

Results: In the case-case analysis <strong>of</strong> DAOA, SNPs rs3916967 (P=<br />

0.014) and rs2391191 (P =0.040), C respectively A allele were both<br />

significantly associated with the non-cognitive dysfunction patients.<br />

Also, they formed a protective haplotype CA (OR=0.77, P=0.067),<br />

and a risk haplotype TG (OR =1.29, P =0.080). In the case-control<br />

analysis <strong>of</strong> DAOA, rs3916967 (P =0.035) and rs239119 (P =0.028),<br />

C respectively A allele were both significantly associated with noncognitive<br />

dysfunction patients compared to controls. Haplotype<br />

analysis showed a protective haplotype (CA, OR= 0.82, P =0.088),<br />

and a risk haplotype (TG, OR =1.22, P =0.090). In the case-case<br />

analysis <strong>of</strong> COMT, SNP rs5993883 (P=0.024), T allele was<br />

significantly associated with non-cognitive dysfunction patients,<br />

while SNP rs165599 (P=0.036), G allele was associated with<br />

cognitive dysfunction. In the case-control analysis <strong>of</strong> COMT, both<br />

SNPs rs5993883 (P=0.0037) and rs165599 (P=0.018), G allele for<br />

both SNPs were associated with cognitive dysfunction.<br />

Conclusions: We found genetic associations between cognitive<br />

function/dysfunction and the DAOA and COMT genes, both in casecase<br />

and case-control analyses. The result is in line with earlier<br />

findings <strong>of</strong> the DAOA (D-amino acid oxidase activator) gene<br />

complex as one <strong>of</strong> the most interesting loci for the major psychiatric<br />

disorders. DAOA is a part <strong>of</strong> central glutamate system and plays an<br />

essential role in the formation <strong>of</strong> memory and neuronal development.<br />

The association <strong>of</strong> DAOA with schizophrenia has led to the<br />

development <strong>of</strong> several potential treatment agents, active in animal<br />

models that produce antipsychotic effects by enhancing NMDA<br />

receptor function. The DAOA gene is located in a region with a<br />

positive linkage peak for bipolar disorder. DAOA is an activator <strong>of</strong><br />

the DAO protein, involved in the metabolism<strong>of</strong> D-serine, which plays<br />

a role in glutamatergic neurotransmission. Glutamate signaling is<br />

involved in brain development and synaptic plasticity, both <strong>of</strong> which<br />

are modified in individuals affected with bipolar disorder.The COMT<br />

gene has been analyzed in several psychiatric disorders because <strong>of</strong> its<br />

effect on cognitive and emotional function. In the last few years a<br />

number <strong>of</strong> studies have investigated the association between COMT<br />

polymorphisms and both suicidal behavior and personality traits.<br />

135<br />

PP132 EMOTIONAL MODULATION OF RESPONSE<br />

INHIBITION IN UNAFFECTED SIBLINGS OF PATIENTS<br />

WITH BIPOLAR I DISORDER<br />

J. Brand(1), T. Goldberg(1,2,3), C. Gopin(1), A. Malhotra(1,2,3), K.<br />

Burdick*(1,2,3)<br />

1. Zucker Hillside Hospital-NSLIJHS 2. The Feinstein Institute for<br />

Medical Research 3. Albert Einstein College <strong>of</strong> Medicine<br />

*kburdick@nshs.edu<br />

Introduction: Bipolar disorder (BPD) research has identified a<br />

number <strong>of</strong> neurocognitive deficits as potential vulnerability markers<br />

<strong>of</strong> the disorder however, very few studies have focused on patterns <strong>of</strong><br />

performance on affective processing tasks (e.g. Affective Go/No-Go<br />

tasks) which may be more closely tied to the pathophysiology <strong>of</strong> the<br />

illness than standard neuropsychological measures. We have<br />

previously demonstrated that stable BPD patients demonstrate a more<br />

liberal response bias to negative affective stimuli than healthy<br />

controls or patients with schizophrenia (Gopin et al., 2011). The<br />

purpose <strong>of</strong> the current study was to expand upon these prior findings<br />

and investigate these patterns in the unaffected siblings <strong>of</strong> patients<br />

with BPD and to compare them with healthy volunteers who do not<br />

have a first degree relative with psychiatric illness.<br />

Methodology: An affective Go/No-Go test was used to evaluate<br />

inhibitory response to negatively-valenced, positively-valenced, and<br />

neutral stimuli in unaffected siblings (n=20) and healthy controls<br />

(n=20). Accuracy (d') and response bias (beta) served as dependent<br />

variables in a series <strong>of</strong> repeated measures ANCOVAs and<br />

independent samples ANOVAs.<br />

Results: We found a non-significant main effect for group when<br />

comparing accuracy performance (d') on the Affective Go/No-Go <strong>of</strong><br />

unaffected siblings vs. unrelated healthy controls [F = .984 (2, 34), p<br />

= .384]. However, very similar to the pattern that we previously<br />

reported in stable BPD patients, unaffected siblings showed a more<br />

liberal response bias (beta) toward negatively valenced stimuli in<br />

comparison with healthy controls [Main effect <strong>of</strong> group: F = 3.97 (2,<br />

34), p = .028].<br />

Conclusions: The current results extend our recent work which<br />

suggested that bipolar patients – as opposed to individuals with<br />

schizophrenia and healthy controls – are more willing to respond to<br />

negative target stimuli (Gopin et al, 2011). These new data,<br />

indicating that unaffected siblings demonstrate an affective<br />

processing bias similar to that seen in the patient group, implicate this<br />

task as a potential endophenotype in bipolar disorder.


PP133 LINKAGE AND ASSOCIATION ANALYSIS OF THE<br />

WFS1GENE<br />

N. Németh*(1), Z. Bognár(1), Z. Elek(1), G. Varga(2), A. Veres-<br />

Székely(2), Z. Rónai(1), M. Sasvári-Székely(2)<br />

1. Semmelweis University 2. Eötvös Lorand University<br />

*nora.nemeth@eok.sote.hu<br />

Introduction: WFS1 codes the wolframin, which is located in the<br />

membrane <strong>of</strong> the endoplasmic reticulum. Loss-<strong>of</strong>-function mutations<br />

<strong>of</strong> this gene are responsible for the Wolfram-syndrome, <strong>of</strong>ten<br />

associated with psychiatric symptoms. Our earlier results showed that<br />

the rare allele <strong>of</strong> the rs1046322 SNP was more frequent in the group<br />

<strong>of</strong> people with a depression (tendency). Here we examined several<br />

SNPs <strong>of</strong> the WFS1 gene to assess linkage disequilibrium in this<br />

region. An association study was also carried out, to investigate the<br />

connection between the WFS1 and the impulsivity endophenotype.<br />

Methodology: PCR based techniques were employed for the<br />

genotype analysis <strong>of</strong> rs1046322 SNP. Impulsivity was measured by<br />

the Barratt Impulsivity Scale. Genotype-phenotype association was<br />

analyzed by ANOVA. Further genotyping for the linkage study was<br />

done by OpenArray real-time PCR Platform. Linkage analysis was<br />

carried out by the GOLD s<strong>of</strong>tware package.<br />

Results: From 680 healthy persons, participants possessing the<br />

rs1046322 A allele, obtained a significant higher score at the Barratt<br />

Impulsivity Scale (p=0,009). In our pilot study the linkage between<br />

the 3 SNPs was determined. Our findings suggested that there was<br />

only a moderate linkage between rs1046322 and rs1046320 SNPs<br />

(D`=0,282) as well as between rs1046322 and rs9457 polymorphisms<br />

(D`=0,302). In contrast, a strong linkage (D`=0,959) was<br />

demonstrated between the rs1046320 and the rs9457 variations.<br />

Conclusions: Our results confirmed the assumption that the<br />

polymorphic variants <strong>of</strong> the WFS1 gene could contribute to the<br />

genetic background <strong>of</strong> psychiatric diseases. Earlier researches<br />

showed the strongest association between diabetes mellitus and the<br />

rs1046320 SNP, but a functional effect <strong>of</strong> this polymorphism has not<br />

yet been proved. Considering our results the biological role <strong>of</strong> the<br />

rs9457 SNP is possible, as it is not only linked to the rs1046320 SNP,<br />

but it was also suggested that the SNP influences the binding site <strong>of</strong> a<br />

miRNA, which can be the background <strong>of</strong> the biological effect.<br />

136<br />

PP134 SCHIZOPHRENIA GWAS FOLLOW-UP<br />

IMPLICATES THE MHC REGION IN COGNITION<br />

J. Walters*(1), G. Donohoe(2), G. Russo(1), S. Dwyer(1), H.<br />

Williams(1), A. Corvin(2), G. Michael(2), I. Giegling(3), D.<br />

Rujescu(3), M. O'Donovan(1), M. Owen (1)<br />

1. Neuroscience and Mental Health Research Institute, MRC Centre<br />

for Neuropsychiatric <strong>Genetics</strong> and Genomics, Cardiff University 2.<br />

Neuropsychiatric <strong>Genetics</strong> Research Group, Department <strong>of</strong><br />

Psychiatry and Institute <strong>of</strong> Molecular Medicine, Trinity College 3.<br />

Molecular and Clinical Neurobiology and Department <strong>of</strong> Psychiatry,<br />

Ludwig- Maximilians-University<br />

*waltersjt@cardiff.ac.uk<br />

Introduction: Genome-wide association studies (GWAS) have<br />

begun to successfully identify novel genetic risk variants and<br />

associated biological pathways in schizophrenia. Three companion<br />

studies recently reported the first genome-wide significant risk<br />

variants for schizophrenia and additional GWAS significant<br />

schizophrenia risk alleles have been discovered by the <strong>Psychiatric</strong><br />

GWAS Consortium. To take these results forward the actions <strong>of</strong> such<br />

polymorphisms need to be identified, be that at the level <strong>of</strong> protein,<br />

cell, neural circuit or phenotype. Neurocognition <strong>of</strong>fers a way <strong>of</strong><br />

exploring the action <strong>of</strong> polymorphisms on behavioural neural circuits<br />

and has the potential to prioritise risk variants that influence<br />

cognition in schizophrenia- a key therapeutic target. In this study we<br />

set out to investigate the influence <strong>of</strong> the genome-wide significant<br />

risk variants on cognitive phenotypes in schizophrenia cases and<br />

healthy controls.<br />

Methodology: In a discovery and replication phase we used two<br />

large large independent European Caucasian samples <strong>of</strong> healthy<br />

controls (totalling n=2500) and cases with DSM-IV schizophrenia<br />

(n=700). Participants in both the discovery and replication samples<br />

underwent comprehensive neurocognitive assessment using<br />

recognised cognitive batteries. A limited number <strong>of</strong> tests were<br />

selected a priori to be equivalent in the two samples and to represent<br />

domains <strong>of</strong> cognition known to be compromised in schizophrenia IQ,<br />

episodic memory, working memory and attention.<br />

With the aim <strong>of</strong> limiting multiple testing a representative panel <strong>of</strong><br />

SNPs was selected to provide coverage for all genome-wide<br />

significant Sz signals thus far identified. Using linear regression we<br />

analyzed associations with cognition for this panel <strong>of</strong> SNPs.<br />

Results: The majority <strong>of</strong> SNPs that are genome-wide significant for<br />

schizophrenia do not demonstrate association with cognition in this<br />

adequately powered design (p>0.05). The MHC region is associated<br />

with cognition such that the risk alleles for schizophrenia are<br />

associated with diminished cognitive funciton (range <strong>of</strong> significant p<br />

values for cognitive tests, p=0.001-0.004). These results replicate in<br />

the same allelic direction in an independent sample (p=0.015).<br />

Conclusions: Recent evidence suggests there is at most limited<br />

overlap between the genetics <strong>of</strong> schizophrenia and cognition. Our<br />

results are consistent with these findings in that the majority <strong>of</strong><br />

genome-wide significant SNPs in schizophrenia do not appear to be<br />

associated with perforamnce across a range <strong>of</strong> cognitive domains.<br />

However SNPs in the MHC region displayed robust association with<br />

cognition, a result that was independently replicated. Whilst the<br />

MHC region is genetically complex, further investigation is<br />

warranted given its apparent dual role in schizophrenia and cognition<br />

and the rich potential to identify novel treatment pathways, which<br />

may include the newly discovered role for the MHC complex in<br />

synaptic development and plasticity.


PP135 BURNOUT CLUSTERS AND SLEEP SYMPTOMS<br />

S. Sulkava(1), S. Schoenauer(2), H. Mannila(3), T. Paunio*(1)<br />

1. National Institute for Health and Welfare 2. University <strong>of</strong> Helsinki<br />

and Helsinki Institute for Information Technology 3. Aalto<br />

University<br />

*tiina.paunio@thl.fi<br />

Introduction: Burnout is a work-related stress syndrome that is<br />

linked to sleep disturbances. The most common measure for burnout<br />

is the Maslach Burnout Inventory (MBI) which covers three<br />

dimensions <strong>of</strong> burnout syndrome: emotional exhaustion (EX),<br />

cynicism (CY) and reduced pr<strong>of</strong>essional self-esteem (PROF). In this<br />

study we explored if naturally occurring MBI clusters correlate with<br />

different aspects <strong>of</strong> sleep.<br />

Methodology: Our Finnish population-based study sample consisted<br />

<strong>of</strong> 3224 workers with information on MBI, sleep length, early<br />

morning awakenings, fatigue and Global Seasonality Score. We<br />

performed clustering analysis for 16 items <strong>of</strong> MBI using k-means<br />

algorithm separately for males and females. The stability <strong>of</strong> the<br />

results was ensured. Correlations <strong>of</strong> clusters and sleep traits were<br />

analysed. Data comprising 550 000 genome-wide distributed single<br />

nucleotide polymorphisms (SNPs) from ~1460 subjects will be<br />

correlated with the obtained cluster solutions.<br />

Results: We found analogous stable 4 cluster solutions for both<br />

sexes. The biggest group ("C-Low ") consisted <strong>of</strong> individuals with<br />

generally low scores. Individuals with symptoms <strong>of</strong> cynicism and<br />

exhaustion were clustered together and formed two clusters based on<br />

the degree <strong>of</strong> the symptoms ("C-Intermediate" and "C-High").<br />

Individuals with problems <strong>of</strong> PROF constituted another cluster ("C-<br />

Pr<strong>of</strong>"). Correlations <strong>of</strong> sleep measures and MBI clusters were highly<br />

significant. Sleep was the shortest and the most fragmented, and<br />

complaints about fatigue were the most common in the cluster C-<br />

High. Individuals in C-Pr<strong>of</strong> had slightly elevated symptom scores<br />

relative to those in C-Low, but lower than among those in C-<br />

Intermediate.<br />

Conclusions: Data mining <strong>of</strong> MBI show stable clusterings separating<br />

individuals with different degrees <strong>of</strong> symptoms from the subscales<br />

EX and CY from the individuals with the most prominent symptoms<br />

from the subscale PROF. Individuals differ in sleep related traits<br />

according to their cluster such that individuals in the C-high and Cintermediate<br />

clusters exhibit most symptoms. Our results support the<br />

view that altered sleep is an important aspect <strong>of</strong> burnout syndrome.<br />

The study will comprise a template for subsequent molecular genetic<br />

analyses.<br />

137<br />

PP136 GENOME-WIDE SUPPORTED RISK VARIANT FOR<br />

SCHIZOPHRENIA IMPACTS ON HIPPOCAMPUS<br />

ACTIVATION DURING CONTEXTUAL FEAR<br />

CONDITIONING<br />

S. Pohlack(1), V. Nieratschker*(2), F. Nees(1), M. Ruttorf(3), S.<br />

Witt(2), M. Rietschel(2), H. Flor(1)<br />

1. Department <strong>of</strong> Cognitive and Clinical Neuroscience,Central<br />

Institute <strong>of</strong> Mental Health 2. Department <strong>of</strong> Genetic Epidemiology in<br />

Psychiatry, Central Institute <strong>of</strong> Mental Health 3. Computer Assisted<br />

Clinical Medicine, Medical Faculty Mannheim, Heidelberg<br />

University<br />

*vanessa.nieratschker@zi-mannheim.de<br />

Introduction: Key features <strong>of</strong> schizophrenia are declarative memory<br />

and contextual processing deficits. Recently, the neurogranin gene<br />

has been linked to schizophrenia in a genome-wide association study.<br />

Neurogranin is abundantly expressed in key regions <strong>of</strong> contextual<br />

learning and discrimination, such as the hippocampus. Further,<br />

abnormal functioning <strong>of</strong> the hippocampal formation as a major site<br />

for cognitive and contextual processing has been found in<br />

schizophrenia. Contextual fear conditioning is heritable (35-45%) and<br />

constitutes a paradigm that is well suited to examine the impact <strong>of</strong> the<br />

genome-wide supported variant rs12807809 on cognitive and<br />

contextual processing.<br />

Methodology: Hence, we employed an imaging genetics approach in<br />

healthy volunteers to investigate the influence <strong>of</strong> rs12807809 on the<br />

hippocampus during a contextual fear conditioning paradigm using<br />

structural as well as functional magnetic resonance imaging (fMRI).<br />

Results: Carriers <strong>of</strong> the schizophrenia risk-allele variant showed<br />

significantly decreased activations in the left hippocampus during<br />

acquisition, indicating impaired hippocampal activity during<br />

contextual learning.<br />

Conclusions: The present results suggest that contextual fear<br />

conditioning might constitute a valuable endophenotype for<br />

schizophrenia research. However, independent replication <strong>of</strong> our<br />

results is necessary.


PP137 PHENOTYPIC CHARACTERIZATION OF<br />

SCHIZOAFFECTIVE DISORDER-BIPOLAR TYPE: A<br />

COMPARISON WITH BIPOLAR I DISORDER<br />

L. Forty*(1), K. Gordon-Smith(1,2), E. Russell(1), L. Jones(2), I.<br />

Jones(1), J. Walters(1), N. Craddock(1)<br />

1. Cardiff University 2. University <strong>of</strong> Birmingham<br />

*fortyl@cf.ac.uk<br />

Introduction: The term schizoaffective disorder is used to describe<br />

the co occurrence <strong>of</strong> symptoms suggesting both schizophrenia and<br />

mood disorders. There have been various definitions <strong>of</strong><br />

schizoaffective disorder and the diagnostic category remains poorly<br />

defined with boundaries between affective disorder, schizoaffective<br />

disorder and schizophrenia remaining indistinct. We have shown<br />

schizoaffective bipolar disorder [SABD] to be <strong>of</strong> particular genetic<br />

interest and found that participants with RDC schizoaffective<br />

disorder - bipolar type stand out genetically from both those<br />

participants with bipolar disorder [BD] and those with schizophrenia<br />

[SCZ]. Here we examine phenotypic differences between SABD and<br />

BD.<br />

Methodology: Diagnostic and clinical ratings, including the OPCRIT<br />

symptom checklist, were undertaken on 1731 individuals collected as<br />

part <strong>of</strong> ongoing molecular genetic studies <strong>of</strong> mood and psychotic<br />

disorders. Narrow (DSM-IV) and broad (Research Diagnostic<br />

Criteria) definitions <strong>of</strong> SABD were employed. For each <strong>of</strong> the<br />

clinical variables being investigated, a separate logistic regression<br />

analysis including specified covariates, was conducted to see if the<br />

selected variable was predictive <strong>of</strong> SABD diagnosis.<br />

Results: Including sex, age at interview, illness duration and method<br />

<strong>of</strong> recruitment as covariates in each logistic regression analysis, the<br />

following variables were predictive <strong>of</strong> a SABD, rather than a BD,<br />

diagnosis: Earlier age at onset <strong>of</strong> illness and psychosis (p


PP139 WHITE-MATTER VOLUME AND GENETIC<br />

ANALYSES OF THE MYELIN OLIGODENDROCYTE<br />

GLYCOPROTEIN (MOG) AND OLIGODENDROCYTE<br />

LINEAGE TRANSCRIPTION FACTOR 2 (OLIG2) GENES IN<br />

PATIENTS WITH PEDIATRIC OBSESSIVE-COMPULSIVE<br />

DISORDER (OCD)<br />

G. Zai*(1,2), P. Arnold(2,3), M. Richter(1,2,4), G. Hanna(5), D.<br />

Rosenberg(6), J. Kennedy(1,2)<br />

1. Neurogenetics Section, Centre for Addiction and Mental Health 2.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Toronto 3. Program in<br />

<strong>Genetics</strong> and Genomic Biology and Department <strong>of</strong> Psychiatry, The<br />

Hospital for Sick Children 4. Department <strong>of</strong> Psychiatry, Sunnybrook<br />

Health Science Centre 5. Department <strong>of</strong> Psychiatry, University <strong>of</strong><br />

Michigan 6. Department <strong>of</strong> Psychiatry, Wayne State University and<br />

The Children's Hospital <strong>of</strong> Michigan<br />

*gwyneth_zai@camh.net<br />

Introduction: Several recent neuroimaging studies have implicated<br />

abnormalities <strong>of</strong> white matter in patients with obsessive-compulsive<br />

disorder (OCD). The myelination genes, myelin oligodendrocyte<br />

glycoprotein (MOG) and oligodendrocyte lineage transcription factor<br />

2 (OLIG2), have previously demonstrated significant association with<br />

this disorder, and are clear candidates for white matter<br />

alterations. The purpose <strong>of</strong> this study is to examine the relationship<br />

between white matter volume in OCD and genetic variations in MOG<br />

and OLIG2 genes.<br />

Methodology: Two polymorphisms in the MOG gene,<br />

MOG(C1334T) and MOG(C10991T), and a polymorphism in the<br />

OLIG2 gene, rs928736, were investigated for association with total<br />

white matter volume as measured using magnetic resonance imaging<br />

(MRI) as well as left/right/total anterior cingulate gyrus and<br />

left/right/total orbit<strong>of</strong>rontal cortex (OFC) white matter volumes in 37<br />

pediatric OCD patients. We compared white matter volumes<br />

between allele and genotype groups for each polymorphism using<br />

Analysis <strong>of</strong> Covariance (ANCOVA).<br />

Results: In our preliminary analysis, a significant relationship was<br />

detected between genotype 2/2 <strong>of</strong> MOG(C10991T) and decreased<br />

total white matter volume (P=0.016) and a trend was also observed<br />

between genotype <strong>of</strong> MOG(C1334T) and white matter volume<br />

(P=0.059). We did not observe significant evidence for association<br />

for the other tested polymorphisms or white matter regions.<br />

Conclusions: Our results showed a correlation between a MOG<br />

genetic variant and white matter volume. This finding is intriguing in<br />

light <strong>of</strong> the important role <strong>of</strong> white matter alteration in the<br />

pathoetiology <strong>of</strong> at least some cases <strong>of</strong> childhood-onset<br />

OCD. Further investigation with larger samples and sub-regional<br />

white matter volume phenotypes is required.<br />

139<br />

PP140 ASSOCIATION STUDY BETWEEN EATING<br />

DISORDER SUB-PHENOTYPES AND SEROTONINERGIC<br />

GENES<br />

B. Camarena*(1), S. Hernandez(1), L. Gonzalez(2), G. Flores(2), L.<br />

Campos(1), V. Vazquez(2), A. Caballero(2)<br />

1. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Dept.<br />

Genética. 2. Instituto Nacional de Psiquiatría Ramón de la Fuente,<br />

Eating Disorders Clinic, Mexico City<br />

*camare@imp.edu.mx<br />

Introduction: Several lines <strong>of</strong> evidence implicate the serotoninergic<br />

system in body weight regulation and more specifically in eating<br />

disorders (ED). Interestingly, psychopathological traits that identify<br />

clinical sub-phenotypes have been associated with a serotoninergic<br />

dysfunction. In particular, a functional polymorphism located on the<br />

promoter region <strong>of</strong> SLC6A4 gene (5-HTTLPR) has been associated<br />

with impulsivity, novelty-seeking and compulsivity in ED. In<br />

addition, the 5-HT1Dβ is involved in the regulation <strong>of</strong> 5-HT release<br />

from serotoninergic neuron terminals in the brain. Therefore,<br />

abnormal function <strong>of</strong> this auto-receptor could be involved in the<br />

serotonin dysfunction suggested in ED. We examined association<br />

between SLC6A4 and 5-HT1Dβ gene polymorphisms and clinical<br />

sub-phenotypes in patients with bulimia and anorexia spectrum eating<br />

disorders using a family-based method.<br />

Methodology: Our sample consisted <strong>of</strong> 168 families, 119 patients<br />

with bulimia-spectrum disorders (41% with BN purge type, 3% with<br />

BN non-purge type, 8% with AN binge-eating/purging type, 19%<br />

with bulimia-spectrum EDNOS) and 49 patients with anorexiaspectrum<br />

disorders (15% with AN restricting type and 14% with<br />

anorexia-spectrum EDNOS) according to DSM-IV criteria. Sample<br />

was recruited from Eating Disorder Clinic <strong>of</strong> INPRFM in Mexico<br />

City. Symptoms were evaluated using EDI scale, Plutchik´s<br />

Impulsivity Scale, Overt Aggression Scale, YB-Cornell, HAM-A,<br />

and HAM-D. DNA was analyzed by PCR and 5’exonuclease assay<br />

using TaqMan allelic discrimination assays for SLC6A4<br />

polymorphisms: 5HTTLPR, rs25531 and rs6355; and 5-HT1Dβ:<br />

rs6296, rs6297 and rs130058. Significance level was adjusted to<br />

p≤0.0083.<br />

Results: FBAT analysis <strong>of</strong> SLC6A4 gene polymorphisms in bulimiaspectrum<br />

families showed a high transmission <strong>of</strong> 5HTTLPR s allele<br />

(z=2.94, p=0.0033). However, 5-HT1Dβ gene polymorphism did not<br />

show linkage disequilibrium in anorexia-spectrum families<br />

(p≥0.008). FBAT quantitative analysis <strong>of</strong> clinical sub-phenotypes in<br />

bulimia-spectrum families <strong>of</strong> SLC6A4 gene polymorphism showed a<br />

preferential transmission <strong>of</strong> 5HTTLPR s allele associated with high<br />

scores <strong>of</strong> impulsivity (z=3.02, p=0.0025) and EDI-bulimia sub-scale<br />

(z=3.6, p=0.00036), and a trend for significance for EDI-body<br />

dissatisfaction sub-scale (z=2.6, p=0.0096). We did not find linkage<br />

disequilibrium between 5-HT1Dβ gene and clinical sub-phenotypes<br />

in bulimia-spectrum families. Finally, FBAT analysis in anorexiaspectrum<br />

families did not show linkage disequilibrium between 5-<br />

HT1Dβ gene polymorphisms and clinical sub-phenotypes.<br />

Conclusions: We replicated the association between 5HTTLPR s<br />

allele and bulimia-spectrum disorders. Also, we found association<br />

between 5HTTLPR s allele and impulsivity in subjects with bulimiaspectrum<br />

disorders. Therefore, our results support the findings that<br />

link serotoninergic disturbances with trait <strong>of</strong> impulsivity or<br />

dysregulation in bulimia patients. Analysis <strong>of</strong> alternative phenotypes<br />

in larger samples is necessary to try to identify genetic variants<br />

involves in ED subtypes.


PP141 GENOME-WIDE ANALYSES OF<br />

NEUROCOGNITIVE/BRAINIMAGING PHENOTYPES<br />

REVEAL ENRICHED ASSOCIATION OF NEURONAL AND<br />

PSYCHIATRIC CANDIDATE GENES<br />

ECIP<br />

P. Lee*(1,2), A. Holmes(3,4), J. Fagerness(1), J. R<strong>of</strong>fman(3,4), S.<br />

Purcell(1,2), R. Buckner(3,4), J. Smoller(1,2)<br />

1. Department <strong>of</strong> Psychiatry, Harvard Medical School 2.<br />

Neurodevelopmental & <strong>Psychiatric</strong> <strong>Genetics</strong> Unit, Center for Human<br />

<strong>Genetics</strong> Research, Massachusetts General Hospital 3. Department <strong>of</strong><br />

Psychology, Center for Brain Science, Harvard University 4.<br />

Athinoula A. Martinos Center for Biomedical Imaging,<br />

Massachusetts General Hospital<br />

*phlee@pngu.mgh.harvard.edu<br />

Introduction: A number <strong>of</strong> recent neuroimaging genetics studies<br />

have reported association <strong>of</strong> psychiatric risk genes with alterations in<br />

the brain structure and function in apparently healthy subjects.<br />

Notable examples include the functional effects <strong>of</strong> two schizophrenia<br />

risk genes, ZNF804A and COMT on the disturbed regional<br />

connectivity in the brain default networks. Nevertheless, most <strong>of</strong><br />

prior imaging genetics work used a candidate gene-based approach,<br />

focusing on only a few, widely studied psychiatric candidate genes.<br />

In this study, we employ an un-biased genome-wide analysis<br />

(GWAS) <strong>of</strong> neurocognitive/brainimaging traits to elucidate genetic<br />

influences on normal variation in brain structure and function, and<br />

ultimately those underlying a psychiatric illness.<br />

Methodology: Our study is based on the Brain Genomics Library<br />

(BGL), a multi-site protocol for genetic studies <strong>of</strong> brain and<br />

behavioral phenotypes. The BGL study enrolled more than 2,200<br />

healthy volunteers in Boston, USA (average age 21.3; female ratio<br />

55.96%). A group <strong>of</strong> 14 endophenotypes were assessed for all<br />

participants, measuring diverse aspects <strong>of</strong> brain function, structure,<br />

and personality/temperament relevant to major psychiatric illnesses.<br />

Initial genotyping was done for 470 subjects <strong>of</strong> European ancestry,<br />

yielding genotype data <strong>of</strong> 1,140,419 SNPs (Illumina OMNI 1M).<br />

After quality control and imputation using the 1000 Genome Project<br />

data, 5,888,968 SNPs with imputation quality scores <strong>of</strong> at least 0.8<br />

were retained for statistical analysis. Logistic regression was<br />

conducted on imputed allele dosages <strong>of</strong> individual SNPs for each <strong>of</strong><br />

14 phenotypes using age, sex, and four multidimensional scaling<br />

(MDS) components as covariates. We also examined enriched<br />

association <strong>of</strong> four brain-function and psychiatric candidate gene sets<br />

using a GWAS-specific pathway analysis tool, INRICH<br />

(http://atgu.mgh.harvard.edu/inrich).<br />

Results: Single-SNP-based logistic regression found no SNPs with<br />

genome wide significant association with any <strong>of</strong> 14 phenotypes.<br />

Interesting findings were made in pathway analyses, which identified<br />

enriched association <strong>of</strong> neuronal-activity genes with two functional<br />

and structural brain-imaging phenotypes: Default Network<br />

Connectivity (p=1.2e-03) and Left Amygdala Volume (p=1.3e-03).<br />

Furthermore, two behavioral phenotypes <strong>of</strong> temperament and<br />

personality, namely the Barratts scale and the Negative Affect<br />

Composite score showed significant association with known<br />

psychiatric candidate genes (p=2.3e-03 and 3.9e-04, respectively).<br />

Conclusions: Using a GWAS analysis <strong>of</strong><br />

neurocognitive/brainimaging phenotypes, we found a potential role <strong>of</strong><br />

neuronal and psychiatric candidate genes in modulating the normal<br />

variations <strong>of</strong> the brain structure, function, and<br />

personality/temperament measures. It is noteworthy that a number <strong>of</strong><br />

recent pathway-based GWAS studies have reported involvement <strong>of</strong><br />

brain-function genes in the pathogenesis <strong>of</strong> schizophrenia, autism,<br />

and bipolar disorder. Our work demonstrates that endophenotypebased<br />

genetics studies <strong>of</strong> healthy subjects may provide a powerful<br />

strategy for elucidating the neurogenetic risk mechanisms underlying<br />

these psychiatric disorders, as well as for understanding the<br />

functional consequences <strong>of</strong> specific genes involved in neuronal<br />

activity.<br />

140<br />

PP142 BRAIN VOLUMETRIC PHENOTYPES AND<br />

SEROTONIN SYSTEM GENETIC VARIATION IN<br />

PEDIATRIC OBSESSIVE-COMPULSIVE DISORDER<br />

P. Arnold*(1,2), V. Sinopoli(1,2), K. Wu(2), P. Easter(3), J.<br />

Kennedy(2,4), G. Hanna(5), D. Rosenberg(3)<br />

1. Hospital for Sick Children 2. University <strong>of</strong> Toronto 3. Wayne State<br />

University and the Children's Hospital <strong>of</strong> Michigan 4. Centre for<br />

Addiction and Mental Health 5. University <strong>of</strong> Michigan<br />

*paul.arnold@sickkids.ca<br />

Introduction: Serotonin system genes have been the most studied<br />

candidate genes in OCD but evidence for association has been<br />

equivocal. Individuals with OCD display regional volumetric brain<br />

abnormalities, which provide promising intermediate phenotypes <strong>of</strong><br />

the disorder (Arnold et al., 2009). In this preliminary study we<br />

examined the association between serotonergic system gene<br />

polymorphisms and volumetric brain region abnormalities in children<br />

with OCD.<br />

Methodology: Volumes <strong>of</strong> brain regions selected for a priori<br />

evidence <strong>of</strong> association with OCD (thalamus, anterior cingulate<br />

cortex (ACC), orbit<strong>of</strong>rontal cortex (OFC), and caudate) were<br />

measured using structural magnetic resonance imaging (sMRI) in 20<br />

psychotropic-naïve pediatric OCD patients (ages 7 to 17). A total <strong>of</strong><br />

40 single nucleotide polymorphisms (SNPs) were analyzed from<br />

three serotonergic candidate genes (serotonin transporter, SLC6A4;<br />

serotonin receptor 2A, HTR2A; and serotonin receptor 1B, HTR1B)<br />

for association with regional volume.<br />

Results: In HTR2A, the C allele <strong>of</strong> single nucleotide polymorphism<br />

(SNP), rs1923886, was found to be significantly associated with<br />

reduction in volume <strong>of</strong> the left putamen (adjusted P-value <strong>of</strong><br />

0.035). The same SNP was nominally associated with total and right<br />

putamen volume (not significant after adjustment for multiple<br />

comparisons).<br />

Conclusions: We identified an association between HTR2A and<br />

volume <strong>of</strong> the putamen in psychotropic-naive children with OCD.<br />

These results, although requiring replication in a larger sample<br />

size, are consistent with previous neuroimaging findings implicating<br />

the basal ganglia in OCD pathophysiology. Structural imaging<br />

phenotypes provide potentially valuable intermediate phenotypes that<br />

may be associated with decreased heterogeneity compared to<br />

complex behaviours, and thereby facilitate the identification <strong>of</strong> risk<br />

genes.


PP143 IMPACT OF CATECHOL-O-<br />

METHYLTRANSFERASE VAL158MET (RS4680) GENOTYPE<br />

ON VERBAL WORKING MEMORY IN WOMEN WITH HIV<br />

E. Sundermann*(1), J. Bishop(2), L. Rubin(1), E. Martin(1), M.<br />

Kreek(3), O. Levran(3), M. Randesi(3), K. Weber(4), M. Cohen(4),<br />

M. Pauline(1)<br />

1. University <strong>of</strong> Illinois at Chicago, Department <strong>of</strong> Psychiatry 2.<br />

University <strong>of</strong> Illinois at Chicago, Department <strong>of</strong> Pharmacy Practice 3.<br />

Rockefeller University, Laboratory <strong>of</strong> the Biology <strong>of</strong> Addictive<br />

Diseases 4. The Core Center at Stroger Hospital <strong>of</strong> Cook County<br />

*esundermann@psych.uic<br />

Introduction: The HIV virus enters the central nervous system<br />

(CNS) leading to neurological, cognitive and behavioral<br />

complications. Even in the era <strong>of</strong> combination antiretroviral therapy,<br />

mild neurocognitive impairment persists in approximately 33% <strong>of</strong><br />

HIV-infected individuals particularly in the domains <strong>of</strong> executive<br />

function and learning and memory. The Val158Met (rs4680) single<br />

nucleotide polymorphism (SNP) <strong>of</strong> the catechol-O-methyltransferase<br />

gene (COMT) was shown to impact executive function through its<br />

effect on dopamine metabolism in healthy individuals as well as<br />

individuals with schizophrenia. HIV infection is associated with<br />

decreased dopaminergic activity. Therefore, this study aims to<br />

determine the effect <strong>of</strong> the Val158Met polymorphism on working<br />

memory performance in women with HIV.<br />

Methodology: Participants included 54 HIV seropositive women (33<br />

Met allele carriers & 21 Val/Val) from the Chicago site <strong>of</strong> the<br />

Women’s Interagency HIV Study. All participants were between the<br />

ages <strong>of</strong> 25 and 64 (mean age = 43.6) and had no history <strong>of</strong> CNS<br />

disease or major psychiatric disorders. The sample was 87% African<br />

American (n=47). Participants completed the0, 1 and 2-back<br />

conditions <strong>of</strong> the N-back test which is an established measure <strong>of</strong><br />

working memory. Hierarchical linear modeling was used to test the<br />

association between the Val158Met SNP and N-back performance<br />

after adjusting for relevant covariates.<br />

Results: A significant effect <strong>of</strong> the Val158Met SNP was found on Nback<br />

performance. Specifically, compared to women with the<br />

Val/Val genotype, Met allele carriers had a significantly higher<br />

percent correct on the 1-back (p


PP145 ESTABLISHMENT OF HOMOGENOUS SUB-GROUPS<br />

IN ALZHEIMER’S DISEASE BY EVALUATING EPISTATIC<br />

INTERACTIONS<br />

S. Prabhu*(1), B. Bagepally(1), M. Varghese(2), M.<br />

Purushottam(2), S. Jain(1,2), O. Mukherjee(1)<br />

1. National Centre for Biological Sciences 2. National Institute <strong>of</strong><br />

Mental Health and Neurosciences<br />

*sujanap@ncbs.res.in<br />

Introduction: Alzheimer’s disease (AD) an irreversible and<br />

progressive neurodegenerative disorder is the most common form <strong>of</strong><br />

dementia in the elderly. This disorder follows a dichotomous age<br />

related model. The early onset form is less frequent and has less<br />

number <strong>of</strong> risk genes associated with it having a higher penetrance.<br />

The late onset AD is genetically heterogeneous and has been found to<br />

have multiple gene <strong>of</strong> small effect interacting to give rise to the late<br />

onset pathology. Linkage and candidate gene studies have previously<br />

shown strong association <strong>of</strong> APOE (apoe 2/3/4) with dementia. The<br />

COMT (Val158Met) enzyme catechol-o-methyltranferase, affects the<br />

cognition process through by metabolic degradation <strong>of</strong> released<br />

dopamine in the frontal cortex. The methionine (Met) allele, produces<br />

enzymes which have a lower level <strong>of</strong> activity, hence lesser<br />

degradation <strong>of</strong> dopamine, ultimately affecting cognition.5HTR2A<br />

(His452Tyr) for our study, the tyrosine (Tyr) form <strong>of</strong> this SNP is<br />

shown to be involved in poorer memory performance.<br />

Methodology: The samples used in this study were collected with<br />

informed consent <strong>of</strong> the patients and also after ethical clearance. In<br />

the present study, genotype data was generated for the three loci for<br />

217 Alzheimer’s type Dementia cases and 89 unaffected subjects<br />

(age). In addition to the genotypic data, quantitative intermediate<br />

phenotypes have been used to capture underlying heritable trait<br />

variation and create more homogenous high-risk and low-risk groups<br />

<strong>of</strong> subjects. Data was initially assessed for single loci association<br />

using chi-square test and then checked for gene x gene interactions as<br />

well as gene x quantitative trait (such as age at assessment,<br />

psychometric scores, brain volumes) interactions using<br />

multidimensional factorial reduction (MDR) and Generalized MDR<br />

(GMDR) respectively. MRI data was available for 43 individuals, 24<br />

cases and 19 controls, <strong>of</strong> the entire sample list.<br />

Results: ApoE 4 allele showed positive association (p


PP147 GENOME-WIDE LINKAGE SCAN OF ANTISOCIAL<br />

BEHAVIOR, DEPRESSION AND GENERAL SUBSTANCE<br />

MISUSE IN THE UCSF FAMILY ALCOHOLISM STUDY<br />

I. Gizer*(1), C. Ehlers(2), C. Vieten(3), H. Feiler(4), D. Gilder(2), K.<br />

Wilhelmsen(5)<br />

1. University <strong>of</strong> Missouri 2. Scripps Research Institute 3. California<br />

Pacific Medical Center 4. Lawrence Berkeley National Laboratory 5.<br />

University <strong>of</strong> North Carolina<br />

*gizeri@missouri.edu<br />

Introduction: Epidemiological and clinical studies suggest that rates<br />

<strong>of</strong> antisocial behavior, depression, and substance misuse are<br />

increased among individuals diagnosed with alcohol dependence<br />

relative to those who are not. Given that each <strong>of</strong> these phenotypes has<br />

demonstrated moderate evidence <strong>of</strong> heritability, the present study<br />

conducted genome-wide linkage scans <strong>of</strong> quantitative measures <strong>of</strong><br />

antisocial behavior, depressive symptoms, and general substance<br />

misuse in the University <strong>of</strong> California at San Francisco Family<br />

Alcoholism Study.<br />

Methodology: Participants included 1647 individuals from 713<br />

families recruited as part <strong>of</strong> the UCSF Family Alcoholism Study.<br />

Select scales from the MMPI-2 were used to assess antisocial<br />

behavior (Antisocial Practices scale: ASP), depressive symptoms<br />

(Depression scale: DEP), and general substance misuse (the revised<br />

MacAndrew Alcoholism scale: MAC-R). Linkage analysis was<br />

conducted using the variance components approach implemented in<br />

SOLAR.<br />

Results: Although no loci achieved genome-wide significance (i.e.,<br />

LOD score > 3.3), suggestive evidence <strong>of</strong> linkage to three genomic<br />

regions was observed that was independent <strong>of</strong> alcohol and cannabis<br />

dependence diagnostic status: (1) evidence <strong>of</strong> linkage between<br />

antisocial behavior and chromosome 13 at 11 cM was observed<br />

(LOD = 2.57), (2) evidence <strong>of</strong> linkage between substance misuse and<br />

chromosome 15 at 47 cM was observed (LOD = 3.25), and (3)<br />

evidence <strong>of</strong> multivariate linkage between antisocial behavior, general<br />

substance misuse, and depressive symptoms and chromosome 17 at<br />

57-58 cM was observed (LOD = 3.16).<br />

Conclusions: The chromosome 17 locus is located in a relatively<br />

gene-rich region and includes several candidate genes, including<br />

SLC6A4, PPP1R1B, PNMT, VAT1, MAPT, NEUROD2, and CRHR1<br />

that have been studied in relation to a broad range <strong>of</strong> psychiatric<br />

disorders such as substance dependence, mood and anxiety disorders,<br />

ADHD, and schizophrenia suggesting potentially broad relations<br />

between this region and psychopathology. The loci on chromosomes<br />

13 and 15 have shown previous evidence <strong>of</strong> linkage to antisocial<br />

behavior and substance dependence, respectively.<br />

143<br />

PP148 RESULTS FROM THE IMAGEN GENE X<br />

NEUROIMAGING STUDY ON REINFORCEMENT-<br />

RELATED BEHAVIOUR IN ADOLESCENTS<br />

G. Schumann*, I. Consortium<br />

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

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

Introduction: In the IMAGEN study we aim to identify the genetic<br />

and neurobiological basis <strong>of</strong> individual variability in impulsivity,<br />

reinforcer sensitivity and emotional reactivity, and to determine their<br />

predictive value for the development <strong>of</strong> frequent psychiatric<br />

disorders.<br />

Methodology: Comprehensive behavioural and neuropsychological<br />

characterization, functional and structural neuroimaging and genomewide<br />

association analyses <strong>of</strong> 2000 14-year-old adolescents are<br />

combined with functional genetics in animal and human models.<br />

Results: In this presentation, we will provide examples <strong>of</strong> recent<br />

candidate gene x neuroimaging analyses, including a study where we<br />

show that the oxytocin receptor polymorphism rs2268494 interacts<br />

with adverse social experiences in influencing fMRI BOLD<br />

responses to angry faces and risk for social-affective problems. Girls<br />

carrying the minor A-allele but not TT- genotype showed decreased<br />

ventral striatal activity after negative experiences, and greater<br />

vulnerability for behavioural problems. Boys with the minor A-allele<br />

had increased medial prefrontal cortex activity and fewer emotional<br />

problems.<br />

Conclusions: Our findings point to a novel neurobehavioural<br />

mechanism through which the OXTR polymorphism rs2268494<br />

interacts with negative experiences in contributing to risk versus<br />

resilience for social-emotional problems in adolescent boys and girls.


POSTER SESSION III<br />

ABSTRACTS:<br />

SPECIAL SESSION ON<br />

SUBSTANCE ABUSE<br />

GENETICS<br />

144


PP149 NEUROTROPHIC TYROSINE KINASE RECEPTOR 2<br />

(NTRK2) GENE POLYMORPHISMS AND NICOTINE<br />

DEPENDENCE<br />

H. Chen*(1), T. Shinkai(1), K. Utsunomiya(1), S. Sakata(1,2), K.<br />

Yamada(1), O. Ohmori(1,3), J. Nakamura(1)<br />

1. University <strong>of</strong> Occupational and Environmental Health 2.<br />

Sagamigaoka Hospital 3. Wakato Hospital<br />

*stbchen@hotmail.com<br />

Introduction: Brain-derived neurotrophic factor (BDNF) is a nerve<br />

growth factor that develops and maintains the neurons and is an<br />

important regulator <strong>of</strong> synaptic plasticity in human brain. Recently,<br />

BDNFgene has been reported to play animportant role in the<br />

development<strong>of</strong> nicotine dependence(ND). BDNFacts through its<br />

high-affinity receptor neurotrophic tyrosine kinase receptor 2<br />

(TrkBor NTRK2) in order to supportthe survival and growth <strong>of</strong><br />

neurons. Animal and in vitro studies showed that either BDNF or<br />

NTRK2 expression were modulated by nicotine. The previous studies<br />

showed a possible association between BDNF or NTRK2and ND. In<br />

the present study, we examinethe association between the<br />

NTRK2gene and ND.<br />

Methodology: We collected a total <strong>of</strong> 673 subjects from one <strong>of</strong> the<br />

Japanese major manufacturing companies (total N = 673), with an<br />

advantage <strong>of</strong> a relatively homogeneous background. Blood samples<br />

and questionnaires with their smoking status were collected. We<br />

selected the rs1187323 (5'-promoter), rs1187329 (intron), and<br />

rs1545285 (intron) <strong>of</strong> NTRK2 geneaccording to a previous<br />

association study, minor allele frequencies or coverage <strong>of</strong> the gene.<br />

Results: We could not find any association between FTND score and<br />

the three NTRK2 polymorphisms in our sample. Interestingly, we<br />

found that after sorting the BDNF Val66Met (which was conducted<br />

in our previous study) genotypes into 2 groups (AA vs. AG+GG), we<br />

found a non-significant trend towards a decreased FTND score in<br />

individuals with NTRK2 (rs1187323) A-allele carrier dose<br />

dependently. This effect was likely only in the BDNF AA-genotypic<br />

group.Stratified analysis <strong>of</strong> the BDNFpolymorphisms showed that the<br />

subjects with A-allele<strong>of</strong> rs1187323 tend to have higher riskforND,<br />

although the statistic result does not reacha significant level (Kruskal-<br />

Wallis Test, p=0.59).<br />

Conclusions: Although we did not find any<br />

significant association between ND and BDNF nor NTRK2 gene<br />

polymorphisms respectively. Our data still suggests that BDNF and<br />

NTRK2 genes may have some interactiveeffect on ND in Japanese<br />

workers.<br />

145<br />

PP150 BIPOLAR DISORDER AND SUBSTANCE ABUSE:<br />

GENETIC AND CLINICAL PREDICTORS OF DEPRESSIVE<br />

OUTCOME<br />

L. Mandelli*(1), M. Mazza(2), G. Martinotti(2), D. Tavian(3), E.<br />

Colombo(2), S. Missaglia(2), M. Di Nicola(2), D. De Ronchi(1), G.<br />

Negri(2), R. Colombo(3), L. Janiri(2), A. Serretti(1)<br />

1. University <strong>of</strong> Bologna, Italy 2. Catholic University <strong>of</strong> Rome 3.<br />

Catholic University <strong>of</strong> Milan<br />

*laura.mandelli@unibo.it<br />

Introduction: Little is known about factors associated with treatment<br />

outcome <strong>of</strong> depression in Bipolar Disorder (BD). In our study we<br />

extensively evaluated a sample <strong>of</strong> BD patients for clinical features,<br />

including substance use disorder (SUD) comorbidity, personality<br />

disorders and traits, and social functioning, together with<br />

polymorphisms in 8 candidate genes for BD and response to<br />

antidepressant treatment.<br />

Methodology: One-hundred and thirty-one BD patients and 65<br />

healthy controls were recruited at the Department <strong>of</strong> Psychiatry and<br />

the Centre <strong>of</strong> Addiction at the Gemelli hospital <strong>of</strong> Rome, Italy.<br />

Patients were characterized for demographic and clinical variables<br />

and evaluated by the Hamilton scales for depression and anxiety<br />

(HAMD, HAMA), the Global assessment <strong>of</strong> functioning scale<br />

(GAF), the Temperament and character inventory (TCI), the Social<br />

adjustment scales, self report (SASS) and the Quality <strong>of</strong> life scale<br />

(QoL). Polymorphisms in the following genes were genotyped in<br />

both BD patients and controls: Serotonin transporter (SLC6A4),<br />

Serotonin receptor 2C (5HTR2C), Tryptophan hydroxylase (TPH),<br />

Dopamine receptor D2 (DRD2,) Dopamine receptor D4 (DRD4),<br />

Brain derived neurotrophic factors, Inteleukin 1 beta (IL1b), Nitric<br />

oxide synthase 1 adaptor protein (NOS1AP), Transient receptor<br />

potential channel 2 (TRPM2). Moreover, 92 patients satisfying<br />

criteria for a depressive episode were entered in a 12-months followup<br />

under naturalistic treatment with antidepressants, mood stabilizers<br />

and/or antipsychotics.<br />

Results: Comorbidity for SUD was more frequent in BD type II and<br />

in patients with a Personality disorder, was associated with poorer<br />

social functioning but did not impact treatment outcome. Clinical<br />

factors associated with response to treatments were baseline severity<br />

and the temperamental trait <strong>of</strong> Harm avoidance. A polymorphism<br />

within 5HTR2C was found associated with BD in case-control<br />

analysis. Trends <strong>of</strong> association were observed between haplotypes in<br />

TPH1 and BD, and IL1b and SUD. The BDNF gene was found to<br />

independently influence the outcome <strong>of</strong> treatment, while variants in<br />

SLC6A4 did influence the outcome in interaction with levels <strong>of</strong> Harm<br />

avoidance.<br />

Conclusions: Data derived from this study confirmed an involvement<br />

<strong>of</strong> 5HTR2C in BD as well as a potential role <strong>of</strong> TPH1. Other genes<br />

were not associated with the risk for BD, while IL1b may play a role<br />

in SUD. SUD and personality disorders did not impact significantly<br />

the outcome <strong>of</strong> bipolar depression, while the temperamental trait <strong>of</strong><br />

Harm avoidance seem to play a critical role. Nevertheless, the effect<br />

<strong>of</strong> Harm avoidance may be mediated by variants in the SLC6A4<br />

gene, while it is independent from the effect <strong>of</strong> other genes. Finally,<br />

BDNF may play a critical role in treatment outcome as well,<br />

independently from other individual features.


PP151 DRD2 HAS BOTH TRAIT-SPECIFIC AND GENERAL<br />

ASSOCIATION WITH MULTIPLE MEASURES OF<br />

ALCOHOL USE AND ABUSE<br />

J. Meyers*(1), E. Nyman(2), A. Loukola(2), R. Rose(3), J.<br />

Kaprio(2), D. Dick(1)<br />

1. Virginia Institute <strong>of</strong> <strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong>, Virginia<br />

Commonwealth University 2. University <strong>of</strong> Helsinki & National<br />

Institute for Health and Welfare 3. Indiana University<br />

*meyers.jacquelyn@gmail.com<br />

Introduction: Alcohol consumption and related problems are a<br />

multi-faceted set <strong>of</strong> complex behaviors. Multivariate analyses <strong>of</strong> twin<br />

and family data a genetic architecture with many different genetic<br />

factors influencing various aspects <strong>of</strong> current alcohol consumption<br />

and problems. Prior analyses conducted within the Finnish<br />

population-based twin sample, Finntwin16 (n=5,238), yielded a twin<br />

model suggesting four latent genetic factors that account for the<br />

genetic variance across seven measures <strong>of</strong> alcohol consumption<br />

(frequency <strong>of</strong> drinking, frequency x quantity, frequency <strong>of</strong> heavy<br />

drinking, frequency <strong>of</strong> intoxication, and maximum drinks in a 24<br />

hour period) and two measures <strong>of</strong> problems (The Rutgers Alcohol<br />

Problem Index and The Mälmö-modified Michigan Alcohol Screen<br />

Test - MmMAST). The first two latent genetic factors loaded onto all<br />

<strong>of</strong> the drinking measures the third latent genetic factor loaded<br />

exclusively onto maximum drinks in a 24 hr period and The<br />

MmMAST and the fourth latent genetic factor loaded onto the two<br />

indices <strong>of</strong> problems.<br />

Methodology: The present study follows up on the complex genetic<br />

architecture seen across these measures in the twin analysis with<br />

measured genotypic data collected on a subset <strong>of</strong> 600 individuals<br />

from this twin sample. We conducted a series <strong>of</strong> genetic association<br />

analyses across nine Dopamine Receptor D2 (DRD2) SNPs testing<br />

the relationship between alcohol use/abuse candidate gene DRD2 and<br />

the four latent genetic factor scores provided by the previously<br />

indicated twin model.<br />

Results: The results follow the model implicated by prior twin<br />

analyses, where some SNPs (e.g., rs4245149) are significantly<br />

associated across all <strong>of</strong> the genetic factor scores (p-values= 0.009-<br />

0.02), but most significantly associated with the general latent genetic<br />

factor score which loads onto all drinking measures (p=0.009), while<br />

other SNPs (e.g. rs1799978) are only associated with specific genetic<br />

factors, in this case the genetic factor which loads uniquely onto<br />

maximum drinks in a 24hr period and the MmMAST(p=0.008).<br />

Conclusions: Our analyses indicate that different measures <strong>of</strong> current<br />

alcohol consumption and alcohol problems show a complex genetic<br />

architecture there is not a single genetic factor responsible for the<br />

phenotypic overlap between different measures <strong>of</strong> consumption and<br />

problem use. Furthermore, careful attention must be paid to the<br />

phenotype in efforts to “replicate” genetic effects across samples or<br />

combine samples for meta-analyses <strong>of</strong> genetic effects influencing<br />

susceptibility to alcohol-related outcomes.<br />

146<br />

PP152 EVIDENCE OF SHARED POLYGENIC RISK AMONG<br />

SMOKING BEHAVIORS AND BODY COMPOSITION<br />

R. Peterson*, X. Chen, J. Chen, B. Webb, H. Maes<br />

Virginia Commonwealth University<br />

*peterson.roseann@gmail.com<br />

Introduction: Obesity and nicotine dependence (ND) represent<br />

complex heterogeneous diseases which pose serious public health<br />

problems, affecting 33 and 20 percent <strong>of</strong> Americans, respectively.<br />

While cross-sectional studies <strong>of</strong> ND are typically supportive <strong>of</strong> a<br />

negative relationship between smoking and body mass index (BMI),<br />

a positive association is supported by the observations that, within<br />

smoking cohorts, heavy smokers tend to be <strong>of</strong> increased bodyweight<br />

compared to light smokers. A growing body <strong>of</strong> literature<br />

demonstrates the utility <strong>of</strong> genome-wide association studies (GWAS)<br />

for identifying single nucleotide polymorphisms (SNP) that<br />

contribute to disease risk. The GWAS approach has been applied to<br />

BMI and smoking behaviors (SB) using sample sizes in the tens <strong>of</strong><br />

thousands and yielded several putative risk variants <strong>of</strong> small effects<br />

on individual traits. Many traits show comorbidity but most studies<br />

do not examine common versus specific variants. The purpose <strong>of</strong> this<br />

study was to investigate whether variants affecting BMI or SB were<br />

common to multiple behaviors or were trait specific.<br />

Methodology: 75 BMI and 54 SB associated SNPs were catalogued<br />

from large-scale GWAS meta-analyses. These variants were tested<br />

for association in n=2,802 (41% African-American) older<br />

community-dwelling adults (68-80 years old) from the Health Aging<br />

and Body Composition study.<br />

Results: Current smokers had significantly lower BMI and<br />

abdominal visceral fat than never or former smokers in males and<br />

females (p


PP153 LATENT CLASS ANALYSIS AND HERITABILITY OF<br />

A DEVELOPMENTAL SUBTYPE OF BIPOLAR DISORDER<br />

B. Sharma*, E. Nwulia<br />

Howard University<br />

*bikashsharma222@hotmail.com<br />

Introduction: To investigate the validity <strong>of</strong> a developmental form <strong>of</strong><br />

bipolar disorder (BD) characterized clinically by early age at BD<br />

onset, alcohol use disorder, rapid-cycling, and suicidal behaviors.<br />

Methodology: Data from participants in the U.S. Bipolar Genomic<br />

Study (BiGS), who met the DSM-IV criteria for bipolar I disorder<br />

(BDI) or Schizoaffective Disorder, Bipolar Type were used to<br />

generate a discrete, one-factor model with two classes. We used<br />

misclassification rates, sensitivity (true positive rates) and 1-<br />

specificity (false-positive rates) tests to determine the quality <strong>of</strong><br />

measurement <strong>of</strong> this trait. Heritability <strong>of</strong> the derived latent class was<br />

then estimated from 635 NIMH bipolar pedigrees.<br />

Results: 926 out <strong>of</strong> 1000 participants in the study had complete<br />

information on age at onset, alcoholism, suicide attempts and rapid<br />

cycling. The prevalence <strong>of</strong> developmental BD (dBD) in the cohort<br />

was 61%. Early age at onset as a single trait has a sensitivity <strong>of</strong><br />

93.3% for dBD and false positive rate (FPR) <strong>of</strong> 68%. Corresponding<br />

values <strong>of</strong> sensitivity and FPR for alcoholism, rapid switching and<br />

suicide attempts are: 58.6% and 28.8%; 74.4% and 29.7%; and<br />

59.7% and 19.4%, respectively. Having all four items gave a positive<br />

predictive value (PPV) <strong>of</strong> 97.1%. Absence <strong>of</strong> all four items has a<br />

negative predictive value (NPV) <strong>of</strong> 96.7%.The global goodness <strong>of</strong> fit<br />

likelihood ratio test (2 degrees <strong>of</strong> freedom) <strong>of</strong> this model was 4.864<br />

(P < 0.09). From a sample <strong>of</strong> 635 BDI families the estimated<br />

heritability <strong>of</strong> dBD was 0.75 (95% CI, 0.44 – 0.87).<br />

Conclusions: Our results demonstrate the validity and heritability <strong>of</strong><br />

dBD, which can be characterized clinically by early age at BD onset,<br />

alcohol use disorder, rapid-cycling, and suicidal behaviors.<br />

Correlation with molecular markers would add further evidence to<br />

the biological validity <strong>of</strong> dBD. Moreover, phenotypes derived<br />

through confirmatory latent variable methods, such as dBD, could<br />

help minimize heterogeneity complicating genetic studies <strong>of</strong> other<br />

complex disorders.<br />

147<br />

PP154 UNIQUE LOCI IN THE CHRNA5/CHRNA3/CHRNB4<br />

GENE CLUSTER ARE ASSOCIATED WITH AGE OF<br />

REGULAR SMOKING<br />

S. Stephens*(1), N. H<strong>of</strong>t(1), S. Hartz(2), N. Saccone(2), L. Bierut(2),<br />

M. Ehringer(1)<br />

1. Institute for Behavioral <strong>Genetics</strong> 2. Washington University St.<br />

Louis<br />

*sarah.h.stephens@colorado.edu<br />

Introduction: Variation in the CHRNA5/CHRNA3/CHRNB4 gene<br />

cluster on chromosome 15 has been associated with nicotine<br />

dependence and smoking behaviors in multiple studies including<br />

genome-wide association studies (Bierut et al. 2007 Berrettini et al.<br />

2008 Caporaso et al. 2009 Thorgeirsson et al. 2008) and four recent<br />

meta-analyses (Furberg et al. 2010 Liu et al. 2010 Saccone et al. 2010<br />

Thorgeirsson et al. 2010). Extensive genotyping <strong>of</strong> this region has<br />

identified statistically independent nicotine dependence association<br />

signals(Saccone et al. 2009)tagged by SNPs rs16969968<br />

(nonsynonomous), rs578776, rs588765, and rs12914008 (rare). Two<br />

additional independent loci have been associated with early initiation<br />

<strong>of</strong> tobacco (Schlaepfer et al. 2008) and age <strong>of</strong> first regular use<br />

(rs684513) (unpublished data).<br />

Methodology: We (as part <strong>of</strong> the GEMINI consortium) designed a<br />

large meta-analysis to examine SNPs in this region for association<br />

with both tobacco age <strong>of</strong> initiation (AOI) and age <strong>of</strong> first regular<br />

tobacco use (AOR). This meta-analysis included ~40,000 subjects<br />

from 6 countries and evaluated 5 SNPs and their correlates including<br />

rs16969968, rs578776, and rs588765. SNPs rs1948 and rs684513<br />

were also examined due to their association with AOI and AOR<br />

(respectively). A unified script (in SAS and R) was developed and<br />

sent to all participating groups for local analysis <strong>of</strong> each data<br />

set. Summary statistics were extracted from the output and metaanalysis<br />

was carried out using PLINK (Purcell et al 2007).<br />

Results: Preliminary results with AOR yielded significant<br />

associations with SNPs rs578776 (Beta = 0.029, p = 0.001) and<br />

rs684513 (Beta = 0.043, p = 0.005). Rs16969968, the most<br />

replicated signal in this region for nicotine dependence and cigarettes<br />

per day, was not significant for AOR.<br />

Conclusions: These results provide important insight into the<br />

complexity <strong>of</strong> the genetic signals in the CHRNA5/A3/B4 gene cluster,<br />

suggesting that independent loci may contribute to the risk for<br />

nicotine dependence (and other drug behaviors) via different<br />

mechanisms.


PP155 TRANSLATIONAL MODELING THE GENETICS OF<br />

ALCOHOL USE DISORDERS AND TREATMENT EFFICACY<br />

IN A NON-HUMAN PRIMATE MODEL ORGANISM<br />

E. Vallender*, C. Sweeney, G. Chen, D. Platt, G. Miller<br />

Harvard Medical School - New England Primate Research Center<br />

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

Introduction: Alcohol use disorders are a major source <strong>of</strong> public<br />

health problems in society with significant health, economic, and<br />

social consequences. Understanding the factors leading to these<br />

disorders and developing treatment options has long been a major<br />

public health initiative. Over the past several decades it has become<br />

clear that alcohol use disorders are polygenic and numerous genes<br />

have been identified that associate with the disease. Among these<br />

genes and variation are OPRM1, SERT, and MAOA. While each <strong>of</strong><br />

these has been associated with alcohol use disorders in the past,<br />

additional studies have had variable success in replicating the<br />

findings in separate populations. This pattern has also been extended<br />

to include the OPRM1 pharmacogenetic response to naltrexone<br />

treatment. As these human studies have progressed, variation has<br />

been identified in rhesus macaques that functionally parallels the<br />

variation observed in humans for each <strong>of</strong> these genes. By using<br />

specific cohorts <strong>of</strong> rhesus macaques as natural genetic models <strong>of</strong><br />

alcohol abuse we can better understand the true genetic factors<br />

underlying individual susceptibility and response to treatment as well<br />

as control for and manipulate environmental interactions.<br />

Methodology: Male rhesus macaques have been genotyped at the<br />

SERT promoter repeat polymorphism, MAOA promoter repeat<br />

polymorphism, OPRM1 C77G, and several other SNPs in<br />

serotonergic and opioidergic genes. These animals are then trained to<br />

self-administer alcohol. Their preference for alcohol and total<br />

consumption is measured at various concentrations. These animals<br />

are then also treated with naltrexone at varying doses and their<br />

responses are measured. These phenotypic measures <strong>of</strong> alcohol<br />

consumption and naltrexone efficacy can then be analyzed in a<br />

genetic context.<br />

Results: The OPRM1 G77 allele is associated with increased alcohol<br />

preference and consumption in rhesus macaques in an additive<br />

fashion. G77 homozygotes show 40% greater alcohol consumption<br />

across concentrations and respond to naltrexone at 30-fold lower<br />

dosage levels. Heterozygotes show intermediate phenotypes. These<br />

results, along with in vitro functional studies, validate and elucidate<br />

human OPRM1 A118G polymorphism studies. This work is also<br />

being extended to additional genes and variation. This helps to<br />

explain a greater percentage <strong>of</strong> the overall variation seen between<br />

animals as well as to explain variation within OPRM1 C77G<br />

genotypes.<br />

Conclusions: Association studies <strong>of</strong> human genetic variation<br />

underlying alcohol use disorders have produced mixed results. Using<br />

a non-human primate model organism that harbors alleles that<br />

functionally parallel disease-associated alleles in humans, it is<br />

possible to clarify these findings and develop better understandings<br />

<strong>of</strong> the relevant genetic basis <strong>of</strong> the disease.<br />

148<br />

PP156 USING GENETIC INFORMATION FROM GENOME-<br />

WIDE ASSOCIATION STUDIES IN RISK PREDICTION FOR<br />

ALCOHOL DEPENDENCE IN TWO SAMPLES<br />

J. Yan*(1), F. Aliev(1), K. Kendler(1), B. Webb(1), M. Schuckit(2),<br />

J. Nurnberger Jr(3), H. Edenberg(4), J. Kramer(5), A. Goate(6), J.<br />

Tischfield(7), D. Dick(1)<br />

1. Virginia Institute for <strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong>, Virginia<br />

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

<strong>of</strong> California-San Diego 3. Department <strong>of</strong> Psychiatry, Indiana<br />

University School <strong>of</strong> Medicine 4. Department <strong>of</strong> Biochemistry and<br />

Molecular Biology, Indiana University School <strong>of</strong> Medicine 5.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Iowa College <strong>of</strong> Medicine 6.<br />

Department <strong>of</strong> Psychiatry, Washington University School <strong>of</strong><br />

Medicine 7. Department <strong>of</strong> <strong>Genetics</strong>, Rutgers University<br />

*jia.jiayan@gmail.com<br />

Introduction: Genome wide association studies (GWAS) <strong>of</strong> alcohol<br />

dependence (AD) have reported numerous associated variants.<br />

However, the clinical validity <strong>of</strong> these genetic variants to<br />

discriminate cases and controls for DSM-IV AD compared to family<br />

history has not been assessed. The Collaborative Study on the<br />

<strong>Genetics</strong> <strong>of</strong> Alcoholism (COGA) and the Study <strong>of</strong> Addiction: Genes<br />

and Environment (SAGE) GWAS samples were used to examine the<br />

aggregate impact <strong>of</strong> multiple genetic variants with small effect sizes<br />

on clinical risk prediction using receiver operating characteristic<br />

(ROC) curve analysis.<br />

Methodology: Subsets <strong>of</strong> the COGA and SAGE samples were used<br />

as gene discovery and validation samples in 3 sets <strong>of</strong> analyses.<br />

Genetic sum scores were created by adding risk alleles <strong>of</strong> associated<br />

SNPs in discovery samples and then assessed for their ability to<br />

discriminate between cases and controls in independent validation<br />

samples. The 3 analyses sets were distinguished based on methods <strong>of</strong><br />

selecting SNPs from GWAS results: (1) SNPs from initial GWAS<br />

results. We used sum scores based on semi-independent SNPs<br />

resulting from GWAS analysis in one half <strong>of</strong> the SAGE sample to<br />

assess discriminative accuracy in the second half <strong>of</strong> the sample. (2)<br />

SNPs from GWAS analysis that are also associated in a second<br />

sample. A subset <strong>of</strong> individuals from the SAGE sample who were not<br />

part <strong>of</strong> COGA was split in half. Sum scores based on SNPs that were<br />

associated in both halves <strong>of</strong> this sample were created and used for<br />

assessing discriminative accuracy in the COGA sample, with the idea<br />

that SNPs that had replicated may be more likely to represent “true<br />

positives” and enhance the predictive ability. (3) SNPs from GWAS<br />

analysis using varying “significance” criteria. Individuals from the<br />

SAGE sample who were not part <strong>of</strong> COGA were combined with the<br />

COGA GWAS sample, and then split randomly in half. Subsets <strong>of</strong><br />

SNPs were selected from GWAS results in one half <strong>of</strong> the sample<br />

based on p-value thresholds <strong>of</strong> 0.001, 0.01, 0.05, 0.1, 0.2, 0.3, 0.4,<br />

and 0.5. Each subset was then used to create sum scores to assess<br />

discriminative accuracy in the other half <strong>of</strong> the sample, testing the<br />

hypothesis that using SNPs meeting less stringent p-value thresholds<br />

may better detect variants <strong>of</strong> small effect. Analyses were repeated<br />

using several random splits <strong>of</strong> the combined SAGE-COGA GWAS<br />

sample in order to account for chance effects.<br />

Results: ROC curve analysis using the first two SNP-selection<br />

methods did not result in significant discriminative ability for any <strong>of</strong><br />

the sum scores, suggesting that the SNPs are not predicting better<br />

than chance. Association <strong>of</strong> the sum scores and the individual SNPs<br />

in the samples that were used for ROC curve analysis did not yield<br />

significant results, which explains the lack <strong>of</strong> significant<br />

discriminative accuracy in these samples. The presence or absence <strong>of</strong><br />

family history for AD was a better classifier <strong>of</strong> case-control status,<br />

with a significant area under the ROC curve (AUC) <strong>of</strong> 0.686 (95% CI<br />

= 0.654, 0.718, p < 0.001) in COGA and 0.614 (95% CI = 0.584,<br />

0.643, p < 0.001) for a parental history <strong>of</strong> AD-related traits in SAGE.<br />

The third SNP selection method using p-value thresholds <strong>of</strong> 0.01 to<br />

0.5 did yield significant AUC estimates ranging from 0.535 for SNPs<br />

with p-value < 0.01 to 0.573 for SNPs with p-value < 0.5. AUC<br />

results did not show a consistent pattern across all random splits <strong>of</strong><br />

the sample.<br />

Conclusions: This study shows that these SNPs have limited clinical<br />

utility and that family history is currently a better predictor <strong>of</strong> AD.<br />

This illustrates the need for further development <strong>of</strong> prediction panels<br />

that incorporate replicated variants contributing to risk for AD.


PP157 EPIGENETIC MODIFICATIONS IN THE WAKE-<br />

PROMOTING BASAL FOREBRAIN MAY CONTRIBUTE TO<br />

INSOMNIA DURING ALCOHOL WITHDRAWAL<br />

S. Lodhi*, R. Sharma, D. DeRoode, M. Thakkar<br />

Harry S Truman Memorial Veterans Hospital, Department <strong>of</strong><br />

Neurology, University <strong>of</strong> Missouri<br />

*shafilodhi@�����.com<br />

Introduction: Insomnia and associated sleep disturbances are<br />

amongst the most pr<strong>of</strong>ound and protracted symptoms<strong>of</strong> alcohol<br />

withdrawal (Roehrs and Roth, 2001). Importantly, convincing<br />

evidence exist to suggest that insomnia observed during alcohol<br />

withdrawal is a predictor <strong>of</strong> relapse to alcoholism (Brower, 2003).<br />

Recently, we have demonstrated that chronic alcohol exposure causes<br />

insomnia by reducing the expression <strong>of</strong> genes involved inadenosine<br />

release and transmission and increasing the activation <strong>of</strong> wakepromoting<br />

neurons in the cholinergic basal forebrain (BF; Sharma et.<br />

al., 2010). Histone acetylation is a key epigenetic mechanism that is<br />

believed to control gene expression. Increased histoneacetylation<br />

opens the chromatin and enhances gene expression, whereas reduced<br />

histone acetylation packs thechromatin and reduces gene expression.<br />

Does chronic ethanol exposure reduce histone acetylation in the BF?<br />

We performed two experiments to address this issue. Our first<br />

experiment examined histone acetylation in the BF <strong>of</strong>ethanol<br />

dependent rats. Our second experiment monitored sleep in ethanol<br />

dependent rats following bilateraladministration <strong>of</strong> the histone<br />

deacetylase (HDAC) inhibitor, Trichostatin-A (TSA) into the BF<br />

Methodology: Experiment 1:Male Sprague Dawley rats (200-300<br />

gm) were divided into two groups: controls (N=5)and experimentals<br />

(N=5). Ethanol dependency was achieved by using the<br />

Majchrowicz’s method as described previously (Sharma et al., 2010).<br />

In brief, at light onset, a priming dose <strong>of</strong> 5 g/Kg <strong>of</strong> ethanol (35% v/v)<br />

wasintragastrically administered to the experimental animals.<br />

Subsequent doses were adjusted based on theintoxication status <strong>of</strong> the<br />

animals and administered every 8 hr for 4 days. Controls were<br />

intragastricallyadministered with isocaloric water (10 ml/Kg) at same<br />

time points for 4 days. On withdrawal day, 12 hr after the last dose <strong>of</strong><br />

ethanol, the rats were euthanized and brains removed, blocked and<br />

the BF region was sectioned andprocessed for acetylated histone<br />

immunohistochemistry. Experiment 2: Under aseptic conditions and<br />

inhalation anesthesia, male Sprague Dawley rats (200-300 gm) were<br />

stereotaxically implanted with sleep recording electrodes along with<br />

bilateral guide cannulas targeted towards the BF (Thakkar et al.,<br />

2010). Following post-operative recovery and habituation, ethanol<br />

treatment was initiated and continued for 4 days as described above.<br />

On withdrawal day, at light onset, the animals were randomly divided<br />

into two groups. Group 1 animals were bilaterally microinjected with<br />

TSA (10 nmol/1µL/each side) into the BF. Group 2 animals were<br />

bilaterally microinjected with the vehicle (10% DMSO/1 uL/each<br />

side). Subsequently undisturbed sleep-wakefulness was continuously<br />

recorded for 24 hr. Results: Experiment 1:Our results suggests that<br />

there was a significant reduction (p


PP159 MULTIVARIATE GENOME-WIDE ASSOCIATION<br />

ANALYSES OF NEUROPHYSIOLOGICAL PHENOTYPES IN<br />

ALCOHOLISM<br />

S. Kang*(1), N. Manz(1), M. Rangaswamy(1), J. Kramer(2), L.<br />

Almasy(3), V. Hesselbrock(4), S. Kuperman(2), J. Nurnberger(5), H.<br />

Edenberg(5), J. Tischfield(6), B. Porjesz(1)<br />

1. SUNY Downstate Medical Center 2. University <strong>of</strong> Iowa 3. Texas<br />

Biomedical Research Institute 4. University <strong>of</strong> Connecticut Health<br />

Center 5. Indiana University School <strong>of</strong> Medicine 6. Rutgers<br />

University<br />

*������������������������<br />

Introduction: Event-related oscillations (EROs) represent highly<br />

heritable neuroelectrical correlates <strong>of</strong> human perception and<br />

cognitive performance, and are markers <strong>of</strong> risk for alcoholism. To<br />

identify genetic variants associated with EROs, we performed a<br />

genome-wide association study (GWAS). While statistical methods<br />

used for GWAS data analyses have been largely univariate (single<br />

phenotypes), multivariate analyses have advantages over univariate<br />

analysis whether or not there is correlation between phenotypes. To<br />

investigate the advantages <strong>of</strong> information from correlated<br />

phenotypes, we conducted multivariate GWAS for delta and theta<br />

ERO and P3 amplitude<br />

Methodology: All neurophysiological phenotype values were<br />

extracted from the target case <strong>of</strong> the Visual Oddball experiment. The<br />

two EROs were calculated at the parietal midline channel (Pz) for the<br />

delta band (1-3 Hz) and the frontal midline channel (Fz) for the theta<br />

band (3-7 Hz). The extracted measures were derived from a 300-700<br />

ms post stimulus window, bounding the visual P3 event. P3<br />

amplitude was measured as the voltage difference between the prestimulus<br />

baseline (100 ms prestimulus) and the largest positive going<br />

peak in the latency window 300-600 ms after stimulus onset at the<br />

parietal midline electrode (Pz). Illumina HumanHap 1M chips in<br />

1399 unrelated European Americans drawn from the Collaborative<br />

Study on the <strong>Genetics</strong> <strong>of</strong> Alcoholism (COGA) were used for analysis.<br />

After quality control procedures, we regressed delta and theta ERO<br />

and P3 amplitude phenotypes on age in each gender and included<br />

alcohol dependence as a covariate.<br />

Results: We compared the p values <strong>of</strong> univariate and multivariate<br />

analyses results for the SNPs in genes previously found to be<br />

associated with alcohol dependence or neurophysiological<br />

phenotypes. SNPs in SNCA, ADH4, TACR3, NPY2R, ACN9, GRM8,<br />

CHRM2, OPRK1, PENK, GABRG3, CHRNA7, PDYN, CDH13 and<br />

PRKG2 genes that showed no significant association with the<br />

neurophysiological phenotypes in univariate analysis were<br />

significantly associated with the phenotypes in multivariate analysis.<br />

For example, univariate analysis p values for rs1316749 in NPY2R<br />

are greater than 0.1 for all three phenotypes, whereas multivariate<br />

analysis p value for this SNP is less than 0.008. Another example is<br />

rs7656323 in PRKG2. The univariate p values for theta, delta, and P3<br />

are 2×10-4, 0.99, and 0.02, respectively. The bivariate p value for<br />

theta and P3 is 4×10-7 while the multivariate p value for theta, delta,<br />

and P3 is 2×10-6.<br />

Conclusions: These results underscore the advantage <strong>of</strong> using<br />

multivariate over univariate GWAS by utilizing the correlated<br />

phenotypic information to prioritize significant genes.<br />

150<br />

PP160 IDENTIFICATION OF RARE VARIANTS UNDER AN<br />

ALCOHOLISM SUSCEPTIBILITY LINKAGE PEAK IN A<br />

COGA FAMILY USING WHOLE EXOME SEQUENCING<br />

M. Kapoor*(1), J. Wang(1), A. Hinrichs(1), S. Bertelsen(1), J.<br />

Budde(1), A. Agrawal(1), J. Tischfield(2), L. Almasy(3), M.<br />

Schuckit(4), L. Bierut(1), A. Goate(1)<br />

1. Washington University School <strong>of</strong> Medicine 2. Department <strong>of</strong><br />

<strong>Genetics</strong>/Human <strong>Genetics</strong> Institute, Rutgers University 3.<br />

Department <strong>of</strong> <strong>Genetics</strong>, Texas Biomedical Research Institute 4.<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> California, San Diego<br />

*kapoorm@psychiatry.wustl.edu<br />

Genetic factors may explain as much as 60% <strong>of</strong> the variance in risk<br />

for alcoholism. Genomewide association studies <strong>of</strong> alcoholism only<br />

accounted for small proportion <strong>of</strong> this variance. We undertook whole<br />

exome sequencing to investigate the possible contribution <strong>of</strong> rare<br />

coding variants to susceptibility to alcohol dependence. Finding<br />

missing heritability through rare variants is conceptually easy, as it<br />

can involve closer scrutiny <strong>of</strong> genes and regions <strong>of</strong> interest.<br />

Combining the power <strong>of</strong> traditional linkage studies with next-gen<br />

sequencing can help find missing heritability in the form <strong>of</strong> rare and<br />

novel variants. Multiple chromosomal regions have been linked to<br />

alcohol dependence and other key phenotypes in our previous studies.<br />

In the present study, we have sequenced a trio (two alcohol<br />

dependent and one non-dependent control) selected from a large<br />

family that has many alcoholic subjects, and showed a very high NPL<br />

score on chromosome 2 with DSM-IV diagnosis. We used the<br />

SureSelect Human All Exon Kit and protocols provided by Agilent to<br />

capture exons, and performed paired-end sequencing on the Illumina<br />

GA2 platform. More than 95% <strong>of</strong> the exome was captured with >8X<br />

read depth. A total <strong>of</strong> 722 variants on chromosome 2 were detected in<br />

the affected parent and <strong>of</strong>fspring, but were not present in the<br />

unaffected parent. Among these, 83 variants lie under linkage peak<br />

and were coding variants. Out <strong>of</strong> 83 variants, 22 variants had minor<br />

allele frequency (MAF) less than 5% in dbSNP and 7 were novel. We<br />

validated these 29 variants by genotyping in rest <strong>of</strong> family members,<br />

and in about 4000 cases and controls. Preliminary association<br />

analysis using these rare and novel variants helped us to identify 6<br />

variants in 6 genes which may be associated with alcohol<br />

dependence. Further we sequenced these 6 genes using the targeted<br />

sequence capture approach in case-control dataset to determine<br />

enrichment <strong>of</strong> non-synonymous rare variants among alcohol<br />

dependents. Functional analysis <strong>of</strong> these non-synonymus variants<br />

present in these genes genes and variants will help us in better<br />

understanding <strong>of</strong> underlying biological mechanisms for alcohol abuse<br />

and dependence.


PP161 EPIGENETIC CONTROL OF GENE EXPRESSION IN<br />

ALCOHOLIC BRAIN<br />

I. Ponomarev*, S. Wang, L. Zhang, R. Harris, R. Mayfield<br />

Waggoner Center for Alcohol and Addiction Research and the College<br />

<strong>of</strong> Pharmacy, University <strong>of</strong> Texas at Austin<br />

*����������������������<br />

Recent evidence from brain transcriptome studies suggested that<br />

changes in gene expression associated with alcohol drinking<br />

phenotypes are, at least in part, controlled by global epigenetic events<br />

(chromatin modifications). We tested this hypothesis by comparing<br />

several epigenetic marks in frontal cortex <strong>of</strong> human alcoholics and<br />

control cases. We first examined DNA methylation status <strong>of</strong> the<br />

endogenous retroviral sequences, Long Terminal Repeats (LTR).<br />

LTRs are actively repressed by DNA methylation, and their<br />

transcriptional activity is a sensitive marker <strong>of</strong> global DNA<br />

hypomethylation. We found that DNA in brains <strong>of</strong> alcoholics is less<br />

methylated, which was consistent with the up-regulation <strong>of</strong> LTR<br />

transcripts. We next tested the hypothesis that up-regulation <strong>of</strong> some<br />

genes in alcoholic brain is associated with more tri-methylation <strong>of</strong><br />

histone 3 at Lysine 4 (H3K4me3), a marker <strong>of</strong> active transcription.<br />

Our chromatin assays provided evidence supporting this hypothesis.<br />

A combination <strong>of</strong> chromatin immunoprecipitation and qRT-PCR<br />

showed more global H3K4me3 in alcoholics. H3K4me3 at the<br />

promoters <strong>of</strong> several genes was also increased in alcoholics. In<br />

addition, we examined the expression <strong>of</strong> several genes that form<br />

transcription co-repression complexes (TCC). TCCs link histone<br />

deacetylase to specific DNA sites at gene promoters, resulting in<br />

histone deacetylation and transcriptional repression <strong>of</strong> the affected<br />

genes. Most genes that form TCCs were up-regulated in alcoholics,<br />

and the number <strong>of</strong> up-regulated TCC genes was significantly greater<br />

than chance (P


PP163 GENETIC TESTING FOR THE SUSCEPTIBILITY OF<br />

ALCOHOL DEPENDENCE: INTEREST AND CONCERNS<br />

D. Scott *, E. Nwulia, V. Headings, J. Kwagyan, G. Cain, V.<br />

Marshall, N. Kalu, C. Williams, R. Taylor<br />

Howard University College <strong>of</strong> Medicine<br />

*d_m_scott2@howard.edu<br />

Introduction: Recent advances in the search for susceptibility genes<br />

<strong>of</strong> alcohol dependence (AD) are generating growing interest for<br />

possible genetic risk assessment <strong>of</strong> at-risk relatives and the general<br />

population. The purpose <strong>of</strong> this study is to examine the level <strong>of</strong><br />

interest and concerns for predictive genetic testing for susceptibility<br />

to AD.<br />

Methodology: Respondents are 304 African American adult<br />

residents <strong>of</strong> Washington, DC metropolitan area who were recruited<br />

through public advertisement. All participants were administered the<br />

Genetic Psycho-Social Implication (G-PSI) questionnaire, which<br />

surveyed their interests in hypothetical genetic testing for AD, as<br />

well as their perception <strong>of</strong> ethical, social and legal concerns<br />

Results: More than 85% <strong>of</strong> participants were interested in predictive<br />

genetic testing; however, persons with higher education (p< 0.002)<br />

and income (p< 0.008) were less willing to receive<br />

testing. Perception <strong>of</strong> AD as a deadly disease (48.60%) and curiosity<br />

to know for their children (47.90%) were most prevalent reasons for<br />

interest in testing. Among those not interested in testing, belief that<br />

they were currently acting to lower their risk was the most prevalent<br />

reason for their decision. Although several concerns were raised, the<br />

most widely expressed concern in the entire sample was accuracy <strong>of</strong><br />

testing (35.50%). Other notable concerns between participants<br />

interested in testing and those not interested in testing were: method<br />

<strong>of</strong> testing (p< 0.01), side effects (p


PP165 CHRNA5-CHRNA3-CHRNB4 GENE CLUSTER IS<br />

ASSOCIATED WITH SEVERAL NICOTINE DEPENDENCE<br />

RELATED TRAITS<br />

J. Wedenoja (1,2,3,4),. Broms(1,4),. Largeau(2,4),. Korhonen(1,4),<br />

J. Pitkäniemi(1,4), K. Vuokko(1), A. Häppölä(1), K. Heikkilä(4), K.<br />

Heikkilä(1), S. Ripatti(2,4), S. Sarin(2), O. Salminen(5), T. Paunio<br />

T(2,4,6), M. Pergadia(7), J. Kaprio(1,2,4), A. Loukola(1,4)<br />

1. Department <strong>of</strong> Public Health, Hjelt Institute, University <strong>of</strong> Helsinki<br />

2. Institute for Molecular Medicine Finland FIMM, University <strong>of</strong><br />

Helsinki and National Institute for Health and Welfare 3. Department<br />

<strong>of</strong> Medical <strong>Genetics</strong>, University <strong>of</strong> Helsinki 4. National Institute for<br />

Health and Welfare 5. Division <strong>of</strong> Pharmacology and Toxicology,<br />

Faculty <strong>of</strong> Pharmacy, University <strong>of</strong> Helsinki 6. Department <strong>of</strong><br />

Psychiatry, Helsinki University Central Hospital 7. Washington<br />

University School <strong>of</strong> Medicine, Saint Louis<br />

Reguar tobacco smoking represents one preventable cause <strong>of</strong><br />

mortality, but is <strong>of</strong>ten associated with psychiatric and substance<br />

disorder co-morbidities. Especially the nicotinic acetylcholine<br />

receptor gene cluster on chromosome 15q24-25 has been established<br />

in nicotine dependence (ND) and other smoking-related traits.<br />

Here, we utilized a study sample <strong>of</strong> twin pairs concordant for history<br />

<strong>of</strong> cigarette smoking as well as their first-degree relatives. This series<br />

with an extensive dataset <strong>of</strong> 30 smoking-related phenotypes was<br />

ascertained from the Finnish Twin Cohort, involving adult twins born<br />

between 1938 and 1957, with baseline in 1975 at age 18 and over.<br />

First, we analyzed 15 tagging SNPs in the CHRNA5-CHRNA3-<br />

CHRNB4 cluster on 15q24-25 in 1428 individuals (mostly<br />

twins) from 735 families. We detected significant associations with a<br />

variety <strong>of</strong> traits measuring ND, the strongest signal emerging for<br />

DSM-IV ND symptoms (best p=0.000009; rs2036527 near<br />

CHRNA5). Additionally, rs11636753 in CHRNB4 showed association<br />

with regular drinking (p=0.003) and the co-morbidity <strong>of</strong> depression<br />

and ND (p=0.003).<br />

Second, we performed genome-wide association analysis in a subset<br />

<strong>of</strong> 1125 twins with 3,858,597 markers from Illumina 670k chip and<br />

HapMap2-based imputation data. In addition to CHRNA5-CHRNA3-<br />

CHRNB4 cluster, strongest signals were detected for DSM-IV ND<br />

symptoms on chromosome 2q33 (best p=0.0000008; near CD28) and<br />

8q21 (best p=0.000001; near STMN2), for cigarettes per day (CPD)<br />

on chromosome 16p12 (best p=0.000000007; near SYT17), and<br />

sensation felt after smoking the first cigarette on chromosome 8p21<br />

(best p=0.0000004; near GFRA2). Interestingly, also association with<br />

number <strong>of</strong> depression symptoms was detected on chromosome 4q31<br />

(best p=0.00000004; in ZNF827).<br />

153<br />

PP166 UNDERSTANDING THE RELATION BETWEEN AGE<br />

AT FIRST DRINK AND DRINKING TO COPE MOTIVES: A<br />

TWIN STUDY<br />

K. Young-Wolff*<br />

University <strong>of</strong> Southern California<br />

*kellyyw@gmail.com<br />

Introduction: Prior research indicates that individuals who begin<br />

drinking at an early age are at a greater risk for using alcohol to cope<br />

with negative mood states; however, the mechanisms underlying this<br />

association remain unclear. One possibility is that early drinking<br />

directly increases risk for drinking to cope (DTC). Alternatively, the<br />

association between early drinking and DTC may be attributable to<br />

overlapping familial sources (shared environmental or genetic<br />

factors). No prior genetically informative study has investigated the<br />

sources <strong>of</strong> covariation underlying the early drinking-DTC<br />

association.<br />

Methodology: Early age at first drink (< age 15) was assessed using<br />

structured clinical interviews in a sample <strong>of</strong> 7566 male and female<br />

participants aged 19-56 years from the Virginia Adult Twin Study <strong>of</strong><br />

<strong>Psychiatric</strong> and Substance Use Disorders. Data on DTC was obtained<br />

using the mood management scale <strong>of</strong> the Alcohol Use Inventory<br />

(AUI). The sources <strong>of</strong> the covariation between early first drink and<br />

DTC were estimated using both a matched case-control analysis <strong>of</strong><br />

twin pairs discordant for early drinking and bivariate twin modeling.<br />

Results: Approximately 22% <strong>of</strong> participants reported drinking before<br />

the age <strong>of</strong> 15 years. Regression analyses indicated that early drinking<br />

onset was associated with significantly higher scores on DTC.<br />

Results from co-twin control analyses and bivariate structural twin<br />

models <strong>of</strong> early drinking onset and DTC indicated that a substantial<br />

portion <strong>of</strong> the early drinking-DTC association was attributable to<br />

overlapping genetic variation.<br />

Conclusions: Individuals who reported they began to drink before<br />

age 15 were more likely to endorse drinking to cope with negative<br />

mood. Our results suggest that this early drinking-coping association<br />

largely reflected genetic factors that contribute to the etiology <strong>of</strong> both<br />

characteristics.


PP167 ASSESSMENT OF KNOWLEDGE AND ATTTIUDE<br />

OF CARE GIVERS OF PEOPLE WITH SCHIZOPHRENIA<br />

AND BIPOLAR DISORDER ABOUT ROLE OF GENETICS IN<br />

DISORDER CAUSATION<br />

Y. Balhara*<br />

Lady Hardinge Medical College And SSK Hospital<br />

*ypsbalhara@gmail.com<br />

Introduction: Role <strong>of</strong> gentic factors in multifactor causation <strong>of</strong><br />

major mental disorders including schizophrenia and bipolar disorders<br />

is well established. However the knowledge and understanding <strong>of</strong> the<br />

care givers <strong>of</strong> those suffering from these conditions has never been<br />

studied in India. Such a knowledge is likely to shape the attitude and<br />

belief <strong>of</strong> these individuals towards the patients and his/her siblings.<br />

Such information would be <strong>of</strong> help in planning ppropriate<br />

psychoeducation for thes eindividuals.<br />

Methodology: We assessed the care givers <strong>of</strong> patients with<br />

schizophrenia or bipolar disorder (as per DSM IV TR) for their<br />

knowledge and understanding on the role <strong>of</strong> genetic factros <strong>of</strong> these<br />

disorders. Consecutive patients with either <strong>of</strong> these conditions were<br />

selected fr the study following informed consent. Total <strong>of</strong> 200 care<br />

givers (100 each <strong>of</strong> schizophrenia and bipolar disorder) were<br />

included in the study. The assessments were carried out using a<br />

questionnaire developed specifically for this purpose. The<br />

questionnaire is aimed at assessmen t<strong>of</strong> knoledge <strong>of</strong> the care givers<br />

on the role <strong>of</strong> genetic factors in causation <strong>of</strong> these to disorders. It also<br />

assessed attitude <strong>of</strong> the care givers towards possibility <strong>of</strong> these<br />

disorders among children <strong>of</strong> the patients. In between group<br />

comparisons were made between the two care givers <strong>of</strong> two<br />

disorders. Non -parametric analyssi was carried out using<br />

MannWhitney U test and Kriskall Wallis test. Level <strong>of</strong> statistical<br />

significance was kept at p< .05.<br />

Results: The findigns <strong>of</strong> the current study bring out interesting facts.<br />

Care givers <strong>of</strong> patients with schizophrenia as well as bipolar disorder<br />

have limited knowledge about role <strong>of</strong> gentic fcators in their<br />

causation. Also the information is limited on heritability <strong>of</strong> these<br />

disorders. This in turn seems to shape their attutude toards the<br />

children <strong>of</strong> the patients.<br />

Conclusions: It is impoartant to assess and intervene regarding the<br />

knowledge and attitude <strong>of</strong> care givers <strong>of</strong> patients with schizophrenia<br />

and bipolar disorders towards role <strong>of</strong> gentic factors in their causation.<br />

154<br />

PP168 STATISTICAL PHASE VERSUS MOLECULAR<br />

PHASE RESOLUTION: HTR2C HAPLOTYPE AND SUICIDE<br />

ATTEMPT IN FEMALE PATIENTS WITH MAJOR<br />

PSYCHOSES<br />

V. de luca*, C. Teo, B. Mckenzie, C. Zai, J. Kennedy<br />

CAMH<br />

*vincenzo_deluca@camh.net<br />

Introduction: Haplotype is the combination <strong>of</strong> phased allele on the<br />

same chromosome. When we genotype a sample <strong>of</strong> unrelated patients<br />

the phase in unknown in the subjects with two or more heterozygous<br />

genotypes. The parental genotype can be helpful to reconstruct the<br />

phase however the molecular techniques available to resolve the<br />

phase are not currently used in studies aimed to develop genetic<br />

testing. The most common statistics used to infer the phase in<br />

unrelated subjects is the PHASE program. The aim <strong>of</strong> this study is to<br />

compare the inferred phase with the phase resolved by the means <strong>of</strong><br />

the ABI 3130 using the same genotype data.<br />

Methodology: We analyzed a sample <strong>of</strong> 250 females with affective<br />

and non affective psychoses assessed for suicide attempt lifetime<br />

using the PHASE program for the HTR2C haplotype.<br />

Results: When we considered the molecular phase none <strong>of</strong> the<br />

haplotypes was associated with suicide attempt (p>0.05). However<br />

many double heterozygous subjects were misdiagnosed in terms <strong>of</strong><br />

haplotype pairs when we used the PHASE algorithm compared<br />

to the molecular phase.<br />

Conclusions: This simple result suggests that future sequencing<br />

analysis should investigate the utility <strong>of</strong> the molecular phase<br />

haplotypes in the development <strong>of</strong> genetic testing in psychiatry


PP169 ALCOHOLIC BEVERAGE PREFERENCE: AN<br />

EPIDEMIOLOGICAL AND TWIN STUDY<br />

ECIP<br />

L. Hack*(1), J. Myers(3), H. Maes(1,2,3), K. Kendler(1,3)<br />

1. Department <strong>of</strong> Human and Molecular <strong>Genetics</strong>, Virginia Institute<br />

for <strong>Psychiatric</strong> and Behavioral 2. Massey Cancer Center, Virginia<br />

Commonwealth University 3. Department <strong>of</strong> Psychiatry, Virginia<br />

Institute for <strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong>, Virginia<br />

Commonwealth University<br />

*hacklm@vcu.edu<br />

Introduction: Alcoholic beverage preference, generally defined as<br />

one’s most heavily or frequently consumed alcoholic beverage, has<br />

been associated with particular lifestyle habits, social consequences,<br />

and medical outcomes. The present study sought to replicate and<br />

expand some <strong>of</strong> these findings in a twin sample by assessing the risk<br />

<strong>of</strong> developing alcohol use disorders (AUDs) and related measures<br />

based on alcoholic beverage preference in both the same individual<br />

and the co-twin. Additionally, while there is an extensive genetic<br />

epidemiological literature on frequency <strong>of</strong> consumption, few studies<br />

have considered specific alcoholic beverage intake when assessing<br />

the magnitude <strong>of</strong> genetic and environmental contributions to<br />

consumption measures. Thus, we assessed the factors contributing to<br />

twin resemblance for most frequently consumed beverage, or<br />

alcoholic beverage preference.<br />

Methodology: The sample consisted <strong>of</strong> 5489 twins (3200 males and<br />

2289 females) from the Virginia Adult Twin Study <strong>of</strong> <strong>Psychiatric</strong> and<br />

Substance Use Disorders (VATSPSUD) identified through the<br />

population-based Virginia Twin Registry. Clinically trained<br />

interviewers collected data on beverage preference (wine, beer, or<br />

spirits), AUDs, consumption measures, and other psychiatric<br />

conditions, including antisocial personality disorder (ASPD),<br />

generalized anxiety disorder (GAD), and major depression (MD).<br />

The authors conducted multivariate regression in SAS and structural<br />

twin modeling in Mx .<br />

Results: The risk <strong>of</strong> AUDs, symptoms <strong>of</strong> these disorders,<br />

consumption measures, and ASPD is lowest in wine preference<br />

drinkers. The significant co-twin findings showed a similiar trend.<br />

Beer drinkers are the most likely to engage in high and frequent<br />

consumption, while distilled spirit drinkers are at the greatest risk for<br />

alcohol dependence, more impairing symptoms <strong>of</strong> AUDs, and ASPD.<br />

For all three preference categories, the tetrachoric correlations<br />

between twins suggest a stronger genetic influence on preference in<br />

females and a stronger shared environmental influence in males.<br />

When we proceeded to formal univariate twin modeling, we found<br />

that genders could be equated for beer and distilled spirits without<br />

loss in model fit but not for wine. We are currently attempting to find<br />

the most appropriate multivariate model for quantity <strong>of</strong> consumption<br />

<strong>of</strong> the three beverages in a second twin sample, the Virginia 30,000<br />

(VA30K). This analysis has proven to be challenging due to the<br />

competitive nature <strong>of</strong> consumption <strong>of</strong> these beverages.<br />

Conclusions: Firstly, preference for particular alcoholic beverages<br />

leads to differential risk for the development <strong>of</strong> AUDs, symptoms <strong>of</strong><br />

these disorders, consumption measures, and ASPD. These findings<br />

have implications for classifying alcohol dependent individuals into<br />

subtypes and for the application <strong>of</strong> personalized treatments for AD.<br />

Secondly, the relationship between preference and differential risk<br />

appears to be influenced by familial factors. Finally, beverage<br />

preference is substantially influenced by genetic and shared<br />

environmental factors, the nature <strong>of</strong> which may differ across the<br />

genders. This finding suggests that refininement <strong>of</strong> our consumption<br />

phenotypes may be beneficial for identifying risk genes in genomewide<br />

association studies.<br />

155<br />

PP170 HUMAN µ-OPIOID RECEPTOR GENE VARIANT<br />

A118G (ASN40ASP): LACK OF ASSOCIATION WITH<br />

ALCOHOL DEPENDENCE OR ALCOHOL CONSUMPTION<br />

N. Rouvinen-Lagerström(1), J. Lahti*(2), H. Alho(1,3), M. Aalto(1,<br />

11), L. Kovanen(1), T. Partonen(1), K. Silander(4), D. Sinclair(1), K.<br />

Räikkönen(2), J. Eriksson(5,6,7,8), A. Palotie(5, 9, 10)<br />

1. Department <strong>of</strong> Mental Health and Substance Abuse Services,<br />

National Institute for Health and Welfare 2. Institute <strong>of</strong> Behavioural<br />

Sciences 3. Research Unit <strong>of</strong> Substance Abuse Medicine, University<br />

<strong>of</strong> Helsinki and Helsinki University Central Hospital 4. Unit <strong>of</strong><br />

Public Health Genomics, National Institute for Health and Welfare,<br />

and Institute for Molecular Medicine FIMM 5. National Institute for<br />

Health and Welfare 6. Department <strong>of</strong> General Practice and Primary<br />

Health Care, University <strong>of</strong> Helsinki 7. Unit <strong>of</strong> General Practice,<br />

Helsinki University Central Hospital 8. Folkhalsan Research Centre,<br />

Biomedicum Helsinki, University <strong>of</strong> Helsinki 9. Wellcome Trust<br />

Sanger Institute 10. Broad Institute <strong>of</strong> Harvard and MIT 11.<br />

Department <strong>of</strong> Psychiatry, South Ostrobothnia Hospital District<br />

*jari.lahti@helsinki.fi<br />

Introduction: The human µ-opioid receptor (Mu Opioid Receptor,<br />

MOR) has been implicated in the reinforcement, tolerance and<br />

withdrawal effect <strong>of</strong> many substances <strong>of</strong> abuse including<br />

alcohol. The OPRM1 gene (Opioid receptor Mu 1 gene) carries a<br />

functional A118G (Asn40Asp) polymorphism. When compared with<br />

the 118A (Asn40) allele, the 118G (Asp40) allele has been reported<br />

to result in increased binding <strong>of</strong> β-endorphin, which is one <strong>of</strong> the<br />

important physiological mediators <strong>of</strong> alcohol-induced<br />

reinforcement. Several studies have linked A118G polymorphism to<br />

individual differences in alcohol-induced stimulation and alcohol<br />

consumption in experimental settings. The molecular epidemiological<br />

studies on association <strong>of</strong> A118G polymorphism and alcohol use<br />

disorders have, however, given contradictory results. The focus <strong>of</strong><br />

this study was to test the possible association <strong>of</strong> A118G<br />

polymorphism and alcohol use disorders and alcohol consumption in<br />

large cohort-based study populations.<br />

Methodology: : The association between the OPRM1 A118G<br />

(Asn40Asp, rs1799971) polymorphism and alcohol use disorders and<br />

alcohol consumption was analyzed using three different populationbased<br />

samples: i) a Finnish cohort study, Health 2000, with 503<br />

participants with DSM-IV diagnosis for alcohol dependence and/or<br />

alcohol abuse and 506 age and sex-matched controls ii) a Finnish<br />

cohort study, FINRISK (N=2360), and iii) the Helsinki Birth Cohort<br />

Study (N= 1384), which lacked diagnosis-based alcohol information,<br />

but included detailed information on alcohol consumption.<br />

Results: We found no statistically significant differences in<br />

genotypic or allelic distribution between controls and subjects with<br />

alcohol dependence or abuse diagnoses. Also no significant effects<br />

were observed between the A118G genotype and alcohol<br />

consumption.<br />

Conclusions: These results suggest that A118G (Asn40Asp)<br />

polymorphism is not associated alcoholism or alcohol consumption<br />

and thus may not have a major effect on the development on alcohol<br />

use disorders.


PP171 FROM SUBSTANCE USE TO SUBSTANCE<br />

DEPENDENCE: THE ROLE OF EXECUTIVE FUNCTIONING<br />

J. Vandever*(1,2), N. Friedman(2), S. Young(2,3), A. Miyake(1), R.<br />

Corley(2), J. Hewitt(1,2), M. Stallings(1,2)<br />

1. Department <strong>of</strong> Psychology and Neuroscience, University <strong>of</strong><br />

Colorado 2. Institute for Behavioral <strong>Genetics</strong>, University <strong>of</strong> Colorado<br />

3. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Colorado<br />

*joanna.vandever@colorado.edu<br />

Introduction: Individual differences/deficits in executive cognitive<br />

functioning (EF) have been hypothesized to play an important role in<br />

substance use disorders. In a study <strong>of</strong> adolescent twins, genetic<br />

influences on substance use and substance dependence vulnerability<br />

were found to be related to cognitive processes common across<br />

inhibiting, updating, and shifting tasks (Common EF; Vandever et al.,<br />

in preparation). Although our measure <strong>of</strong> substance use (number <strong>of</strong><br />

substances used repeatedly) was less heritable than our measure <strong>of</strong><br />

substance dependence vulnerability (average dependence symptoms,<br />

or total number <strong>of</strong> DSM-IV criteria endorsed divided by the number<br />

<strong>of</strong> substances used repeatedly), the genetic correlation between<br />

substance use and Common EF was greater in magnitude than the<br />

genetic correlation between substance dependence vulnerability and<br />

Common EF. This preliminary finding suggests that genetic variation<br />

in substance ‘use’ may be more strongly related to Common EF than<br />

genetic variation in dependence vulnerability. For the present study<br />

we followed up on this finding by using more detailed measures <strong>of</strong><br />

substance use histories to better understand the role <strong>of</strong> executive<br />

functions across the developmental course <strong>of</strong> substance<br />

use/dependence.<br />

Methodology: We conducted a biometrical analysis <strong>of</strong> adolescent<br />

twins (16–18 years) who had completed a battery <strong>of</strong> nine laboratory<br />

assessments <strong>of</strong> executive functioning and a structured clinical<br />

interview on substance use. Three substance-related transitions were<br />

identified: 1) use versus no use; 2) among users, problem use versus<br />

no problems (assessed by the endorsement <strong>of</strong> DSM-IV ‘symptoms’<br />

<strong>of</strong> dependence but no dependence diagnosis); and 3) among problem<br />

users, the progression to substance dependence (assessed by meeting<br />

DSM-IV criteria for dependence on at least one substance). For initial<br />

analyses factor scores were obtained for Common EF.<br />

Results: Consistent with our expectations, mean levels <strong>of</strong> Common<br />

EF were significantly lower when comparing users to non-users; but<br />

no significant mean differences were found when comparing problem<br />

users to non-problem users, and when comparing problem users with<br />

those progressing to dependence.<br />

Conclusions: Our findings are consistent with the hypothesis that<br />

individual differences/deficits in executive function distinguish<br />

between users and non-users (i.e., who initiates substance use and<br />

who doesn’t), but may play a lesser role in the transitions to problem<br />

use and dependence. However, the equivalence <strong>of</strong> cross-trait twin<br />

correlations between Common EF and these categorically scored<br />

substance transitions indicate that environmental influences, rather<br />

than genetic factors, underlie these associations. Analyses utilizing a<br />

latent measure <strong>of</strong> Common EF and continuous measures <strong>of</strong> substance<br />

use stages will be explored.<br />

156<br />

PP172 META-ANALYSIS OF SEROTONIN TRANSPORTER<br />

GENE PROMOTER POLYMORPHISM (5-HTTLPR)<br />

ASSOCIATION WITH ANTIDEPRESSANT EFFICACY<br />

S. Porcelli*, C. Fabbri, A. Serretti<br />

Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna<br />

*stefano.porcelli@yahoo.it<br />

Introduction: In the last decade the serotonin transporter gene<br />

promoter polymorphism (HTTLPR) was likely the most studied<br />

genetic variant as predictor <strong>of</strong> antidepressant response. Nevertheless<br />

results are not consistent across studies and previous meta-analysis,<br />

since various factors seem to modulate its effect on antidepressant<br />

response.<br />

Methodology: We systematically reviewed literature, selecting 18<br />

studies performed on Caucasians (3746 subjects)and 12 on Asians<br />

(1429 subjects). We tested two phenotypes - remission and response<br />

rates - and two genotype comparisons - ll versus ls/ss and ss versus<br />

ll/ls - using the Cochrane review manager. Evaluations were<br />

performed separately for SSRIs and mixed/other drugs and for<br />

Caucasians and Asians. Possible clinical modulators were<br />

investigated.<br />

Results: In Caucasians, we found an association between l allele and<br />

both response (p=0.004), and remission (p=0.004) in the SSRI group.<br />

Only a marginal association between l allele and remission (p=0.04)<br />

survived pooling together mixed antidepressant treatments. In Asians,<br />

a small effect <strong>of</strong> HTTLPR on remission for mixed antidepressants<br />

was detected (p=0.01). Gender, age and age at onset modulated the<br />

association in Caucasians. Gender, age and depression severity at<br />

baseline modulated the association in Asians.<br />

Conclusions: In Caucasians HTTLPR is likely a predictor <strong>of</strong><br />

antidepressant response and remission, whilst in Asians it does not<br />

appear to play a major role.


PP173<br />

Withdrawn<br />

157<br />

PP174 INTERACTION BETWEEN COMT HAPLOTYPES<br />

AND CANNABIS USE ON THE SEVERITY OF PSYCHOTIC<br />

SYMPTOMS<br />

M. Zanoni*(1), C. Bonetto(1), A. Lasalvia(1), D. Crist<strong>of</strong>alo(1), E.<br />

Ira(1), K. De Santi(1), V. Marangon(2), R. Riolo(3), D. Collier(4), M.<br />

Ruggeri(1), S. Tosato(1)<br />

1. Department <strong>of</strong> Public Health and Community Medicine, Section <strong>of</strong><br />

Psychiatry and Clinical Psychology, University <strong>of</strong> Verona 2. CMHC<br />

Mirano 3. CMHC Camposampiero 4. Social, Genetic and<br />

Developmental Psychiatry, Institute <strong>of</strong> Psychiatry<br />

*martina.zanoni@univr.it<br />

Introduction: One <strong>of</strong> the environmental factors extensively studied<br />

in relation to schizophrenia susceptibility is cannabis use, at least<br />

among genetically vulnerable individuals. One attractive gene to<br />

analyze in a gene×environment interaction is catechol-Omethyltransferase<br />

(COMT), which is involved in the catabolism <strong>of</strong><br />

catecholamines and is the main enzyme controlling the metabolism <strong>of</strong><br />

dopamine in prefrontal cortex. Three different common functional<br />

haplotypes have been described in COMT, called low pain sensibility<br />

(LPS), average pain sensibility (APS), and high pain sensibility<br />

(HPS), with APS and HPS haplotypes showing significant reductions<br />

in enzymatic activity in comparison to the LPS haplotype. It was<br />

found that high activity haplotypes (LPS) fastly reduce the levels <strong>of</strong><br />

dopamine in the prefrontal cortex, leading to worse negative<br />

symptoms. In the current work, in a first episode psychosis cohort,<br />

our aim is to demonstrate that cannabis users carrying LPS COMT<br />

haplotype experience worse negative symptoms than cannabis<br />

users carrying at least one <strong>of</strong> the high activity haplotypes (either HPS<br />

or APS). Moreover, among LPS haplotype carriers, we hypothesize<br />

that cannabis users experience worse negative symptoms than non<br />

users.<br />

Methodology: The study has been conducted in the broader<br />

framework <strong>of</strong> the Psychosis Incident Cohort Outcome Study<br />

(PICOS), a multisite research conducted in Veneto (Italy), aiming at<br />

characterizing first-episode psychosis patients. We used a case-only<br />

design comprising about 200 first-episode psychosis patients.<br />

Psychopathology was collected using PANSS and information on<br />

drug use in the year before the first contact with the <strong>Psychiatric</strong><br />

Service for psychosis was collected by using the CDUS (Clinical<br />

Drug Use Scale) (information was obtained from both the subject and<br />

the relatives and interviewed the case-manager when needed).<br />

Genetic analyses are being performed investigating COMT<br />

haplotypes as defined by rs4680, rs4633, rs4818, and rs6269.<br />

Results: Two hundred Caucasian patients (92% from Italy and 8%<br />

from East Europe, mainly Romania) gave their DNA for the genetic<br />

analysis. This sample included 49 (24.5%) patients with<br />

schizophrenia (ICD10-F20), 108 (54.0%) with non schizophrenic non<br />

affective psychosis (ICD10-F21-F29), and 43 (21.5%) with affective<br />

psychosis (ICD10-F30-F32). 51% are males with a mean age <strong>of</strong> 30<br />

years. At present the genotyping analyses are still ongoing and no<br />

preliminary results are available, yet.<br />

Conclusions: Our study may help to clarify the role <strong>of</strong> genetic<br />

interaction in the clinical presentation <strong>of</strong> psychoses at the onset. In<br />

particular, while previous studies mainly investigated the impact <strong>of</strong><br />

GxE interaction in either schizophrenia or bipolar disorder, to our<br />

knowledge this is the first study focusing on first-episode psychosis<br />

patients, thus encompassing diagnostical boundaries. Moreover, the<br />

perspective <strong>of</strong> a symptom-based approach is attractive because it may<br />

provide a model for studying the heterogeneity <strong>of</strong> psychosis,<br />

enhancing the ability to identify the underlying pathophysiology <strong>of</strong><br />

the illness.


PP175 DNA METHYLATION PROFILE IN THE HUMAN<br />

PERIOD 1 GENE PROMOTER AND ITS RELATION TO<br />

STRESS AND ALCOHOL DRINKING<br />

C. Wong*, F. Carvalho, B. Ruggeri, J. Mill, S. Desrivières, G.<br />

Schumann<br />

MRC Social, Genetic & Developmental Psychiatry Centre, Institute<br />

<strong>of</strong> Psychiatry, King's College London<br />

*c_peng.wong@kcl.ac.uk<br />

Introduction: Research over decades has demonstrated the<br />

importance <strong>of</strong> circadian rhythms in controlling gene expression. Our<br />

recent analyses have indicated that the Period (PER) genes might be<br />

involved in regulating alcohol drinking behaviour, in addition to their<br />

role in circadian rhythms. Mutation <strong>of</strong> Per2 results in increased<br />

alcohol drinking and mutation <strong>of</strong> Per1 enhances stress-induced<br />

alcohol drinking in mice, for which the data supports the association<br />

studies conducted in humans (Spanagel et al., 2005 Li et al.,<br />

submitted). Moreover, gene expressions <strong>of</strong> Per1 and Per2 are known<br />

to be modulated by alcohol, and that the expression <strong>of</strong> Per1 is also<br />

modulated by stress. These data suggest Per1 may participate in the<br />

adaptations <strong>of</strong> environmental factors such as stress, via various<br />

genetic and epigenetic mechanisms. To test this hypothesis, we<br />

investigate the relationship between the variations in DNA<br />

methylation pr<strong>of</strong>ile at the Per1 promoter and the stress- and alcoholrelated<br />

behaviours in humans.<br />

Methodology: We took a sub-sample <strong>of</strong> n=700 14-year old<br />

adolescents from the FP6-funded IMAGEN project, for which<br />

genetic, neuroimaging and behavioural data such as stressful life<br />

events and alcohol drinking are available. Whole blood DNA samples<br />

<strong>of</strong> these individuals were used to analyse DNA methylation patterns<br />

in the PER1 promoter region using Sequenom © MALDI-TOF<br />

technology.<br />

Results: In our first step, we analysed methylation status <strong>of</strong> a small<br />

region within the Per1 promoter. We identified variations at 6 CpG<br />

units with mean methylation levels ranging from 0.16 to 0.49, up to a<br />

maximum methylation level <strong>of</strong> 0.77. Inter-individual variations in<br />

methylation at some CpG units were found. We are currently<br />

analysing the relationship between methylations at the identified CpG<br />

units and genetic variations around the analysed region. In addition,<br />

we are conducting association analyses <strong>of</strong> methylation and brain<br />

activation pr<strong>of</strong>iles subjected to impulsivity- and emotion-related<br />

neuroimaging tasks among the same 700 individuals.<br />

Conclusions: We found inter-individual variations in DNA<br />

methylation patterns within the promoter <strong>of</strong> PER1. At present, we are<br />

investigating the significance <strong>of</strong> such variations in stress- and alcohol<br />

drinking-related behaviours, using the behavioural and neuroimaging<br />

phenotypes available in the IMAGEN project. The comparison <strong>of</strong><br />

methylation levels at the PER1 promoter region will provide insight<br />

on the importance <strong>of</strong> epigenetic modification on PER1 function and<br />

its impact on human behaviour.<br />

158<br />

PP176 ASSOCIATION STUDY OF GABRG2<br />

POLYMORPHISMS WITH ALCOHOL USE AND SUICIDAL<br />

BEHAVIOUR IN SCHIZOPHRENIA PATIENTS<br />

C. Zai*, A. Tiwari, G. Zai, V. de Luca, J. Kennedy<br />

CAMH<br />

*cchzai@yahoo.ca<br />

Introduction: γ-aminobutyric acid (GABA) is produced in areas <strong>of</strong><br />

the brain implicated in schizophrenia, and several genes coding for<br />

GABAA subunits, including GABRG2 encoding the γ2 subunit, are<br />

clustered at 5q31-q35, a chromosomal region associated with<br />

schizophrenia in genome scan studies. We recently reported<br />

GABRG2 to be associated with schizophrenia in our schizophrenia<br />

case-control and family samples (Zai et al, 2009).<br />

Methodology: We tested eight single nucleotide polymorphisms<br />

spanning GABRG2 for association with alcohol use, and with suicidal<br />

behaviour, and with both combined in our schizophrenia sample <strong>of</strong><br />

European ancestry (N=179).<br />

Results: We did not find GABRG2 to be significantly associated<br />

with history <strong>of</strong> alcohol dependence or suicide attempt. We found<br />

rs209356 to be associated with history <strong>of</strong> combined alcohol<br />

dependence and suicide attempt (p=0.03). We also found an<br />

interaction between history <strong>of</strong> alcohol dependence and rs211037 in<br />

severity <strong>of</strong> suicidal behaviour (p=0.04).<br />

Conclusions: Taken together, the results <strong>of</strong> the present study suggest<br />

GABRG2 may be involved in suicidal behaviour and alcohol use in<br />

schizophrenia patients, but replications are required. These results<br />

may help in the discovery <strong>of</strong> novel treatments for alcoholism and/or<br />

prevention <strong>of</strong> suicide. We plan to analyze other GABA system genes<br />

with these phenotypes.


PP177 A PROMOTER VARIANT IN THE NET (SLC6A2)<br />

GENE PREDICTS BOLD RESPONSE TO GUSTATORY<br />

ALCOHOL CUES<br />

S. Blaine*(1), H. Haughey(2), E. Claus(1), K. Hutchinson(1)<br />

1. 1The MIND Research Network. 2. 2University <strong>of</strong> Virginia School<br />

<strong>of</strong> Medicine, Dept. <strong>of</strong> Psychiatry and Neurobehavioral Sciences<br />

*sblaine@mrn.org<br />

Because alterations within the SLC6A2 gene contribute to differences<br />

in craving for alcohol, the present study sought to examine the<br />

influence <strong>of</strong> allelic variations in the SLC6A2 gene on variation in<br />

neural responses to alcohol cues. The SLC6A2 gene encodes for the<br />

sodium-dependent noradrenaline (NET) transporter. NET transports<br />

norepinephrine and dopamine from the synapse back to presynaptic<br />

cytosol, for later storage and release. We collected DNA samples and<br />

fMRI data using a cue-elicited craving paradigm in 326 heavy<br />

drinkers. The sample included heavy drinkers, with average alcohol<br />

dependence lifetime and current symptom counts <strong>of</strong> 6 ± 3, mean<br />

Alcohol Dependence Scale score 18 ±8, and mean Alcohol Use<br />

Disorders Identification Test score <strong>of</strong> 19 ±8. The pattern <strong>of</strong> BOLD<br />

activity in response to the alcohol taste cue task replicated previous<br />

findings, with activation in the alcohol minus litchi contrast mainly in<br />

the pathways where NET transporters are highly expressed. These<br />

pathways include connections among the brain stem, amygdala,<br />

hippocampus, caudate, and frontal medial gyri. BOLD activation in<br />

incentive and control regions in response to gustatory alcohol cues<br />

was predicted by 13 <strong>of</strong> the 60 SNPs in the SLC6A2 gene tested. Each<br />

<strong>of</strong> the SNPs associated with BOLD activation are in one <strong>of</strong> two LD<br />

blocks, one at the 3’ end <strong>of</strong> the gene and one toward the 5’ end <strong>of</strong> the<br />

gene. One SNP that predicted BOLD response, rs2242446, is a<br />

promoter variant in the gene that has been previously linked to<br />

medication response. These findings replicate and extend recent<br />

research suggesting that genetic variation at the NET locus may<br />

underlie the expression <strong>of</strong> alcohol phenotypes, including craving<br />

responses.<br />

159<br />

PP178 ALCOHOL MISUSE IN BIPOLAR DISORDER A<br />

SYSTEMATIC REVIEW AND META-ANALYSIS OF<br />

COMORBIDITY RATES: IMPLICATIONS FOR GENETIC<br />

ANALYSIS<br />

A. Di Florio, M. van den Bree, N. Craddock<br />

MRC Centre for Neuropsychiatric <strong>Genetics</strong> & Genomics<br />

Introduction: One <strong>of</strong> the specific objectives <strong>of</strong> the <strong>Psychiatric</strong><br />

GWAS Consortium is the analysis <strong>of</strong> comorbidities between major<br />

psychiatric disorders and alcohol/substances use disorders. The<br />

possibility <strong>of</strong> heterogeneity and bias across studies is an important<br />

issue in the definition <strong>of</strong> phenotypes suitable for genetic analyses. Of<br />

all DSM IV axis I disorders, bipolar disorder (BD) has been reported<br />

to be the most strongly linked with alcohol use disorders (AUD).<br />

AIM: To assess the current state <strong>of</strong> knowledge on the prevalence <strong>of</strong><br />

AUD in people affected by BD and to explore possible sources <strong>of</strong><br />

heterogeneity.<br />

Methodology: Studies reporting the comorbidity or co-occurrence<br />

rates <strong>of</strong> AUD in people affected by BD were identified through<br />

database searches. Meta-analytic techniques were employed to<br />

aggregate data on lifetime comorbidity. Possible sources <strong>of</strong><br />

heterogeneity were explored using univariate and multivariate<br />

random effects meta-regression. Funnel plots were employed for<br />

diagnosing certain forms <strong>of</strong> publication bias.<br />

Results: Of 1639 studies initially identified, 41 included in the<br />

systematic review. 12 papers from the ancestor research were<br />

selected and included. Data on 9535 subjects with BD were included<br />

in the meta-analysis. Overall, AUD affected more than 1 in 3 subjects<br />

with BD, although a large variability across studies was reported.<br />

Variability was largely explained by geographical location and<br />

methodological differences across studies. Gender affected<br />

comorbidity rates as well, with AUD rates 1.5-2 times higher in men<br />

than women.<br />

Conclusions: The heterogenic nature <strong>of</strong> comorbidity between BD<br />

and AUD is underscored by the current literature. Analyses aimed to<br />

identify convincing genotype-phenotype associations should account<br />

for the high variability in the clinical assessment across samples. This<br />

meta-analysis produced results which corroborate the findings <strong>of</strong> a<br />

great deal <strong>of</strong> the previous work in this field, which has found a high<br />

prevalence <strong>of</strong> AUD in BD and a strong association between male<br />

gender and AUD. However, we reported a large variability between<br />

studies that has not previously been described. Moreover, metaregression<br />

analyses indicated a significant effect <strong>of</strong> methods <strong>of</strong><br />

ascertainment and geographical location <strong>of</strong> the studies in the<br />

prevalence estimates. These results seem to be consistent with other<br />

research on AUD in the general population.


PP179 ASSOCIATION BETWEEN THE NOREPINEPHRINE<br />

TRANSPORTER GENE AND ALCOHOL DEPENDENCE IN<br />

EUROPEAN AMERICANS<br />

H. Haughey*, C. Seneviratne, A. A.S. Joshi, B. Johnson, M. Li<br />

University <strong>of</strong> Virginia School <strong>of</strong> Medicine, Dept. <strong>of</strong> Psychiatry and<br />

Neurobehavioral Sciences<br />

*hmh8f@virginia.edu<br />

Introduction: Alcohol dependence is a chronic, complex, polygenic<br />

debilitating disease that causes economic, personal, and family<br />

hardship worldwide. Individual differences in response to the acute<br />

effects <strong>of</strong> alcohol, as well as risk for alcohol dependence (AD), are<br />

known to be influenced by both genetic (G) and environmental (E)<br />

factors, along with their GxE interactions. Past and present<br />

preclinical and clinical studies have suggested that the norepinephrine<br />

(NE) system plays a role in alcohol consumption, sensitivity, craving,<br />

and may affect drug and alcohol reward and dependence. To date,<br />

there are both negative and positive association studies <strong>of</strong> the NE<br />

transporter (NET; slc6a2) gene and alcohol dependence. A most<br />

recent study in a large cohort <strong>of</strong> German subjects has found a positive<br />

association between alcohol dependence and slc6a2. Therefore, the<br />

purpose <strong>of</strong> the present study was to replicate these findings in a<br />

sample <strong>of</strong> Americans <strong>of</strong> European descent.<br />

Methodology: We tested two promoter SNPs and 6 other NET SNPs<br />

one from each haplotype block to interrogate the NET gene region<br />

(rs192303, rs47958, rs36030, rs1800887, rs36021, rs11648486,<br />

rs17841327, rs2242446). The study included 271 AD and 327 control<br />

subjects whose population admixture was tested with a panel <strong>of</strong> 24<br />

ancestral informative markers. The AD was diagnosed using DSM-<br />

ІV, and those with co-morbid Axis 1 diagnoses other than nicotine<br />

dependence were excluded. Associations <strong>of</strong> the SNPs with AD were<br />

assessed at both the individual SNP and haplotype levels.<br />

Results: Results demonstrated a gender specific association <strong>of</strong> two<br />

SNPs with AD at the individual SNP level. The interaction between<br />

male gender and A allele and AA genotype <strong>of</strong> NET intron 1 SNP<br />

rs17841327 were significantly associated with AD (X 2 =5.443,<br />

P=0.020 and X 2 = 4.855, P=0.028, respectively). Additionally, the<br />

interaction between male gender and C allele <strong>of</strong> another intron 1 SNP<br />

rs36030showed a trend for developing AD (P=0.074).<br />

Conclusions: : Taken together, these data suggest that<br />

polymorphisms within the NET gene are significantly associated with<br />

the development <strong>of</strong> AD in Caucasian men. The gender differences<br />

that we detected warrant further examination <strong>of</strong> gender specific<br />

autonomic system differences associated with alcohol addiction.<br />

(Research supported by NIAAA grant K01-AA015331(HMH),<br />

Grants 7 U10 AA011776-10, 1 N01 AA001016-000, 7 R01<br />

AA010522-12, and 5 R01 AA012964-06 (BAJ) and Grants R01<br />

DA012844 and R01 DA013783 (MDL).<br />

160<br />

PP180 ABSENCE OF SIGNIFICANT ASSOCIATION OF<br />

ALLELE A9 OF DOPAMINE TRANSPORTER GENE<br />

DAT1/SLC6A3 WITH ALCOHOL WITHDRAWAL SEIZURE<br />

OR DELIRIUM TREMENS IN THE INDIAN POPULATION:<br />

A CASE-CONTROL ASSOCIATION STUDY<br />

B. Kishore*(1, 2), P. Murthy(1), S. Jain(1), V. Benegal(1), K.<br />

Thennarasu(1), M. Purushottam(1), D. Subhashree(1), H. Kiran(1)<br />

1. Department <strong>of</strong> Psychiatry, National Institute <strong>of</strong> Mental Health and<br />

Neurosciences 2. 2North Western Mental Health<br />

*brijkishore@aol.com<br />

Introduction: The allele A9 <strong>of</strong> dopamine transporter gene (DAT1;<br />

SLC6A3) has been implicated for its role in complicated alcohol<br />

withdrawal as manifested by alcohol withdrawal seizures (AWS) and<br />

delirium tremens (DT). However there have been both positive and<br />

negative studies in literature. We investigated the role <strong>of</strong> the DAT1<br />

gene in an Indian alcohol dependent population for the first time.<br />

Methodology: A group <strong>of</strong> 106 alcohol dependence cases (52 alcohol<br />

dependent males with simple alcohol withdrawal and 54 with<br />

complicated alcohol withdrawal) and 104 normal healthy male<br />

controls were genotyped and compared in a case-control design, after<br />

assessing them with strict inclusion and exclusion criteria and<br />

informed consent.<br />

Results: In contrast to the positive studies showing higher frequency<br />

<strong>of</strong> allele A9 carriers in alcohol dependent patients with complicated<br />

withdrawal, our study in the Indian population reveals that the<br />

frequency <strong>of</strong> allele A9 carriers [ƒ (A9+)] was nominally higher in<br />

simple alcohol withdrawal group [ƒ (A9+) = 0.25] rather than<br />

complicated alcohol withdrawal group [ƒ (A9+) = 0.22] and normal<br />

healthy male controls [ƒ (A9+) = 0.22]. Our results also show that the<br />

frequency <strong>of</strong> individuals carrying the allele A9 [ƒ (A9+)] was higher<br />

in group <strong>of</strong> alcoholics [ƒ (A9+) =0.27], compared to the normal<br />

healthy controls [ƒ (A9+) =0.22], but the association was not<br />

statistically significant.<br />

Conclusions: To conclude there is absence <strong>of</strong> any significant<br />

association <strong>of</strong> allele A9 <strong>of</strong> DAT1 VNTR polymorphism with<br />

complicated alcohol withdrawal or alcohol dependence cases in<br />

Indian population. We propose further studies with larger sample<br />

sizes including female alcohol dependent patients as well as familybased<br />

studies to assess the role <strong>of</strong> DAT1 VNTR polymorphism in<br />

complicated alcohol withdrawal and alcohol dependence.


PP181 A PILOT STUDY ON THE MOLECULAR EFFECTS<br />

OF DEPRESSION AND ALCOHOLISM ON NEURAL STEM<br />

CELLS<br />

C. May, P. Sathyan, S. Majumder<br />

The University <strong>of</strong> Texas MD Anderson Cancer Center, The<br />

University <strong>of</strong> Texas Graduate School <strong>of</strong> Biomedical Sciences<br />

Depression and alcoholism are both mental illnesses that are poorly<br />

understood at the molecular level. Here we began to examine the<br />

effect <strong>of</strong> these two illnesses on neural stem cells (NSCs). Here we<br />

show that a novel function <strong>of</strong> the transcriptional repressor REST is to<br />

maintain the self-renewal and multipotency <strong>of</strong> mouse NSCs. Primary<br />

neurospheres obtained from E12 brains <strong>of</strong> transgenic mice expressing<br />

Egfp through the Rest promoter/enhancer sequences (Rest-Egfp)<br />

showed the co-expression <strong>of</strong> REST, EGFP and NSC stemness<br />

markers.<br />

Culturing NSCs under differentiation conditions decreased REST and<br />

stemness marker expression and increased the expression <strong>of</strong><br />

differentiation markers <strong>of</strong> neuronal, glial, and oligodendrocyte<br />

lineages, indicating that REST-expressing NSCs are multipotent.<br />

RESTpositive, but not REST-negative NSCs, showed self-renewal<br />

for several passages. Knockdown <strong>of</strong> REST in REST-positive NSCs<br />

increased REST target gene expression and decreased their selfrenewal<br />

capacity. Thus, a new function <strong>of</strong> REST is to maintain<br />

normal NSC self-renewal and multipotency.<br />

We further show that NSCs subjected to treatments mimicking either<br />

a depressive environment (i.e. cortisol) decreased REST levels, while<br />

NSCs subjected to treatments mimicking alcoholic environment<br />

increased REST levels. This suggests that though on the behavioral<br />

level these two diseases may have overlapping syndromes,<br />

molecularly they affect neural stem cells differently and that<br />

alcoholic environment may induce NSC self-renewal, an effect not<br />

studied before.This work was partly supported by grants from the<br />

NCI (CA97124 and CA81255 to S.M.).<br />

161<br />

PP182 SEROTONIN TRANSPORTER MRNA LEVELS IN LL<br />

GENOTYPE CARRIERS AS A GENOMIC BIOMARKER FOR<br />

QUANTITATIVE ASSESSMENT OF DRINKING SEVERITY<br />

IN ALCOHOLICS<br />

C. Seneviratne(1), N. Ait-Daoud(1), X. Wang(2), M. Li(1), B.<br />

Johnson(1)<br />

1. Departments <strong>of</strong> Psychiatry and Neurobehavioral Sciences 2. Public<br />

Health Sciences,<br />

Introduction: Paucity <strong>of</strong> sensitive biomarkers to quantify transient<br />

changes in alcohol consumption level remains a critical barrier for the<br />

development <strong>of</strong> efficacious therapeutic agents to treat alcoholism.<br />

Recently, in an 11-week, randomized, placebo-controlled, double<br />

blind trial <strong>of</strong> 283 alcohol dependent individuals, Johnson et al (2011) 1<br />

demonstrated that ondansetron was efficacious at reducing the<br />

severity <strong>of</strong> drinking (measured as drinks/drinking day, DDD) in<br />

alcoholics carrying the LL compared with the LS/SS genotype <strong>of</strong><br />

serotonin transporter gene, 5’-HTTLPR.<br />

Methodology: Using peripheral blood samples from a cohort <strong>of</strong> 41 <strong>of</strong><br />

these subjects, we determined if there was a relationship between<br />

mRNA expression level <strong>of</strong> the 5’-HTTLPR genotypes (measured at<br />

weeks 0, 4, and 11) and self-reported alcohol consumption following<br />

treatment with either ondansetron (4 µg/kg twice daily; N=19) or<br />

placebo (N=21). Using a mixed-effects linear regression model, we<br />

analyzed the effects <strong>of</strong> DDD and 5-HTTLPR genotypes, on mRNA<br />

expression levels within and between the ondansetron and placebo<br />

groups.<br />

Results: We found a significant three-way interaction effect <strong>of</strong> DDD,<br />

5′-HTTLPR genotypes, and treatment, on mRNA expression levels<br />

(p=0.0396). Among ondansetron but not placebo recipients there was<br />

a significant interaction between DDD and 5′-HTTLPR genotype<br />

(p=0.0385 and p=0.7938, respectively). In the ondansetron group,<br />

DDD was associated positively with mRNA levels at a greater rate <strong>of</strong><br />

expression alteration per standard drink in those with the LL<br />

genotype (slope=+1.1698 in Ln scale).<br />

Conclusions: We suggest that the combination <strong>of</strong> 5-HTTLPR - LL<br />

genotype and 5’-HTTLPR mRNA expression levels might be a<br />

promising and novel biomarker to quantify drinking severity in<br />

alcoholics treated with ondansetron.


PP183<br />

WITHDRAWN<br />

162<br />

PP184 THE GENETICS OF RECREATIONAL DRUGS<br />

ABUSE IN BIPOLAR PATIENTS INVESTIGATED<br />

THROUGH A GENOME WIDE MOLECULAR PATHWAY<br />

ANALYSIS<br />

A. Drago, A. Serretti<br />

Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna<br />

Introduction: Addiction to recreational drugs is a major health issue<br />

worldwide 1. <strong>Genetics</strong> may shape the risk <strong>of</strong> addiction to recreational<br />

drugs, and consistent genetic evidence is gathering 2. Knowing in<br />

advance who holds a genetic liability to addiction may help clinicians<br />

to tailor relevant clinical choices. This issue may be crucial in<br />

specific subpopulation <strong>of</strong> patients for whom addiction to drugs may<br />

worsen the prognosis. Bipolar patients may be at risk<br />

Methodology: We analyzed the STEP-BP sample 4 at enrollment.<br />

Having a present or a past history <strong>of</strong> abuse for opiates, LSD, cocaine<br />

or amphetamines was the principal outcome. A three step analysis<br />

was conducted: first, a genome wide molecular pathway analysis<br />

identified the molecular pathways differentially regulated in bipolar<br />

patients with or without addiction to recreational drugs. Second, the<br />

genes within those molecular pathways were ranked according to the<br />

strength <strong>of</strong> association with the principal outcome and their relevance<br />

within each pathway. Third, one gene per pathway was further<br />

genome wide investigated for epistatic interactions. R and plink<br />

served for the analysis.<br />

Results: 1821 subjects were initially enrolled and genotyped. 1697<br />

subjects had compete data on drug addiction, 115 out <strong>of</strong> whom were<br />

recreational drugs abusers. 366125 SNPs were available for analysis<br />

after quality check. Pathway associated with cell signaling<br />

(hsa04080, hsa02010 and hsa04020), cell movement (hsa04514,<br />

hsa04512, hsa04510 and hsa04810), cell replication and cell death<br />

(hsa04210, hsa05222 and hsa05215) were significantly associated<br />

with the principal outcome. Hsa04080, hsa04514 and hsa04510 were<br />

further investigated. Epistatic interactions <strong>of</strong> interest were:<br />

rs11644075 ( GRIN2A a glutamatergic receptor) vs rs7702070 (less<br />

than 1kb away from the GDNF (glial cell derived neurotrophic<br />

factor)) (OR = 17.41), rs2267787 ( GRIN2A ) vs rs6837383 (SEPT11<br />

septin 11, involved in cytokinesis and vesicle trafficking)) (OR =<br />

15.48), rs9932138 ( GRIN2A ) vs rs2070699 ( EDN1 endothelin 1)<br />

(OR=15.93); rs919751 ( PDGFRB platelet-derived growth factor<br />

receptor) vs rs7945377 ( SHANK2 ankyrin) (OR=15.67), rs12365943<br />

(less than 2 kb from DDX10 RNA elicase) vs rs1864971 ( PDGFRB )<br />

(OR=13.15), rs1864971 ( PDGFRB ) vs rs9948162 ( LOC100130480<br />

hypothetical protein ) (OR=18.51); rs10748138 ( CNTN1 contactin 1)<br />

vs rs557725 (less than 1kb from the KCNU1 potassium channel)<br />

(OR=13.87), rs11178018 ( CNTN1 ) vs rs1855640 (less than 1kb<br />

from ANKRD30A ankyrin) (OR=14.34), rs11610114 ( CNTN1 ) vs<br />

rs2714016 ( SLC39A11 metal ion transporter) (OR = 14.00). All<br />

associations had p levels < 1e-4 (default for plink genome wide<br />

epistatic interactions)<br />

Conclusions: We conducted a genome wide molecular pathway<br />

analysis in a sample <strong>of</strong> bipolar patients in order to investigate the<br />

genetics <strong>of</strong> addiction in this subpopulation. We brought some results<br />

that enlarge the genetic perspective on the disorder including genes<br />

related to cell movement and cell survive. These results are partially<br />

consistent with previous findings as well, as for example glutamate 5<br />

and the ankyrins


PP185 CHARACTERIZATION OF THE NEURON-SPECIFIC<br />

ENOLASE GENE (ENO2) AS A SUSCEPTIBLE GENE OF<br />

HEROIN ADDICTION<br />

H. Liao(1), D. Liao(2), M. Cheng(3), C. Lai(4), C. Chen(4)<br />

1. Institute <strong>of</strong> Biotechnology and Graduate Program <strong>of</strong><br />

Biotechnology in Medicine, National Tsing-Hua University 2. Bali<br />

<strong>Psychiatric</strong> Center, Department <strong>of</strong> Health, Executive Yuan 3.<br />

Department <strong>of</strong> Psychiatry, Yuli Mental Health Research Center, Yuli<br />

Veterans Hospital 4. Division <strong>of</strong> Mental Health and Addiction<br />

Medicine, Institute <strong>of</strong> Population Health Sciences, National Health<br />

Research Institutes<br />

Heroin addiction is a complex mental disorder resulting from<br />

interactions between genetic and environmental factors. Identifying<br />

the susceptible genes <strong>of</strong> heroin addiction is essential for better<br />

treatment and prevention <strong>of</strong> heroin abuse and addiction. We<br />

previously found the expression <strong>of</strong> neuron-specific enolase gene<br />

(ENO2) in the lymphoblastoid cell lines (LCL) was down-regulated<br />

in heroin addicts in a gene expression pr<strong>of</strong>iling analysis. We verified<br />

this finding using real-time quantitative PCR and Western blot<br />

analysis in this study. To assess whether the ENO2 gene is associated<br />

with heroin, we compared the allele and genotype frequencies <strong>of</strong> 3<br />

single nucleotide polymorphisms (SNPs, rs11064464, rs3213433 and<br />

rs10849541) between 532 male heroin addicts and 369 male controls.<br />

No significant differences in the allele or genotype frequencies <strong>of</strong><br />

these 3 SNPs were detected between these two groups. Nevertheless,<br />

we identified a haplotype (T-C-G) derived from these 3 SNPs<br />

significantly under-represented in heroin addicts compared to control<br />

group (72.7% vs 75.9%, p


PP187 INCUBATION OF HEROIN CRAVING IS NOT<br />

ASSOCIATED WITH ALTERATIONS IN BDNF, TRKB, AND<br />

MECP2 EXPRESSION AND SIGNALING IN NUCLEUS<br />

ACCUMBENS, DORSAL STRIATUM, AND MEDIAL<br />

PREFRONTAL CORTEX<br />

Q. Liu, F. Theberge, S. Fanous, C. Pickens, E. Goldart, B. Hope, Y.<br />

Shaham<br />

BRC/NIDA/IRP<br />

Introduction: Nucleus accumbens (NAc) BDNF signaling plays an<br />

important role in incubation <strong>of</strong> cocaine craving (time-dependent<br />

increases in cue-induced cocaine seeking after withdrawal). Here, we<br />

studied whether incubation <strong>of</strong> heroin craving is associated with<br />

alterations <strong>of</strong> expression in BDNF and phosphorylation <strong>of</strong> TrkB (the<br />

BDNF receptor) in NAc, as well as dorsal striatum (DS), and medial<br />

prefrontal cortex (mPFC) and DNA methylaiton <strong>of</strong> BDNF promoters<br />

in central amygdala (CEA). We also assessed expression and<br />

phosphorylation changes <strong>of</strong> MeCP2, a transcription suppressor that<br />

modulates BDNF expression.<br />

Methodology: We trained groups <strong>of</strong> rats to self-administer heroin<br />

(0.075 mg/kg/infusions, 6 h/d for 10 d) or saline (a control<br />

condition). After 1, 11 and 30 withdrawal days, we dissected the<br />

NAc, DS, and mPFC and measured protein levels <strong>of</strong> BDNF, TrkB,<br />

TrkB phosphorylation (pY816 site), MeCP2, and MeCP2<br />

phosphorylation (pS80 and pS421sites). In other groups <strong>of</strong> rats, we<br />

assessed cue-induced heroin seeking in 30-min extinction tests after<br />

1, 11, or 30 withdrawal days.<br />

Results: As in previous studies, cue-induced heroin seeking<br />

progressively increased or incubated after withdrawal. This<br />

incubation was not associated with changes in either protein or<br />

mRNA levels <strong>of</strong> BDNF and MeCP2 in NAc, DS, and mPFC. BDNF<br />

promoters are hypomethylated in CEA <strong>of</strong> both heroin and saline<br />

treated rats.<br />

Conclusions: Incubation <strong>of</strong> heroin craving is not associated with<br />

changes in BDNF and MeCP2 signaling in NAc, DS, and<br />

mPFC. Based on previous studies on the role <strong>of</strong> BDNF in incubation<br />

<strong>of</strong> cocaine craving, the present data suggest that the cellular<br />

mechanisms <strong>of</strong> incubation <strong>of</strong> cocaine and heroin craving are likely<br />

different.<br />

164<br />

PP188 COCAINE REGULATION OF THE CART GENE IS<br />

MODULATED IN PART BY DIFFERENTIAL REGULATION<br />

OF THE NRSF TRANSCRIPTION FACTOR<br />

P. Myers, S. Gillies, K. Haddley, H. Burrell, S. Vasiliou, V. Bubb,<br />

A. Warburton, A. Savage, J. Quinn<br />

Molecular and Clinical Pharmacology, Institute <strong>of</strong> Translational<br />

Medicine, University <strong>of</strong> Liverpool<br />

Introduction: The CART (cocaine and amphetamine-regulated<br />

transcript) peptides are widely distributed in the CNS and are<br />

involved in regulating multiple processes, including modulation <strong>of</strong><br />

dopamine and endocrine systems, aspects <strong>of</strong> reward, food intake and<br />

addiction. CART has recently been identified as a transcriptional<br />

target for negative regulation by Neuron Restrictive Silencing Factor<br />

(NRSF), also termed Repressor Element-1 Silencing Transcription<br />

factor (REST). NRSF is a factor we have demonstrated as a key<br />

regulator <strong>of</strong> other neuropeptides including Substance P, NKB and<br />

AVP. NRSF is also implicated in adult neurogenesis, epilepsy,<br />

cognition and Scitzophrenia. Cocaine treatment is known to upregulate<br />

the expression <strong>of</strong> CART, as well as a second NRSFregulated<br />

neuronal gene, brain derived neurotrophic factor (BDNF).<br />

However, despite our current knowledge <strong>of</strong> cocaine-mediated<br />

regulation <strong>of</strong> CART and BDNF, it has yet to be clarified whether<br />

cocaine modulates NRSF. We set out to investigate the impact <strong>of</strong><br />

cocaine treatment upon NRSF and CART expression in the human<br />

SH-SY5Y neuroblastoma cell line.<br />

Methodology: Cells from the human neuroblastoma cell line SH-<br />

SY5Ywere treated with 1µM and 10µM cocaine, at 4hrs and 24hrs,<br />

followed by measurement <strong>of</strong> NRSF and CART mRNA expression<br />

through real time polymerase chain reaction (RT-PCR). NRSF<br />

mRNA expression was further investigated using quantitative<br />

polymerase chain reaction (qPCR). We proceeded with an<br />

investigation into the binding <strong>of</strong> NRSF to its recognition sequence<br />

(NRSE) within the CART and BDNF promoter regions in response to<br />

cocaine; through chromatin immunoprecipitation (ChIP).<br />

Results: Cocaine treatment was found to significantly decrease<br />

NRSF mRNA expression, in an exposure-duration and concentration<br />

dependant manner. This reduction in NRSF correlated with an<br />

increase in CART mRNA. Furthermore, we demonstrate by<br />

chromatin immunoprecipitation, that cocaine treatment markedly<br />

reduces NRSF binding to both the CART and BDNF promoter<br />

Conclusions: These findings indicate that cocaine has the capacity to<br />

increase expression <strong>of</strong> CART via modulation <strong>of</strong> the NRSF<br />

transcriptional regulatory system. Together with our previous data on<br />

NRSF modulation <strong>of</strong> several neuropeptides and transmitters we are<br />

beginning to understand the functional significance <strong>of</strong> the network <strong>of</strong><br />

genes modulated by NRSF in response to various challenges. This<br />

allows for a better understanding <strong>of</strong> the combinatorial action <strong>of</strong><br />

peptides in regulatory processes such as addiction, endocrine<br />

function, the stress response, depression, epilepsy and modulation <strong>of</strong><br />

dopamine systems.


PP189 GENE-GENE INTERACTION IN DIFFERENT<br />

BIOLOGICAL PATHWAYS IN A SAMPLE OF<br />

CRACK/COCAINE BRAZILIAN PATIENTS<br />

A. Negrão(1), A. Pereira(2), C. Guindalini(3), G. Messas(1), R.<br />

Laranjeira(4), J. Krieger(2), H. Vallada(1)<br />

1. Program <strong>of</strong> <strong>Genetics</strong> and Pharmacogenetics, Department and<br />

Institute <strong>of</strong> Psychiatry, University <strong>of</strong> São Paulo Medical School 2.<br />

Laboratory <strong>of</strong> Genetic and Molecular Cardiology/LIM-13, Heart<br />

Institute (InCor), University <strong>of</strong> Sao Paulo Medical School 3.<br />

Department <strong>of</strong> Psychobiology, Universidade Federal de São Paulo<br />

(UNIFESP) 4. Alcohol and Drug Research Unit, Psychiatry<br />

Department, Federal University <strong>of</strong> São Paulo<br />

Genetic susceptibility to crack/cocaine addiction is less studied than<br />

other drug addictions although there are several single-locus positive<br />

association studies. Similarly to other complex disorders, odds ratios<br />

derived from those studies are low and there is a paucity <strong>of</strong> studies in<br />

the field investigating gene-gene interactions. Clinical and<br />

experimental data indicate the expression <strong>of</strong> genes associated with<br />

dopaminergic brain reward systems as well as biological rhythms,<br />

cortical modulation <strong>of</strong> mesolimbic function, pharmacokinetics <strong>of</strong><br />

cocaine among others that may render subjects susceptible to cocaine<br />

addiction. We used nonparametric analytical methods to unravel<br />

gene-gene interactions in genes associated with different biochemical<br />

pathways in patients with crack/cocaine addiction. A total <strong>of</strong> 699<br />

inpatients and outpatient who met ICD10 diagnosis <strong>of</strong> cocaine<br />

dependence and screened unaffected normal controls (n=886) were<br />

genotyped for 29 markes int the following genes: Dopamine receptor<br />

D2 (DRD2), Dopamine transporter gene (DAT), Catechol-Omethyltransferase<br />

(COMT), Dopamine beta-hydroxylase (DBH), Gprotein-coupled<br />

receptor kinase 3 (GRK3), Period (PER) 1 and 2;<br />

and, Gluthatione-S-tranferese-Pi (GSTP1). To examine potential<br />

gene-gene interactions we applied the Multifactor Dimensionality<br />

Reduction (MDR) and parameters were set to ten-fold leave-one-out<br />

crossvalidation and a search <strong>of</strong> all possible four loci interactions. A<br />

model that contained one marker each <strong>of</strong> the DAT/DBH/PER/GRK3<br />

genes had the best fit with a crossvalidation consistency <strong>of</strong> 9/10 and a<br />

testing balance accuracy <strong>of</strong> 61% and resulted in a odds ratio <strong>of</strong> 3.9<br />

(3.1 - 4.7). Markers in those genes had failed to be detected in the<br />

past as being associated with the cocaine status in previous papers<br />

using this very same sample. Graphical representations <strong>of</strong> this<br />

epistatic model in a entropy generated model in the same MDR<br />

s<strong>of</strong>tware showed that DBH and PER interaction contributed to a<br />

decreased level <strong>of</strong> entropy whereas the DAT and GRK3 genes had<br />

main effects that did not contribute in the same way to the epsitatic<br />

result generated by the program. We demonstrate that analytical tools<br />

that addressed epistasis can detect loci that were not associated with<br />

cocaine addiction in single-locus studies and can broaden the picture<br />

<strong>of</strong> susceptibility genes in cocaine addiction.<br />

165<br />

PP190 GENOME-WIDE ASSOCIATION STUDY (GWAS) OF<br />

ALCOHOL DEPENDENCE (AD) AND RELATED TRAITS IN<br />

THE IRISH AFFECTED SIB PAIR STUDY OF ALCOHOL<br />

DEPENDENCE (IASPSAD)<br />

A. Adkins*(1), L. Hack(1), B. Webb(2), M. Riemers(3), D.<br />

Patterson(5), D. Walsh(6), C. Prescott(4), D. Dick(1,2), K.<br />

Kendler(1,2), B. Riley(1,2)<br />

1. Virginia Institute for <strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong>,<br />

Department <strong>of</strong> Human and Molecular <strong>Genetics</strong>, Virginia<br />

Commonwealth University 2. Virginia Institute for <strong>Psychiatric</strong> and<br />

Behavioral <strong>Genetics</strong>, Department <strong>of</strong> Psychiatry, Virginia<br />

Commonwealth University 3. Department <strong>of</strong> Biostatistics, Virginia<br />

Commonwealth University 4. Department <strong>of</strong> Psychology, University<br />

<strong>of</strong> Southern California 5. Shaftesbury Square Hospital 6. Health<br />

Research Board<br />

*adkinsae@vcu.edu<br />

Introduction: The powerful, systematic, and unbiased genome-wide<br />

association study (GWAS) has been successful in identifying<br />

replicated susceptibility variants for numerous complex diseases,<br />

including psychiatric disorders. Only 3 individually genotyped casecontrol<br />

GWAS <strong>of</strong> AD have been published. We report here the<br />

completion <strong>of</strong> data collection and QC for a fourth in samples from the<br />

IASPSAD.Our study has the advantage that our sample is clinically<br />

homogeneous and richly phenotyped. In addition to being a<br />

reasonably powered individual GWAS <strong>of</strong> AD, our study will also<br />

increase by ~20% the total genotyped case/control data available for<br />

AD.<br />

Methodology: Our sample consists <strong>of</strong> 1050 Irish cases and affected<br />

siblings drawn from the IASPSAD and diagnosed using DSM-IV<br />

criteria. The majority <strong>of</strong> the sample was genotyped on the Affymetrix<br />

V6.0 array, while promising markers will be genotyped using<br />

Taqman assays in the remaining cases. 2000 Irish population controls<br />

were genotyped on the same platform by the Wellcome Trust Case<br />

Control Consortium 2 (WTCCC2) and the Broad Institute.<br />

Results: 99.7% <strong>of</strong> our sample has Birdseed call rates >95%. A<br />

rigorous, novel QC method developed at the Virginia Institute for<br />

<strong>Psychiatric</strong> and Behavioral <strong>Genetics</strong> involving factor analysis <strong>of</strong> QC<br />

metrics and visual inspection <strong>of</strong> false color microarray images is<br />

being applied to our data. Primary analysis will include allelic<br />

association testing for AD using maximum likelihood methods to<br />

correct for the non-independence <strong>of</strong> sibs and false discovery rate to<br />

correct for multiple testing, and will be completed Spring-Summer<br />

2011. Our full sample has > 78% power to detect effect sizes for AD<br />

<strong>of</strong> 1.2-1.3 with minor allele frequencies <strong>of</strong> 0.2-0.3. Through<br />

collaborations, we have access to multiple independent AD GWAS<br />

samples to attempt replication <strong>of</strong> our findings and conduct a metaanalysis.<br />

Secondary analyses will include testing for association with<br />

several quantitative traits within AD cases, including initial<br />

sensitivity, tolerance, maximum daily drinks, and a factor score for<br />

withdrawal.<br />

Conclusions: Along with previous AD GWAS data, our data will<br />

provide the opportunity to identify replicable associations that can be<br />

followed up with functional studies in model organisms within the<br />

context <strong>of</strong> the Virginia Commonwealth University Alcohol Research<br />

Center (VCU-ARC).


PP191 BDNF MODERATES THE ASSOCIATION BETWEEN<br />

MATERNAL PRENATAL SMOKING AND OFFSPRING<br />

EXTERNALIZING BUT NOT INTERNALIZING DISORDERS<br />

A. Talati*, M. Weissman, S. Hodge, P. Wickramaratne, S. Hamilton<br />

Columbia University Medical Center<br />

*at2071@columbia.edu<br />

Introduction:<br />

Introduction: Maternal smoking during pregnancy is associated with<br />

a number <strong>of</strong> adverse psychiatric and behavioral outcomes among<br />

their <strong>of</strong>fspring1-2. The biological underpinnings <strong>of</strong> these associations<br />

however remain unclear. We examined the role <strong>of</strong> the brain derived<br />

neurotrophic factor (BDNF) in this pathway, based on the cumulative<br />

evidence that (a) BDNF plays a critical role in neuronal survival and<br />

differentiation, processes disrupted by overstimulation <strong>of</strong> nicotinic<br />

receptors during fetal development; (b) nicotine exposure may be<br />

able to regulate BDNF gene expression via epigenetic modification<br />

(methylation)3; and (c) BDNF levels correlate inversely with the<br />

level <strong>of</strong> nicotine exposure among adult smokers 4. We tested the<br />

hypothesis that subjects who were exposed to prenatal smoking and<br />

had lower BDNF function would be at the greatest risk for aberrant<br />

neurocircuit development, and thus show greater rates <strong>of</strong> disorders<br />

related to behavioral control.<br />

Methodology: The analysis is based on 221 <strong>of</strong>fspring (70 exposed;<br />

141 unexposed) who were followed for approximately 20 years<br />

through four assessment waves. Prenatal history, which included<br />

information on maternal smoking and other substance use during<br />

pregnancy, was assessed at the first wave (year 1). Exposure was<br />

classified based on whether or not the mother smoked 10 or more<br />

cigarettes daily while pregnant. Offspring outcomes were then<br />

measured prospectively at years 2, 10 and 20 using the age<br />

ageappropriate version <strong>of</strong> the Schedule for Affective Disorders and<br />

Schizophrenia (SADS). Final diagnoses were made using the bestestimate<br />

procedure, blind to exposure status or previous assessments.<br />

BDNF activity was assessed by the genotype at the functional<br />

polymorphism rs6265 (val66met) which is associated with<br />

differential secretion levels <strong>of</strong> the protein; four additional single<br />

nucleotide polymorphisms (SNPs) were also examined: rs4923463,<br />

rs11030104, rs7103411, and rs2049045.<br />

Results: Prenatal smoke exposure was associated with increased risk<br />

for drug use disorder (DUD), alcohol use disorder (AUD), and<br />

conduct disorder (CD), but not with major depressive disorder<br />

(MDD) or any anxiety disorder. BDNF gene variation was not<br />

independently associated with any outcomes. However, <strong>of</strong>fspring<br />

who had the low-functioning BDNF variant (defined as having one or<br />

more met encoding alleles at rs6265) and were exposed in utero,<br />

showed a two- to three-fold increase in the rates rates <strong>of</strong> AUD (p =<br />

.03), DUD (p = .09), and CD (p = .005), than <strong>of</strong>fspring who were<br />

val/val and/or unexposed. Similar patterns were obtained for the other<br />

tested SNPs except rs7103411. There were no gene-by-exposure<br />

interactions for MDD or anxiety outcomes, for nonpsychopathological<br />

drug or alcohol use. The above associations could<br />

not be accounted for by maternal smoking outside <strong>of</strong> pregnancy,<br />

alcohol use, or antisocial personality.<br />

Conclusions: BDNF plays an important role in fetal development <strong>of</strong><br />

neural circuitry. Offspring who already have lower levels BDNF and<br />

are then exposed to further prenatal insults such as smoke exposure<br />

may be most vulnerable to disorders that depend on these circuits. It<br />

should be borne in mind that these results point to a general<br />

vulnerability to a spectrum <strong>of</strong> externalizing problems rather a specific<br />

DSM disorder. Future research should therefore continue to examine<br />

multiple outcomes, as well as outcomes at multiple thresholds, in<br />

order to further delineate the specificity <strong>of</strong> these mechanisms.<br />

166<br />

PP192 GENETIC VARIATION IN HEROIN DEPENDENCE,<br />

BINGE DRINKING AND CIGARETTE SMOKING<br />

C. Barta*(1), N. Nemeth(1), Z. Demetrovics(2), R. Urban(2), J.<br />

Farkas(2), N. Torma(2), A. Eisinger(2), A. Magi(2), M. Kapitany-<br />

Foveny(2), P. Sarkozy(3), M. Sasvari-Szekely(1)<br />

1. Institute <strong>of</strong> Medical Chemistry, Molecular Biology and<br />

Pathobiochemistry, Semmelweis University 2. Institute <strong>of</strong><br />

Psychology, Eotvos Lorand University 3. Department <strong>of</strong><br />

Measurement and Information Systems, Budapest University <strong>of</strong><br />

Technology and Economics<br />

*csaba.barta@eok.sote.hu<br />

Introduction: Neural networks within the reward system play a key<br />

role in the development <strong>of</strong> substance dependence. Variations in<br />

candidate genes coding for receptors, transporters and metabolizing<br />

enzymes involved in these pathways have been implicated for<br />

association with substance use disorder.<br />

Methodology: A total <strong>of</strong> 32 SNPs were genotyped on the Applied<br />

Biosystems Open Array Real-Time PCR Platform in three Hungarian<br />

cohorts. N=300 heroin dependent patients (HD) on methadone or<br />

suboxone maintenance therapy, N=300 university students (US)<br />

phenotypically characterized in detail for alcohol consumption habits<br />

and N=300 high school students aged 16 to 18 (HS) phenotypically<br />

characterized in detail for smoking habits. The studied genetic<br />

variations included SNPs in cholinergic receptors (CHRNA3/4/5,<br />

CHRNB3), dopaminergic (DRD2, DRD4), serotonergic, opioid,<br />

cannabinoid and GABAergic receptors, transporters and metabolizing<br />

enzymes, alcohol and aldehyde dehydrogenase genes, as well as<br />

some HPA axis gene variants.<br />

Results: Significant genetic effects were observed within the three<br />

groups. Certain genotypes were related to good versus poor response<br />

to substitution therapy. SNPs influencing alcohol drinking patterns<br />

and smoking habits were also identified.<br />

Conclusions: These results might prove useful in the individualized<br />

choice <strong>of</strong> therapy for heroin dependent patients or to prevent the<br />

development <strong>of</strong> substance use disorders in healthy adolescents and<br />

young adults.<br />

The European Union and the Europen Social Fund has provided<br />

financial support under the grant agreement No.: TAMOP 4.2.1./B-<br />

09/1/KMR-2010-0003.<br />

The study was co-funded by National Grants: OTKA F-46788 and<br />

KAB-KT-10-0016


PP193 GENETIC VARIATION, ADHD SYMPTOMS, AND<br />

SMOKING PHENOTYPES: A PRELIMINARY STUDY<br />

L. Bidwell*(1), M. Garrett(1), F. McClernon(1), B. Fuemmeler(1),<br />

R. Williams(1), A. Ashley-Koch(1), S. Kollins(1)<br />

1. Brown University 2. Duke University Medical Center<br />

*cinnamon_bidwell@brown.edu<br />

Introduction: Symptoms <strong>of</strong> attention-deficit hyperactivity disorder<br />

(ADHD) are a significant risk factor for smoking behaviors,<br />

including earlier age <strong>of</strong> initiation and likelihood <strong>of</strong> progression to<br />

regular use. Several converging lines <strong>of</strong> work suggest that the high<br />

rates <strong>of</strong> smoking in the presence <strong>of</strong> ADHD symptoms may be related<br />

to common genetic vulnerabilities that increase risk for both nicotine<br />

dependence and ADHD. Therefore, it may be informative to include<br />

ADHD symptoms in genetic studies <strong>of</strong> smoking. We conducted an<br />

exploratory study to assess whether ADHD symptoms interact with<br />

genetic variation to predict smoking phenotypes in a large<br />

epidemiological sample.<br />

Methodology: Participants were a subsample <strong>of</strong> 1,900 unrelated<br />

individuals with genotype data drawn from the National Longitudinal<br />

Study <strong>of</strong> Adolescent Health (Add Health), a nationally representative<br />

sample <strong>of</strong> adolescents followed from 1995 to 2002. Linear<br />

regression was used to examine relationships among self-reported<br />

ADHD symptoms, genotype, and smoking phenotypes, including<br />

self-reported initial reactions to cigarettes and lifetime history <strong>of</strong><br />

regular smoking.<br />

Results: No main effect for any <strong>of</strong> these polymorphisms was<br />

observed in predicting smoking phenotypes. DRD2 and MAOA<br />

polymorphisms both interacted with ADHD symptoms to<br />

significantly predict early smoking experiences and risk for lifetime<br />

smoking. Interactions with ADHD symptoms and SLC6A4, DRD4,<br />

andCYP2A6 genes in predicting smoking phenotypes were also<br />

found.<br />

Conclusions: These findings, while preliminary, suggest that<br />

genotypes associated with dopamine neurotransmission and nicotine<br />

metabolism interact with ADHD symptoms to influence early and<br />

later smoking risk. Theseresults suggest that ADHD symptoms<br />

qualify genetic associations with smoking phenotypes in important<br />

ways and should be accounted for in genetic studies <strong>of</strong> smoking.<br />

167


POSTER SESSION IV<br />

ABSTRACTS<br />

168


STATISTICAL GENETICS, GENETIC<br />

EPIDEMIOLOGY<br />

PP194 GENOME-WIDE INTERACTION ANALYSIS IN A<br />

DANISH POPULATION-BASED SCHIZOPHRENIA SAMPLE<br />

ECIP<br />

L. Foldager*(1,2), J. Grove(2,3), D. Demontis(3), M. Hollegaard(4),<br />

T. Ørnt<strong>of</strong>t(5,6), M. Didriksen(7), D. Hougaard(4), C. Wiuf(2), P.<br />

Mortensen(8), O. Mors(1), A. Børglum(1,3)<br />

1. Centre for <strong>Psychiatric</strong> Research, Aarhus University Hospital 2.<br />

Bioinformatics Research Centre, Aarhus University 3. Inst. <strong>of</strong><br />

Biomedicine, Dept. <strong>of</strong> Human <strong>Genetics</strong>, Aarhus University 4. Section<br />

<strong>of</strong> Neonatal Screening and Hormones, Dept. Clinical Biochemistry<br />

and Immunology, Statens Serum Institut 5. Dept. <strong>of</strong> Molecular<br />

Medicine, Aarhus University Hospital 6. AROS Applied<br />

Biotechnology A/S 7. Synaptic transmission, H. Lundbeck A/S 8.<br />

National Centre for Register-based Research, Aarhus University<br />

*leslfold@rm.dk<br />

Introduction: In a search for single nucleotide polymorphism (SNP)<br />

interactions in a Danish genome-wide association study <strong>of</strong><br />

schizophrenia (SZ) we applied a number <strong>of</strong> genome-wide interaction<br />

strategies. The aim was to 1) complement the single-marker analysis,<br />

2) pursue interesting pathways, and 3) compare various interaction<br />

analysis methods.<br />

Methodology: The case sample consisted <strong>of</strong> Danish citizens born in<br />

May 1981 or later who had a diagnosis <strong>of</strong> SZ (ICD-10 F20) in the<br />

Danish registries as <strong>of</strong> May 2007. One-to-one matched controls were<br />

selected as follows: same birth date, same gender, not the same<br />

mother, resident in Denmark (and alive) and without a registered<br />

diagnosis <strong>of</strong> SZ at the time <strong>of</strong> the first SZ diagnosis <strong>of</strong> the case. DNA<br />

was extracted using whole-genome-amplification from neonatal dried<br />

blood spot samples obtained from the Danish Newborn Screening<br />

Biobank and genotyped using the Illumina Infinium HD Human610-<br />

Quad BeadChip. Quality control (QC) excluded subjects having a call<br />

rate less than 0.97, and SNPs with call rate less than 0.99, minor<br />

allele frequency less than 0.0015 or Hardy-Weinberg equilibrium P<br />

value less than 0.0001. A total <strong>of</strong> 869 schizophrenia cases (397<br />

females and 472 males) and an equivalent number <strong>of</strong> matched<br />

controls remained for the analyses. We only consider SNPs from<br />

autosomes in which a total <strong>of</strong> 527,833 SNPs remained after QC.<br />

Single-marker additive analysis (trend test) and test for additive<br />

interaction between pairs <strong>of</strong> markers were carried out with<br />

conditional logistic regression using Stata. Population stratification<br />

was accounted for by inclusion <strong>of</strong> the first principal component<br />

obtained by Eigenstrat on a pruned subset <strong>of</strong> 42,388 autosomal SNPs<br />

not in linkage disequilibrium (r2


PP196 MATHEMATICALLY-BASED INTEGRATION OF<br />

MULTIPLE HETEROGENEOUS GENETIC DATA SETS<br />

J. Bukszar*(1), A. Khachane(1), K. Aberg(1), Y. Liu(2), J.<br />

McClay(1), P. Sullivan(2), E. van den Oord(1)<br />

1. Center for Biomarker Research and Personalized Medicine, School<br />

<strong>of</strong> Pharmacy, Virginia Commonwealth University 2. Department <strong>of</strong><br />

<strong>Genetics</strong>, University <strong>of</strong> North Carolina at Chapel Hill<br />

*jbukszar@vcu.edu<br />

Introduction: We present MIND, a novel mathematically-based<br />

framework/s<strong>of</strong>tware that integrates information from multiple<br />

heterogeneous existing data sets into a novel data collection in order<br />

to identify markers associated with a complex disease. MIND can<br />

integrate existing data sets generated by any kind <strong>of</strong> technology (e.g.,<br />

expression arrays, proteomics, GWAS) or activity (e.g., actual data<br />

collection, literature search, construction <strong>of</strong> disease-specific<br />

biochemical networks). Furthermore, external data may provide<br />

information at any genetic level ranging from individual variants<br />

(e.g., SNPs), genes (e.g., literature search), groups <strong>of</strong> genes (e.g.,<br />

pathways), or entire chromosomal segments (e.g., linkage studies or<br />

targeted next-generation sequencing). A unique feature <strong>of</strong> MIND is<br />

that it is entirely based on a solid mathematical foundation, which<br />

ensures substantially higher accuracy than widely used heuristic<br />

methods.<br />

Methodology: First, we developed the mathematical formulas that<br />

provide us with the exact posterior probability that a marker is<br />

associated with a certain disease based on the information in different<br />

type <strong>of</strong> data sets. We then formulated a parametrization that allowed<br />

us to collapse many model parameters into a single parameter, for<br />

which we developed a precise estimator. We implemented the<br />

method in a freely-available and user-friendly R package, called<br />

MIND (Mathematically-based Integration <strong>of</strong> heterogeNeous Data).<br />

Moreover, we developed tools that assess whether an existing data set<br />

is informative or not, and tools that allow us to measure the increase<br />

in total information by adding a new data set.<br />

Results: Through simulations and cross-validation analyses<br />

involving GWAS meta-analyses <strong>of</strong> psychiatric disorders as well as an<br />

actual replication study <strong>of</strong> over 6,500 SNPs in 6,300 schizophrenic<br />

patients and their family members we show that MIND 1) is accurate,<br />

2) outperforms marker selection for follow up studies based on pvalues,<br />

and 3) identifies effects that would otherwise require<br />

replication <strong>of</strong> over 20 times as many markers.<br />

Conclusions: MIND is able to make use <strong>of</strong> the multi-dimensional<br />

data that is become available from the dramatically growing<br />

repositories and other data bases to detect effects that would<br />

otherwise require prohibitive large sample sizes or replication<br />

studies.<br />

170<br />

PP197 EVIDENCE FOR ASSOCIATION OF PRO-<br />

INFLAMMATORY CYTOKINE GENE<br />

POLYMORPHISMS WITH SCHIZOPHRENIA IN SOUTH<br />

INDIAN POPULATION<br />

L. Srinivas*(1), N. V.(1), C. Nair(2), P. Allencherry(3), M.<br />

Banerjee(1)<br />

1. Human Molecular <strong>Genetics</strong> Laboratory, Rajiv Gandhi Centre for<br />

Biotechnology 2. Nair's Hospital 3. Mental Health Centre<br />

*lekshmys@gmail.com<br />

Introduction: Schizophrenia is a severe and debilitating mental<br />

disorder with a lifetime risk <strong>of</strong> about 1%, characterized by<br />

hallucinations, delusions and cognitive deficits, with heritability<br />

estimated at up to 80%. It is a complex disorder which may involve<br />

multiple genes with mild to moderate effect and non-genetic risk<br />

factors like environmental and psychological assaults. Cytokines,<br />

regulators <strong>of</strong> immune/inflammatory reactions and brain development,<br />

emerge as part <strong>of</strong> a common pathway <strong>of</strong> genetic and environmental<br />

components <strong>of</strong> schizophrenia. They are secreted by various cells and<br />

act as signals between cells to regulate the immune response to injury<br />

and infection. In addition to providing communication between<br />

immune cells, specific cytokines play a role in signaling the brain to<br />

produce neurochemical, neuroendocrine, neuroimmune, and<br />

behavioral changes. They strongly influence the dopaminergic,<br />

noradrenergic, and serotonergic neurotransmission. Considering these<br />

factors, cytokines are implicated as a contributing factor for<br />

schizophrenia. Functional polymorphisms in cytokine genes may<br />

predispose individuals to schizophrenia. Genome-wide association<br />

studies in schizophrenia have found significant associations with<br />

several single nucleotide polymorphisms (SNPs) across the major<br />

histocompatibility complex (MHC) region reinforcing an immune<br />

component to schizophrenia risk.<br />

Methodology: We performed a case-control association study using<br />

248 patients from Kerala, South India and 244 normal healthy<br />

ethnically matched controls. We screened 23 SNPs and 1 VNTR<br />

polymorphism from 10 cytokine genes (IL1A, IL1B, IL1RN, IL3,<br />

IL4, IL6, IL10, IFNG, TNFA and TGFB1). Genomic DNA was<br />

isolated from subjects and genotyping was carried out by PCR-RFLP,<br />

Taqman allelic discrimination assay and Kaspar assays. Data analysis<br />

including allelic, genotypic, haplotypic and diplotypic frequencies<br />

were calculated and compared using Unphased v3.0.13 s<strong>of</strong>tware. LD<br />

between SNPs was also computed using Unphased. Hardy Weinberg<br />

equilibrium (HWE) was calculated and LD plots were generated<br />

using Haploview v4.1.<br />

Results: Functional polymorphisms in pro-inflammatory cytokine<br />

genes IL1A, IL6, TNFA and IFNG were found to be associated with<br />

schizophrenia. IL1A polymorphism -889G/A (rs1800587) was found<br />

to be associated with schizophrenia at the allelic (p= 0.0269),<br />

genotypic (p=0.0412), haplotypic and diplotypic levels. IL6 -572 G/C<br />

(rs1800796) was found to be associated with the disease at the<br />

genotypic (p=0.0038) and at the diplotypic level (p=0.001). IL6 -174<br />

G/C (rs1800795) was found to be associated at the allelic (p=0.0374),<br />

genotypic (p=0.0339), haplotypic (p=0.02) and diplotypic levels<br />

(p=0.001). TNFA -238G/A (rs361525) polymorphism was also<br />

significantly associated (allelic p=0.0216, genotypic p=0.0063).<br />

Statistically significant association was also observed with IFNG<br />

+3232 C/T (rs2069718) at the genotypic level (P= 0.0096). We also<br />

found a very strong protective effect <strong>of</strong> this polymorphism at the<br />

haplotypic (p=9.23E-07) and diplotypic (p=1.51E-07) levels.<br />

Significant differences were observed between the LD plots <strong>of</strong> IL-6<br />

and IFNG in cases and controls.<br />

Conclusions: Our findings <strong>of</strong> association <strong>of</strong> cytokine gene<br />

polymorphisms with schizophrenia support the hypothesis that<br />

genetically determined changes in cytokine regulation may contribute<br />

to the pathogenesis <strong>of</strong> schizophrenia confirming a role <strong>of</strong> immune<br />

genes in disease susceptibility


PP198 D-AMINO ACID OXIDASE ACTIVATOR GENE<br />

(DAOA) VARIATION AFFECTS CEREBROSPINAL FLUID<br />

HOMOVANILLIC ACID CONCENTRATION IN HEALTHY<br />

CAUCASIANS<br />

D. Andreou*(1), P. Saetre(1), T. Werge(2), O. Andreassen(3,4), I.<br />

Agartz (1,5,6), G. Sedvall(1), H. Hall(1,7), L. Terenius(1), E.<br />

Jönsson(1)<br />

1. Department <strong>of</strong> Clinical Neuroscience, HUBIN project, Karolinska<br />

Institutet and Hospital, R5:00, SE-171 76 2. Research Institute <strong>of</strong><br />

Biological Psychiatry, Mental Health Center Sct. Hans, Copenhagen<br />

University Hospital 3. TOP project, Division <strong>of</strong> Psychiatry, Ullevål<br />

University Hospital, University <strong>of</strong> Oslo 4. TOP project, Institute <strong>of</strong><br />

Clinical Medicine, Psychiatry section Vinderen, University <strong>of</strong> Oslo 5.<br />

Department <strong>of</strong> Psychiatry, Diakonhjemmet Hospital 6. Institute <strong>of</strong><br />

Psychiatry, University <strong>of</strong> Oslo 7. Department <strong>of</strong> Public Health and<br />

Caring Sciences, Uppsala University<br />

*dimitrios.andreou@sll.se<br />

Introduction: Variation in the D-amino acid oxidase activator gene<br />

(DAOA) has been associated with schizophrenia, schizophreniarelated<br />

characteristics, major depression and bipolar disorder. The<br />

DAOA protein has been detected in various parts <strong>of</strong> the central<br />

nervous system and modulates the function <strong>of</strong> D-amino oxidase<br />

(DAO), an enzyme that catalyzes the oxidative deamination <strong>of</strong> Dserine<br />

and D-3,4-dihydroxyphenylalanine (D-DOPA). D-serine is<br />

implicated in glutamatergic transmission, whereas D-DOPA is<br />

converted to L-3,4-DOPA, a precursor <strong>of</strong> dopamine and<br />

noradrenaline. We hypothesized that DAOA polymorphisms are<br />

associated with dopamine, serotonin and noradrenaline turnover in<br />

the human brain.<br />

Methodology: Unrelated healthy Caucasian participants (n=132)<br />

were included in the study. Genomic DNA was extracted from whole<br />

blood samples and four single nucleotide polymorphisms (rs2391191,<br />

rs778294, rs3918342, rs1421292) previously reported to be<br />

associated with schizophrenia,were genotyped using the Illumina<br />

Bead Station 500GX and the 1536-plex Illumina Golden Gate assay.<br />

Cerebrospinal fluid (CSF) samples were drawn by lumbar puncture<br />

and the concentrations <strong>of</strong> the major dopamine metabolite<br />

homovanillic acid (HVA), the major serotonin metabolite 5hydroxyindoleacetic<br />

acid (5-HIAA), and the major noradrenaline<br />

metabolite 3-methoxy-4-hydroxyphenylglycol (MHPG) were<br />

measured with mass fragmentography. To test allele association<br />

between DAOA SNPs and CSF monoamine metabolite<br />

concentrations, each monoamine metabolite was analyzed with a<br />

general linear model where the concentration was modeled as a linear<br />

function <strong>of</strong> the allele count <strong>of</strong> each SNP separately and one or more<br />

covariates<br />

Results: Two <strong>of</strong> the investigated DAOA polymorphisms, rs3918342<br />

(corrected p-value=0.013) and rs1421292 (corrected p-value=0.043),<br />

were found to be significantly associated with CSF HVA<br />

concentrations. Rs1421292 was nominally associated with CSF 5-<br />

HIAA concentrations. None <strong>of</strong> the SNPs were associated with MHPG<br />

concentrations. Rs3918342 and rs1421292 were in strong linkage<br />

disequilibrium with each other and no additional effect <strong>of</strong> rs1421292<br />

on HVA concentration, on top <strong>of</strong> rs3918342, was observed. Carriers<br />

<strong>of</strong> the rs3918342 T allele had 50 nmol/l lower HVA mean<br />

concentration compared to C homozygotes.<br />

Conclusions: Two DAOA polymorphisms, likely to capture the same<br />

signal, were significantly associated with CSF HVA concentrations,<br />

indicating altered dopamine turnover in healthy human subjects. This<br />

association may indirectly explain the replicated effect <strong>of</strong> DAOA on<br />

human behavior and psychiatric symptomatology. Further research is<br />

needed in order to reveal whether the association between DAOA and<br />

the dopamine system is also present in patients with schizophrenia<br />

and other psychiatric disorders.<br />

171<br />

PP199 ROBUST CALLING OF RARE AND POTENTIALLY<br />

COMMON CNVS IN SPECIFIC GENOMIC REGIONS<br />

D. Ivanov*, E. Rees, M. O'Donovan, M. Owen, G. Kirov<br />

Cardiff University<br />

*ivanovdk2@cf.ac.uk<br />

Introduction: Copy number variants (CNVs) account for a relatively<br />

large proportion <strong>of</strong> the human genetic variation and have been<br />

implicated as risk factors for common and rare disorders (Craddock,<br />

Hurles et al. 2010). Successfully identifying genuine CNVs is a nontrivial<br />

task (Baker 2010 Craddock, Hurles et al. 2010). Several<br />

methods have been proposed to normalize data and reduce interindividual<br />

variation, although no single method works for every CNV<br />

(Craddock, Hurles et al. 2010 Dellinger, Saw et al. 2010). Here we<br />

propose an improved analytical approach for calling <strong>of</strong> CNVs with a<br />

relatively high degree <strong>of</strong> confidence.<br />

Methodology: The method consists <strong>of</strong> genome-wide normalization<br />

<strong>of</strong> inter-individual variation, by using normalized intensity ratios (i.e.<br />

Log 2 Ratio derived from SNP arrays), followed by a genome-wide<br />

normalization <strong>of</strong> individual inter-probe variation. The method is an<br />

extension <strong>of</strong> the MeZOD method, by McCarthy et al (McCarthy,<br />

Makarov et al. 2009). Median z-scores for specific regions <strong>of</strong> interest<br />

were subsequently used to produce intensity histograms, where<br />

outliers represent potential CNVs. Although the method relies on<br />

calculating median z-scores for specific regions <strong>of</strong> interest, unlimited<br />

number <strong>of</strong> regions could be examined using the same normalized<br />

data. Furthermore, an automatic approach <strong>of</strong> CNV calling could be<br />

utilized by the use <strong>of</strong> normalized median z-scores.<br />

Results: The method was successfully used to identify schizophrenia<br />

CNV loci in 662 affected proband-parent trios. 270 CNVs were<br />

tested for validation with Agilent aCGH custom arrays. A<br />

conservative approach in calling CNVs with the z-score method<br />

resulted in 2.4% false positives and 5% false negatives, whereas a<br />

non-conservative approach produced 7.7% and 1.9% false positive<br />

and false negative rates respectively.<br />

Conclusions: We conclude that this method allows users to make<br />

highly confident predictions for the validity <strong>of</strong> any CNV, called by<br />

different CNV-calling algorithms, or for specific regions <strong>of</strong> interest.<br />

Programs for using this method are freely available at:<br />

http://x004.psycm.uwcm.ac.uk/~dobril/z_scores_cnvs/


PP200 THE IMPACT OF THE PHENOTYPIC<br />

HETEROGENEITY ON GENOME-WIDE ASSOCIATION<br />

STUDIES IN COMPLEX PSYCHIATRIC DISEASES: A<br />

SIMULATION CASE-CONTROL STUDY<br />

M. Manchia*, J. Cullis, M. Alda<br />

Department <strong>of</strong> Psychiatry, Dalhousie University 5909 Veterans'<br />

Memorial Lane Room 3089, A.J. Lane Bldg, B3H 2E2<br />

*mirko.manchia@dal.ca<br />

Introduction: Findings from genome wide association studies<br />

(GWAS) <strong>of</strong> psychiatric disorders have identified genetic risk variants<br />

that explain only a fraction <strong>of</strong> the total phenotypic variance.<br />

Researchers have hypothesized that part <strong>of</strong> the missing heritability <strong>of</strong><br />

complex diseases could be attributed to yet undetected rare risk<br />

variants with larger effect sizes than common variants. Another<br />

possible explanation is that <strong>of</strong> phenotypic heterogeneity affecting<br />

GWAS findings. <strong>Psychiatric</strong> diseases are characterized by clinical<br />

heterogeneity that can influence the accuracy <strong>of</strong> phenotyping in<br />

genetic studies. For instance, imprecise phenotyping can reduce the<br />

estimate <strong>of</strong> the effect sizes <strong>of</strong> the associated variants. In bipolar<br />

disorder (BD), researchers have attempted to reduce clinical<br />

heterogeneity by using subphenotypes such as early-onset BD,<br />

presence <strong>of</strong> mood incongruent psychosis, or response to lithium<br />

treatment. Increased homogeneity <strong>of</strong> such group <strong>of</strong> patients can<br />

improve statistical power <strong>of</strong> detecting true association signals in<br />

genetic studies.<br />

Methodology: To study the impact <strong>of</strong> phenotypic heterogeneity on<br />

GWAS findings, we simulated case-control data assuming increasing<br />

(up to 90%) phenotypic admixture (β) in cases. For each level <strong>of</strong><br />

admixture, we calculated the minimum sample size needed to achieve<br />

90% <strong>of</strong> the statistical power requested to detect association signals at<br />

P < 5 x 10 -8 . Simulations were performed testing different genotype<br />

relative risks (GRRs) (1.1, 1.2, 1.3, 1.5, 2, 5), for varying population<br />

prevalence (F = 0.001, 0.01, 0.05, 0.1), under dominant and<br />

multiplicative genetic models using a compiled script run in R<br />

environment.<br />

Results: Admixture significantly affected the sample size required to<br />

reach a power <strong>of</strong> 90% for significant threshold set at P < 5 x 10 -8 . We<br />

observed that: 1) heterogeneity caused a significant loss <strong>of</strong> statistical<br />

power that was disproportionately larger than the admixture: increase<br />

in a proportion <strong>of</strong> “non-cases” resulted in a non-linear increase <strong>of</strong> the<br />

sample size needed to achieve 90% <strong>of</strong> statistical power. For instance,<br />

admixture set at 50% required at a minimum three times the sample<br />

size needed to achieve the same statistical power without admixture<br />

2) heterogeneity caused a marked reduction <strong>of</strong> the estimated effect<br />

size reflected in decrease <strong>of</strong> estimated odds ratios. This effect size<br />

reduction was also non-linear in relation to the degree <strong>of</strong> admixture.<br />

Conclusions: These findings from a simulated case-control study<br />

provide evidence on the impact that phenotypic heterogeneity can<br />

exert on GWAS findings. The absence <strong>of</strong> admixture ensures the need<br />

<strong>of</strong> significantly smaller sample sizes to achieve the same statistical<br />

power <strong>of</strong> larger sample sizes with medium-high level <strong>of</strong> phenotypic<br />

heterogeneity. The accuracy in the phenotypic assessment and the use<br />

<strong>of</strong> subgroups such as responders to lithium treatment in BD can<br />

significantly decrease the number <strong>of</strong> cases needed to be collected in<br />

order to achieve sufficient statistical power to detect association<br />

signals in GWAS.<br />

172<br />

PP201 FAMILY-BASED ASSOCIATION ANALYSIS OF<br />

GENES INVOLVED IN SYNAPTIC PLASTICITY AND<br />

AUTISM<br />

R. Sasanfar*(1,2,), R. Siburian(2), M. Ghadami(3), S. Haddad(2), A.<br />

Tolouei(4), B. Galloway(2), M. Rostami(5), S. Chang(6), A.<br />

Mahmoodizadeh(4), S. Santangelo(1,2,6)<br />

1. Harvard Medical School 2. Massachusetts General Hospital 3.<br />

Research Institute for Education 4. Special Education Organization 5.<br />

Kariminejad-Najmabadi Pathology & <strong>Genetics</strong> Center 6. Harvard<br />

School <strong>of</strong> Public Health<br />

*rsasanfar@pngu.mgh.harvard.edu<br />

Introduction: A wide variety <strong>of</strong> evidence, using many different<br />

behavioral paradigms, indicates a broad role for the involvement <strong>of</strong><br />

the extracellular signaling-regulated kinases (ERK) pathway in<br />

synaptic plasticity and consequently learning and memory.<br />

Disruption <strong>of</strong> neuronal protein synthesis through ERK pathway is the<br />

shared point <strong>of</strong> several essential pathways associated with autism<br />

including the mTOR pathway. Therefore, aberrant regulation <strong>of</strong> the<br />

ERK pathway is a potential cause for deficits associated with autism.<br />

Methodology: Family-based association analysis was performed<br />

using 700 individuals from 200 nuclear families from Iran with at<br />

least one autistic child. In total, 175 SNPs were genotyped for all six<br />

genes using Sequenom iPLEX. TDT analyses were performed with<br />

Plink s<strong>of</strong>tware. Quality control procedures applied to the data<br />

included rejection <strong>of</strong> any SNP markers with missingness greater than<br />

0.15, MAF less than 0.01, HWE p-value less than 2.8*10 -4 , and<br />

Mendelian Error rate <strong>of</strong> greater than 0.1. Families with a Mendelian<br />

Error rate <strong>of</strong> greater than 0.05 were also removed. After QC, 150<br />

SNPs and 672 individuals from 194 nuclear families were analyzed.<br />

Results: We failed to identify a significant association with these six<br />

genes and ASD. The best result was a nominally significant<br />

association <strong>of</strong> two markers from GRM1(rs9403771, p=.047, and<br />

rs2024589, p=.049) and one marker from GRIN2A (rs11645219,<br />

p=.046). Analysis is ongoing using Plink and FBAT to explore<br />

potential multimarker associations and gender effects.<br />

Conclusions: Although the sample size for this investigation is small<br />

and slightly underpowered (For a significance level <strong>of</strong> 0.05, there is<br />

78% power to detect a heterozygote GRR <strong>of</strong> 2.0 at D’ = 0.8 for q =<br />

0.1), our results do not support the involvement <strong>of</strong> these genes in<br />

conferring risk for ASD.


PP202 HOW IS MET/MET GENOTYPE OF THE<br />

CATECHOL-O-METHYL-TRANSFERASE VAL 108/158 MET<br />

POLYMORPHISM ASSOCIATED WITH SUICIDE VICTIMS<br />

FROM SLOVENIA?<br />

A. Videtič Paska*(1), P. Pregelj(2), M. Nikolac(3), T. Zupanc(4), G.<br />

Nedic(3), D. Muck Seler(3), R. Komel(1), N. Pivac(3)<br />

1. Institute <strong>of</strong> Biochemistry, Faculty <strong>of</strong> Medicine, University <strong>of</strong><br />

Ljubljana 2. University <strong>Psychiatric</strong> Hospital Ljubljana 3. Laboratory<br />

for Molecular Neuropsychiatry, Division <strong>of</strong> Molecular Medicine,<br />

Rudjer Boskovic Institute 4. Institute <strong>of</strong> Forensic Medicine, Faculty<br />

<strong>of</strong> Medicine, University <strong>of</strong> Ljubljana<br />

*alja.videtic@mf.uni-lj.si<br />

Introduction: Slovenia is one <strong>of</strong> the countries with the highest<br />

suicide rate in the world. In the year 2007 suicide rate was 21.5<br />

suicide victims per 100.000 citizens. So far, the results from the<br />

association studies <strong>of</strong> the various candidate genes for suicide are<br />

contradictory, with a lot <strong>of</strong> either positive associations or lack <strong>of</strong><br />

associations. The most <strong>of</strong> the studies suggest the implication <strong>of</strong><br />

specific signaling pathways, like serotonergic or catecholaminergic<br />

pathway, in suicide. ΦAmong the later the genes for dopaminergic<br />

receptors DRD2 and DRD4, dopamine transporter, dopamine-betahydroxylase,<br />

DOPA decarboxylase, aromatic L amino acid<br />

decarboxylase and catechol-o-methyl-transferase (COMT), are<br />

frequently studied candidate genes for suicide. It was hypothesized<br />

that COMT Val 108/158 Met polymorphism is associated with suicide,<br />

independently <strong>of</strong> psychiatric disorders. COMT is an enzyme that<br />

inactivates catecholamines (dopamine, norepinephrine and<br />

epinephrine) by thetransfer <strong>of</strong> a methyl group which leads to<br />

degradation <strong>of</strong> the molecule. A functional polymorphism on the<br />

COMT gene, COMT Val 108/158 Met,a G to A nucleotide transition<br />

resulting in valine (Val) to methionine (Met) substitution, was<br />

associated with psychiatric disorders related to high suicide risk and<br />

to suicidal behavior.<br />

Methodology: In a study comprising <strong>of</strong> 356 samples from suicide<br />

victims and 198 samples from control non-suicidal victims who died<br />

from traffic accidents, COMT Val 108/158 Met polymorphism was<br />

evaluated. To our knowledge we were the first to test the association<br />

<strong>of</strong> completed suicide and COMT Val 108/158 Met polymorphism<br />

hypothesis on Caucasian population.<br />

Results: The results revealed significant differences in the<br />

frequencies <strong>of</strong> the COMT Val 108/158 Met variants in male and female<br />

suicide victims and male and female control groups. There were<br />

significant differences in the frequencies <strong>of</strong> the Val/Val, Val/Met and<br />

Met/Met genotypes, Val and Met alleles, or Val carriers and the<br />

homozygous Met/Met genotype between male control and male<br />

suicide victims, or male control and male violent suicide victims. On<br />

the other hand, there was no significant difference in the frequency <strong>of</strong><br />

COMT Val 108/158 Met variants between female control and female<br />

suicide victims. Male carriers <strong>of</strong> the Met/Met genotype were detected<br />

more frequently in male control, than in male suicide victims.<br />

Conclusions: Our results, obtained on the sample from homogenous<br />

unrelated Caucasians, originating from Slovenia, suggest that suicide<br />

victims were less frequently carriers <strong>of</strong> the Met/Met genotype than<br />

control subjects, and that Met/Met genotype <strong>of</strong> the COMT<br />

Val 108/158 Met might have a protective role against suicide. However,<br />

we are suggesting further studies in order to confirm our results.<br />

173<br />

PP203 MENTAL HEALTH, GENDER AND SIBLING<br />

FERTILITY<br />

R. Power*(1), P. Lichtenstein(2), P. McGuffin(1), C. Lewis(1), A.<br />

Svensson(2)<br />

1. Institute <strong>of</strong> Psychiatry 2. Karolinska Institutet<br />

*robert.r.power@kcl.ac.uk<br />

Introduction: The combination <strong>of</strong> high heritability, high prevalence<br />

and a large decrease in fecundity compared to the general population<br />

means psychiatric disorders should face strong purifying selection.<br />

However almost every psychiatric disorder is sexually dimorphic and<br />

so will be exposed to sexual selection. Since fecundity is reduced, it<br />

is clear that negative health in the individual is not outweighed by<br />

increased ability to attract mates or have children. However selection<br />

may affect the frequency <strong>of</strong> sexually antagonistic genes, those that<br />

increase the fitness <strong>of</strong> one sex at the expense <strong>of</strong> the other. To give a<br />

possible example, autism (found more commonly in males) might<br />

result from the burden <strong>of</strong> several female beneficial alleles. This<br />

would lead to balancing selection on these genetic variants, keeping<br />

them at a stable frequency in the population.<br />

Methodology: Using the Swedish Multi-Generation Register and<br />

Hospital Discharge Register we examined evidence for sexually<br />

antagonistic genes in 4 psychiatric disorders. Fertility <strong>of</strong> affected<br />

individuals and their siblings was calculated as a ratio to that <strong>of</strong> the<br />

general population. The hypothesis being that individuals with<br />

disorders common in males should have sisters with increased fitness<br />

and brothers with decreased fitness, with the opposite being true for<br />

those disorders common in females. We chose anorexia nervosa and<br />

depression as female biased disorders (M:F ratios <strong>of</strong> 1:10 and 1:2<br />

respectively), and substance abuse and autism as male biased<br />

disorders (both 4:1). We combined previously explored inpatient<br />

records from 1973 to 2009 with outpatient records from 2001. A birth<br />

cohort including all individuals born between 1950 and 1980 was<br />

selected, to balance inclusion in all aspects <strong>of</strong> the dataset, diagnostic<br />

reliability, and likelihood <strong>of</strong> completing reproductive cycle. The total<br />

cohort included 4.4 million individuals, and ICD codes were used to<br />

identify affected individuals and their siblings.<br />

Results: Anorexia nervosa had a prevalence <strong>of</strong> 0.12% and sibling<br />

frequency <strong>of</strong> 0.17%. Depression had a prevalence <strong>of</strong> 3.44% and<br />

sibling frequency <strong>of</strong> 4.5%. Autism had a prevalence <strong>of</strong> 0.26% and a<br />

sibling frequency <strong>of</strong> 0.35%. Substance abuse, including alcoholism,<br />

had a prevalence <strong>of</strong> 1.74% and a sibling frequency <strong>of</strong> 2.39%. Using<br />

generalized estimating equations to account for within family<br />

correlation and year <strong>of</strong> birth, a fertility ratio <strong>of</strong> affected individuals<br />

and their siblings compared to the general population was calculated.<br />

In the four disorders the fertility <strong>of</strong> affected individuals varied but<br />

always differs from the general population significantly: a ratio <strong>of</strong><br />

1.01 for depression compared to general population was found 0.61<br />

for anorexia 0.79 for substance abuse and 0.29 for autism. For<br />

siblings there was a general trend, with brothers' having consistently<br />

significantly lower fertility than the general population (0.77-0.95)<br />

and sisters having significantly increased fertility (1.14-1.27).<br />

Conclusions: This similar trend across all disorders, whether more<br />

common in males or females, was not what we expected to find in the<br />

presence <strong>of</strong> sexually antagonistic genes. For several <strong>of</strong> these<br />

disorders we believe these to be the first results on the impact on<br />

siblings' fertility, and while we have yet to fully explore the cause it<br />

is clear that sex seems to play a crucial role in the direction <strong>of</strong> effect.<br />

Better understanding <strong>of</strong> the differential effects <strong>of</strong> the genes for<br />

psychiatric disorders will hopefully lead to a better understanding <strong>of</strong><br />

the genetic architecture <strong>of</strong> these disorders and the identification <strong>of</strong> the<br />

causal variants.


PP204 STRONG STABILITY IN GENETIC INFLUENCES ON<br />

AUTISTIC TRAITS IN CHILDHOOD: A LONGITUDINAL<br />

STUDY OF MORE THAN 6000 TWIN PAIRS<br />

K. Holmboe*(1), V. Hallett(1), F. Happé(1), R. Plomin(1), A.<br />

Ronald(2)<br />

1. MRC Social, Genetic and Developmental Psychiatry Centre,<br />

Institute <strong>of</strong> Psychiatry, King's College London 2. Department <strong>of</strong><br />

Psychological Sciences, Birkbeck, University <strong>of</strong> London<br />

*karla.holmboe@gmail.com<br />

Introduction: Autism spectrum disorders (ASDs) constitute a set <strong>of</strong><br />

developmental disorders characterized by deficits in the three core<br />

domains <strong>of</strong> social interaction, communication, and restricted,<br />

repetitive and stereotyped behaviors and interests. Sub-clinical<br />

autistic traits are detectable and show quantitative variation in the<br />

general population. Previous studies have found autistic traits in the<br />

general population to be moderately to highly heritable and to show<br />

modest environmental influences from infancy to early adulthood.<br />

However, it is not known whether the genetic and environmental<br />

influences on autistic traits that are important at one age are the same<br />

as at other ages. Answering this question is important in order to<br />

understand the developmental course <strong>of</strong> autistic traits and the causal<br />

influences that determine this course. The present study is the first<br />

population-based twin study to investigate the question <strong>of</strong> causal<br />

influences on the longitudinal development <strong>of</strong> autistic traits across<br />

middle childhood.<br />

Methodology: The sample included 6,280 twin pairs from the Twins<br />

Early Development Study (a United Kingdom general population<br />

study). Male and female twins were assessed on the same measure <strong>of</strong><br />

autistic traits (CAST: Childhood Asperger Syndrome Test; Scott,<br />

Baron-Cohen, Bolton, & Brayne, 2002) by teachers at ages 9 and 12<br />

years, and by parents at ages 8, 9 and 12 years. Data were subjected<br />

to structural equation modeling, and parameter estimates were<br />

derived from the best-fitting model. A Cholesky decomposition<br />

model was employed.<br />

Results: The parent data indicated high stability <strong>of</strong> autistic traits<br />

between ages 8, 9 and 12 for both males and females (r = .57-.67),<br />

whereas stability was more modest in the teacher data (r = .25-.35).<br />

The best-fitting model for the parent data involved additive genetic<br />

influences (A), reflecting the additive effect <strong>of</strong> alleles influencing<br />

autistic traits; nonadditive genetic influences (D), reflecting<br />

interactions between alleles at the same locus; and nonshared<br />

environmental influences (E), which impact on each individual and<br />

also include measurement error. The best-fitting model for the<br />

teacher data also included A and E, and a modest shared environment<br />

component (C), reflecting environmental influences that make<br />

children growing up in the same family similar. High heritability was<br />

observed in the parent data. There was also some difference between<br />

sexes, with males showing nonadditive genetic effects, which females<br />

did not. Teacher ratings showed moderate heritability and nonshared<br />

environmental influences and small effects <strong>of</strong> the shared<br />

environment. The longitudinal modeling revealed high genetic<br />

correlations, suggesting substantial stability in the genetic influences<br />

on autistic traits between 8 and 12 years <strong>of</strong> age.<br />

Conclusions: In conclusion, it is well known that ASDs are highly<br />

heritable and that diagnoses tend to be stable across the life span.<br />

Here we present the first evidence from a longitudinal populationbased<br />

twin study demonstrating that individual differences in autistic<br />

traits in the general population are also stable and highly heritable,<br />

and that this stability is due, in large part, to similar genetic<br />

influences operating across middle childhood.<br />

174<br />

PP205 EEG WAVE PATTERNS IN BIPOLAR DISORDER<br />

AND SCHIZOPHRENIA: COMPUTATIONAL<br />

DISCRIMINATION DURING GO/NOGO PARADIGMS<br />

Z. Syed*(1), M. Kamali(1), P. Deldin(1), L. O'Donnell(1), J.<br />

Chun(2), M. McInnis(1)<br />

1. University <strong>of</strong> Michigan 2. Harvard University<br />

*zhs@umich.edu<br />

Introduction: Bipolar disorder (BD) and schizophrenia (SZ) have<br />

considerable overlapping features and are characterized by deficits in<br />

response inhibition and social cognition relative to controls (CT). We<br />

explored the ability <strong>of</strong> sophisticated computational models using L1regularized<br />

L2-loss support vector machine (SVM) classification <strong>of</strong><br />

emotional Go/NoGo P300 peak amplitudes to distinguish between<br />

SZ, BD and CT patients.<br />

Methodology: 25 BD, 19 SZ and 20 CT patients were presented with<br />

a target (Go) or distractor (NoGo) emotion. In half the blocks one<br />

type <strong>of</strong> emotional face (angry, sad, happy) served as target while<br />

neutral faces were distractors. In the other half <strong>of</strong> the blocks the roles<br />

<strong>of</strong> targets and distracters were reversed. A total <strong>of</strong> 12 blocks with 600<br />

trials took about 20-25 minutes. Neurophysiological testing was<br />

completed while measuring electroencephalograhy (EEG) and eventrelated<br />

brain potentials were obtained from the EEG data. For each<br />

pair <strong>of</strong> categories out <strong>of</strong> BD, SZ and CT, a separate L1-regularized<br />

L2-loss SVM was trained and evaluated using leave-one-out crossvalidation<br />

on the average peak P300 amplitudes across all laterality x<br />

caudality x blocks (3 x 3 x 12 = 108 features). SVM parameters were<br />

learned by further cross-validation within the data in each iteration<br />

excluding the held out set.<br />

Results: L1-regularized L2-loss SVM classification <strong>of</strong> peak P300<br />

amplitudes produced a highly accurate diagnosis <strong>of</strong> SZ vs. CT [area<br />

under the receiver operating characteristic curve (AUROC): 0.984,<br />

precision: 94.7%, recall: 90.0%] and SZ vs. BP [AUROC: 0.983,<br />

precision: 92.3%, recall: 96.0%]. The SVM-based approach achieved<br />

moderate accuracy for the BP vs. CT case [AUROC: 0.720,<br />

precision: 72.7%, recall: 64.0%].<br />

Conclusions: Computational diagnostic models using emotion<br />

modulated event-related brain potentials can form the basis <strong>of</strong><br />

accurate discrimination between BP, SZ, and CT patients. These<br />

models can generalize to unseen test data not used in model<br />

development, and characterize differences between psychiatric<br />

patients and controls, as well as between different forms <strong>of</strong><br />

psychiatric disease. There are considerable implications for genetics<br />

research in characterizing the distinction between bipolar disorder<br />

and schizophrenia.


PP206 DEPRESSION AND THE VAL66MET<br />

POLYMORPHISM INFLUENCE THE SERUM BDNF LEVEL<br />

H. Buttenschøn*(1), B. Elfving(1), L. Foldager(1,2), P. Poulsen(1),<br />

J. Andersen(3), J. Bonde(4), M. Grynderup(5), A. Hansen(6), H.<br />

Kolstad(5), A. Kærgaard(3), L. Kærlev(5,7), S. Mikkelsen(4), J.<br />

Thomsen(4), A. Børglum(1,8), G. Wegener(1), O. Mors(1)<br />

1. Centre for <strong>Psychiatric</strong> Research, Aarhus University Hospital,<br />

Risskov 2. Bioinformatics Research Centre, Aarhus University 3.<br />

Danish Ramazzini Center, Department <strong>of</strong> Occupational Medicine,<br />

Regional Hospital 4. Department <strong>of</strong> Occupational Medicine,<br />

Bispebjerg University Hospital 5. Danish Ramazzini Center,<br />

Department <strong>of</strong> Occupational Medicine, Aarhus University Hospital 6.<br />

National Research Centre for the Work Environment 7. Center for<br />

National Clinical Databases South, Odense University Hospital 8.<br />

Institute <strong>of</strong> Biomedicine, Dept <strong>of</strong> Human <strong>Genetics</strong>, Aarhus<br />

University<br />

*henrbutt@email.dk<br />

Introduction: Depression may involve neurodegeneration and<br />

aberrant neuronal network function. One <strong>of</strong> the most extensively<br />

investigated targets with respect to brain neuroplasticity is brainderived<br />

neurotrophic factor (BDNF). Several studies have reported a<br />

difference in plasma and serum level <strong>of</strong> BDNF for individuals with<br />

depression and healthy controls. BDNF has been suggested as a<br />

candidate gene for depression.<br />

Methodology: The purpose <strong>of</strong> the present study was to perform a<br />

thorough investigation <strong>of</strong> BDNF using 163 individuals with<br />

depression and 290 healthy individuals. All individuals returned a<br />

completed questionnaire and participated in a semi-structured<br />

diagnostic interview. We included socio-demographic variables and<br />

health indicators. We determined serum BDNF levels and<br />

successfully analyzed six SNPs for association with depression. To<br />

assess independent determinants <strong>of</strong> BDNF we applied multiple linear<br />

regression models.<br />

Results: Several variables were correlated with the serum BDNF<br />

level. Age at diagnostic interview was the most significantly<br />

associated variable. We also observed a significant difference in<br />

serum BDNF level between cases and controls. However, in contrast<br />

to other studies we observed the BDNF level to be higher among<br />

cases compared to controls. A significant interaction between sex and<br />

the Val66Met polymorphism was furthermore observed. In the<br />

genetic analyses none <strong>of</strong> the genotyped SNPs were associated with<br />

depression.<br />

Conclusions: We conclude that the serum BDNF level is influenced<br />

by several factors including age, depression, the interaction between<br />

sex, and Val66Met polymorphism.<br />

175<br />

PP207 EXPLORATION OF GENETIC RISK,<br />

SCHIZOPHRENIA AND BRAIN VOLUME<br />

G. Russo*(1), A. Vasquez(2,3), M. Rijpkema(4), G. Fernández(4,5),<br />

H. Brunner(2), P. Holmans(1), B. Franke(2,3), O. Michael (1), O.<br />

Michael(1)<br />

1. MRC Centre fr Neuropsychiatric <strong>Genetics</strong> and Genomics 2.<br />

Department <strong>of</strong> Human <strong>Genetics</strong>, Institute for Genetic and Metabolic<br />

Disorders, Center for Neuroscience, Radboud University Nijmegen<br />

Medical Center 3. Department <strong>of</strong> Psychiatry, Donders Institute for<br />

Brain, Cognition and Behavior, Center for Neuroscience, Radboud<br />

University Nijmegen Medical Center 4. Donders Institute for Brain,<br />

Cognition and Behavior, Center for Cognitive Neuroimaging,<br />

Radboud University Nijmegen 5. Department <strong>of</strong> Neurology, Donders<br />

Institute for Brain, Cognition and Behavior, Center for Neuroscience,<br />

Radboud University Nijmegen Medical Center<br />

*russog@cf.ac.uk<br />

Introduction: Whole and regional brain volumes and psychiatric<br />

phenotypes have shared familial/genetic factors. The present study<br />

investigates the relationship between common genetic risk factors for<br />

schizophrenia and brain volumes based on a) the data from the<br />

Schizophrenia <strong>Psychiatric</strong> Genome-Wide Association Study (GWAS)<br />

Consortium (PGC) and b) the Brain Genetic Imaging (BIG) sample<br />

<strong>of</strong> 926 healthy individuals for whom the brain volumes <strong>of</strong> 11 brain<br />

regions have been measured via Magnetic Resonance Imaging<br />

(MRI). Our aim was to test the hypothesis that single nucleotide<br />

polymorphisms (SNPs) associated with the risk <strong>of</strong> SZ are also related<br />

to the variance <strong>of</strong> the volumes <strong>of</strong> brain structures in healthy<br />

individuals, and that the risk variants might consistently implicate the<br />

same brain structure.<br />

Methodology: We investigated the 10 independent, genome-wide<br />

significant SNPs identified by the PGC. A correlation analysis based<br />

on singular value decomposition <strong>of</strong> the matrix <strong>of</strong> the 11 brain<br />

measures was performed to appropriately correct for multiple testing.<br />

With this method, we retrieved 3 independent measures. A polygenic<br />

score analysis was also performed to evaluate the cumulative effect<br />

<strong>of</strong> large collections <strong>of</strong> SNPs enriched for risk alleles. For each brain<br />

volume, alleles were selected from the BIG GWAS that surpassed a<br />

range <strong>of</strong> arbitrary association thresholds (0.5, 0.1, 0.01, and 0.001)<br />

and these were used to try to predict SZ state in a subset <strong>of</strong> the PGC<br />

dataset.<br />

Results: With 10 independent SNPs, there are 30 independent tests,<br />

and an effective significance threshold <strong>of</strong> P = 0.0017. Before multiple<br />

testing correction, four SNPs were nominally significantly associated<br />

(p < 0.05) with at least one brain region, there being a total <strong>of</strong> 10<br />

nominally significant findings. The most significant associations<br />

were to rs11191580 at chromosome 10 which was associated with<br />

reduced volume for 5 brain regions. Three <strong>of</strong> these signals remain<br />

significant after correction for multiple testing (accumbens, P =<br />

0.00076 globuspallidus, P = 0.00060 hippocampus, P = 0.0010). SNP<br />

rs11191580 maps to NT5C2 (5'- nucleotidase, cytosolic II), but the<br />

signal is not precisely located to this gene as a result <strong>of</strong> extensive LD<br />

in the region. None <strong>of</strong> the other associations survived correction for<br />

multiple testing. In the polygenic analyses, no prediction was<br />

statistically significant after multiple testing correction, for which<br />

permutations and singular value decomposition gave very similar<br />

results.<br />

Conclusions: Our study is consistent with the hypothesis that the<br />

association between schizophrenia and rs11191580 may be mediated<br />

through effects on brain development that are visible at the structural<br />

level, although pleiotropy may be another explanation. We did not<br />

find evidence that in general, the polygenic risk for schizophrenia is<br />

shared with genetic variance in brain volumes. The size <strong>of</strong> our sample<br />

might well be underpowered to detect such common genetic factors.<br />

It is also possible that volumes as measured in this study are not the<br />

best endophenotypes for the disorder, and that more consistent<br />

evidence for shared genetic risk between schizophrenia and brain<br />

structure will require more sophisticated analyses, for example<br />

through hypothesis free voxel based morphometry, studies which are<br />

currently under way.


PP208 FAMILIAL CLUSTERING OF SCHIZOPHRENIA,<br />

BIPOLAR DISORDER AND MAJOR DEPRESSIVE<br />

DISORDER<br />

M. Aukes*(1,2), W. Laan(2), J. Buizer-Voskamp(1,3), F.<br />

Termorshuizen(2), E. Hennekam(4), H. Smeets(2), R. Oph<strong>of</strong>f(1,3,5),<br />

M. Boks(1,2), R. Kahn(1)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience, Department <strong>of</strong><br />

Psychiatry, University Medical Center Utrecht 2. Julius Center for<br />

Health Sciences and Primary Care, University Medical Center<br />

Utrecht 3. Department <strong>of</strong> Medical <strong>Genetics</strong>, University Medical<br />

Center Utrecht 4. Department <strong>of</strong> Clinical <strong>Genetics</strong>, University<br />

Medical Center Utrecht 5. Center for Neurobehavioral <strong>Genetics</strong>,<br />

University <strong>of</strong> California<br />

*m.aukes@umcutrecht.nl<br />

Introduction: Recent population-based studies suggest that<br />

schizophrenia and bipolar disorder share part <strong>of</strong> their genetic<br />

vulnerability. Several molecular genetic studies have indeed<br />

identified both shared and distinct genetic variants that are associated<br />

with these disorders. Others suggest that a wide range <strong>of</strong> mental<br />

disorders cluster in families. We investigated familial clustering <strong>of</strong><br />

schizophrenia, bipolar disorder, and major depressive disorder.<br />

Methodology: By combining data from the <strong>Psychiatric</strong> Case Register<br />

Middle Netherlands and Statistics Netherlands we obtained<br />

information on 4,673 affected probands and 18,692 matched<br />

population controls. We calculated the relative risks (RRs) for having<br />

an affected sibling using the population matched group <strong>of</strong> controls as<br />

a reference group.<br />

Results: Probands with schizophrenia had relative risks for having a<br />

sibling with schizophrenia <strong>of</strong> 3.50 (95% CI: 2.42–5.06), with bipolar<br />

disorder <strong>of</strong> 1.46 (0.53-4.03) and with major depressive disorder <strong>of</strong><br />

(3.05, 2.47-3.77) compared to a reference proband. Probands affected<br />

with bipolar disorder have a RR <strong>of</strong> 5.33 (2.15-13.22) for having a<br />

sibling with bipolar disorder and <strong>of</strong> 1.59 (0.66-3.84) for having a<br />

sibling with schizophrenia compared to a reference proband.<br />

Probands affected with major depressive disorder also have increased<br />

risk for having a sibling with schizophrenia (RR: 2.05, 1.56-2.69)<br />

compared to a reference proband, which was similar to the risk for<br />

having a sib with major depressive disorder (RR: 1.78, 1.52-2.09) or<br />

bipolar disorder (RR: 1.89, 1.12-3.17).<br />

Conclusions: Overall our findings are consistent with other large<br />

population-based studies, in that they show the same decrease (about<br />

tw<strong>of</strong>old) for cross-wise schizophrenia-bipolar disorder risk compared<br />

to within diagnoses risk. This reiterates the fact that although there<br />

may be a shared familial vulnerabilitybetween schizophrenia and<br />

bipolar disorder, risk is considerably lower across diagnostic entities.<br />

However, for major depressive disorder risk was similar within and<br />

across diagnostic entities. These data as well as previous reports are<br />

consistent with a model in which there are underlying genetic or<br />

environmental factors that give rise to broad, as well as specific,<br />

familial vulnerabilities to psychiatric disorders.<br />

176<br />

PP209 T(9;17) (Q33.2;Q25.3) TRANSLOCATION REVEALS<br />

THE ASSOCIATION BETWEEN BIPOLAR DISORDER AND<br />

RNF213 AS WELL AS RPTOR GENES<br />

ECIP<br />

A. Rajkumar*(1,2), J. Christensen(1), I. Jacobsen(1), J. Pallesen(1),<br />

D. Demontis(1), J. Grove(1), Z. Tümer(3), N. Tommerup(3), A.<br />

McQuillin(4), H. Gurling(4), O. Mors(2), A. Borglum(1,2)<br />

1. Department <strong>of</strong> Human <strong>Genetics</strong>, Aarhus University, 2. Center for<br />

<strong>Psychiatric</strong> Research, Aarhus University hospital, Risskov 3.<br />

Department <strong>of</strong> Cellular and Molecular Medicine, Copenhagen<br />

University, 4. Department <strong>of</strong> Mental Health Sciences, University<br />

College London,<br />

*antoprajkumar@yahoo.com<br />

Introduction: Rare copy number variants and other chromosomal<br />

aberrations increase the risk for major psychotic disorders. Patients<br />

with bipolar affective disorder (BD) and chromosomal aberrations<br />

provide the impetus to investigate the association between BD and<br />

the genes within those chromosomal breakpoint regions. Cross<br />

linking Danish <strong>Psychiatric</strong> Case Registry and Danish Cytogenetic<br />

Case Registry identified a patient with a t(917) (q33.2q25.3)<br />

translocation and BD as well as alcohol dependence syndrome. We<br />

aimed to identify the chromosomal breakpoint regions in that patient<br />

and to investigate the association between BD and the single<br />

nucleotide polymorphisms (SNPs) <strong>of</strong> known protein coding genes<br />

within those regions in a larger population.<br />

Methodology: We employed Fluorescent In Situ Hybridization<br />

(FISH) to narrow down the breakpoint regions in that patient. We<br />

used Ensembl and Biomart web tools to identify the genes within<br />

those regions, which code for proteins, expressed in brain. We<br />

accessed WTCCC, UCL and STEP-BD data and analyzed the allelic<br />

and genotypic associations between BD and the SNP <strong>of</strong> those genes,<br />

using Plink v1.07.<br />

Results: We narrowed down 9q33.2and 17q25.3 breakpoint regions<br />

within 112 KB (124,202,476-124,315,433) and 3l8 KB (78,267,202-<br />

78,585,422) size, respectively. We identified three protein coding<br />

genes (FLJ35220, RNF213 and RPTOR) in those regions. When we<br />

analyzed the WTCCC data for the SNP <strong>of</strong> those genes, rs8072229,<br />

rs4602089 and rs12601089 in RPTOR (Regulatory associated Protein<br />

<strong>of</strong> MTOR, complex 1) had significant allelic and genotypic<br />

associations with BD. When we analyzed WTCCC, UCL and STEP-<br />

BD data together, rs7223701 and rs8359 in RNF213 (ring finger<br />

protein 213) and rs4561525, rs11150864, rs8072229, rs4602089,<br />

rs12601089, rs2138125, rs9911574 and rs7216306 in RPTOR were<br />

significantly associated with BD (p < 0.05).<br />

Conclusions: RPTORregulates cell growth and survival in response<br />

to nutrient and hormonal signals. RPTOR and RNF213 may<br />

contribute towards the complex etiology <strong>of</strong> BD. Further studies<br />

investigating the underlying molecular pathways are desired.


PP210 KNOWN LIPID GENE VARIANTS CONSTITUTES A<br />

MOLECULAR LINK BETWEEN REGULATION OF SLEEP<br />

AND LIPID METABOLISM<br />

H. Ollila*(1), S. Utge(1), E. Kronholm(2), T. Paunio(1,3)<br />

1. Public Health Genomics Unit and Institute for Molecular Medicine<br />

FIMM 2. Department <strong>of</strong> Chronic Disease Prevention, Population<br />

Studies Unit, National Institute for Health and Welfare 3. Department<br />

<strong>of</strong> Psychiatry, University <strong>of</strong> Helsinki<br />

*hanna.m.ollila@helsinki.fi<br />

Introduction: Epidemiological studies suggest a relationship<br />

between lipid metabolism and regulation <strong>of</strong> sleep. In addition,<br />

inactivation <strong>of</strong> circadian genes induces insulin resistance and<br />

hyperlipidemia. We hypothesized that regulation <strong>of</strong> sleep length and<br />

lipid metabolism is partially conducted by the same genes and aimed<br />

to identify such genes in humans.<br />

Methodology: We studied the association <strong>of</strong> total sleep time (TST)<br />

with 60 genetic variants that had been previously associated with<br />

lipid traits. The analyses were performed in a Finnish population<br />

based sample comprising 6334 participants and replicated in 2189<br />

twins. Finally, RNA expression from mononuclear leucocytes was<br />

measured in 10 healthy volunteers before and after partial sleep<br />

restriction (4 h sleep per night for 5 days).<br />

Results: TST explained a small proportion <strong>of</strong> the variation in the<br />

lipid traits. The genetic analysis identified variants near that<br />

independently contributed to both blood lipid levels and to TST. The<br />

strongest finding was replicated in twins. Meta-analysis further<br />

strengthened the association and the association was not lost when<br />

adjusted for lipid levels. RNA expression levels <strong>of</strong> the studied genes<br />

were changed after the sleep restriction period in healthy individuals.<br />

Conclusions: The results further support the connection between<br />

sleep and lipid metabolism on genetic level. The shared genetic<br />

background <strong>of</strong> these phenomena may at least partially explain the<br />

well-established connection between diseases involving disrupted<br />

metabolic processes such as cardiovascular disease, obesity and type<br />

2-diabetes and sleep.<br />

177<br />

PP211 ASSOCIATION STUDY OF VESICULAR<br />

MONOAMINE TRANSPORTER 1 (VMAT1) GENE WITH<br />

BIPOLAR DISORDER IN A KOREAN POPULATION<br />

Y. Kim *(1,2,3), J. Song(1,2), S. Kim (1,2), Y. Ahn (1,2,3)<br />

1. Department <strong>of</strong> Psychiatry and Behavioral Science, Seoul National<br />

University College <strong>of</strong> Medicine 2. Department <strong>of</strong> Neuropsychiatry,<br />

Seoul National University Hospital 3. Institute <strong>of</strong> Human Behavioral<br />

Medicine, Seoul National University College <strong>of</strong> Medicine<br />

*kys@snu.ac.kr<br />

Introduction: Vesicular monoamine transporters (VMATs) are<br />

responsible for the uptake <strong>of</strong> cytosolic monoamines into synaptic<br />

vesicles in monoaminergic neurons. Variations in the VMAT1 gene<br />

might affect transporter function and/or expression <strong>of</strong> vesicular<br />

monoamine transporters and might be involved in the etiology <strong>of</strong><br />

psychiatric disorders. The vesicular monoamine transporter 1 gene<br />

(VMAT1/SLC18A1) maps to the shared bipolar<br />

disorder/schizophrenia susceptibility locus on chromosome 8p21.<br />

Methodology: We investigated the association <strong>of</strong> the SLC18A1,<br />

which were suggested as potential susceptibility genes for bipolar<br />

disorder. Along with 350 healthy individuals, 352 bipolar patients<br />

were analyzed.<br />

Results: All <strong>of</strong> the five examined single-nucleotide polymorphisms<br />

(SNPs) <strong>of</strong> SLC18A1 (rs988713, rs2270641, rs2270637, rs1390938,<br />

rs17092104) showed no significant association with bipolar disorder,<br />

although rs988713 revealed a significant genotypic association with<br />

bipolar disorder. However, the ‘‘C-G’’ haplotype <strong>of</strong> rs1390938 and<br />

rs17092104 was overrrepresented in the patients with bipolar disorder<br />

(P=0.019, OR=1.287), while the “G-G” haplotype was<br />

underrepresented (P=0.03, OR=0.745).<br />

Conclusions: These results support a possible role <strong>of</strong> the SLC18A1 in<br />

bipolar disorder in Korean population. Further studies with larger<br />

samples and phenotypes are necessary.


PP212 PORTRAIT OF SCHIZOPHRENIA IN PSYCHIATRIC<br />

PATIENTS IN THE CITY OF MANAUS<br />

A. Britto Junior*(1), H. Telles Ribeiro(2), L. Fajardo(3), A. Lima de<br />

Souza(4)<br />

1. Amazonas State University, School <strong>of</strong> Health Science 2.<br />

Amazonas State University, Lab. <strong>of</strong> Genomics and Proteomics 3.<br />

Amazonas State University, Eduardo Ribeiro <strong>Psychiatric</strong> Hospital 4.<br />

Amazonas State University, Lab. <strong>of</strong> Human <strong>Genetics</strong><br />

*ademarbritto@gmail.com<br />

Introduction: Schizophrenia (ESQ) is an intriguing psychiatric<br />

illness, which has a prevalence <strong>of</strong> about 1% and generally affects<br />

people during the height <strong>of</strong> their production processes.The ESQ, as<br />

well as other complex diseases, seems to be caused by a number <strong>of</strong><br />

factors, including environmental and multifactorial inheritance.<br />

According to the multifactor model, there are several genes involved<br />

in this disorder that may act additively and suffer environmental<br />

influences, increasing susceptibility to this disease and/or worsening<br />

the patient's condition is a threshold genetic influenced by the<br />

environment.<br />

Methodology: Our research tryed to overview Manaus, the largest<br />

city in northern Brazil, with approximately 2 million habitants. This<br />

research is the first held in the Amazon that aims to study the genetic<br />

patterns <strong>of</strong> this disease. Investigations were carried out in cases <strong>of</strong><br />

ESQ in the family <strong>of</strong> 17 diagnosed patients from the Eduardo Ribeiro<br />

<strong>Psychiatric</strong> Center, through interviews with them, relatives or<br />

guardians, which served as information for the pedigrees construction<br />

for each family <strong>of</strong> the patients who agreed to participate.<br />

Results: From the data analysis we made 17 families pedigrees,<br />

containing 3-5 generations each, with 15-47 individuals in a total<br />

universe <strong>of</strong> 463 people, 38 (23 men and 15 women) <strong>of</strong> those<br />

diagnosed with schizophrenia.<br />

Conclusions: The risk <strong>of</strong> developing schizophrenia was increased in<br />

relatives <strong>of</strong> probands, and more pronounced for 1st degree and lower<br />

in the 2nd. Depression appears as one <strong>of</strong> the most frequent phenotype<br />

in affected and their relatives in Manaus, followed by bipolar<br />

disorder.<br />

178<br />

PP213 MAPPING RECESSIVE RISK VARIANTS FOR<br />

SCHIZOPHRENIA IN AN INBRED POPULATION<br />

H. Mansour*(1,2), J. Wood(1), K. Chowdari(1), A. Watson(1), W.<br />

Fathi(2), M. Elassy(2), I. Ali(2), A. Eissa(2), A. Yassin(2), H.<br />

Salah(2), S. Tobar(2), H. Elsayed(2), W. El-Bahaei(2), Z. Gomaa(2),<br />

V. Nimgaonkar(1,3)<br />

1. Department <strong>of</strong> Psychiatry, University <strong>of</strong> Pittsburgh School <strong>of</strong><br />

Medicine, Western <strong>Psychiatric</strong> Institute and Clinic, Pittsburgh,<br />

Pennsylvania 2. Department <strong>of</strong> Psychiatry, Mansoura University<br />

School <strong>of</strong> Medicine, Mansoura, Egypt 3. Department <strong>of</strong> Human<br />

<strong>Genetics</strong>, Graduate School <strong>of</strong> Public Health, University <strong>of</strong> Pittsburgh,<br />

Pittsburgh, Pennsylvania<br />

*mansourha@upmc.edu<br />

Introduction: Mapping rare genetic variants in outbred populations<br />

requires very large samples, but may be more facile in inbred<br />

populations by genotyping a handful <strong>of</strong> patients using homozygosity<br />

mapping (HM). Ancestral chromosomal segments flanking disease<br />

mutations are passed down identical by descent, (IBD). For recessive<br />

diseases, the IBD segments are by definition homozygous by descent<br />

(HBD). We are evaluating the utility <strong>of</strong> HM for Schizophrenia (SZ),<br />

because our recent studies have shown that SZ is associated with<br />

consanguinity in Egypt (Mansour et al, 2010).<br />

Methodology: Egyptian patients with SZ (n=93, DSM-IV) and adult<br />

controls (n=87) were recruited and genotyped using the Affymetrix<br />

6.0 SNP array. Following quality control procedures, we estimated<br />

homozygous segments using PLINK s<strong>of</strong>tware, to be confirmed using<br />

Beagle s<strong>of</strong>tware.<br />

Results: Consistent with published data, consanguineous individuals<br />

are more likely to have longer homozygous segments the mean<br />

homozygous segment length is greater among consanguineous<br />

patients compared with either control groups or non-consanguineous<br />

patients (Figure 1). Several homozygous segments are more frequent<br />

among cases. As we refine our analyses, we will be guided by (i)<br />

genomic regions identified through recent genome-wide association<br />

studies (ii) genomic regions implicated in autism spectrum disorders<br />

(since some risk variants are shared between autism and SZ).<br />

Conclusions: Consanguineous SZ cases are more likely to have<br />

longer homozygous segments. Our analysis showed an HBD<br />

segment on 15q region that shows intriguing case-control<br />

differences. HBD analysis may help detect recessively inherited<br />

chromosomal regions in SZ.


PP214 CANDIDATE GENES FOR AGGRESSION AND<br />

ANTISOCIAL BEHAVIOR: A META-ANALYSIS OF THE 5-<br />

HTTLPR AND MAOA-UVNTR<br />

C. Ficks*, I. Waldman<br />

Emory University<br />

*courtney.ficks@gmail.com<br />

Introduction: Antisocial behavior (behavioral tendencies that reflect<br />

disregard for social norms and the rights <strong>of</strong> others) is exhibited in<br />

elevated levels in clinical populations and associated with negative<br />

outcomes, such as violent crime, alcoholism, and drug use. Genetic<br />

influences have been estimated to explain approximately 50% <strong>of</strong> the<br />

variance in antisocial phenotypes. Variation within the serotonergic<br />

system has been associated with aggression in animals and humans,<br />

leading to hypotheses that individual differences within serotonergic<br />

system genes may underlie antisocial phenotypes. Nonetheless,<br />

candidate gene studies examining the main effects <strong>of</strong> common<br />

variation within serotonergic system genes have yielded small,<br />

inconsistent findings thus far.<br />

Methodology: We conducted a meta-analysis <strong>of</strong> associations<br />

between antisocial behavior and the two most commonly examined<br />

serotonergic system variants: the serotonin transporter-linked<br />

polymorphic region (5-HTTLPR) <strong>of</strong> the serotonin transporter gene<br />

and a 30 bp VNTR within the promoter <strong>of</strong> the monoamine oxidase A<br />

gene (MAOA-uVNTR). Specifically, we examined: 1) whether the<br />

putative "risk” alleles <strong>of</strong> each marker had significant main effects on<br />

antisocial phenotypes across studies, 2) whether there was significant<br />

heterogeneity in study effect sizes, and 3) whether demographic or<br />

methodological differences contributed to the observed heterogeneity<br />

across studies.<br />

Results: We found small but significant main effects for the 5-<br />

HTTLPR (OR = 1.389, p < 0.001, 95% CI [1.179 - 1.637]), but not<br />

the MAOA-uVNTR (OR = 1.063, p = 0.433, 95% CI [0.913 – 1.237]).<br />

In addition, there was significant heterogeneity in effect sizes for<br />

both markers. Regression analyses suggested that this heterogeneity<br />

could be partially explained by sample age, sex and ethnic<br />

composition.<br />

Conclusions: Overall, it appears that the most commonly examined<br />

serotonergic variants contribute only a small portion <strong>of</strong> the variance<br />

in antisocial phenotypes, and it is thus important that future studies<br />

focus on additional common and rare variants within these and other<br />

genes known to affect serotonergic function.<br />

179<br />

PP215 GENDER SPECIFIC ASSOCIATION OF<br />

TSNAX/DISC1 LOCUS FOR SCHIZOPHRENIA AND<br />

BIPOLAR AFFECTIVE DISORDER IN SOUTH INDIAN<br />

POPULATION<br />

R. Anjanappa (1), M. Purushottam(1), K. Halagur BhogeGowda (1),<br />

V. Manduva (1), N. Krishna(1), S. Kallahalli Jayramu (1), J.<br />

Yemmiganur Chandrash(1), S. Ghosh(2), S. Jain*(1)<br />

1. Molecular <strong>Genetics</strong> Laboratory, Department <strong>of</strong> Psychiatry,<br />

National Institute <strong>of</strong> Mental Health and Neurosciences 2. Human<br />

<strong>Genetics</strong> Unit, Indian Statistical Institute (ISI), 203 B.T. Road,<br />

*sjain.nimhans@gmail.com<br />

Introduction: Schizophrenia (SCZ) and bipolar affective disorder<br />

(BPAD) are severe psychiatric disorders, each affecting<br />

approximately 1% <strong>of</strong> the population worldwide. Family, twin and<br />

adoption studies confirm that genetic factors play a significant role in<br />

the etiology <strong>of</strong> these disorders. Genetic studies have implicated the<br />

TSNAX/DISC1 in SCZ, BPAD and major depression. This study<br />

was performed to assess the possible involvement <strong>of</strong> TSNAX/DISC1<br />

locus in the etiology <strong>of</strong> BPAD and SCZ in the southern Indian<br />

population.<br />

Methodology: We genotyped seven SNPs from TSNAX/DISC1<br />

region in 1252 individuals (419 BPAD patients, 408 SCZ patients<br />

and 425 controls). Association between disease and genotype/allele<br />

frequencies was tested by logistic regression analysis and Chi-square<br />

test.<br />

Results: Logistic regression analysis did not show significance with<br />

the SNPs studied for the gender combined samples. However,<br />

significant allelic association was observed with BPAD as well as the<br />

pooled phenotype <strong>of</strong> BPAD or SCZ within the female subjects for the<br />

rs766288. Two locus analysis <strong>of</strong> rs766288 and rs821616 showed C-A<br />

as a risk combination, whereas A-A and A-T appeared as a protective<br />

combination in female cases compared to female<br />

controls. Significant genotypic association was also seen in BPAD<br />

females, SCZ males and in pooled male cases for rs766288. No<br />

positive association was detected for other SNPs studied.<br />

Conclusions: Our results provide further evidence for sex-dependent<br />

effects <strong>of</strong> the TSNAX/DISC1 locus in the etiology <strong>of</strong> SCZ and<br />

BPAD in our sample set.


PP216 EXPLORING GENOTYPE-PHENOTYPE<br />

RELATIONSHIPS IN PSYCHIATRIC DISORDERS USING<br />

LATENT SEMANTIC ANALYSIS<br />

R. Breuer*(1), M. Schubert(2), J. Frank(1), M. Mattheisen (3,4,8), F.<br />

Degenhardt(4,9), T. Mühleisen(4,9), T. Consortium(5), S.<br />

Cichon(4,6,9), M. Nöthen(4,9), T. Schulze(7), M. Rietschel(1)<br />

1. Department <strong>of</strong> Genetic Epidemiology in Psychiatry, Central<br />

Institute <strong>of</strong> Mental Health (ZI) Mannheim, University <strong>of</strong> Heidelberg<br />

2. Database Group, Institute for Informatics, Ludwig-Maximilians-<br />

University 3. Department <strong>of</strong> Biostatistics, Harvard School <strong>of</strong> Public<br />

Health 4. Department <strong>of</strong> Genomics, Life & Brain Center, University<br />

<strong>of</strong> Bonn 5. Department <strong>of</strong> Psychiatry, University <strong>of</strong> California 6.<br />

Institute <strong>of</strong> Neuroscience and Medicine (INM-1), Research Centre<br />

Juelich 7. Department <strong>of</strong> Psychiatry and Psychotherapy, University<br />

Medical Center, Georg-August-University 8. Institut <strong>of</strong> Medical<br />

Biometry, Informatics, and Epidemiology, University <strong>of</strong> Bonn 9.<br />

Institute for Human <strong>Genetics</strong>, University <strong>of</strong> Bonn<br />

*rene.breuer@zi-mannheim.de<br />

Introduction: Schizophrenia, bipolar disorder, and major depression<br />

are severe, complex psychiatric disorders with a life-time prevalence<br />

varying between 0.5-1% for schizophrenia and bipolar disorder up to<br />

about 15% for major depression. Heritability estimates range between<br />

50-80% for schizophrenia and bipolar disorder and 30-75% for major<br />

depression. Genome-wide association studies (GWAS) for these three<br />

disorders have so far identified only a few genome-wide significant<br />

associations with small effects. Recently it was shown for human<br />

height that when taking the information content <strong>of</strong> all tested SNPs<br />

into consideration they can explain roughly half <strong>of</strong> the reported<br />

heritability. Nevertheless, the question which specific genotype<br />

combinations relate to specific phenotype clusters remains a<br />

challenging and competitive task which requires further approaches.<br />

Methodology: Here, we introduce latent semantic analysis, originally<br />

applied to text mining (Deerwester et al., 1990), as a novel approach<br />

to analyse genome-wide data with respect to various phenotypic<br />

traits. The main principle <strong>of</strong> this approach is the usage <strong>of</strong> implicit<br />

higher-order structures in the association <strong>of</strong> genetic factors with<br />

phenotypic traits. The particular technique used is singular-value<br />

decomposition, which decomposes genotypes and phenotypes into a<br />

set <strong>of</strong> meaningful orthogonal factors. The decomposition step is<br />

further used to remove noisy implicit structures. In a train-test-set<br />

framework, we explore the ability <strong>of</strong> this approach to predict the<br />

presence <strong>of</strong> phenotypic traits in patients with bipolar disorder based<br />

on genotype data.<br />

Results: Latest results from the application to over 3000 patients out<br />

<strong>of</strong> 3 independent bipolar datasets will be presented: the Genetic<br />

Association Information Network (GAIN) sample, the Translational<br />

Genomics Research Institute (TGEN) sample, and our own German<br />

(BoMa) sample.<br />

Conclusions: An adequate accuracy <strong>of</strong> this approach should lead to<br />

genotype-phenotype clusters allowing the identification <strong>of</strong> genetically<br />

more homogeneous subgroups <strong>of</strong> patients.<br />

180<br />

PP217 GENOME-WIDE SIGNIFICANT INTERACTION BY<br />

CYTOMEGALOVIRUS WITH RESPECT TO RISK OF<br />

SCHIZOPHRENIA<br />

J. Grove*(1,2), D. Demontis(1), M. Hollegaard(3), C. Pedersen(4), T.<br />

Ornt<strong>of</strong>t(5,6), R. Yolken(7), M. Didriksen(8), D. Hougaard(3), C. Wiuf(2), O.<br />

Mors(9), P. Mortensen(4), A. Borglum(1,9)<br />

1. Inst. <strong>of</strong> Biomedicine, Depts. <strong>of</strong> Human <strong>Genetics</strong>, Aarhus University 2.<br />

Bioinformatics Research Centre, Aarhus University 3. Dept. <strong>of</strong> Clinical<br />

Biochemistry and Immunology, Statens Serum Institut 4. National Centre for<br />

Register-based Research, Aarhus University 5. Dept. <strong>of</strong> Molecular Medicine,<br />

Aarhus University Hospital 6. AROS Applied Biotechnology A/S 7. The<br />

Johns Hopkins University School <strong>of</strong> Medicine, Stanley Division <strong>of</strong><br />

Developmental Neurovirology 8. Synaptic Transmission, H. Lundbeck A/S 9.<br />

Centre for <strong>Psychiatric</strong> Research, Aarhus University Hospital<br />

*grove@humgen.au.dk<br />

Introduction: Reports <strong>of</strong> viral contribution to the aetiology <strong>of</strong> schizophrenia<br />

(SZ) goes back as far as when psychoses with schizophrenia-like symptoms<br />

were linked to the 1918 influenza pandemic. More recently there have been<br />

many well documented reports <strong>of</strong> the association <strong>of</strong> other viruses to SZ.<br />

Infection by cytomegalovirus (CMV) which is <strong>of</strong> the herpes family, is the<br />

most frequent congenital infection, and has been implicated in a number <strong>of</strong><br />

papers for playing a role in the aetiology <strong>of</strong> schizophrenia (SZ). So far little is<br />

known as to what role CMV may have in the causal pathways leading to SZ.<br />

We carried out a genome-wide environment interaction study with the goal <strong>of</strong><br />

detecting interaction with respect to SZ between maternal CMV infection and<br />

the individual SNPs in a GWAS.<br />

Methodology: The study was conducted on a population based, matched<br />

case-control sample. Cases consist <strong>of</strong> all Danish citizens born from May 1981<br />

who has a SZ (ICD-10 F20) in the Danish <strong>Psychiatric</strong> Central Registry as <strong>of</strong><br />

May 2007. Controls were matched 1-1 to cases by gender and birth date and<br />

selected among those who were not <strong>of</strong> the same mother as the proband,<br />

resident in Denmark, alive and without a SZ diagnosis at the time the proband<br />

got its first diagnosis and became case. Neonatal dried blood spot samples<br />

were obtained from the Danish Newborn Screening Biobank. DNA was<br />

extracted, whole-genome-amplified and subsequently genotyped on the<br />

Illumina Infinium HD Human610-Quad BeadChip. In the quality control<br />

subjects with call rates below 0.97 were excluded, as were SNPs with call rate<br />


PP218 ON 2X2 AND 2X3 CONTINGENCY TABLES CHI-<br />

SQUARE TESTS FOR RARE VARIANTS<br />

F. Aliev*<br />

Virginia Commonwealth University<br />

*faliev@vcu.edu<br />

Introduction: Genome-wide association studies (GWASs) have<br />

successfully identified thousands <strong>of</strong> common genetic variants, mainly<br />

common single nucleotide polymorphisms (SNPs), associated with<br />

complex traits, including many common diseases. The power <strong>of</strong><br />

association analyses depend on minor allele frequencies (MAF) <strong>of</strong><br />

SNPs. For low MAF power <strong>of</strong> the association test also depends on<br />

contingency table’s approximation with Chi-square distribution. For<br />

small sample sizes this divergence can be handled by using Fisher’s<br />

exact test. For big sample sizes use <strong>of</strong> Fisher’s exact test is<br />

complicated because <strong>of</strong> big factorials in the test statistics. In this<br />

study we estimated difference between Chi-square statistics and exact<br />

distribution <strong>of</strong> the contingency table.<br />

Methodology: We used approximation theory to derive the test<br />

statstics asymptoticv distribution and to find the inflation term from<br />

Chi-square distribution. These estimates allowed us to re-scale Chisquare<br />

distribution and find exact p-values to avoid inflated results.<br />

Then we generated and tested different case-control samples with<br />

MAF’s changing from 0.5% to 5% with same and different case and<br />

control sample sizes.<br />

Results: Theoretical and simulations results show that when minor<br />

allele counts are small in the population there is a shift in asymptotic<br />

Chi-square statistics which results in inflated p-values. The length <strong>of</strong><br />

the shift depends on minor allele counts and as a result p-values <strong>of</strong><br />

the associations look smaller than exact p-values.<br />

Conclusions: Our study shows that correction for small MAF has to<br />

be made to Chi-squared statistics to get more precisious p-values.<br />

181<br />

PP219 NEUREGULIN AND DOPAMINERGIC GENES: RISK<br />

FACTORS FOR SCHIZOPHRENIA IN MALAYSIAN<br />

POPULATION<br />

S. Tee*, P. Tang, H. Loh<br />

Universiti Tunku Abdul Rahman<br />

*teesf@utar.edu.my<br />

Introduction: Schizophrenia is a complex psychotic disorder with a<br />

lifetime prevalence <strong>of</strong> approximately 1%. Susceptibility <strong>of</strong> some<br />

genes at many genetic locations with small penetrance is suggestive<br />

<strong>of</strong> thismultigenicdisorder. Molecular components <strong>of</strong> the neuregulin<br />

genes and dopaminergic systems may play an important role in the<br />

pathophysiology <strong>of</strong> SCZ. Dopamine D3 receptors (DRD3)<br />

concentrated in limbic regions <strong>of</strong> the brain may be particularly<br />

relevant to SCZ. The enzyme catechol-O-methyltransferase (COMT)<br />

is crucial for the inactivation <strong>of</strong> prefrontal dopamine. Besides, the<br />

role <strong>of</strong> the neuregulin system has been implicated in the etiology <strong>of</strong><br />

SCZ. Substantial studies have placed the neuregulin genes (NRG1),<br />

DRD3, COMT and in the limelight as candidategenes for human<br />

psychotic disorders.<br />

Methodology: In this study, we assessed the associations between<br />

SCZ and single nucleotide polymorphisms (SNPs) <strong>of</strong> NRG1, Ser9Gly<br />

DRD3, COMT rs4680 and with Malaysia cases-control. A total <strong>of</strong><br />

267 schizophrenic patients and 250 healthy controls were recruited.<br />

Polymerase chain reaction-restriction fragment length polymorphism<br />

(PCR-RFLP) was performed to genotype these three SNPs and<br />

analyzed with HWE and Chi-square test.<br />

Results: Our results suggest that the polymorphisms at theNRG1,<br />

DRD3 and COMT rs4680 genes are susceptible to SCZ. Individual<br />

SNP analysisshowed that bothgenotype and allele frequencies <strong>of</strong>these<br />

three SNPs were significantly (p < 0.05) associated with SCZ in the<br />

Malaysian population.<br />

Conclusions: Further studies are needed to validate the role <strong>of</strong> these<br />

polymorphismsalthough positive associations among NRG1, Ser9Gly<br />

DRD3 and COMT rs4680 and polymorphisms were found in the<br />

Malaysian population.


FUNCTIONAL GENOMICS & MODEL<br />

ORGANISMS<br />

PP220 NO ASSOCIATION OF COMT VAL158MET<br />

POLYMORPHISM WITH SUICIDAL BEHAVIOR: META-<br />

ANALYSIS AND NEW DATA<br />

S. Pool García*(1), I. Juárez Rojp(2), T. Frias-Ramón(2), D.<br />

Bermudez Ocaña(2), A. Jimenez Santos(2), M. Villar Soto(3), M.<br />

Sanchez(1), B. Camarena Medellin(4), A. Genis(5), N. Humberto(5),<br />

C. Tovilla-Zarate(2)<br />

1. Hospital General de Comalcalco, Tabasco. Secretaria de Salud 2.<br />

Universidad Juárez Autónoma de Tabasco, División<br />

Multidisciplinaria de Comalcalco 3. Hospital de alta especialidad<br />

“Gustavo A. Rovirosa P" 4. Departamento de Genética Psiquiátrica,<br />

Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz” 5.<br />

Grupo de estudios medicos y familiar Carracci<br />

*shepoga70@hotmail.com<br />

Introduction: The polymorphism COMTval158met has been<br />

associated with suicidal behavior in cases-control and meta-analysis<br />

study but results and conclusions remain controversial. The objective<br />

<strong>of</strong> this study we examined the association between COMT val158met<br />

and suicide behavior in a cases-control and for assess the combined<br />

evidence we realized a meta-analysis study<br />

Methodology: We conducted a cases-control study with 105 patients<br />

with suicide attempter and 236 controls. Subsequently we performed<br />

a meta-analysis <strong>of</strong> published genetic association by searching through<br />

Medline, PubMed and Wev <strong>of</strong> Science databases<br />

Results: No significant differences were found in the distribution <strong>of</strong><br />

alleles (χ2=0.33, 1 df, p=0.56), or genotypes (χ2=2.36, 2 df, p=0.26).<br />

The meta-analysis comprising 12 association studies (including the<br />

present one) showed that the risk allele COMTmet <strong>of</strong><br />

COMTval158/met is not associated with suicidal behavior (OR: 1.09,<br />

95% CI: 0.97-1.23) in the absence <strong>of</strong> heterogeneity (OR: 1.09, 95%<br />

CI: 0.97-1.23)<br />

Conclusions: Our results showed no association between<br />

COMTval158/met and suicidal behavior. However, more studies are<br />

necessary to determine an association between COMT and suicidal<br />

behavior<br />

182<br />

PP221 GENOME-WIDE EXPRESSION PROFILING OF<br />

WHOLE BLOOD FROM HEAVY CANNABIS USERS AND<br />

CONTROLS REVEALS DIFFERENTIAL REGULATION OF<br />

TWO RELEVANT GENES<br />

C. Schubart*(1), S. de Jong(3), E. Janson(3), W. van Gastel(1), I.<br />

Sommer(1), R. Kahn(1), R. Oph<strong>of</strong>f(2,3), M. Boks(1)<br />

1. Rudolf Magnus Institute <strong>of</strong> Neuroscience, University Medical<br />

Centre Utrecht 2. UCLA Center for Neurobehavioral <strong>Genetics</strong> 3.<br />

Dept <strong>of</strong> Medical <strong>Genetics</strong>, University Medical Centre Utrecht<br />

*c.schubart@umcutrecht.nl<br />

Introduction: With over 165 million users worldwide, cannabis is<br />

the most frequently used illicit drug and is associated with a variety<br />

<strong>of</strong> adverse effects <strong>of</strong> which many are (neuro-) psychiatric.<br />

Particularly the association with psychotic disorders is clinically<br />

meaningful and reproduced repeatedly. However, little is known<br />

about the biological mechanisms by that underlie these associations.<br />

Gene-expression pr<strong>of</strong>iling is a valuable hypothesis generating tool to<br />

unravel the biological mechanisms underlying the adverse effects<br />

associated with cannabis use. We here report a genome wide<br />

expression pr<strong>of</strong>iling study in whole blood from heavy cannabis users<br />

and controls that reveals differential regulation <strong>of</strong> two relevant genes.<br />

Methodology: We selected 190 subjects, 90 heavy users and 100<br />

cannabis naïve subjects. All subjects were assessed using<br />

standardized structure clinical interviews (SCID, CIDI). Genomewide<br />

RNA expression pr<strong>of</strong>iling was obtained with HumanRef-12<br />

arrays using Illumina’s standard protocol. Gene expression data were<br />

transformed, normalized and filtered. Gene-expression was tested for<br />

association with cannabis status (with age, gender, smoking and other<br />

drug use as covariates) using linear model regression analysis<br />

implemented in the limma R package. FDR correction for multiple<br />

testing was applied.<br />

Results: The expression <strong>of</strong> two transcripts CX3CR1 (LogFold<br />

Change -0.42456) and PPFIA2 (LogFold Change 0.168589) was<br />

significantly associated with cannabis use.<br />

Conclusions: The chemokine receptor CX3CR1 is expressed on<br />

neurons and may play a role in neuronal survival. Two previous<br />

studies involved the CX3C receptor-ligand system in gene expression<br />

changes induced by cannabinoids in mouse brain and isolated<br />

astrocytes. PPFIA2 encodes a protein that is a member <strong>of</strong> the LAR<br />

protein-tyrosine phosphatase-interacting protein (liprin) family. In<br />

interaction with liprins, these proteins are known to moderate axon<br />

guidance. Moreover liprin proteins are thought to be involved in<br />

synapse maturation. We are not aware <strong>of</strong> previous studies that<br />

investigate genome wide gene-expression gene expression changes in<br />

whole blood associated with cannabis use in humans. The two<br />

transcripts that were found to be genome wide differentially<br />

expressed in heavy cannabis users compared to cannabis naïve<br />

individuals are plausible candidates to help us further understand the<br />

etiology <strong>of</strong> (psychiatric-) adverse effects associated with cannabis<br />

use.


PP222 ENHANCED EXPRESSION OF TRANSCRIPTION<br />

FACTOR SP1 IN POSTMORTEM BRAIN TISSUES AND<br />

ALTERED REGULATION OF SEVERAL AUTISM<br />

CANDIDATE GENES<br />

T. Ismail*(1), A. Ayyappan(2), K. Nakamura(1), S. Suda(3), H.<br />

Matsuzaki(2), N. Mori(1,2)<br />

1. Department <strong>of</strong> Psychiatry and Neurology, Hamamatsu University<br />

School <strong>of</strong> Medicine 2. Research Centre for Child Mental<br />

Development, Hamamatsu University School <strong>of</strong> Medicine 3.<br />

Department <strong>of</strong> Psychiatry, Jichi Medical University<br />

*thanseem@hama-med.ac.jp<br />

Introduction: The prevailing hypothesis suggests autism as a<br />

polygenic disease, in which, multiple genes, each with a small effect,<br />

are involved in predisposition to the development <strong>of</strong> the disorder.<br />

Thegeneral view,points totheinvolvement <strong>of</strong> epigenetic mechanisms<br />

in autism, where susceptibility genes interplay with environmental<br />

factors as well as with other modulating genes and, due to such<br />

complexity, variable clinical features are incurred. Specificity protein<br />

1 (Sp1) is a zinc finger (Cys2/Hys2) DNA-binding transcription factor<br />

that binds to and acts through the GC-box promoter elements <strong>of</strong><br />

genes. This ubiquitous transcription factor is known to regulate the<br />

expression <strong>of</strong> a variety <strong>of</strong> genes. Interestingly, Sp1 binding sites have<br />

been reported in the promoter regions <strong>of</strong> several genes, which are<br />

implicated in autism. Since pr<strong>of</strong>ound changes in gene expression can<br />

result from abnormalities in the concentrations <strong>of</strong> sequence-specific<br />

transcription factors, we hypothesize that Sp1 might be a regulator <strong>of</strong><br />

the susceptibility genes <strong>of</strong> autism abnormalities <strong>of</strong> Sp1 may account<br />

for the heterogeneity observed in this disorder.To examine this<br />

hypothesis, we investigated whether the Sp1 gene is abnormally<br />

expressed in autism brains, and consequently, can affect the<br />

expressions <strong>of</strong> potential candidate genes implicated in this disorder.<br />

Methodology: The brain samples were obtained from the Autism<br />

Tissue Program and the Harvard Brain Tissue Resource Centre. The<br />

mRNA expressions <strong>of</strong> Sp1 and that <strong>of</strong> reported autism candidate<br />

genes (with promoterbindingsites for Sp1) were analyzed in the<br />

postmortem brain tissues<strong>of</strong> autism patients (n=8)and healthy controls<br />

(n=13)with q-RTPCR using TaqMan gene expression probes.<br />

Furthermore, alterations in the autism gene expressions were studied<br />

in the SK-NH neuronal cell lines upon (i) Sp1/DNA binding<br />

inhibition using mithramycinA, and (ii) Sp1 gene silencing by RNAi.<br />

Results: We observed elevated expression <strong>of</strong> Sp1 in the anterior<br />

cingulate gyrus (ACG) <strong>of</strong> autism patients (p=0.010). We also<br />

observed altered expressions <strong>of</strong> several autism genes. GABRB3 and<br />

HTR2A showed reduced expressions, while CD38, ITGB3, MAOA,<br />

MECP2, OXTR and PTEN showed elevated expressions in autism. In<br />

SK-N-SH cells, OXTR, PTEN and RELN showed reduced expressions<br />

upon Sp1/DNA binding inhibition and Sp1 silencing.<br />

Conclusions: The results indicate that transcription factor Sp1 may<br />

bedysfunctional in autism. Anelevated expression <strong>of</strong> Sp1 in autism<br />

indicates the possible involvement <strong>of</strong> this transcription factor in the<br />

gene expression alterations underlying the pathology <strong>of</strong> this<br />

disorder.Consequently, the expressions <strong>of</strong> potential autism candidate<br />

genes, OXTR, PTEN and RELN, regulated by Sp1, could be affected.<br />

The diverse downstream pathways mediated by the Sp1-regulated<br />

genes, along with the environmental- and intracellular signal- related<br />

regulation <strong>of</strong> Sp1, could explain the complex phenotypes associated<br />

with autism.<br />

183<br />

PP223 NEUROPHYSIOLOGICAL FEATURES OF<br />

PERCEPTION OF EMOTIONAL STIMULI AND THEIR<br />

RELATIONSHIP TO THE GENETIC FACTOR IN<br />

SCHIZOPHRENIA<br />

A. Arkhipov*<br />

Institute <strong>of</strong> Higher Nervous Activity and Neurophysiology RAS<br />

*arhip76@bk.ru<br />

Introduction: It is assumed that in schizophrenia there is an<br />

infringement <strong>of</strong> the spontaneous and evoked brain activity during<br />

perception <strong>of</strong> emotional stimuli. In the psychotic period, there is a<br />

significant violation <strong>of</strong> the neural networks associated with<br />

perception and processing <strong>of</strong> emotional stimuli, but their remission is<br />

some recovery. Genetic factors influence the severity <strong>of</strong> pathological<br />

rearrangements <strong>of</strong> neural networks in schizophrenia as a psychotic<br />

period and during remission.<br />

Methodology: Methods were used:The PANSS. Registration and<br />

processing <strong>of</strong> EEG during perception <strong>of</strong> pleasant, neutral, and<br />

threatening visual stimuli.Fence venous blood for DNA extraction by<br />

phenol-chlor<strong>of</strong>orm method (Maniatis 1984). Polymerase chain<br />

reaction (PCR) to determine allelic polymorphism: DAT-1 gene<br />

dopamine transporter, EAAT-2 glutamate transporter gene, GAT-3<br />

GABA transporter gene, Val66Met gene BDNF. Statistical methods:<br />

ANOVA, Wilcoxon's test, Mann-Whitney test, correlation analysis.<br />

Results: The combination <strong>of</strong> some <strong>of</strong> the changes <strong>of</strong> allelic<br />

polymorphism <strong>of</strong> DAT-1 gene dopamine transporter, EAAT-2<br />

glutamate transporter gene, GAT-3 GABA transporter gene,<br />

Val66Met BDNF gene determinesneurophysiological disorders in<br />

schizophrenia.<br />

Conclusions: Nature <strong>of</strong> the rhythm <strong>of</strong> the brain in different ranges <strong>of</strong><br />

cortical and subcortical structures in subjects with schizophrenia in<br />

acute psychotic period, different from the remission period and differ<br />

from healthy, which is caused by genetic factors. The stronger the<br />

neurophysiological changes in the psychotic period, the less stable<br />

remission, the severity is caused by genetic factors.


PP224 MOLECULAR AND CELLULAR<br />

CHARACTERISATION OF A BRAIN-SPECIFIC<br />

TRANSCRIPTIONAL REGULATOR<br />

A. Kepa*, S. Desrivières<br />

MRC-SGDP Centre, Institute <strong>of</strong> Psychiatry, Kings College London<br />

*agnieszka.kepa@kcl.ac.uk<br />

Introduction: Abnormalities in brain development and maladaptive<br />

plasticity underlie a range <strong>of</strong> neurodevelopmental and neurological<br />

disorders. Therefore, there is a need to identify genes and pathways<br />

involved in these processes. Transcription factors (TFs) play a pivotal<br />

role in brain development by directing the formation <strong>of</strong> neurons and<br />

glia from uncommitted progenitor cells and by controlling expression<br />

<strong>of</strong> neural-specific genes. The myelin transcription factor 1- like<br />

(MYT1L) gene product, a transcription factor specifically expressed<br />

in the brain, has recently been shown to be one <strong>of</strong> three factors<br />

sufficient to transform fibroblasts into neurons, suggesting a central<br />

role for this protein neural cell function. Our recent quantitative trait<br />

loci (eQTL) analyses have revealed networks <strong>of</strong> putative downstream<br />

target genes through which MYT1L might regulate neurite<br />

development and morphogenesis. We aim at validating these findings<br />

using appropriate cellular models. We also aim at better<br />

understanding the biological function <strong>of</strong> this gene by identifying<br />

putative upstream regulators.<br />

Methodology: We will express MYT1L as native or recombinant<br />

protein (with a C-terminal V5 epitope) into human neural (SH-SY5Y,<br />

neural progenitor) and non-neural (HEK293T) cell lines. Effects <strong>of</strong><br />

enhanced MYT1L expression on mRNA levels <strong>of</strong> putative<br />

downstream targets will be measured by real-time RT-PCR. Cells<br />

expressing recombinant MYT1L will be used to identify MYT1Lcontaining<br />

protein complexes following immunoprecipitation with<br />

anti-V5 antibody, elution co-immunoprecipitating proteins and their<br />

separation <strong>of</strong> by SDS-PAGE. Identification <strong>of</strong> isolated protein bands<br />

will be performed by liquid chromatography-mass spectrometry.<br />

Results: We have constructed lentiviral expression vectors in which<br />

MYT1L is expressed either as a native protein or fused to a Cterminal<br />

V5 epitope. These viruses are currently used to transduce<br />

various lines <strong>of</strong> human cells. RNA extracts from these cells will be<br />

used for the identification downstream targets <strong>of</strong> MYT1L. Protein<br />

extracts <strong>of</strong> MYT1L-V5 expressing cells will be used for<br />

immunoprecipations. Results <strong>of</strong> these experiments will be presented.<br />

Conclusions: Identification <strong>of</strong> genes and pathways acting upstream<br />

and downstream <strong>of</strong> MYT1L will help us to understand the<br />

mechanisms by which this gene regulates neural differentiation. Such<br />

findings are expected to provide a greater understanding <strong>of</strong> normal<br />

brain development and have important implications by identifying<br />

genes and genes pathways that might be deregulated in<br />

neurodevelopmental and neurological disorders.<br />

184<br />

PP225 A NOVEL, DIET-INDUCED, MURINE MODEL OF<br />

OBSESSIVE-COMPULSIVE DISORDER<br />

D. Trujillano*, J. McDonald, M. Gratacós, M. Dierssen, X. Estivill<br />

Genes and Disease Program, Center for Genomic Regulation (CRG-<br />

UPF)<br />

*daniel.trujillano@crg.eu<br />

Introduction: The main purpose for the development <strong>of</strong> animal<br />

models for Obsessive-Compulsive Disorder (OCD) is to use them for<br />

the study <strong>of</strong> the neural mechanisms underlying this disorder and<br />

developing novel treatments. The majority <strong>of</strong> these OCD animal<br />

models involve acute, drug-induced symptom provocation or genetic<br />

alterations associated with stereotypies or anxiety. Here we present a<br />

novel, diet-induced OCD murine model that demonstrates multiple<br />

OCD-like behaviors.<br />

Methodology: Young mice were given free choice between standard<br />

chow and a palatable, chocolate diet using the PHECOMP (Panlab)<br />

food and drink monitoring system. Cases and controls were tested for<br />

multiple OCD-like behaviors, including marble burying and nestlet<br />

shredding.<br />

Results: Two weeks after the start <strong>of</strong> the experiment, treated mice<br />

developed a compulsive feeding pattern accompanied by the<br />

apparition <strong>of</strong> multiple OCD-like behaviors. This behavioral data is<br />

supported by molecular data suggesting a deregulation in the<br />

expression <strong>of</strong> various serotonin and dopamine receptors in different<br />

areas <strong>of</strong> the brain, including the striatum, the frontal cortex and the<br />

thalamus.<br />

Conclusions: This is a novel, multiple-symptom, diet-induced OCD<br />

murine model that shows behavioral changes accompanied by<br />

biochemical changes in brain regions directly involved in the<br />

pathophysiology <strong>of</strong> OCD. Moreover, this OCD model also warns us<br />

about the physical and mental consequences <strong>of</strong> the abuse <strong>of</strong> a<br />

cafeteria diet.


PP226 FUNCTIONAL STUDY OF THE<br />

ENDOCANNABINOID SYSTEM USING POSTMORTEM<br />

BRAINS AND RODENT MODELS OF FEAR AND STRESS<br />

K. Choi*(1), G. Xing(1), T. Le(1), J. McGuire(1), L. Zhang(1), H.<br />

Li(1), L. Johnson(1), M. Webster(2), D. Benedek(1), R. Ursano(1)<br />

1. Dept. Psychiatry, Uniformed Services University <strong>of</strong> the Health<br />

Sciences 2. Stanley Medical Research Inst.<br />

*kwang.choi@usuhs.mil<br />

Introduction: Posttraumatic stress disorder (PTSD) is an anxiety<br />

disorder that affects approximately 7% <strong>of</strong> the population in the US.<br />

Core symptoms <strong>of</strong> PTSD include re-experiencing,<br />

avoidance/numbing and hyper-arousal. Individuals with PTSD<br />

experience significant functional impairment such as occupational<br />

and social dysfunction, as well as physical and mental health<br />

problems. There is evidence to suggest the endocannabinoid system<br />

is involved in mood and anxiety disorders and may be an important<br />

drug target for the treatment <strong>of</strong> PTSD. Animal and human studies<br />

indicate that endocannabinoid receptors (CNR1 and CNR2) may<br />

regulate fear conditioning/extinction and peri-traumatic dissociation<br />

symptoms.<br />

Methodology: We investigated expression pr<strong>of</strong>iles <strong>of</strong> CNR1 and<br />

CNR2 in the prefrontal cortex (PFC) <strong>of</strong> humans and rodents using<br />

data from gene expression microarrays and quantitative PCR. The<br />

subjects include a developmental cohort <strong>of</strong> normal individuals<br />

ranging in age from birth to 50 years <strong>of</strong> age, a cohort <strong>of</strong> mood<br />

disorder patients and unaffected age-matched controls, rats exposed<br />

to restraint and tail-shock stress and mice selectively bred for high<br />

and low fear phenotypes.<br />

Results: We found that the expression pr<strong>of</strong>iles <strong>of</strong> endocannabinoid<br />

receptors are age-dependent and disease-specific. For example,<br />

CNR1, but not CNR2, levels gradually decrease in the PFC during<br />

postnatal brain development. CNR1 mRNA levels decrease following<br />

exposure to restraint and tail-shock stress, whereas CNR2 levels<br />

appear to increase, in the PFC <strong>of</strong> female rats. In mood disorder<br />

patients, CNR2 levels were differentially expressed between bipolar<br />

disorder and major depression. A study is in progress to measure<br />

mRNA levels <strong>of</strong> CNR1 and CNR2 in the PFC <strong>of</strong> high and low fear<br />

mice following fear conditioning and extinction <strong>of</strong> fear memory.<br />

Conclusions: Our results suggest that expression levels <strong>of</strong><br />

endocannabinoid receptors in the brain are altered following exposure<br />

to stress and anxiety. Our translational approach combines data from<br />

animal models <strong>of</strong> stress and from postmortem brains with mood<br />

disorders to enhance our understanding on the molecular mechanisms<br />

that may underlie these disorders. More research is needed to<br />

determine the functional significance <strong>of</strong> the endocannabinoid system<br />

in individuals with stress and anxiety disorders.<br />

185<br />

PP227 KNOCKDOWN OF PBRM1, A PUTATIVE RISK GENE<br />

FOR MOOD DISORDER AND SCHIZOPHRENIA, INDUCES<br />

WIDESPREAD ALTERATIONS IN GENE EXPRESSION,<br />

CELLULAR MORPHOLOGY, AND PROLIFERATION<br />

X. Jiang*, S. Detera-Wadleigh, F. McMahon<br />

NIMH<br />

*sjiang1@mail.nih.gov<br />

Introduction: A meta-analysis <strong>of</strong> genome-wide association studies<br />

(GWAS) in mood disorders found the strongest evidence <strong>of</strong><br />

association at a synonymous SNP in PBRM1 (McMahon et al<br />

2010). The same SNP has been shown to be associated with<br />

schizophrenia (Williams et al. 2011). Although several genes map to<br />

the same ~350 kb linkage disequilibrium region on chromosome<br />

3p21.1, in GWAS "the causal gene will <strong>of</strong>ten be located near the<br />

most strongly associated DNA variant"(Allen et al. 2010), so we<br />

have undertaken initial functional studies <strong>of</strong> PBRM1.<br />

Methodology: We employed a loss-<strong>of</strong>-function paradigm by<br />

generating a stable knockdown <strong>of</strong> PBRM1in both HeLa and SHSY5Y<br />

neuroblastoma cells via lentiviral-mediated transduction <strong>of</strong> short<br />

hairpin (sh) RNA. Differential gene expression pr<strong>of</strong>iles examined<br />

using the Illumina Human HT 12v3 Expression Bead Array.<br />

Results: PBRM1 knockdown generated 129 overexpressed and 70<br />

underexpressed genes.Notably, PBRM1 knockdown induced a<br />

significant downregulation <strong>of</strong> PDE4B, which encodes cAMP-specific<br />

3’5’ phosphodiesterase 4B. This protein is known to interact with<br />

DISC1 (Millar et al. 2005), and altered phosphodiesterase signaling<br />

has been reported in both mood disorders and schizophrenia. We<br />

further found that the underexpression <strong>of</strong> PDE4B after PBRM1<br />

knockdown was partially reversed by valproate, consistent with its<br />

therapeutic role in bipolar disorder. PBRM1 encodes polybromo1<br />

(aka BAF180), which isthe chromatin binding component <strong>of</strong> the<br />

Brahma-associated factor (BAF) nucleosome remodeling complex<br />

(Ho & Crabtree 2010). It is therefore interesting that <strong>of</strong> the 27<br />

histone-related genes that were represented on the array, 9 were<br />

differentially expressed after PBRM1 knockdown, and all <strong>of</strong> these<br />

were upregulated at least 1.8-fold. HeLa and SHSY5Y cells with<br />

stable PBRM1 knockdown displayed strikingly slower proliferation<br />

and altered cellular morphology,suggesting a possible role for this<br />

gene in cell cycle control and apoptosis.<br />

Conclusions: If PBRM1 plays a causal role in mood disorders and<br />

schizophrenia, this role may be mediated by widespread changes in<br />

the expression <strong>of</strong> genes involved in signal transduction, chromatin<br />

remodeling, and cellular development. This study demonstrates the<br />

value <strong>of</strong> GWAS findings in directing studies to diverse biological<br />

processes potentially involved in disease etiology.


PP228 GLOBAL TRANSCRIPTOMIC AND PROTEOMIC<br />

CHARACTERIZATION OF ANTIDEPRESSANT DRUGS AND<br />

STRESS RESPONSE IN A MOUSE MODEL OF DEPRESSION<br />

K. Malki, R. Uher, J. Payo-Cano, E. Binder, K. Aitchinson, I. Craig,<br />

J. Campbell*, P. McGuffin, L. Schalkwyk<br />

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

*james.campbell@kcl.ac.uk<br />

Introduction: Selective serotonin reuptake inhibitors (SSRIs) and<br />

Noradrenergic reuptake inhibitors(NRI’s) represent two class <strong>of</strong><br />

antidepressant drugs with varying pharmacodynamic and<br />

pharmacokinetic mode <strong>of</strong> action that have been shown to be clinically<br />

effective in the treatment <strong>of</strong> depression. However biomarkers with<br />

the power to predict responders to each drug class are not yet<br />

established and prescription still follows a trial and error protocol.<br />

The GENDEP study was designed to uncover genetic markers <strong>of</strong><br />

antidepressant response by integrating findings from a large scale<br />

human pharmacogentic, in-vitro and animal study using the same two<br />

antidepressants across all <strong>of</strong> its treatment arms. In this paper we<br />

report findings from from the mouse arm <strong>of</strong> the GENDEP project.<br />

Methodology: Expression pr<strong>of</strong>iles derived from the hippocampus<br />

tissue <strong>of</strong> 4 inbred mouse strains (DBA, FVB, C57 and 129) <strong>of</strong> both<br />

sexes, treated with varying dose (chronic and acute) <strong>of</strong> either<br />

nortriptyline (NRI) or escitalopram(SSRI) and subjected to two<br />

different types <strong>of</strong> depressogenic protocols (maternal Deprivation and<br />

Chronic Mild Stress) were analysed using the Affymetrix MOE 430<br />

V2 expression chip. Quantitative proteomic analyses were undertaken<br />

on hippocampal tissue from a subset (18) <strong>of</strong> the 144 experimental<br />

groups used for the transcriptomic analysis. Complementary<br />

approaches, namely two dimensional polyacrylamide gel<br />

electrophoresis (2DE) and isobaric tandem mass tagging (TMT),<br />

were applied to the selected experimental cells.Multivariate and<br />

machine learning methods were used to uncover candidate genes and<br />

genes products differentially expressed in response to the two drugs<br />

tested.<br />

Results: The results highlighted significant strain and stress related<br />

differences across both transcriptomic and proteomic data sets. The<br />

most compelling results is the identification <strong>of</strong> three gene products<br />

involved in serotonergic (PXBD5, YHWAH, SLC25A4) and one in<br />

noradrenergic antidepressant action (PXBD6). Specifically YWHAB<br />

was significantly up regulated in response to SSRI treatment. It<br />

belongs to a large family <strong>of</strong> 14-3-3 proteins that participate in a<br />

number <strong>of</strong> important cellular processes implicated in depression and<br />

antidepressant treatment including cell signalling, apoptosis and the<br />

stress response. It is both a brain specific and highly conserved<br />

(>99%) between currently studied eukaryotes including drosophila,<br />

yeast, human rat and mouse. SLC25A4 is part <strong>of</strong> the mitochondrial<br />

carrier subfamily. Recent studies have suggested that mitochondrial<br />

dysfunction may play a role in the pathogenesis <strong>of</strong> unipolar<br />

depression and the SLC25A4 gene and has previously been<br />

implicated in this process.<br />

Conclusions: The search for candidate genes is limited by both our<br />

incomplete understanding <strong>of</strong> the aetiology <strong>of</strong> depression and the<br />

mechanism <strong>of</strong> action <strong>of</strong> therapeutics drugs. Animal models allow for<br />

a hypothesis free search for genetic variants associated with treatment<br />

response and allow for gene expression to be measured in depressionrelevant<br />

brain regions. However mRNA analysis alone cannot<br />

capture information on post transcriptional and regulatory factors or<br />

predict the effect <strong>of</strong> transcriptional control on protein translation, the<br />

effect <strong>of</strong> gene-gene interaction and other post-transcriptional<br />

factors. Proteomic approaches are therefore useful to complement<br />

the analysis at the transcriptome level to obtain a more<br />

comprehensive characterisation across the functional<br />

genomics. Candidate genes emerging from this study will be<br />

used to inform prioritisation in human pharmacogentic studies.<br />

186<br />

OTHER<br />

PP229 NO ASSOCIATION BETWEEN THE HTR1A GENE<br />

AND SUICIDAL BEHAVIOR: A META-ANALYSIS<br />

M. Rivera Angles*(1), D. Bermudez Ocaña(1), I. Juárez Rojop(1),<br />

B. Camarena Medellin(2), A. Genis(3), T. Frias Ramón(1), H.<br />

Nicolini(3), C. Tovilla-Zarate(1)<br />

1. Universidad Juaréz Autónoma de Tabasco, División Académica<br />

Multidisciplinaria de Comalcalco 2. Departamento de Genética<br />

Psiquiátrica, Instituto Nacional de Psiquiatría “Ramón de la Fuente<br />

Muñiz”, 3. Grupo de Estudios Medicos y familiares Carracci<br />

*miriam_angles@hotmail.com<br />

Introduction: Dysfunction <strong>of</strong> serotonin 1A receptors (HTR1A) may<br />

play a role in the genesis <strong>of</strong> suicidal behavior. We studied the<br />

association between a functional polymorphism in the gene HTR1A<br />

and suicidal behavior<br />

Methodology: We performed a meta-analysis <strong>of</strong> published genetic<br />

association studies by searching through Medline, PubMed and Web<br />

<strong>of</strong> Science databases to analyze a possible correlation between the<br />

rs6295 polymorphism and suicidal behavior in different populations<br />

Results: Four were the eligible studies comprising a total <strong>of</strong> nine<br />

hundred and fifty seven patients with suicidal behavior and nine<br />

hundred and fifty seven controls. The allele G <strong>of</strong> the rs6295<br />

polymorphism could not be associated with suicidal behavior<br />

(Random-effects model: OR:1.08 95%CI:0.80-1.45 p(Z)=0.80) in<br />

presence <strong>of</strong> heterogeneity (Q=17.84, df =4, p=0.0013). In a second<br />

analysis that presented no heterogeneity, a negative association was<br />

also observed (OR: 0.94 95%CI: 0.79-1.13 p(Z)=0.99).<br />

Conclusions: To our knowledge, the present study is the first metaanalysis<br />

looking for a correlation between rs6295 <strong>of</strong> HTR1A and<br />

suicidal behavior. Our results showed no association between<br />

HTR1A and suicidal behavior. However, more studies are necessary<br />

that include other populations, as well as larger samples


PP230 GLUTAMATERGIC PATHWAY DISSECTION AND<br />

INDICATIONS FOR GENETIC ASSOCIATION STUDIES IN<br />

MAJOR DEPRESSIVE DISORDER<br />

C. Crisafulli*(1), A. Drago(2), A. Sidoti(1), D. De Ronchi(2), A.<br />

Serretti(2)<br />

1. Department <strong>of</strong> Biomorphology and Biotechologies, Division <strong>of</strong><br />

Biology and <strong>Genetics</strong>, University <strong>of</strong> Messina 2. Institute <strong>of</strong><br />

Psychiatry, University <strong>of</strong> Bologna<br />

*ccrisafulli@unime.it<br />

Introduction: The glutamatergic pathway has been consistently<br />

involved in the physiopathology <strong>of</strong> depressive disorder, however a<br />

complete dissection and integration <strong>of</strong> its role in the context <strong>of</strong> other<br />

known mechanisms is lacking. We summarized and integrated the<br />

evidence <strong>of</strong> various levels <strong>of</strong> interaction between glutamatergic and<br />

monoaminergic pathways to provide a detailed list <strong>of</strong> possible<br />

candidates with practical suggestions for association studies planning.<br />

Methodology: We identified six molecular pathways, some <strong>of</strong> which<br />

with specific regional distribution within the brain. From the six<br />

pathways we identified the key proteins and their coding genes.<br />

Hapmap database (hapmap.ncbi.nlm.nih.gov) served for the<br />

identification <strong>of</strong> the tagging variations. Pubmed database<br />

(http://www.ncbi.nlm.nih.gov) served for the identification <strong>of</strong> all the<br />

validated variations and functional variations for each candidate. All<br />

the identified validated variations for each candidate were forced as<br />

input in the Hapmap dataset in order to retrieve the maximum<br />

coverage <strong>of</strong> each candidate under a tagging approach.<br />

Results: Reviewing the lines <strong>of</strong> evidence <strong>of</strong> two fundamental<br />

theories <strong>of</strong> major depressive disorder (MDD) we disentangled six<br />

main metabolic pathways that could be pivotal in the<br />

pathophysiology <strong>of</strong> the disease: the CAM kinase II, the MAP kinase<br />

and ERK, PKA and PKC, the inositols pathway, the NO cascade, the<br />

axon guidance and actin cytoskeleton and the cellular death pathway.<br />

Conclusions: We obtained a list <strong>of</strong> pivotal enzymes out <strong>of</strong> which we<br />

numbered the covering genetic variations. Such coverage may<br />

provide a genetic framework to serve future genetic association<br />

studies.<br />

187<br />

PP231 INVESTIGATING GENE EXPRESSION CHANGES IN<br />

THE HIPPOCAMPUS AND HYPOTHALAMUS FOLLOWING<br />

A MATERNAL SEPARATION PARADIGM IN TWO INBRED<br />

STRAINS OF MICE<br />

T. Powell*, R. Kember, J. Mill, C. Fernandes, L. Schalkwyk<br />

King's College London<br />

*timothy.powell@kcl.ac.uk<br />

Introduction: Gene-environment interaction (GxE) studies have<br />

shown that the existence <strong>of</strong> susceptibility genes (G) in the presence <strong>of</strong><br />

early life stress (E) increases the chances <strong>of</strong> developing later life<br />

depression. The current study aimed to further investigate the effects<br />

<strong>of</strong> early-life stress using a maternal separation paradigm.<br />

Methodology: Two inbred strains <strong>of</strong> mice, C57BL/6J and DBA/2J,<br />

were used to establish whether different genetic strains (G) in the<br />

presence <strong>of</strong> the same early life stress (E) showed differential gene<br />

expression (GxE) in two key brain regions related to the<br />

pathophysiology <strong>of</strong> depression. Mice were assigned to either a<br />

control (n=22) or separated group (n=18). Separated groups<br />

underwent maternal separation at postnatal day 9 for 24hours, control<br />

and separated groups then underwent behavioural testing at week 12<br />

(discussed briefly) and culling at week 15. The hippocampus and<br />

hypothalamus were dissected and RNA was extracted and used for in<br />

vitro cDNA synthesis. cDNA was then fragmented and hybridised for<br />

use on Affymetrix Gene Chip microarrays.<br />

Results: Results discussed include any notable gene-environment<br />

interactions as well as strain-specific and tissue-specific expression<br />

differences.<br />

Conclusions: Using mouse models to manipulate environments at<br />

different developmental stages in relation to genetic backgrounds is<br />

useful in gaining a unique insight in the complicated pathological<br />

processes involved in disorders such as depression.


PP232 SEQUENTIAL SCREENING OF MUTATIONS<br />

MEDIATED BY ALU IN SCHIZOPHRENIA<br />

M. Ueno*, A. Akahane, T. Hata, S. Nanko<br />

Teikyo University School <strong>of</strong> Medicine<br />

*mueno@med.teikyo-u.ac.jp<br />

Introduction: In previous reports, we have identified the de novo<br />

mutations mediated by mobile elements in the patients with<br />

schizophrenia. These dynamics <strong>of</strong> mobile elements is not fully fixed,<br />

and might be progressively after event onset. Herein, we have<br />

attempted the sequential screening <strong>of</strong> mutations mediated by mobile<br />

element Alu in patients.<br />

Methodology: DNA samples were obtained from a patient and a<br />

control subject. Sequential collection <strong>of</strong> blood was performed at<br />

intervals <strong>of</strong> 5 years. At first screening, the microarray assay using<br />

Alu-PCR products from each sample was performed to detect the<br />

mutated genes. Second, Fluorescent in situ hybridization(FISH)<br />

analysis was performed on combed DNA using AluYa5 probe and<br />

BAC probes which were containing the genes screened by<br />

microarray. This study was done under the approval <strong>of</strong> the Ethical<br />

Committee for Genetic Research, Teikyo University School <strong>of</strong><br />

Medicine.<br />

Results: We have identified the progressive mutations mediated by<br />

Alu. The microarray assay allows us to clear the characteristic <strong>of</strong><br />

patient genome. The 24 <strong>of</strong> 26 progressive mutations in the patient<br />

were deletion types. Moreover, FISH analysis on TPM4 gene with<br />

progressive deletions showed the decreasing signals <strong>of</strong> Alu probe in<br />

patient.<br />

Conclusions: Our results suggested that the dynamics <strong>of</strong> Alu might<br />

be one <strong>of</strong> the risk factors for schizophrenia. Recently, the dynamics<br />

<strong>of</strong> mobile element L1 have been reported to contribute to the<br />

neuroplasticity in adult brain. Further research using sequential<br />

screenings <strong>of</strong> various mutations mediated by mobile elements is<br />

needed to clarify the etiology in schizophrenia.<br />

188<br />

PP233 CENTRAL CNS NEONATAL LESION IN VENTRAL<br />

HIPPOCAMPUS MAY ALTER PERIPHERAL<br />

LYMPHOCYTE DOPAMINE GENE EXPRESSION<br />

A. Genis*(1,4), C. Lopéz-Rubalcava (2), J. Flores(4), P. Mendoza(3),<br />

P. Ostrosky(3), H. Nicolini(1,4)<br />

1. Universidad Autónoma de la Ciudad de México 2. Centro de<br />

Investigación y de Estudios Avanzados del IPN 3. Instituto de<br />

Ciencias Biomédicas 4. Servicios de atención psiquiátrica<br />

*genis76@yahoo.com<br />

Introduction: The animal model <strong>of</strong> neonatal lesion in the ventral<br />

hippocampus (NLVH) is a wide validated animal model used to study<br />

schizophrenia from a neurodevelopmental focus. This animal model<br />

is also used to investigate how neonatal lesion may alter genetic<br />

expression <strong>of</strong> dopaminergic receptors. Some researchers have<br />

observed changes in the expression <strong>of</strong> drd2/drd3 in patients with<br />

schizophrenia in lymphocytes <strong>of</strong> peripheral blood. These studies have<br />

shown that schizophrenic patients have increased expression <strong>of</strong><br />

mRNA-drd2. Literature indicates that NLVH causes improper<br />

maturation <strong>of</strong> inter-neurons from the prefrontal cortex, that may<br />

produce a hyperactive or irregularly active cortex. This condition has<br />

been proposed as a mechanism underlying dysfunction in the PFC<br />

and other cortical areas that may explain some schizophrenic<br />

characteristics. Studying CNS tissue in schizophrenic patients is not<br />

easy, however, peripheral markers that can be easily obtained and<br />

that could be related to the disease process are necessary.<br />

Methodology: NLVH procedure was applied to male Wistar rats at<br />

5-7 days after birth. At postnatal days 45 and 90, locomotor activity,<br />

social interaction, object recognitionand prepulse inhibition were<br />

analyzed. In addition drd2 and drd3 expression in lymphocytes were<br />

measure. All behaviors and receptor expressions were compared with<br />

those <strong>of</strong> sham the operated group.<br />

Results: Quantification <strong>of</strong> drd2 showed significant over-expression<br />

with respect to the control sample in contrast drd3 expression didn't<br />

change neither at 45 or 90 days <strong>of</strong> age. In 90 days old rats, an<br />

increase in locomotor activity and a reduction in memory, prepulse<br />

inhibition and social interaction behaviors in comparison to the sham<br />

group were observed. The drd2 expression was observed only in the<br />

experimental group with 90 days-age. It is important to indicate that<br />

although the five dopaminergic receptors are present in lymphocytes<br />

<strong>of</strong> peripheral blood, the expression <strong>of</strong> drd2 is not observed in healthy<br />

animals without manipulation or sham operated group nevertheless<br />

when these animals reach adulthood, drd2 is expresses mainly due to<br />

the response <strong>of</strong> the lesion performed at neonatal time.<br />

Conclusions: This change in drd2 expression could serve in the<br />

future as a molecular marker for schizophrenia, since expression in<br />

peripheral blood <strong>of</strong> rats with NLVH correlate with rat's<br />

schizophrenic-like behaviors that reflect some symptoms <strong>of</strong> the<br />

disease.


PP234 GENE EXPRESSION PROFILE OF PERIPHERAL<br />

BLOOD DISCRIMINATES PATIENTS WITH<br />

SCHIZOPHRENIA SUBMITTED TO DISTINCT<br />

TREATMENTS<br />

A. Silva*(1), H. Brentani(2), C. Pereira(3), D. Carraro(1), R.<br />

Puga(1), B. Mello(1), M. Maschieto(1), P. Belmonte-de-Abreu(4), J.<br />

Palha(5)<br />

1. Research Center, A C Camargo Hospital 2. Institute <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Sao Paulo, Medical School 3. Institute <strong>of</strong> Mathematics<br />

and Statistics, University <strong>of</strong> Sao Paulo 4. Department <strong>of</strong> Psychiatry,<br />

Federal University <strong>of</strong> Rio Grande do Sul 5. Life and Health Sciences<br />

Research Institute, School <strong>of</strong> Health Science, University <strong>of</strong> Minho<br />

*aderbs@gmail.com<br />

Introduction: Schizophrenia is a debilitating psychiatric disorder<br />

considered as a “two-hit” disease, with a neurodevelopmental<br />

impairment followed by an additional hit in early adulthood. Both<br />

events are regulated by hormonal (like thyroid and retinols) and<br />

signal transduction pathways (like Wnt signaling pathway), which<br />

have already been identified in post-mortem brain studies <strong>of</strong><br />

schizophrenic individuals. While brain studies are fitted for<br />

pathology assessment post-mortem, there is not suitable material for<br />

searches <strong>of</strong> a schizophrenia “diagnostic signature” in live patients.<br />

There is still a requirement to identify a feasible material to be used<br />

in the investigation <strong>of</strong> molecular markers useful for diagnosis in<br />

schizophrenia patients.<br />

Methodology: Gene expression pr<strong>of</strong>ile <strong>of</strong> blood from twenty eight<br />

males with DSM-IV schizophrenia under three drug regimens<br />

(clozapine, risperidone or haloperidol), and ten healthy controls were<br />

analyzed in a cDNA microarray platform containing 1,364 genes<br />

implicated with hormonal and signal transduction pathways.<br />

Significant Analysis <strong>of</strong> microarray using TMEV s<strong>of</strong>tware identified<br />

differentially expressed genes.<br />

Results: Seven genes were differentially expressed between controls<br />

and patients with different treatments, and 28 between healthy<br />

controls and specific treatments. CTNNA1, GSTM3, HOXA-13 and<br />

KRT18 were commonly altered in all analyses. Furthermore, principal<br />

component analysis based on the differentially expressed genes<br />

expression values discriminated clozapine- (CLO), risperidone-<br />

(RIS), haloperidol-treated (HAL) patients and the healthy controls<br />

(Cont) (Figure 1).<br />

Conclusions: These results suggest that it is feasible to use peripheral<br />

blood, an accessible material, from patients for studying<br />

schizophrenia. Also, this study revealed genes which expression<br />

might be modulated by drug treatments.<br />

189<br />

PP235 APOLIPOPROTEIN H (APO H) AS NOVEL<br />

BIOMARKER FOR SCHIZOPHRENIA<br />

H. Loh*(1), T. Chow(2)<br />

1. Centre for Healthcare Science and Technology, Faculty <strong>of</strong><br />

Engineering and Science, Universiti Tunku Abdul Rahman 2.<br />

Department <strong>of</strong> Science, Faculty <strong>of</strong> Engineering & Science, Universiti<br />

Tunku Abdul Rahman<br />

*hcloh@utar.edu.my<br />

Introduction: Biomarkers for Schizophrenia (SCZ) are essential to<br />

facilitate disease diagnosis ideally at early stages, monitor disease<br />

progression and assess response to existing and future treatments.<br />

Application <strong>of</strong> proteomics to the human brain, cerebrospinal fluid and<br />

serum has greatly hastened the unbiased and high-throughput<br />

searches for novel biomarkers.<br />

Methodology: Serumproteomes <strong>of</strong> 40 patients and 40 controls were<br />

investigated using two-dimensional gel electrophoresis (2-DE) and<br />

matrix-assisted laser desorption ionization time-<strong>of</strong>-flight/time-<strong>of</strong>flight<br />

mass spectrometry (MALDI-ToF/ToF MS). The novel<br />

biomarker found was further validated using Western Blot.<br />

Results: Three protein spots were found to be altered in patients<br />

compared to controls. Among these acute phase proteins,<br />

apolipoprotein H (ApoH, p = 0.00346) showed significant<br />

overexpression in patients whilst apolipoprotein A-I (ApoA-I, p =<br />

0.00365) and haptoglobin (p = 0.00307) were found down-regulated<br />

in patients compared to controls. ApoA-I and haptoglobin were<br />

previously found to be associated with SCZ. However to the best <strong>of</strong><br />

our knowledge, ApoH is a novel serum marker for SCZ. To validate<br />

the result, western blotting with anti-ApoH to control and patient<br />

serums (n = 8) which were randomly selected from the tested group.<br />

As expected, a single specific band <strong>of</strong> molecular weight<br />

approximately 38 kDa was detected in membrane probed with anti-<br />

ApoH antibody in sera tested. In comparison, the expression level <strong>of</strong><br />

ApoH in patient’s serum is significantly higher than normal control<br />

group (p < 0.0298). In order to double confirm the differential<br />

expression <strong>of</strong> ApoH, the same experiment was repeated with the<br />

pooled sera <strong>of</strong> normal and patient group. A similar trend <strong>of</strong> results<br />

was obtained where patient group significantly showed the higher<br />

expression level <strong>of</strong> ApoH protein as compared with the normal<br />

control group (p < 0.0306).<br />

Conclusions: These findings suggest that increased level <strong>of</strong> ApoH<br />

might be associated with the pathology <strong>of</strong> SCZ. Hence ApoH serves<br />

as novel biomarkers for SCZ as it was not reported elsewhere up-todate.


PP236 PAST HISTORY OF MENTAL DISORDERS<br />

ASSOCIATES WITH LONGER PERIPHERAL BLOOD CELL<br />

TELOMERES IN ELDERLY ADULTS: THE HELSINKI<br />

BIRTH COHORT STUDY (HBCS)<br />

K. Savolainen*(1), K. Räikkönen(1), L. Kananen(2,3), E.<br />

Kajantie(4,5), I. Hovatta(2,3,6), M. Lahti(1), J. Eriksson(4,7,8,9,10)<br />

1. Institute <strong>of</strong> Behavioural Sciences, University <strong>of</strong> Helsinki 2.<br />

Research Programs Unit, Molecular Neurology, Biomedicum-<br />

Helsinki, University <strong>of</strong> Helsinki 3. Department <strong>of</strong> Medical <strong>Genetics</strong>,<br />

Haartman Institute, Faculty <strong>of</strong> Medicine, University <strong>of</strong> Helsinki 4.<br />

Diabetes Prevention Unit, Department <strong>of</strong> Chronic Disease<br />

Prevention, National Institution for Health and Welfare 5. Hospital<br />

for Children and Adolescents, Helsinki University <strong>of</strong> Central<br />

Hospital 6. Department <strong>of</strong> Mental Health and Substance Abuse<br />

Services, National Institute for Health and Welfare 7. Folkhälsan<br />

Research Centre 8. Unit <strong>of</strong> General Practice, Helsinki University <strong>of</strong><br />

Central Hospital 9. Vasa Central Hospital 10. Department <strong>of</strong> General<br />

Practice and Primary Health Care, University <strong>of</strong> Helsinki<br />

*katri.savolainen@helsinki.fi<br />

Introduction: Accelerated telomere length (TL) shortening has<br />

previously been linked with mental disorders including schizophrenia<br />

and mood disorders. Mental disorders are also associated with early<br />

mortality. Yet, it remains unclear if accelerated TL shortening<br />

characterize patients who have diagnosed for mental disorders in the<br />

past, and survived till older age.<br />

Methodology: The participants were 61.5 (SD=2.9, Range=56.6-<br />

69.8) year-old women (n=1051) and men (n=905) from the HBCS,<br />

born between years 1934-1944. TL from peripheral blood cells was<br />

measured using real-time quantitative PCR method. Individuals with<br />

mental disorders (n=116) severe enough to warrant hospitalization<br />

were identified from Finnish Hospital Discharge Register. The<br />

average time from last mental disorder hospitalization to<br />

measurement <strong>of</strong> TL was 12.9 (SD=8.9, Range=0.3-31.3) years.<br />

Results: Participants hospitalized for any mental or substance use<br />

disorders had longer TL than non-hospitalized controls (age and sex<br />

controlled p-values


PP238 OVERALL EFFECT OF BDNF VAL66MET<br />

POLYMORPHISM ON BODY MASS INDEX AND BIPOLAR<br />

DISORDER<br />

M. Rivera*(1), G. Hosang(1), S. Cohen-Woods(1), G. Breen(1), R.<br />

Investigators(1), F. Tozzi(2), J. Perry(2), P. Muglia(2), C. Lewis(1),<br />

I. Craig(1), J. Vincent(3), J. Strauss(3), P. McGuffin(1), A. Farmer(1)<br />

1. MRC SGDP Centre, Institute <strong>of</strong> Psychiatry, King's College<br />

London 2. GlaxoSmithKline Research and Development, Medical<br />

<strong>Genetics</strong>, Clinical Pharmacology and Discovery Medicine 3.<br />

Neurogenetics Section, Centre for Addiction and Mental Health,<br />

Department <strong>of</strong> Psychiatry, University <strong>of</strong> Toronto<br />

*margarita.rivera_sanchez@kcl.ac.uk<br />

Introduction: Associations between body mass index (BMI) or<br />

obesity and psychiatric disorders have been reported in numerous<br />

studies. The BDNF Val66Met polymorphism has been associated<br />

with depression and bipolar disorder, as well as obesity and BMI.<br />

The aim <strong>of</strong> this study is to explore the genetic influence <strong>of</strong> the BDNF<br />

Val66Met polymorphism in relation to BMI in a sample <strong>of</strong> bipolar<br />

disorder cases and psychiatrically healthy controls.<br />

Methodology: The sample is part <strong>of</strong> the Bipolar Association Case-<br />

Control Study (BACC) and consists <strong>of</strong> approximately 1,000 bipolar<br />

cases and 1,400 controls. ICD-10 bipolar diagnoses were made using<br />

the SCAN interview. The controls were screened for lifetime absence<br />

<strong>of</strong> any psychiatric disorders. The sample was genotyped for the<br />

BDNF Val66Met polymorphism (rs6265). Participants’ weight and<br />

height were used to calculate BMI, defined as weight (kilograms) /<br />

height (metres) 2 .<br />

Results: Linear regressions were performed to test for association<br />

between BDNF Val66Met variant and BMI in the whole sample and<br />

separatelyin the different clinical groups. We found an overall<br />

association between this polymorphism and BMI in the whole<br />

sample, mainly attributable to the control group.<br />

Conclusions: We have investigated the influence <strong>of</strong> the BDNF<br />

Val66Met variant on BMI in a large clinical sample <strong>of</strong> bipolar<br />

patients and controls. There are two mainly findings, first this<br />

polymorphism was significantly associated with bipolar disorder,<br />

second the BDNF variant was significantly related to BMI. This is the<br />

first study investigating the effect <strong>of</strong> the BDNF Val66Met<br />

polymorphism on BMI within the context <strong>of</strong> bipolar disorder.<br />

191<br />

PP239 ASSOCIATION ANALYSES OF TUMOR NECROSIS<br />

FACTOR ALPHA GENE POLYMORPHISMS WITH<br />

OBSESSIVE-COMPULSIVE AND TIC DISORDERS<br />

K. Emese*(1), N. Zs<strong>of</strong>ia(1), K. Eszter(2), T. Zsanett(2), K.<br />

Gergely(1), S. Maria(1)<br />

1. Institute <strong>of</strong> Medical Chemistry, Molecular Biology and<br />

Pathobiochemistry, Semmelweis University 2. Vadaskert Child and<br />

Adolescent <strong>Psychiatric</strong> Clinic<br />

*mesi10@gmail.com<br />

Introduction: In a minority <strong>of</strong> cases, children are diagnosed with<br />

Tourette syndrome (TS) and obsessive-compulsive disorder (OCD)<br />

after streptococcal infections. The pro-inflammatory cytokine, tumor<br />

necrosis factor alpha (TNF) is hypothesized to play a role in these<br />

autoimmune processes. Therefore, the possible genetic vulnerability<br />

<strong>of</strong> children with TS and OCD diagnoses were tested for TNF gene<br />

promoter polymorphisms with possible effect on gene expression.<br />

Methodology: A total <strong>of</strong> 214 children (male 80.6%, average age at<br />

the onset <strong>of</strong> the disorder 10.3 ± 3.6) with main diagnosis <strong>of</strong> TS<br />

(N=113) or OCD (N=101) participated in the case-control study. The<br />

diagnoses were obtained by the DSM-IV criteria. The control sample<br />

consisted <strong>of</strong> 273 healthy young adults (average age 22.1 ± 6.4). Both<br />

groups were ethnically homogenous (Hungarian, Caucasian origin).<br />

Tic and obsessive-compulsive symptoms were also assessed by the<br />

MINI-kid (Mini-International Neuropsychiatric Interview, child<br />

version) in an extended child psychiatry sample (N=338).<br />

Polymorphisms <strong>of</strong> the TNF-alpha gene were the following: -238 A/G<br />

(rs361525), -308 A/G (rs1800629), and -863 A/C (rs1800630). Chisquare<br />

analyses were carried out for the main diagnoses vs control<br />

group, as well as for tic and obsessive-compulsive symptom present<br />

vs absent groups.<br />

Results: The genetic association analyses <strong>of</strong> the main diagnostic<br />

categories showed an increase <strong>of</strong> the -238 A-allele frequency in both<br />

the TS and OCD groups compared to the control group. The -308 Aallele<br />

frequency was decreased in the TS group (vs control group)<br />

and in the tic-present group (vs tic absent group). No association was<br />

found with the -863 A/C polymorphism.<br />

Conclusions: The preliminary analyses <strong>of</strong> TNF gene promoter<br />

polymorphisms in our child psychiatry sample suggest that the TNF -<br />

238 A-allele might act as a risk (vulnerability) factor in the TS-OCD<br />

spectrum and the -308 A-allele might act as a protective (resilience)<br />

factor in Tourette syndrome.<br />

This work was supported by Hungarian Scientific Research Fund<br />

(OTKA) F67784 and CK80289.


PP240 NICOTINE DEPENDENCE AND DOPAMINE<br />

METABOLISM ENZYME GENES<br />

T. Shinkai*(1), H. Chen(1), K. Utsunomiya(1), K. Yamada(1), S.<br />

Sakata(1,2), O. Ohmori(1,3), J. Nakamura(1)<br />

1. University <strong>of</strong> Occupational and Environmental Health 2.<br />

Sagamigaoka Hospital 3. Wakato Hospital<br />

*shinkai@med.uoeh-u.ac.jp<br />

Introduction: Cigarette smoking increases the risk <strong>of</strong> various health<br />

problems, including cancer, cardiovascular and pulmonary disorders,<br />

making smoking the leading cause <strong>of</strong> preventable death in the world.<br />

Twin and family studies have indicated significant genetic<br />

contributions to smoking behaviors. Although genetic associations to<br />

nicotine dependence have been widely investigated, the relevance <strong>of</strong><br />

individual genetic variations for smoking behavior remains unknown.<br />

The noteworthy candidate genes are involved in the dopamine<br />

pathways for the brain reward system.<br />

Methodology: We examined an association <strong>of</strong> polymorphisms at two<br />

dopamine metabolism enzymes (i.e., tyrosine hydroxylase, dopamine<br />

beta-hydroxylase) and their relation to nicotine dependence in a total<br />

<strong>of</strong> nearly 700 healthy workers in a Japanese heavy industry company.<br />

Smoking behavior was assessed by a questionnaire including<br />

Fagerström Test for Nicotine Dependence (FTND) and Tobacco<br />

Dependence Screener (TDS).<br />

Results: Although our preliminary data does not suggest significant<br />

associations with nicotine dependence, analysis with complete data<br />

set is currently undergoing to clarify the relationship between the<br />

dopamine system genes and nicotine dependence.<br />

Conclusions: It is thought that dopamine is critically involved in<br />

drug addiction processes. Scientific knowledge on dopamine system<br />

accumulated in pharmacogenetic studies may lead to a solution to<br />

prevent and to treat nicotine dependence.<br />

192<br />

PP241 ASSOCIATION STUDY BETWEEN VITAMIN D<br />

RECEPTOR POLYMORPHISM AND ALZHEIMER’S<br />

DISEASE<br />

K. Yamada*(1), T. Shinkai(1), K. Utsunomiya(1), S. Sakata(1,2), H.<br />

Chen(1), J. Nakamura(1)<br />

1. University <strong>of</strong> Occupational and Environmental Health 2.<br />

Sagamigaoka Hospital<br />

*hokuto_no_ken_1225@yahoo.co.jp<br />

Introduction: In the central nervous system (CNS), Vitamin D 3<br />

plays a key role in neuroprotection against oxidative stress, calcium<br />

homeostasis, and production <strong>of</strong> neurotrophins. The single nucleotide<br />

polymorphisms (SNPs) in the vitamin D receptor (VDR) gene which<br />

causes altered affinity to vitamin D3 may thus be related to<br />

neurodegenerative diseases. Gezen-Aket al (2007) reported a positive<br />

association between two SNPs in the VDR gene (ApaI: rs7975232<br />

and TaqI: rs731236) and late-onset Alzheimer's disease (LOAD) in a<br />

Turkish sample (104 LOAD cases and 109 controls). To our<br />

knowledge, no Japanese study has conducted in terms <strong>of</strong> this<br />

association.<br />

Methodology: In the present study, we examined an association<br />

between the above mentioned two SNPs and LOAD in a Japanese<br />

sample (100 LOAD cases and 200 controls).<br />

Results: Although our data does not suggest a robust association,<br />

analysis with complete data set is currently undergoing to clarify the<br />

relationship between the VDR gene and LOAD.<br />

Conclusions: Our study provides evidence for a possible link<br />

between LOAD and vitamin D.


PP242 ASSOCIATION STUDY BETWEEN A SINGLE<br />

NUCLEOTIDE POLYMORPHISM IN ZNF804A GENE AND<br />

ATTENTION DEFICIT HYPERACTIVITY DISORDER<br />

X. Xu*(1), G. Breen(1), L. Luo(2), B. Sun(2), C. Chen(3, 4, 5), Y.<br />

Huang(4, 5), Y. Wu(4, 5), P. Asherson(1)<br />

1. MRC Social, Genetic and Developmental Psychiatry Centre,<br />

Institute <strong>of</strong> Psychiatry, King's College London 2. School <strong>of</strong><br />

Medicine, King's College London 3. Department <strong>of</strong> Psychiatry,<br />

Chang Gung Memorial Hospital 4. Chang Gung University School <strong>of</strong><br />

Medicine 5. Division <strong>of</strong> Mental Health & Drug Abuse Research,<br />

National Health Research Institutes<br />

*xiaohui.xu@kcl.ac.uk<br />

Introduction: Attention-deficit hyperactivity disorder (ADHD) is<br />

one <strong>of</strong> the most frequent and heritable childhood behavioural<br />

disorders characterised by age inappropriate levels <strong>of</strong> hyperactivity,<br />

impulsivity and inattentiveness. There was evidence showing a strong<br />

genetic contribution to ADHD in adoption, family and twin study.<br />

Polymorphic variants in several genes involved in regulation <strong>of</strong><br />

dopamine and related neurotransmitter pathways are reported to be<br />

associated with ADHD. The most investigated candidate genes were<br />

DAT1, DRD4, COMT, MAOA and DBH. Recently genome-wide<br />

association (GWA) study has been conducted on ADHD and<br />

psychiatric disorders. ADHD and psychiatric disorders share many<br />

features clinically. Balog et al. (2010) investigated the relationship<br />

between single nucleotide polymorphism (SNP) rs1344706 in<br />

ZNF804A and attention in 200 healthy volunteers. Their study found<br />

a significant association with the executive control network <strong>of</strong><br />

attention: the A/A genotype and the A-allele were associated with<br />

increased reaction time. One recent study by Landaas et al (2011)<br />

examined six SNPs in CACNA1C, ANK3, MYO5B, TSPAN8 and<br />

ZNF804A genes in adult ADHD, and concluded that the six SNPs<br />

including rs1344706 in ZNF804A with strong evidence <strong>of</strong> association<br />

in bipolar (BD) GWA studies seem unlikely shared risk variants<br />

between ADHD and BD. In this study we examine the role <strong>of</strong><br />

rs1344706 polymorphism in two clinical family-based ADHD<br />

samples from the UK and Taiwan.<br />

Methodology: To investigate the association between the<br />

polymorphism (rs1344706) in ZNF804A and ADHD, two family<br />

samples <strong>of</strong> ADHD probands from the United Kingdom (n = 180) and<br />

Taiwan (n = 212) were genotyped using a TaqMan SNP genotyping<br />

assay on a 7900HT sequence detection system (Applied Biosystems)<br />

and analysed using within-family transmission disequilibrium test<br />

(TDT).<br />

Results: No significant associations were found between rs1344706<br />

polymorphism and ADHD in either <strong>of</strong> the family samples from<br />

Taiwan (P = 0.91) and UK (P = 0.43). Even combining the two<br />

family datasets together the A allele <strong>of</strong> rs1344706 SNP was still not<br />

significantly over-transmitted to the affected probands (P = 0.50).<br />

Conclusions: In this study we used family-based ADHD data in the<br />

UK and Taiwanese population to test for an association between<br />

rs1344706 SNP in ZNF804A gene and ADHD. Our results showed<br />

that the rs1344706 SNP with strong evidence <strong>of</strong> association in bipolar<br />

disorder (BD) GWA studies does not have significant association in<br />

ADHD for UK and Taiwanese population.<br />

193<br />

PP243 REPLICATION OF CNTNAP2 IN A LONGITUDINAL<br />

SAMPLE WITH AND WITHOUT LANGUAGE IMPAIRMENT<br />

K. Mueller*(1), J. Murray(2), B. Tomblin(1)<br />

1. University <strong>of</strong> Iowa, Department <strong>of</strong> Communication Sciences and<br />

Disorders 2. University <strong>of</strong> Iowa, Department <strong>of</strong> Pediatrics<br />

*kathryn-mueller@uiowa.edu<br />

Introduction: Specific Language Impairment (SLI [MIM 602081])<br />

is a relatively common developmental disorder characterized by<br />

difficulty in acquiring language in the absence <strong>of</strong> cognitive or sensory<br />

impairments, and despite adequate experience and educational<br />

opportunities (Tomblin, et al., 1996). The disorder is associated with<br />

deficits in academic achievement, elevated rates <strong>of</strong> unemployment,<br />

and impairments in socio-emotional development. Although there is a<br />

significant heritable component, molecular studies have been slow to<br />

identify genes involved in the neurobiology <strong>of</strong> language<br />

development, and little research exists on common genetic variants<br />

that contribute to individual differences in language ability. One<br />

candidate, CNTNAP2, has been associated with language<br />

impairments both in children with SLI and in children with autism<br />

(Alarcón, et al., 2008; Vernes, et al., 2008). CNTNAP2 encodes a<br />

neurexin, a transmembrane protein that mediates cell–cell<br />

interactions in the developing nervous system. The gene is highly<br />

expressed in the cerebral cortex. In addition, CNTNAP2 is in the<br />

same neurobiological pathway as FOXP2, a gene that has been welldocumented<br />

in the development <strong>of</strong> speech and language. Following<br />

findings from Vernes et al. (2008) and Whitehouse et al. (2011), we<br />

hypothesized that variants in the gene would also influence a broader<br />

range <strong>of</strong> language abilities in a large epidemiological sample <strong>of</strong><br />

children with and without language impairment.<br />

Methodology: Participants (n=604) and their families were recruited<br />

in Iowa and Illinois from a longitudinal study <strong>of</strong> language<br />

development and disorders (Child Language Research Centre, The<br />

University <strong>of</strong> Iowa). An initial pool <strong>of</strong> participants (n=7,218) was<br />

screened in kindergarten as part <strong>of</strong> a cross-sectional epidemiological<br />

study. A stratified-cluster sampling method was used to invite<br />

individuals to participate in the diagnostic phase <strong>of</strong> the study.<br />

Children who failed the language screening test, plus a random<br />

sample who passed (~33%), were administered a more<br />

comprehensive set <strong>of</strong> language and cognitive assessments as a<br />

diagnostic battery for SLI (n=2,009). Children completed five<br />

language subtests from the Test <strong>of</strong> Oral Language Development-2 P:<br />

Picture Vocabulary, Oral Vocabulary, Grammatic Understanding,<br />

Sentence Imitation, and Grammatic Completion, as well as the test <strong>of</strong><br />

Word Articulation (Newcomer & Hammill, 1988). Narrative<br />

comprehension and production were assessed using the Listening to<br />

Paragraphs subtest <strong>of</strong> the CELF-III (Semel, et al., 1987) and a spoken<br />

story generation task by Catts & Fey (1998). Scores were converted<br />

into composite ability scores for the present study. Children with SLI<br />

were defined as those whose standardized language scores fell 1.15<br />

SD below the mean for their age. Because <strong>of</strong> the way the sample was<br />

ascertained, phenotype data followed a normal pattern <strong>of</strong> distribution.<br />

DNA was obtained from blood, buccal swabs, and saliva for 601 <strong>of</strong><br />

the probands, their parents and siblings (total n=1,482). Samples<br />

were genotyped using Taqman Pre-designed Genotyping Assays<br />

under standard reaction conditions (Applied Biosystems, Foster City,<br />

CA). Potential confounds arising from population admixture were<br />

eliminated by restricting analysis to the Caucasian sample only.<br />

Allelic variation was determined via the Sequence Detection Systems<br />

2.2 s<strong>of</strong>tware (Applied Biosystems). PLINK was used to identify<br />

Mendelian errors, estimate allele frequencies, test for Hardy-<br />

Weinberg equilibrium, and perform association analyses.<br />

Results: Significant association was found for a single SNP,<br />

rs2710117, across a range <strong>of</strong> language abilities, including language<br />

diagnosis (LDIAG, p


PP244 ETHICAL CONCERNS ABOUT PSYCHIATRIC<br />

GENETIC STUDIES: ITEM FUNCTIONING AND BIAS OF<br />

QUESTIONNAIRE ABOUT GENETIC RISK<br />

M. Hipolito*(1), M. Malik(1), W. Lawson(1), J. Nurnberger, Jr(2),<br />

E. Nwulia(1)<br />

1. Howard University 2. Indiana University<br />

*mhipolito@howard.edu<br />

Introduction: High level <strong>of</strong> perceived concerns surrounding<br />

unethical use <strong>of</strong> genetic data could hinder participation in psychiatric<br />

genetic studies. However, there is a paucity <strong>of</strong> questionnaires for<br />

examination <strong>of</strong> ethical concerns to participation in psychiatric genetic<br />

studies. Of the few available, robustness to differential ethnic bias<br />

equating for severity <strong>of</strong> concerns have not been demonstrated. We<br />

investigated the quality <strong>of</strong> Questionnaire on Genetic Research (QGR)<br />

for measuring the extent <strong>of</strong> perceived concern in a population with<br />

Bipolar disorder, and examined the robustness <strong>of</strong> the response items<br />

to differential bias by ethnic status.<br />

Methodology: Six questions measured the degree <strong>of</strong> concerns about<br />

negative consequences from psychiatric genetic studies in the QGR.<br />

Each question has 5 response categories corresponding to increasing<br />

degree <strong>of</strong> concern, ranging from “not at all” concerned to “very<br />

concerned.” We applied polytomous item response models (IRM)<br />

with latent constructs for analysis <strong>of</strong> complete responses by<br />

respondents.<br />

Results: Most items <strong>of</strong> the QGR provided reliable measure <strong>of</strong> the<br />

extent <strong>of</strong> concerns. The instrument also validated the findings <strong>of</strong><br />

greater concern about genetic studies among American Blacks<br />

compared to Whites. Item bias was observed for concerns about<br />

racism, procreation and privacy, but adjusting for these latter 3 items<br />

in a sensitivity analysis did not substantially change the association<br />

between Blacks and level <strong>of</strong> concern.<br />

Conclusions: For the most part, the QGR is a reliable measure <strong>of</strong> the<br />

extent <strong>of</strong> perceived concern, based on response patterns to its six<br />

items. However, cautious interpretations <strong>of</strong> some items are warranted<br />

when the ethnicity <strong>of</strong> the study sample is diverse, due to vulnerability<br />

to response bias.<br />

194<br />

PP245 ALZHEIMER’S DISEASE DIAGNOSTICS BASED ON<br />

MULTI-MODAL DATA SETS<br />

M. Sattlecker*(1), K. Lunnon(1), P. Proitsi(1), A. Stewart(3), S.<br />

Williams(3), G. Coppola(3), D. Geschwind(3), A. Hodges(1,2), S.<br />

Lovestone(1,2), R. Dobson(1)<br />

1. NIHR Biomedical Research Centre for Mental Health, South<br />

London and Maudsley NHS Foundation Trust & Institute <strong>of</strong><br />

Psychiatry, Kings College London 2. MRC Centre for<br />

Neurodegeneration, King's College London 3. SomaLogic, Inc. 2945<br />

Wilderness Place<br />

*martina.sattlecker@kcl.ac.uk<br />

Introduction: There is a need for non-invasive biomarkers in<br />

Alzheimer's disease (AD), not only to diagnose the disease state but<br />

also for stratification and experimental medicine as well as providing<br />

an insight into disease pathogenesis. In this study we focus on the<br />

potential <strong>of</strong> blood plasma protein and RNA levels as markers for AD<br />

but describe the added benefit <strong>of</strong> including other measures such as<br />

ApoE4 dosage and sMRI measures using data derived from the EU<br />

funded AddNeuroMed Alzheimer's biomarker project.<br />

Methodology: The AddNeuroMed project has produced a rich<br />

variety <strong>of</strong> datasets including peripheral blood whole transcriptome<br />

RNA measures, 822 protein plasma measures (with SomaLogic, US<br />

Biotech), sMRI, whole genome SNP genotypesand a rich set <strong>of</strong><br />

clinical phenotypes that includes rates <strong>of</strong> cognitive decline (MMSE,<br />

CDR and ADAS-Cog). These data are available on over 300 patients<br />

comprising AD cases, normal elderly controls and as well as patients<br />

suffering from mild cognitive impairment (MCI), <strong>of</strong> which<br />

approximately half converted to AD within two years <strong>of</strong> baseline<br />

measures. We developedpredictive models that included, but went<br />

beyond standard diagnostic comparisons, identifying predictive<br />

models <strong>of</strong> prodromal AD and rate <strong>of</strong> decline using the multi-modal<br />

datasets available<br />

Results: We found that the combination <strong>of</strong> different data types<br />

drastically improves the predictive power <strong>of</strong> models built for AD. A<br />

model integrating gene expression and sMRI yielded an accuracy <strong>of</strong><br />

81% for controls and 100% for MCI cases (controls vs. MCI), 76%<br />

for controls and 83% for AD samples (controls vs. AD), and 92% for<br />

MCI samples and 72% for AD samples (MCI vs. AD). Furthermore<br />

we found that regression models using protein plasma measurements<br />

demonstrate high potential for predicting the rate <strong>of</strong> decline in AD<br />

patients.<br />

Conclusions: This study demonstrated that classification and<br />

regression models integrating multi-modal data sets resulted in<br />

improved accuracy in comparison to models using single data types.<br />

Such models show potential for the future application <strong>of</strong> non-invasive<br />

biomarkers in AD clinical trials and the clinic. However, there are<br />

many considerations to be aware <strong>of</strong> and explored. These include<br />

identifying biomarkers that are specific and reflect the many routes to<br />

the same pathology.


PP246 PURINERGIC RECEPTOR P2RX7 GENE<br />

POLYMORPHISMS: ASSOCIATION STUDY WITH<br />

DEPRESSIVE SYMPTOMS AND MOLECULAR GENETIC<br />

ANALYSIS<br />

Z. Nemoda*(1), O. Abdul Rahman(1), A. Vereczkei(1), A.<br />

Somogyi(2), G. Faludi(3), M. Sasvari-Szekely(1)<br />

1. Department <strong>of</strong> Medical Chemistry, Molecular Biology and<br />

Pathobiochemistry, Semmelweis University 2. 2nd Department <strong>of</strong><br />

Internal Medicine, Semmelweis University 3. Department <strong>of</strong><br />

Psychiatry, Kútvölgyi Clinical Centre, Semmelweis University<br />

*zs<strong>of</strong>ia.nemoda@eok.sote.hu<br />

Introduction: The Gln460Arg polymorphism (rs2230912) <strong>of</strong> the<br />

P2RX7 (purinergic receptor P2X, ligand-gated ion channel, 7) gene<br />

has been indicated in major depressive and bipolar disorder (MDD,<br />

BPD). In our previous studies we reported increased anxiety and<br />

depression scores assessed by the Hospital Anxiety and Depression<br />

Scale (HADS) in patients carrying the 460Arg-variant (G-allele) in<br />

psychiatric and diabetic patient samples. However, recently published<br />

data did not support the role <strong>of</strong> this P2RX7 A/G single nucleotide<br />

polymorphism (SNP). Therefore, we analyzed SNPs in the 3’<br />

untranslated region (UTR) <strong>of</strong> the P2RX7 gene which might affect<br />

putative miRNA binding and are in linkage with the Gln460Arg.<br />

Methodology: The in silico search for polymorphisms in putative<br />

microRNA target sites was carried out by the PolymiRTS<br />

(http://compbio.utmem.edu/miRSNP/) and the Patrocles<br />

(http://www.patrocles.org/) databases. Three A/C SNPs were<br />

identified as possible candidates in the P2RX7 3’ UTR region at<br />

positions 2164-2166, 689 bp downstream from the Gln460Arg. Since<br />

there was only one source <strong>of</strong> information about the 2166 A/C SNP<br />

(rs28969482) indicating 5% minor allele frequency, but the<br />

sequencing <strong>of</strong> 22 random DNA samples did not show polymorphic<br />

site in our patient population, only the 2164 A/C SNP (rs1653625)<br />

and the 2165 A/C SNP (rs1718106) were analyzed. These two SNPs<br />

were genotyped by RFLP technique using primers with mismatches<br />

to create restriction enzyme cleavage sites. The Haploview program<br />

indicated high linkage among these SNPs and Gln460Arg<br />

(rs2230912). In the genetic association analyses the depressive and<br />

anxiety scales <strong>of</strong> the HADS questionnaire was used both in the<br />

psychiatric patient sample (N=154, women 74.7%, average age<br />

47.9±10.8) and in the diabetic patient sample (N=246, women 50.4%,<br />

average age 58.2±13.3). In the molecular analyses HEK cells were<br />

transfected by the pMIR vector constructs containing the P2RX7 3’<br />

UTR downstream <strong>of</strong> the luciferase gene.<br />

Results: The 2164 A/C showed an association with the severity<br />

scores <strong>of</strong> anxiety and depressive symptoms in both populations:<br />

carrying the 2164 A-allele showed a step-wise increase in the 3<br />

genotype groups (depressed patients: anxiety: p=0.017, depression:<br />

p


PHARMACOGENOMICS<br />

PP248 MESOLIMBIC HYPODOPAMINERGIC FUNCTION A<br />

POTENTIAL NUTRIGENOMIC THERAPEUTIC TARGET<br />

FOR DRUG CRAVING AND RELAPSE<br />

K. Blum*(1), M. Miller(2), K. Perrine(3), Y. Liu(1), J. Gordano(4),<br />

M. Oscar-Berman(5)<br />

1. University <strong>of</strong> Florida 2. LifeStream, Inc 3. Cornel University 4. G<br />

&G Holistic Addiction Treatment Center 5. Boston University<br />

*drd2gene@aol.com<br />

Introduction: Dopamine plays an important role in drug seeking<br />

behavior. It is well established that Substance Use Disorder (SUD) is<br />

inheritable and runs in families. Hypodopaminergic function is a<br />

major culprit in multiple addictive behaviors and is regulated by a<br />

number <strong>of</strong> reward genes.<br />

Methodology: In this study we evaluated a known natural<br />

Dopaminergic agonist KB220IV along with oral[KB220] variants<br />

directed to overcoming hypodopaminergic function by assessing pre<br />

and post chronic symptoms and gene testing.<br />

Results: In our first experiment we found a significant reduction <strong>of</strong><br />

chronic symptoms as measured by Chronic Abstinence Symptom<br />

Severity [CASS] Scale for both IV and oral compared to only oral<br />

administration. Specifically, the IV and oral group did significantly<br />

better than the oral only group over the first week as well as over the<br />

30 day period. In the second experiment consisting <strong>of</strong> 129 subjects<br />

receiving both IV and orals three factors with eigenvalues greater<br />

than one were extracted for the baseline CASS variables. Three<br />

scales were constructed based on this factor analysis: Emotion,<br />

Somatic, and Cognitive. Paired sample t-tests between the pretreatment<br />

scales and the post-treatment scales were calculated. All<br />

three scales showed significant declines (p=.00001) from pre- to<br />

post-treatment: t=19.1 for Emotion, t=16.1 for Somatic, and t=14.9<br />

for Cognitive. In a two year follow-up on 23 subjects who underwent<br />

KB220IV therapy (at least five IV treatments over a 7 day period)<br />

plus orals for at least 30days: 21 (91%) were sober at 6 months with<br />

19 (82%) having no relapse 19 (82%) were sober at one -year with 18<br />

(78%) having no relapse 21 (91%) were sober at two-years post –<br />

treatment with 16 (70%) having no relapse. We also found that in two<br />

independent treatment centers ( heroin addicts in China<br />

Psychistimulant abusers in Florida) that 100% <strong>of</strong> all subjects tested<br />

for 6 polymorphic genes involved in dopaminergic neurotransmission<br />

had at least one risk allele and 74% <strong>of</strong> these subjects had moderate to<br />

high genetic risk as measured by the Genetic Addiction Risk Score<br />

(GARS) test. Moreover, through fMRI we are also showing<br />

significant dopaminergic activation <strong>of</strong> a number <strong>of</strong> dopaminergic<br />

brain regions with KB220Z compared to placebo.<br />

Conclusions: While awaiting additional required research, we<br />

cautiously propose that KB220IV therapy (a putative dopaminergic<br />

agonist) may provide early important therapeutic outcomes in<br />

residential treatment programs. These data agree with our recent<br />

KB220Z neuroimaging studies showing qEEG regulation <strong>of</strong> the<br />

cingulate gyrus abnormal dysynchronization in protracted abstinent<br />

psychostimulant addicts especially in DRD2 A1 allele carriers. We<br />

have published earlier showing an increase in alpha activity and an<br />

increase in low beta bands using KB220Z. These results suggest that<br />

following extensive further investigation KB220Z and KB220IV may<br />

be important therapeutic agents to combat drug craving behavior and<br />

potentially relapse by targeting hypodopaminergic deficits<br />

196<br />

PP249 CORRELATION BETWEEN AMPLIFIABLE HUMAN<br />

SALIVA-DERIVED DNA AND AFFYMETRIX ® DMET <br />

ARRAY GENOTYPING CALL RATE IN PATIENTS WITH<br />

SEVERE PSYCHIATRIC DISORDERS<br />

K. Nelson*(1), E. Ehli(1, 2), P. Huizenga(1), T. Soundy(2), G.<br />

Davies(1, 2), Y. Hu(1)<br />

1. Avera Institute for Human Behavioral <strong>Genetics</strong>, Avera McKennan<br />

Hospital and University Health Center 2. Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> South Dakota, Sanford School <strong>of</strong> Medicine<br />

*kelly.nelson@avera.org<br />

Introduction: Affymetrix ® DMET TM (drug metabolism enzymes and<br />

transporters) array is the first assay to <strong>of</strong>fer a comprehensive<br />

representation <strong>of</strong> known pharmacokinetic markers and enables fast<br />

discovery and measurement <strong>of</strong> genetic variation associated with drug<br />

response. Human saliva is an important source for DNA as it is a<br />

painless, convenient, and inexpensive collection method. There is<br />

concern, however, <strong>of</strong> point source microbial contamination inherent<br />

in the human saliva and how it may interfere with array genotyping<br />

call rates (a critical quality control standard). To date, there are no<br />

such reports demonstrating the effect <strong>of</strong> DNA derived from human<br />

saliva on the DMET array genotyping call rates.<br />

Methodology: Saliva samples (n=42) were collected from patients<br />

with mental disorders at the South Dakota Developmental Center.<br />

After genomic DNA extraction, we used a commercially available<br />

RNase P kit to identify the percentage <strong>of</strong> amplifiable human DNA in<br />

each sample. These samples were processed on DMET<br />

arrays following the manufacturer's protocol. A sample subset (n=7)<br />

was also processed on arrays at varying concentrations to determine<br />

the effect DNA concentration had on genotyping call rates. All<br />

samples were analyzed following the manufacturer's<br />

instructions using DMET console s<strong>of</strong>tware with Fixed and Dynamic<br />

Genotype Boundary settings.<br />

Results: Genomic samples containing >28.1% <strong>of</strong> amplifiable human<br />

DNA passed the genotyping call rate threshold <strong>of</strong> 98% when<br />

analyzed by DMET console s<strong>of</strong>tware with Dynamic Genotype<br />

Boundaries. When analyzed using DMET console s<strong>of</strong>tware with<br />

Fixed Genotype Boundaries, 82.4% <strong>of</strong> saliva samples containing<br />

>41.6% <strong>of</strong> amplifiable human DNA passed the genotyping call rate<br />

threshold. Overall, a higher percent <strong>of</strong> amplifiable human<br />

DNA present in the saliva sample had a correspondingly higher<br />

genotyping call rate regardless <strong>of</strong> which Boundary analysis<br />

setting was utilized. Increasing the concentration <strong>of</strong> the extracted<br />

saliva-derived DNA prior to DMET use did not significantly<br />

(p>0.05) improve the genotyping call rate. We believe this was due<br />

in part to the increasing ratio <strong>of</strong> competing microbial DNA remaining<br />

in the saliva derived DNA sample.<br />

Conclusions: Currently, we are genotyping blood-derived DNA from<br />

the same individuals in this study to compare saliva and bloodderived<br />

DNA performance on DMET arrays. While blood-derived<br />

DNA continues to be the standard, this study may provide<br />

a screening protocol and thus a potential improvement to the<br />

genotyping call rate for human saliva DNA samples as a source<br />

material for DMET arrays.


PP250 INFLUENCE OF TPH2, DAOA AND BDNF VARIANTS<br />

ON DIAGNOSIS AND RESPONSE TO TREATMENT IN<br />

PATIENTS WITH MAJOR DEPRESSION, BIPOLAR<br />

DISORDER AND SCHIZOPHRENIA<br />

A. Chiesa(1), C. Pae(2,3), D. De Ronchi(1), A. Serretti*(1)<br />

1. Istitute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna 2. Catholic University<br />

<strong>of</strong> Korea College <strong>of</strong> Medicine 3. Department <strong>of</strong> Psychiatry and<br />

Behavioral Sciences<br />

*albertopnl@yahoo.it<br />

Introduction: Current evidence suggests that several genes including<br />

the tryptophan hydroxylase 2 (TPH2), the D-amino acid oxidase<br />

activator (DAOA) and the brain derived neurotrophic factor (BDNF)<br />

could be involved in the etiology and response to treatment <strong>of</strong> several<br />

psychiatric disorders. Accordingly, we aimed to investigate whether<br />

some SNPs within these genes, including rs4570625, rs10748185,<br />

rs11179027, rs1386498, rs4469933, rs17110747 (TPH2), rs3916966,<br />

rs3916967, rs2391191, rs3916968, rs7139958, rs9558571, rs778293<br />

(DAOA), rs2030324, rs7103873, rs10835210, rs11030101 and rs6265<br />

(BDNF) could be associated with major depression (MD), bipolar<br />

disorder (BD) and schizophrenia and whether they could predict<br />

clinical outcomes in Korean in-patients treated with antidepressants,<br />

mood stabilizers and antipsychotics respectively.<br />

Methodology: One hundred forty five patients with MD, 132 patients<br />

with BD, 221 patients with schizophrenia and 170 psychiatrically<br />

healthy controls were genotyped for the SNPs mentioned above.<br />

Baseline and final clinical measures, including MADRS, YMRS and<br />

PANSS scores for patients with MD, BD and schizophrenia<br />

respectively were recorded.<br />

Results: Rs10835210 CA and rs11030101 AT genotype frequencies<br />

were higher in BD and schizophrenia patients than in healthy and<br />

MD subjects. In patients with MD, rs4570625-rs10748185 G-A<br />

haplotype was associated with higher endpoint MADRS severity.<br />

Additionally, Rs7139958 AA and rs9558571 TT genotypes as well as<br />

rs7139958 A and rs9558571 T alleles were associated with higher<br />

baseline PANSS positive subscale scores in patients with<br />

schizophrenia.<br />

Conclusions: Some associations have been observed between genetic<br />

variants under investigation and specific psychiatric disorders and<br />

clinical outcomes. However, current evidence is far from being<br />

conclusive and further replications are required.<br />

197<br />

PP251 A PRELIMINARY INVESTIGATION OF CREB1,<br />

PTGS2, GRIK4 AND GNB3 POLYMORPHISMS ON<br />

TREATMENT RESISTANCE IN MAJOR DEPRESSION<br />

A. Serretti*(1), A. Chiesa(1), C. Crisafulli(2), R. Calati(1), I.<br />

Massat(3), S. Linotte(4), S. Kasper(5), I. Antonijevic(6), C.<br />

Forray(6), L. Snyder(6), J. Bollen(7), J. Zohar(8), D. De Ronchi(1),<br />

D. Souery(9), J. Mendlewicz(4)<br />

1. Institute <strong>of</strong> Psychiatry, University <strong>of</strong> Bologna 2. University <strong>of</strong><br />

Messina 3. Neurological Experimental Laboratory 4. Universite´<br />

Libre de Bruxelles 5. Department <strong>of</strong> Psychiatry and Psychotherapy,<br />

Medical University 6. Translational Research, Lundbeck Research 7.<br />

Sint-Truiden, <strong>Psychiatric</strong> Center 8. Chaim Sheba Medical Center 9.<br />

Laboratoire de Psychologie Medicale, Universite´ Libre de Bruxelles<br />

and Psy Pluriel, Centre Europe´en de Psychologie Medicale<br />

*albertopnl@yahoo.it<br />

Introduction: Several genes including those coding for the<br />

transcription factor cyclic adenosine monophosphate response<br />

element binding (CREB1) protein, the prostaglandin-endoperoxide<br />

synthase enzyme 2 (PTGS2), the glutamate ionotropic kainate 4<br />

receptor (GRIK4) and the guanine nucleotide binding protein beta<br />

polypeptide 3 (GNB3) have been repeatedly involved in the aetiology<br />

and response to treatments <strong>of</strong> major depression (MD). Accordingly,<br />

The aim <strong>of</strong> the present paper is to investigate whether a set <strong>of</strong> single<br />

nucleotide polymorphisms (SNPs) within CREB1 (rs2709376,<br />

rs2253206, rs7569963, rs7594560, rs4675690),PTGS2(rs4648276,<br />

rs2066826 and rs689466), GRIK4 (rs1954787) and GNB3 (rs5443)<br />

geneswas associated with MD as well with antidepressant response,<br />

remission and treatment resistance.<br />

Methodology: 194 MD patients and 76 healthy controls treated with<br />

antidepressants at adequate doses for at least 4 weeks were genotyped<br />

for CREB1, PTGS2, GRIK4and GNB3SNPs. Response, remission and<br />

treatment resistance were recorded.<br />

Results: A allele <strong>of</strong> rs7569963 as well as rs2253206-rs7569963 A-A<br />

and rs7569963-rs4675690 A-C haplotypes in CREB1 were<br />

significantly associated with treatment resistance. No significant<br />

association was observed between any <strong>of</strong> the remaining genotypic,<br />

allelic and haplotypic variants under investigation and outcomes <strong>of</strong><br />

interest.<br />

Conclusions: Some genetic polymorphisms in CREB1 could<br />

influence treatment resistance to antidepressants. On the other hand,<br />

there is no evidence suggesting any association between CREB1,<br />

PTGS2, GRIK4and GNB3andtreatment response or remission and<br />

with MD. However, on account <strong>of</strong> the several limitations <strong>of</strong> the<br />

present study including a relatively small sample size and the<br />

incomplete coverage <strong>of</strong> genes under investigation, further research is<br />

needed.


PP252 PHARMACOGENETICS: FROM RESEARCH TO<br />

CLINICAL PRACTICE-COST-UTILITY ANALYSIS<br />

P. Olgiati*(1), A. Serretti(1), E. Bajo(2), M. Bigelli(2)<br />

1. Department <strong>of</strong> Psychiatry - University <strong>of</strong> Bologna 2. Department<br />

<strong>of</strong> Management and Economy - University <strong>of</strong> Bologna<br />

*polgiati@hotmail.com<br />

Introduction: Pharmacogenetics may represent a valuable tool to<br />

improve drug choice by identifying likely responders a priori,<br />

however the exact benefit in routine activity has been scarcely<br />

investigated.We performed a cost-utility analysis <strong>of</strong> incorporating 5-<br />

HTTLPR genetic test in antidepressant treatment selection.<br />

Methodology: We implemented a theoretical analytic model to<br />

compare the cost-utility <strong>of</strong> two treatment strategies for major<br />

depressive disorder (MDD) within a 12-weeks trial: A. SSRI<br />

monotherapy B. alternative administration <strong>of</strong> serotonergic or<br />

noradrenergic antidepressants under guidance from 5-HTTLPR<br />

polymorphism. Patients with the long-allele, who are more likely to<br />

respond to SSRI treatment, received citalopram whereas s-s allele<br />

carriers were treated with noradrenergic dopaminergic agent<br />

bupropion. The model was based on Italian Mental Health setting.<br />

Costs (international dollars <strong>of</strong> 2010) were derived from <strong>World</strong> Health<br />

Organization CHOICE project and Italian <strong>of</strong>ficial sources. The effect<br />

size <strong>of</strong> 5-HTTLPR test was estimated from a meta-analysis <strong>of</strong><br />

pharmacogenetic trials.<br />

Results: The use <strong>of</strong> genetic test increased remitters by 5%, with an<br />

estimated gain <strong>of</strong> 0.07 Quality-Adjusted Life Weeks. This projected<br />

ICER to $ 3,269 (largely above cost-effectiveness threshold), but<br />

only for the worse case scenario, while the distribution was largely<br />

below threshold. Sensitivity analysis demonstrated the strongest<br />

impact <strong>of</strong> genetic test cost on ICER. Quality <strong>of</strong> life (HRQL) assigned<br />

to untreated depression and 5-HTTLPR effect size were less<br />

important contributors. Modeling variations in these parameters,<br />

pharmacogenetic approach became cost-effective. Under base-case<br />

conditions genetic test should cost $75 or less to drop below costeffectiveness<br />

threshold. In severe depression with low HRQL the cost<br />

<strong>of</strong> genetic test could raise to $270.<br />

Conclusions: To date performing a pharmacogenetic test before<br />

starting antidepressant treatment may be moderately cost-effective in<br />

selected groups <strong>of</strong> patients with severe and disabling MDD.<br />

Pharmacogenetics may become available for clinical practice if<br />

prediction power <strong>of</strong> tests is improved and costs drop to $50 - $100.<br />

198<br />

PP253 ASSOCIATION BETWEEN COMT POLYMORPHISMS<br />

AND CLINICAL RESPONSE TO RISPERIDONE<br />

TREATMENT: A PHARMACOGENETIC STUDY<br />

Q. Zhao(1,2,3), B. Liu(1,2,3), J. Zhang(1,2,3), L. Wang(1,2,3), X.<br />

Li(1,2,3), Y. Wang(1,2,3), L. He(1,2,3), G. He(1,2,3)<br />

1. Bio-X Center, Key Laboratory for the <strong>Genetics</strong> <strong>of</strong> Developmental<br />

and Neuropsychiatric Disorders (Ministry <strong>of</strong> Education), Shanghai<br />

Jiao Tong University 2. Institutes <strong>of</strong> Biomedical Sciences, Fudan<br />

University, 138 Yixueyuan Road 3. Institute for Nutritional Sciences,<br />

Shanghai Institutes <strong>of</strong> Biological Sciences, Chinese Academy <strong>of</strong><br />

Sciences, 320 Yueyang Road<br />

Introduction: Catechol-O-methyl transferase (COMT) is one <strong>of</strong> the<br />

genes which confer susceptibility to schizophrenia, because <strong>of</strong> its role<br />

in neurotransmitter metabolism and its location in the high risk<br />

schizophrenia related region 22q11. Recent studies also found that<br />

COMT functional polymorphisms influenced individual response to<br />

anti-psychotic medication. Our aim in this study was to explore the<br />

influence <strong>of</strong> COMT polymorphisms on pharmaceutical response to<br />

risperidone in the Chinese population.<br />

Methodology: 130 Chinese schizophrenia patients (85 females) were<br />

enrolled in the study. Clinical efficacy was determined using Brief<br />

<strong>Psychiatric</strong> Rating Scores (BPRS). We genotyped 10 single<br />

nucleotide polymorphisms (SNPs) <strong>of</strong> COMT in our patients and reexamined<br />

them for association with changes in BPRS scores after 8<br />

weeks <strong>of</strong> riperidone monotherapy. Written informed consent was<br />

obtained from either the participants or the participants' legal<br />

representatives after the research aims and procedures were fully<br />

explained. The study protocol was reviewed and approved by the<br />

Shanghai Ethical Committee <strong>of</strong> Human Genetic Resources.<br />

Results: Statistical analysis revealed an association between an<br />

upstream COMT SNP, rs9606186 and scores reduction <strong>of</strong> BPRS in<br />

all patients and male subgroup, but not in female subgroup (Allele<br />

analysis: p=0.055 for all, p=0.012 for male Genotype analysis:<br />

p=0.046 for all, p=0.020 for male, uncorrected). Haplotype analysis<br />

showed a highly significant association between COMT SNP<br />

haplotypes and drug efficiency in our patient sample (p


PP254 GENOME-WIDE ASSOCIATION MAPPING OF LOCI<br />

FOR ANTIPSYCHOTIC-INDUCED EXTRAPYRAMIDAL<br />

SYMPTOMS IN MICE<br />

J. Crowley*(1,2), Y. Kim(2), C. Quackenbush(2), A. Pratt(2), D.<br />

Adkins(3), E. van den Oord(3,4), M. Bogue(5), H. McLeod(1), P.<br />

Sullivan(2,6)<br />

1. Institute for Pharmacogenomics and Individualized Therapy 2.<br />

Department <strong>of</strong> <strong>Genetics</strong> 3. Center for Biomarker Research and<br />

Personalized Medicine 4. Virginia Institute for <strong>Psychiatric</strong> and<br />

Behavioral <strong>Genetics</strong> 5. The Jackson Laboratory 6. Department <strong>of</strong><br />

Psychiatry<br />

*crowley@unc.edu<br />

Introduction: Tardive dyskinesia (TD) is a debilitating,<br />

unpredictable and <strong>of</strong>ten irreversible side effect resulting from chronic<br />

treatment with a typical antipsychotic agent, such as haloperidol. It is<br />

characterized by repetitive, involuntary, purposeless movements<br />

primarily <strong>of</strong> the or<strong>of</strong>acial region. Human pharmacogenomic studies<br />

<strong>of</strong> TD are needed to allow personalized treatment, but have been<br />

underpowered by the relatively small number <strong>of</strong> patient samples<br />

available.<br />

Methodology: Therefore, we used a validated mouse model <strong>of</strong> TD<br />

(vacuous chewing movements, VCMs) for a systems genetics<br />

analysis geared toward detecting genetic predictors <strong>of</strong> TD in human<br />

patients. Previous work from our laboratory has shown that<br />

susceptibility to chronic haloperidol-induced VCMs is a highly<br />

heritable trait in mice, under human-like steady state drug levels. In<br />

the current study, phenotypic data from 27 inbred strains chronically<br />

treated with haloperidol were subject to a comprehensive genomic<br />

analysis involving 426,493 SNPs, 4,047 CNVs, brain gene<br />

expression, along with gene network and bioinformatic analysis.<br />

Results: Our results identified ~50 genes that we expect to have high<br />

prior probabilities for association with haloperidol-induced TD, most<br />

<strong>of</strong> which have never been tested for association with human TD. For<br />

example, among our top hits were two glutamate receptors (Grin1,<br />

Grin2a), a regulator <strong>of</strong> brain development (Zic4) and an indirect<br />

target <strong>of</strong> haloperidol (Drd1a) that has not been as well studied as the<br />

direct target, Drd2.<br />

Conclusions: Not only is 50 genes a manageable number for<br />

candidate-gene pharmacogenetic studies in human patients, but it also<br />

provides a logical rationale for focusing on just a small fraction<br />

(0.2%) <strong>of</strong> the genomic search space, reducing the multiple testing<br />

penalty 500-fold.<br />

199<br />

PP255 RELAXIN POLYMORPHISMS ASSOCIATED WITH<br />

METABOLIC DISTURBANCE IN PATIENTS TREATED<br />

WITH ANTIPSYCHOTICS<br />

M. Arranz*(1), O. Skrobot(2), V. Kay(3), J. de Leon(4), A.<br />

Blakemore(5), J. Munro(6)<br />

1. Institute <strong>of</strong> psychiatry-King's College London 2. Institute <strong>of</strong><br />

Clinical Neurosciences-University <strong>of</strong> Bristol 3. Insitute <strong>of</strong><br />

Neurology-UCL 4. University <strong>of</strong> Kentucky 5. Imperial College 6.<br />

Optimal Medicine<br />

*maria.arranz@kcl.ac.uk<br />

Introduction: People with schizophrenia have an increased risk <strong>of</strong><br />

metabolic syndrome, with consequent elevated morbidity and<br />

mortality largely due to cardiovascular disease. Metabolic disorders<br />

comprise obesity, dyslipidemia and elevated levels <strong>of</strong> triglycerides,<br />

hypertension and disturbed insulin and glucose metabolism.The<br />

elevated risk <strong>of</strong> metabolic syndrome in individuals suffering from<br />

schizophrenia is believed to be multi-factorial, related to a genetic<br />

predisposition, lifestyle characteristics and treatment with<br />

antipsychotic medications. Relaxin 3 (RLN3, also known as INSL7)<br />

is a recently identified member <strong>of</strong> the insulin/relaxin superfamily that<br />

plays a role in the regulation <strong>of</strong> appetite and body weight<br />

control.RLN3 stimulates relaxin-3 receptor 1 (relaxin/insulin-like<br />

family peptide receptor 3, RXFP3) and relaxin receptor 2<br />

(relaxin/insulin-like family peptide receptor 4, RXFP4).<br />

Methodology: We have investigated the role <strong>of</strong> ten polymorphisms<br />

in these genes (RLN3 rs12327666, rs1982632, and rs7249702,<br />

RLN3R1 rs42868, rs6861957, rs7702361, and rs35399, and RLN3R2<br />

rs11264422, rs1018730and rs12124383) in the occurrence <strong>of</strong><br />

metabolic syndrome phenotypes (obesity, diabetes,<br />

hypercholesterolemia, hypertrigyceridemia, and hypertension) in a<br />

cross-sectional cohort <strong>of</strong> 419 US Caucasian patients treated with<br />

antipsychotic drugs.<br />

Results: We found several associations between relaxin<br />

polymorphisms and hypecholesterolemia (p


PP256 A MULTIDISCIPLINARY PHARMACOGENOMIC<br />

TREATMENT APPROACH TO REDUCE MEDICATION<br />

BURDEN AND IMPROVE SUBJECT OUTCOMES IN A<br />

RURAL DEVELOPMENTAL CENTER<br />

K. Bohlen*(1,2), B. Bradley(3), E. Kutscher(3,4,5), T. Soundy(3,4),<br />

E. Ehli(1,2), Y. Hu(1,2), G. Davies(1,2,3,4,5)<br />

1. Avera Research Inst 2. Avera Inst for Human Behavioral <strong>Genetics</strong><br />

3. Avera Behavioral Health Ctr 4. Univ <strong>of</strong> South Dakota Department<br />

<strong>of</strong> Psychiatry 5. South Dakota State Univ<br />

*krista.bohlen@avera.org<br />

Introduction: Saliva and blood samples were collected from<br />

residents at a developmental center in South Dakota. The<br />

samples were genotyped using the Affymetrix Drug Metabolism<br />

Enzyme and Transporter (DMET) array. In addition, variable<br />

number tandem repeats (VNT-R) in various psychiatric candidate<br />

genes were also genotyped to assess subject pharmacogenomics<br />

(Avera Health IRB 2009.053). Residents <strong>of</strong> this center suffer from<br />

psychiatric disorders, mental retardation, and other debilitating<br />

syndromes. The aim <strong>of</strong> this project is to improve patient outcomes<br />

through treatment regimens based on pharmacogenomic analysis.<br />

Methodology: This non-randomized, retrospective and prospective<br />

comparison, pro<strong>of</strong>-in-concept study utilized the DMET array which<br />

analyzes 1,936 drug metabolism markers and transporters on<br />

225 genes. A multidisciplinary team consisting <strong>of</strong> a psychiatrist,<br />

pharmacists, a medical geneticist, and genetics lab scientists<br />

collaborated on this project to provide a database for analysis <strong>of</strong><br />

DMET SNP results. The pharmacists and psychiatrist evaluated the<br />

current medication regimens, past medications and effects, DSM IV<br />

axes, AIMS scores, vital signs, and laboratory values for clinical<br />

recommendations guided by pharmacogenomic results. The primary<br />

polymorphisms analyzed were in CYP 450 enzymes including<br />

CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and<br />

CYP3A4. Additional analyses <strong>of</strong> a SNP in COMT, the 5HTTLPR<br />

polymorphism in SERT, and DRD4 48-bp VNT-R in exon 3 were<br />

also evaluated. The medical geneticist and lab staff designed primers<br />

for PCR analysis to clarify ambiguous allele calls by the DMET array<br />

and provide confirmatory results. Inconclusive results due to<br />

unknown alleles resulted in no recommendation given. The<br />

medications were evaluated in regards to the genomic results and<br />

resultant enzyme function. A concise recommendation document is<br />

provided to the treatment team which consists <strong>of</strong> drug-drug<br />

interactions, pharmacogenetic considerations, therapeutic duplication,<br />

drug monitoring, and other considerations (including<br />

pharmacoeconomic implications).<br />

Results: Fourty-two buccal-derived DNA and blood-derived DNA<br />

have been run on the DMET array. Using the genetic data, a<br />

personalized approach to medication management has been analyzed<br />

in order to make clinical recommendations. The treatment team has<br />

the discretion as to which recommendations to implement or<br />

disregard. The first recommendations were given in February 2011.<br />

For example, one <strong>of</strong> the patients is a CYP1A2 fast metabolizer<br />

(inducible) who is on olanzapine. In a group <strong>of</strong> 10 patients, 4 are fast<br />

(inducible) metabolizers for CYP1A2. In that same group, 1 is a poor<br />

metabolizer for CYP2D6, 4 are intermediate, and 5 are normal.<br />

The patients will be assessed at six months following the treatment<br />

team recommendations. Any <strong>of</strong> those<br />

recommendations implemented will be evaluated for impact on the<br />

patient's outcomes as well as degree <strong>of</strong> implementation.<br />

Conclusions: The multidisciplinary team approach has provided<br />

insight from the psychiatrist, evaluation <strong>of</strong> pharmacogenomic results<br />

by the pharmacists, and expertise from the genetics lab staff. We<br />

hope to impact patients' lives by lessening the treatment burden,<br />

decreasing adverse effects, and ultimately improving patient<br />

outcomes through personalized medicine.<br />

200<br />

PP257 ANALYSIS OF 34 CANDIDATE GENES IN<br />

BUPROPION AND PLACEBO REMISSION<br />

A. Tiwari*(1), A. Yeo(2), M. Chiano(2), F. Tozzi(3), G. Sajeev(1),<br />

C. Zai(1), T. Arenovich(1), D. Müller(1), J. Ascher(4), J. Kennedy(1)<br />

1. Neurogenetics Section, Neuroscience Department, Centre for<br />

Addiction and Mental Health 2. GlaxoSmithKline 3.<br />

GlaxoSmithKline 4. GlaxoSmithKline<br />

*arun_tiwari@camh.net<br />

Introduction: There is considerable variability in the rate <strong>of</strong><br />

response and remission following treatment with antidepressant drugs<br />

or placebo in patients. Bupropion, a commonly prescribed<br />

antidepressant, is a dual norepinephrine and dopamine transporter<br />

inhibitor.<br />

Methodology: A total <strong>of</strong> 532 tagging single nucleotide<br />

polymorphisms (SNPs) in 34 candidate genes (monoaminergic<br />

pathways and Hypothalamic-pituitary-adrenal axis) were investigated<br />

for association with remission and response in patients with major<br />

depressive disorder (MDD). These patients were from four GSK<br />

funded clinical trials in the USA in which they were treated with<br />

either bupropion (n=319) or placebo (n=257). Analyses were<br />

performed using conditional logistic regression. Gene-wide<br />

corrections for multiple testing were carried out.<br />

Results: Significant association was observed for remission<br />

following treatment with bupropion with a SNP within the serotonin<br />

receptor 2A gene (HTR2A, rs2770296, p corrected=0.02) and marginal<br />

evidence for association within the angiotensin converting enzyme<br />

gene (ACE, rs8075924, pcorrected=0.054). Response to bupropion<br />

treatment was also significantly associated with a SNP in the<br />

dopamine transporter gene (SLC6A3; rs6347, pcorrected=0.013) and<br />

marginally associated with another variant within the vesicular<br />

monoamine transporter 2 gene (SLC18A2; rs363225, pcorrected=0.059).<br />

Among the patients that received placebo, marginal association for<br />

remission was observed between a SNP in HTR2A (rs2296972,<br />

pcorrected=0.055) as well as in the serotonin transporter (SLC6A4,<br />

rs4251417, pcorrected=0.050). Placebo response was associated with<br />

SNPs in the glucocorticoid receptor gene (NR3C1; rs1048261,<br />

pcorrected=0.040) and monoamine oxidase A gene (MAOA; rs6609257,<br />

pcorrected=0.046).<br />

Conclusions: These results suggest an important role for HTR2A in<br />

remission to both bupropion and placebo treatments. In accordance<br />

with bupropion pharmacology, response may be associated with<br />

genetic variation in dopamine and vesicular monoamine transporters.<br />

Placebo response was associated with polymorphisms inNR3C1 and<br />

MAOA.


PP258 PHENOTYPIC DEFINITION OF RESPONSE TO<br />

LITHIUM TREATMENT IN THE CONLIGEN CONSORTIUM<br />

M. Manchia*(1), M. Bauer(2), F. McMahon(3), T. Schulze(4), M.<br />

Alda(1)<br />

1. Department <strong>of</strong> Psychiatry, Dalhousie University 2. Department <strong>of</strong><br />

Psychiatry and Psychotherapy, Universitätsklinikum Carl Gustav<br />

Carus, Technische Universität 3. Human <strong>Genetics</strong> Branch, Intramural<br />

Research Program, National Institute <strong>of</strong> Mental Health, NIH, US<br />

DHHS 4. Department <strong>of</strong> Psychiatry and Psychotherapy, University<br />

Medical Center, Georg-August-Universität 5. Consortium on Lithium<br />

<strong>Genetics</strong> (ConLiGen)<br />

*mirko.manchia@dal.ca<br />

Introduction: Bipolar disorder (BD) is a psychiatric illness with a<br />

complex pattern <strong>of</strong> genetic susceptibility. Innovative approaches such<br />

as genome wide association studies (GWAS) in BD have identified<br />

mostly variants with small effect on disease risk. It is possible that<br />

individually rare variants with relatively larger effect than common<br />

variants may account for a large fraction <strong>of</strong> the genetic susceptibility.<br />

The lack <strong>of</strong> conclusive genetic findings might be also related to the<br />

phenotypic heterogeneity presented by BD. In this context, studying<br />

more homogenous subgroups <strong>of</strong> patients, such as responders to<br />

lithium treatment, might represent an effective strategy for<br />

identifying genetic risk variants <strong>of</strong> the illness as well as determinants<br />

<strong>of</strong> treatment response. The Consortium on Lithium <strong>Genetics</strong><br />

(ConLiGen) has been created as an international multi-centre<br />

collaboration investigating the genetic basis <strong>of</strong> response to lithium<br />

treatment. Currently, ConLiGen is undertaking a GWAS on a sample<br />

<strong>of</strong> ~1,500 BD patients characterized for lithium response. In this<br />

context, it is important to assess the key phenotypic measures and the<br />

response to long-term lithium treatment reliability across the<br />

participating centres.<br />

Methodology: We measured the inter-rater reliability <strong>of</strong> the response<br />

definition based on a 0-10 scale published and validated previously<br />

(Gr<strong>of</strong> et al. 2002, Garnham et al. 2007). Fifty-eight investigators from<br />

20 participating sites rated a set <strong>of</strong> 12 standardized case vignettes. We<br />

analyzed the concordance <strong>of</strong> ratings by means <strong>of</strong> Kappa (κ) and intraclass<br />

correlation coefficient (ICC). In addition, we performed mixture<br />

analysis <strong>of</strong> the empirical distribution <strong>of</strong> the total scale score available<br />

for 1,336 patients using minimum message length (MML) and<br />

frequentist Gaussian modeling. For the latter analysis we chose the<br />

best fitting model according to the lowest values <strong>of</strong> Akaike<br />

Information Criterion (AIC) and Bayesian Information Criterion<br />

(BIC).<br />

Results: Kappa scores and ICC showed a substantial level <strong>of</strong><br />

agreement across sites (κ = 0.68, z = 96.1; single measure ICC =<br />

0.74). Frequentist mixture modelling <strong>of</strong> score distribution suggested<br />

the presence <strong>of</strong> a best fitting model <strong>of</strong> three normal curves (2<br />

component: AIC = 6,744.6, BIC = 6,756.4; 3 components: AIC<br />

6,600.5, BIC = 6,631.7; 4 components: AIC = 6,604.5; BIC =<br />

6,646.1) with the following parameters: non responders (mean =<br />

0.76, SD = 1.14, proportion = 0.38), partial responders (mean = 4.6,<br />

SD = 1.14, proportion = 0.29) and full responders (mean = 8.3, SD =<br />

1.14, proportion = 0.33). The three component model was confirmed<br />

by MML analysis. Both models suggested the cut-<strong>of</strong>f for full<br />

response at ≥7.<br />

Conclusions: These findings indicate a substantial inter-rater<br />

reliability <strong>of</strong> assessments <strong>of</strong> the response to lithium and provide<br />

analytical support for the definition <strong>of</strong> phenotypic definition <strong>of</strong><br />

lithium response. The results <strong>of</strong> the inter-rater reliability analysis <strong>of</strong><br />

additional 16 standardized vignettes, currently rated by ConLiGen<br />

investigators, will be presented at the conference.<br />

201<br />

PP259 GENETIC ASSOCIATION STUDY BETWEEN<br />

PROOPIOMELANOCORTIN AND MELANOCORTIN-4<br />

RECEPTOR GENE POLYMORPHISMS WITH<br />

ANTIPSYCHOTIC INDUCED WEIGHT GAIN<br />

N. Chowdhury*(1), A. Tiwari(1), R. Souza(4), C. Zai(1), S.<br />

Shaikh(1), S. Chen(1), F. Liu(1), J. Lieberman(2), H. Meltzer(3), D.<br />

Müller(1), J. Kennedy(1)<br />

1. The Centre for Addiction and Mental Health 2. Department <strong>of</strong><br />

Psychiatry Columbia University and the New York State <strong>Psychiatric</strong><br />

Institute 3. <strong>Psychiatric</strong> Hospital at Vanderbilt University 4. Health<br />

Sciences Graduate Program, Universidade do Extremo Sul<br />

Catarinense<br />

*nabilah.chowdhury@utoronto.ca<br />

Introduction: Antipsychotic induced weight gain (AIWG) may<br />

result in the metabolic<br />

syndrome in schizophrenia patients. The leptin-melanocortin pathway<br />

is important in food intake regulation and energy homeostasis. A<br />

recent genome wide associationstudy <strong>of</strong> (AIWG) found the highest<br />

main peak in the region <strong>of</strong> the melanocortin 4 receptor (MC4R) gene,<br />

at the downstream marker rs489693. The precursor polypeptide<br />

proopiomelanocortin (POMC) is cleaved to produce α-melanocyte<br />

stimulating hormone, which stimulates MC4R to regulate food intake<br />

and energy expenditure in the hypothalamus. Thus, we investigated<br />

the potential role <strong>of</strong> MC4R and POMC variants with antipsychotic<br />

induced weight gain<br />

Methodology: Four MC4R SNPs (rs2229616, rs17782313,<br />

rs11872992, rs8087522) and two POMC SNPs were genotyped in<br />

237 patients who underwent treatment for chronic schizophrenia and<br />

were evaluated for AIWG for up to 14 weeks. We compared weight<br />

change (%) across genotypic groups using analysis <strong>of</strong> variance and<br />

covariance for all SNPs (r2 ≥ 0.8).Variants were genotyped using<br />

ABI TaqMan assays. In the case <strong>of</strong> a positive association, we<br />

investigated in silico and in vitro for functional relevance <strong>of</strong> the SNP.<br />

Results: The rs2229616 SNP was monomorphic in our population<br />

and thus excluded from analyses. No significant genotypic or allelic<br />

associations were found between the MC4R rs11872992,<br />

rs17782313 or rs8087522 polymorphisms or the POMC rs1042571<br />

and rs6713532 polymorphisms and weight gain (p > 0.05). However,<br />

a subsequent analysis showed that patients <strong>of</strong> European ancestry who<br />

were carriers <strong>of</strong> the MC4R rs8087522 A-allele (AG+AA) on<br />

clozapine gained significantly more weight than non-carriers<br />

(p=0.027). Electrophoretic mobility shift assay suggested that the<br />

presence <strong>of</strong> the A-allele appears to create a transcription factorbinding<br />

site.<br />

Conclusions: In this study, we observed that the rs8087522 SNP <strong>of</strong><br />

the MC4R gene may be associated with AIWG in schizophrenia<br />

patients <strong>of</strong> European origin. This SNP is present in the promoter,<br />

alters transcription factor binding and therefore may affect MC4R<br />

expression. This observation warrants replication in an independent<br />

sample.


PP260 ASSOCIATION STUDY OF FUNCTIONAL<br />

POLYMORPHISMS IN NPY GENE WITH ANTIPSYCHOTIC-<br />

INDUCED BODY WEIGHT GAIN<br />

E. Brandl*(1), A. Tiwari(1), O. Likhodi(1), H. Meltzer(2), J.<br />

Kennedy(1), D. Müller(1)<br />

1. Neurogenetics Section, Centre for Addiction and Mental Health 2.<br />

2<strong>Psychiatric</strong> Hospital, Vanderbilt University<br />

*eva_brandl@camh.net<br />

Introduction: Significant body weight gain (BWG) is a serious sideeffect<br />

<strong>of</strong> a number <strong>of</strong> antipsychotic drugs. Previous studies have<br />

demonstrated an influence <strong>of</strong> clozapine, but not haloperidol on<br />

Neuropeptide Y (NPY) expression in the brain. Since NPY is a potent<br />

orexigenic peptide stimulating food intake and genetic variation <strong>of</strong><br />

the gene has been shown to influence development <strong>of</strong> obesity, we<br />

investigated the impact <strong>of</strong> NPY polymorphisms on antipsychoticinduced<br />

BWG.<br />

Methodology: We analyzed four polymorphisms in the NPY gene<br />

(rs16147, rs16475, rs5573 and rs5574) in schizophrenia subjects<br />

(n=80), undergoing first exposure to clozapine for 6 weeks.<br />

Association was tested using analysis <strong>of</strong> covariance with change (%)<br />

from baseline weight as the dependent variable and baseline body<br />

weight as covariate.<br />

Results: A significant association <strong>of</strong> rs16147 genotype with weight<br />

change was observed (p=.002) in patients <strong>of</strong> European ancestry.<br />

Carriers <strong>of</strong> the C-allele gained significantly more weight compared to<br />

individuals with TT-genotype (TC+CC vs. TT; 5.61±5.4 vs.<br />

0.32±4.8). Similarly, two other polymorphisms (rs5573 and rs5574)<br />

were also significantly associated with weight change (p=.009 and<br />

p=.022). The three associated polymorphisms are in high linkage<br />

disequilibrium with each other. There was no association <strong>of</strong> rs16475<br />

(p=.654).<br />

Conclusions: Our observation <strong>of</strong> association <strong>of</strong> NPY polymorphisms<br />

gives further evidence for a genetic influence on antipsychoticinduced<br />

BWG. The polymorphism rs16147, present in the promoter<br />

region, has been previously described to affect NPY expression both<br />

in vivo and in vitro. However, these results warrant replication in<br />

independent and larger samples.<br />

202<br />

PP261 GENETIC VARIATION IN BDNF IS ASSOCIATED<br />

WITH ANTIPSYCHOTIC TREATMENT RESISTANCE IN<br />

PATIENTS WITH SCHIZOPHRENIA<br />

J. Zhang*(1), T. Lencz(1), S. Geisler(1), P. DeRosse(1), E.<br />

Bromet(2), A. Malhotra(1)<br />

1. Zucker Hillside Hospital 2. Brook University School <strong>of</strong> Medicine<br />

*jzhang1@nshs.edu<br />

Introduction: Antipsychotic drugs are the mainstay <strong>of</strong> treatment for<br />

schizophrenia. However, a substantial proportion <strong>of</strong> patients are<br />

poorly responsive or resistant to treatment resistant to first-line<br />

treatments, and clozapine treatment is therefore <strong>of</strong>ten indicated.<br />

Therefore, we and others have used clozapine treatment as a proxy<br />

phenotype for antipsychotic treatment resistance in pharmacogenetic<br />

studies that aim to identify predictors <strong>of</strong> antipsychotic treatment<br />

resistance.<br />

Methodology: We assessed 89 schizophrenia patients clinically<br />

assigned to clozapine treatment versus 190 patients that were not<br />

selected for clozapine treatment All were Caucasians. 64.5% were<br />

males. To minimize multiple comparisons, we tested a set <strong>of</strong> 74<br />

candidate genes that were associated with all-cause discontinuation at<br />

the p


AUTHOR INDEX<br />

203


A<br />

A.S. Joshi, A., 160<br />

Aalto, M., 155<br />

Abdellaoui, A., 29<br />

Abdul Rahman, O., 195<br />

Aberg, K., 23, 44, 53, 170<br />

Abramson, L., 119<br />

Adachi, Y., 68<br />

Adkins, A., 62, 165<br />

Adkins, D., 199<br />

Agartz, I., 85, 120, 171<br />

Aggen, S., 88<br />

Agrawal, A., 56, 150<br />

Aguilar, A., 125<br />

Ahmadkhaniha, H., 109<br />

Ahn , Y., 133, 177<br />

Aitchinson, K., 186<br />

Ait-­‐Daoud, N., 161<br />

Ajit, D., 152<br />

Akahane, A., 188<br />

Akiskal, H., 127<br />

Akterin, S., 62, 97<br />

Akula, N., 40, 55, 59, 64<br />

Alasaari, J., 110<br />

Alda, M., 3, 50, 172, 201<br />

Aleksic, B., 68<br />

Aleksic, B., 70, 75<br />

Aleman, A., 115<br />

Alexander, M., 52<br />

Alho, H., 155<br />

Ali, I., 178<br />

Aliev, F., 62, 148, 181<br />

Al-­‐Janabi, T., 42<br />

Allencherry, P., 170<br />

Almasy, L., 46, 47, 126, 150<br />

Alonso, P., 80<br />

Alth<strong>of</strong>f, R., 29<br />

Amdur, R., 23<br />

Ameis, S., 48<br />

Amelang, M., 122<br />

Amin, N., 87<br />

Anbazhagan, P., 42<br />

Andersen, J., 175<br />

Andreasen, N., 101<br />

Andreassen, O., 16, 85, 120,<br />

171<br />

Andreou, D., 171<br />

Anjanappa , R., 179<br />

Anney, R., 92<br />

Antal, P., 123<br />

Antonijevic, I., 197<br />

Appel, K., 169<br />

Apud, J., 48<br />

Aqil, A., 83<br />

Arad, M., 31<br />

Ardau, R., 50<br />

Arenovich, T., 200<br />

Arias-­‐Vásquez, A., 83, 87<br />

Arkhipov, A., 183<br />

Arnold, P., 139, 140<br />

Arnold, S., 34<br />

Arranz, M., 51, 199<br />

Arun, M., 107<br />

Ascher, J., 200<br />

Asherson, P., 74, 113, 193<br />

Ashley-­‐Koch, A., 167<br />

Aslanidou, P., 63<br />

Athanasiu, L., 85, 120<br />

Aukes, M., 117, 125, 176<br />

Avrabos, C., 5<br />

Avramopoulos, D., 104, 122<br />

Axelsen, M., 101<br />

Ayalew, M., 66<br />

Ayub, M., 100<br />

Ayyappan, A., 124, 183<br />

Azzouz, F., 5<br />

B<br />

Bacanu, S., 22<br />

Backlund, L., 130<br />

Badner, J., 21, 22, 90<br />

Bagepally, B., 142<br />

Bailey, M., 77<br />

Bajo, E., 198<br />

Baker, M., 109<br />

Bakker, S., 125, 130<br />

Balachandar, V., 107<br />

Balaraman, Y., 66<br />

Balhara, Y., 154<br />

Bamne, M., 64<br />

Banaschewski, T., 74, 113<br />

Banerjee, M., 170<br />

Banno, M., 68<br />

Barbacioru, C., 45<br />

Barnes, C., 190<br />

Barr, C., 45, 109<br />

Barrett, J., 25<br />

Barta, C., 166<br />

Bartels, M., 29<br />

Bassett, A., 128<br />

Bauer, M., 201<br />

Bayés, À., 16<br />

Becker, T., 87<br />

Belmonte-­‐de-­‐Abreu, P., 189<br />

Benedek, D., 185<br />

Benegal, V., 160<br />

Bennett, V., 30<br />

Bergen, S., 62, 118<br />

Berlim, M., 8<br />

Bermudez Ocaña, D., 182,<br />

186<br />

Berrettini, W., 5<br />

Berry-­‐Scott, E., 31<br />

Bertelsen, S., 150<br />

Bettinger, J., 62<br />

Bialosuknia, S., 69<br />

Bickeböller, H., 116<br />

Bidwell, L., 167<br />

Biernacka, J., 49<br />

Bierut, L., 17, 19, 79, 126,<br />

147, 150<br />

Bigelli, M., 198<br />

Binder, E., 9, 35, 36, 39, 49,<br />

52, 123, 186<br />

Birkenaes, A., 85<br />

Bishop, J., 141<br />

Bitsios, P., 32, 114<br />

204<br />

Björk, K., 45<br />

Blackwood, D., 73, 100<br />

Blaine, S., 159<br />

Blake, D., 20, 42<br />

Blakemore, A., 199<br />

Blangero, J., 46, 47, 103<br />

Blaya, C., 131<br />

Bloom, R., 89, 91<br />

Blum, K., 196<br />

Bochdanovits, Z., 115<br />

Bognár, Z., 136<br />

Bogue, M., 199<br />

Bohlen, K., 200<br />

Bohn, B., 87<br />

Boks, M., 117, 125, 176, 182<br />

Bollen, J., 197<br />

Bonde, J., 175<br />

Bonetto, C., 157<br />

Boomsma, D., 29, 87<br />

Boos, H., 130<br />

Borglum, A., 176, 180<br />

Børglum, A., 78, 95, 169,<br />

175<br />

Bortoluzzi, A., 131<br />

Bosa, V., 131<br />

Böttcher, Y., 15<br />

Bowers, S., 62<br />

Braaten, E., 129<br />

Bradley, B., 36, 200<br />

Bramon, E., 51, 112<br />

Brand, J., 135<br />

Brandl, E., 40, 202<br />

Bravo, H., 55<br />

Bray, N., 21<br />

Breakspear, M., 4<br />

Breen, G., 96, 191, 193<br />

Breetvelt, E., 125, 128<br />

Brentani, H., 189<br />

Breuer , R., 85<br />

Breuer, R., 72, 180<br />

Brij , K., 152<br />

Britto Junior, A., 178<br />

Brockmöller, J., 116<br />

Bromet, E., 202<br />

Broms, ., 153<br />

Brown, A., 85, 120<br />

Brunner, H., 175<br />

Brusniak, W., 120<br />

Bubb, V., 46, 164<br />

Bucholz, K., 56, 126<br />

Buckner, R., 140<br />

Budde, J., 150<br />

Budde, M., 116<br />

Bui, E., 107<br />

Buitelaar, J., 52, 74<br />

Buizer-­‐Voskamp, J., 176<br />

Bukszár, J., 23, 44, 53, 170<br />

Bulik, C., 98<br />

Burdick, K., 15, 135<br />

Burdick, M., 33, 83<br />

Burke, A., 37<br />

Burrell, H., 164<br />

Buttenschøn, H., 175<br />

Buxbaum, J., 26<br />

Byerley, W., 90<br />

C<br />

Caballero, A., 139<br />

Cahn, W., 130<br />

Cai, X., 5, 31<br />

Cain, G., 152<br />

Cairns, M., 92<br />

Calati, R., 197<br />

Callicott, J., 33, 48, 83, 142<br />

Camarena Medellin, B., 182,<br />

186<br />

Camarena, B., 125, 139<br />

Campbell, J., 186<br />

Campos, L., 139<br />

Cannon, T., 89<br />

Cantor, R., 117<br />

Carless, M., 46, 47, 103<br />

Carlson, G., 34<br />

Carr, G., 33<br />

Carrard, A., 68<br />

Carraro, D., 189<br />

Carter, C., 121<br />

Carvalho, F., 158<br />

Casey, B., 36<br />

Casey, D., 163<br />

Castelao, E., 88<br />

Castellani, C., 90<br />

Castro R, R., 104<br />

Chai, Y., 49<br />

Chakrabortty, A., 57<br />

Chakravarti, A., 13, 54<br />

Chambert, K., 15, 16, 55, 56,<br />

62, 80, 101, 129<br />

Chan, W., 72<br />

Chang, S., 172<br />

Chapman, J., 60<br />

Chatterjee, N., 59<br />

Chen, C., 21, 22, 163, 193<br />

Chen, D., 59<br />

Chen, G., 89, 148<br />

Chen, H., 145, 192<br />

Chen, J., 22, 33, 83, 146<br />

Chen, L., 17, 79<br />

Chen, P., 122<br />

Chen, Q., 48, 142<br />

Chen, S., 71, 201<br />

Chen, X., 22, 24, 146<br />

Chen, Y., 131<br />

Cheng, A., 59<br />

Cheng, L., 21, 22<br />

Cheng, M., 163<br />

Chiano, M., 200<br />

Chiesa, A., 197<br />

Chillotti, C., 50<br />

Chitkara, N., 44<br />

Cho, M., 63<br />

Choi, K., 55, 185<br />

Chow, E., 128<br />

Chow, T., 189<br />

Chowdari, K., 178<br />

Chowdhury, N., 40, 201<br />

Christensen, J., 176<br />

Christ<strong>of</strong>orou, A., 93<br />

Chun, J., 174


Cichon, S., 15, 16, 59, 72, 78,<br />

85, 87, 91, 93, 180<br />

Clark, A., 23<br />

Claus, E., 159<br />

Cohen, M., 141<br />

Cohen, O., 69<br />

Cohen-­‐Woods, S., 96, 191<br />

Coleman, M., 112<br />

Collier, D., 16, 98, 134, 157<br />

Collins, A., 89, 91, 99<br />

Colombo, E., 145<br />

Colombo, R., 145<br />

Congiu, D., 50<br />

Conrad, D., 190<br />

Cooper, P., 134<br />

Coppola, G., 194<br />

Corley, R., 156<br />

Cormican, P., 92<br />

Cornelisse, L., 26<br />

Corry, J., 4<br />

Corvin, A., 15, 20, 92, 136<br />

Coryell, W., 31<br />

Costa, M., 50<br />

Cotsapas, C., 28<br />

Courchesne, E., 121<br />

Cox, N., 22<br />

Craddock, N., 47, 58, 59, 89,<br />

138, 159, 190<br />

Craig, I., 96, 186, 191<br />

Crawley, J., 83<br />

Crespo , J., 132<br />

Creswell, C., 134<br />

Crisafulli, C., 187, 197<br />

Crisponi, L., 72<br />

Crist<strong>of</strong>alo, D., 157<br />

Crow, T., 111, 195<br />

Crowley, J., 76, 199<br />

Cullis, J., 172<br />

Curran, J., 46, 47<br />

Currier, D., 37<br />

Curtis, D., 57<br />

Czibere, L., 5<br />

Czisch, M., 9<br />

D<br />

D'Aiuto, L., 64<br />

Dai, N., 81<br />

Daly, M., 26, 28<br />

Dao, D., 31<br />

Darvasi, A., 96<br />

Dauwan, M., 117<br />

Davies, G., 93<br />

Davies, G., 29, 51, 196, 200<br />

de Geus, E., 29, 87<br />

de Jong, S., 182<br />

de Leon, J., 199<br />

De Luca, V., 102, 111, 154,<br />

158<br />

De Ronchi, D., 145, 187, 197<br />

De Santi, K., 157<br />

De Souza Silva, M., 43<br />

Deary, I., 93<br />

Degenhardt, F., 72, 85, 87,<br />

180<br />

Degenhardt, L., 56<br />

Del Zompo, M., 50<br />

Deldin, P., 174<br />

Demenescu, L., 115<br />

Demetrovics, Z., 123, 166<br />

Demirakca, T., 120<br />

DeModena, A., 39<br />

Demontis, D., 78, 95, 169,<br />

176, 180<br />

Dempster, E., 45<br />

Derks, E., 58, 73<br />

DeRoode, D., 149<br />

DeRosse, P., 202<br />

Desrivières, S., 43, 158, 184<br />

Desta, Z., 49<br />

Detera-­‐Wadleigh, S., 55, 64,<br />

185<br />

Devlin, B., 26<br />

Di Florio, A., 159<br />

Di Maio, R., 64<br />

Di Nicola, M., 145<br />

Dick, D., 61, 62, 146, 148,<br />

165<br />

Dickinson, D., 83<br />

Didriksen, M., 95, 169, 180<br />

Diener, C., 120<br />

Dierssen, M., 184<br />

Dijkstra, T., 26<br />

Dixson, L., 9<br />

Djurovic, S., 85, 120<br />

do Prado, C., 114<br />

Dobson, R., 21, 61, 194<br />

Dolan, C., 29<br />

Domenici, E., 7<br />

Donner, J., 76<br />

Donohoe, G., 20, 92, 136<br />

Doty, N., 129<br />

Doucette, S., 3<br />

Doyle, A., 129<br />

Drago, A., 162, 187<br />

Drain, C., 3<br />

Drews, M., 49<br />

Drigalenko, E., 53, 79<br />

Drury, S., 112<br />

Duan, J., 53, 79, 94<br />

Duffy, A., 3<br />

Duggirala, R., 46, 47<br />

Duong, L., 129<br />

Dwyer, S., 77, 84, 136<br />

Dyer, T., 46, 47, 103<br />

E<br />

Easter, P., 140<br />

Easton, A., 43<br />

Ebstein, R., 74<br />

Edenberg, H., 126, 148, 150<br />

Eder, M., 5<br />

Ehlers, C., 143<br />

Ehli, E., 29, 51, 196, 200<br />

Ehringer, M., 147<br />

Eisinger, A., 166<br />

Eissa, A., 178<br />

Elassy, M., 178<br />

El-­‐Bahaei, W., 178<br />

Elek, Z., 134, 136<br />

Eley, T., 134<br />

205<br />

Elfving, B., 175<br />

el-­‐Guebaly, N., 163<br />

Elhaik, E., 54<br />

Ellonen, P., 6<br />

Elsayed, H., 178<br />

Elves, R., 98<br />

Elvevaag, B., 83<br />

Emese, K., 191<br />

Ende, G., 120<br />

Engberg, G., 130<br />

Enoch, M., 71<br />

Erhardt, A., 6<br />

Erhardt, S., 130<br />

Erickson, J., 63<br />

Erickson, L., 83<br />

Eriksson, J., 155, 190<br />

Erk, S., 32<br />

Escaramís , G., 132<br />

Espeseth, T., 93<br />

Essex, M., 126<br />

Estivill, 132<br />

Estivill, X., 80, 184<br />

Eszter, K., 191<br />

Etemad, B., 50<br />

Eubanks, J., 45<br />

Evgrafov, O., 23<br />

F<br />

Fabbri, C., 156<br />

Fagerness, J., 140<br />

Fahey, C., 92<br />

Fajardo, L., 178<br />

Falkai, P., 116<br />

Faludi, G., 134, 195<br />

Fanous, A., 23, 24<br />

Fanous, S., 164<br />

Faraone, S., 65, 69, 74, 97,<br />

113<br />

Farkas, J., 166<br />

Farmer, A., 59, 96, 134, 191<br />

Fasmer, O., 127<br />

Fathi, W., 178<br />

Faundez, V., 33<br />

Feiler, H., 143<br />

Feng, Y., 45<br />

Fernandes, A., 43<br />

Fernandes, C., 43, 93, 187<br />

Fernandez, E., 16<br />

Fernández, G., 175<br />

Ficks, C., 179<br />

Fisher, C., 3<br />

Flockhart, D., 49<br />

Flor, H., 120, 137<br />

Flores, G., 139<br />

Flores, J., 188<br />

Foldager, L., 95, 169, 175<br />

Forray, C., 197<br />

Forty, L., 47, 59, 89, 138<br />

Fox, N., 112<br />

Fox, P., 47<br />

Frank, J., 72, 122, 180<br />

Franke, B., 52, 87, 97, 175<br />

Frankland, A., 4<br />

Franziska Degenhardt, M.,<br />

16<br />

Freda, J., 53, 79, 94<br />

Freeman, N., 40<br />

Freudenberg, J., 105<br />

Freudenberg-­‐Hua, Y., 105<br />

Freyberger, H., 169<br />

Frias-­‐Ramón, T., 182, 186<br />

Friderici, K., 93<br />

Friedman, N., 156<br />

Frisén, L., 130, 135<br />

Fromer, M., 55, 101<br />

Fuemmeler, B., 167<br />

Fukumoto, M., 117, 118<br />

Furlong, S., 92<br />

G<br />

Galfalvy, H., 37<br />

Gallagher, L., 92<br />

Gallego, J., 44<br />

Galloway, B., 172<br />

Gálvez, V., 132<br />

Gamazon, E., 22<br />

Gandal, M., 34<br />

Garrett, M., 167<br />

Gaudi, S., 88<br />

Geels, L., 29<br />

Geisler, S., 202<br />

Gejman, P., 53, 79, 94<br />

Genis, A., 182, 186, 188<br />

Genome Study, B., 127<br />

Gentile, J., 44<br />

Georgieva, L., 80<br />

Gergely, K., 191<br />

Gerrish, A., 60<br />

Gershon, E., 5, 15, 21, 22<br />

Gerstner, T., 87<br />

Geschwind, D., 194<br />

Geyer, M., 66<br />

Ghadami, M., 172<br />

Ghadiri, M., 109<br />

Ghosh, S., 57, 179<br />

Giakoumaki, S., 32, 114<br />

Gibbs, R., 26, 76<br />

Giegling, I., 136<br />

Giese, K., 43<br />

Gilder, D., 143<br />

Gill, M., 20, 74, 92, 113<br />

Gillies, S., 164<br />

Gizer, I., 143<br />

Glahn, D., 46, 47, 103<br />

Glatt, C., 36<br />

Glatt, S., 69, 121<br />

Glennon, J., 52<br />

Glessner, J., 70<br />

Glowinski, A., 5<br />

Goate, A., 126, 148, 150<br />

Goes, F., 54, 95<br />

Goldani, M., 131<br />

Goldart, E., 164<br />

Goldberg, T., 135<br />

Goldman, D., 13, 37, 71<br />

Goldsmith, H., 126<br />

Goldstein, J., 118<br />

Gomaa, Z., 178<br />

Gomez, L., 45<br />

Gonzalez, L., 139


Gopin, C., 135<br />

Gordano, J., 196<br />

Gordon, M., 44<br />

Gordon-­‐Smith, K., 47, 59,<br />

89, 138<br />

Göring, H., 47, 53, 79<br />

Gould, T., 31<br />

Grabe, H., 169<br />

Grant, S., 11, 16<br />

Gratacòs, M., 80, 132, 184<br />

Greco, D., 6<br />

Green, E., 47, 59, 89<br />

Green, M., 4<br />

Greenwood, T., 127<br />

Gregersen, P., 105<br />

Grennan, K., 22<br />

Griebel, M., 87<br />

Grigoriou, E., 63<br />

Groen-­‐Blokhuis, M., 29<br />

Gr<strong>of</strong>, P., 3<br />

Grove, J., 78, 95, 169, 176,<br />

180<br />

Grozeva, D., 15, 47, 59, 80,<br />

89, 190<br />

Gruber, O., 116<br />

Grynderup, M., 175<br />

Guennel, T., 22<br />

Guha, S., 96<br />

Guindalini, C., 165<br />

Guipponi, M., 7<br />

Gupta, A., 25<br />

Gupta, J., 66<br />

Gur, R., 34<br />

Gurling, H., 176<br />

Gustafsson, O., 120<br />

Gutiérrez-­‐Zotes , A., 132<br />

H<br />

Haavik, J., 127<br />

Hack, L., 155, 165<br />

Haddad, S., 172<br />

Haddley, K., 46, 164<br />

Hadzi-­‐Pavlovic, D., 4<br />

Haenisch, B., 87<br />

Hakonarson, H., 70<br />

Halagur BhogeGowda , K.,<br />

179<br />

Hall, H., 171<br />

Hall, M., 112<br />

Hallett, V., 174<br />

Hamilton, G., 77<br />

Hamilton, S., 166<br />

Hamshere, M., 47, 74, 89<br />

Hänisch, B., 72<br />

Hanna, G., 139, 140<br />

Hansell, N., 124<br />

Hansen, A., 175<br />

Hansen, T., 129<br />

Happé, F., 174<br />

Häppölä, A., 153<br />

Hargreaves, A., 20<br />

Härmä, M., 110<br />

Harold, D., 60<br />

Harris, R., 151<br />

Hartz, S., 19, 147<br />

Hashimoto, R., 70, 117, 118<br />

Hassan, A., 111<br />

Hata, T., 188<br />

Hatton, D., 99<br />

Hattori, E., 81<br />

Haughey, H., 159, 160<br />

Hauser, J., 7<br />

Hawi, Z., 74<br />

He, D., 53, 79, 94<br />

He, G., 71, 198<br />

He, L., 71, 198<br />

Headings, V., 152<br />

Heath, A., 56<br />

Heath, B., 64<br />

Hebbring, S., 49<br />

Hedemand, A., 78<br />

Heikkilä, K., 153<br />

Henders, A., 56<br />

Hennekam, E., 176<br />

Herms, S., 85, 87<br />

Hernández, S., 125, 139<br />

Heskes, T., 26<br />

Hesselbrock, V., 150<br />

Hettema, J., 88<br />

Hewitt, J., 156<br />

Hill, E., 129<br />

Hill, M., 21<br />

Hilliard, C., 76<br />

Hinrichs, A., 150<br />

Hipolito, M., 108, 194<br />

Hiroshi, U., 70<br />

Hodge, S., 166<br />

Hodges, A., 194<br />

Hodgkinson, C., 37<br />

Hoeben, W., 125<br />

H<strong>of</strong>t, N., 147<br />

Höhn, D., 9<br />

Hollegaard, M., 78, 95, 169,<br />

180<br />

Hollingworth, P., 60<br />

Holmans, P., 16, 58, 73, 74,<br />

175<br />

Holmboe, K., 174<br />

Holmes, A., 7, 140<br />

Holsboer, F., 9, 49, 52, 85<br />

Hoogendijk, W., 115<br />

Hope, B., 164<br />

Horrocks, J., 3<br />

Hosang, G., 191<br />

Hottenga, J., 29, 87<br />

Hougaard, D., 78, 95, 169,<br />

180<br />

Hovatta, I., 6, 76, 190<br />

Hu, X., 7<br />

Hu, Y., 29, 51, 196, 200<br />

Huang, K., 110<br />

Huang, Y., 37, 193<br />

Hudson, J., 134<br />

Hudziak, J., 29, 51<br />

Huganir, R., 104<br />

Huizenga, P., 29, 196<br />

Hukic, D., 135<br />

Hulse, G., 82<br />

Hulsh<strong>of</strong>f Pol, H., 130<br />

Hultman, C., 44, 55, 56, 62,<br />

89, 97, 101<br />

206<br />

Humberto, N., 182<br />

Hunter, R., 77<br />

Hurles, M., 190<br />

Hutchinson, K., 159<br />

Hyde, J., 119<br />

Icay, K., 6<br />

Iidaka, T., 68<br />

Ikeda, M., 15, 68, 70, 75, 80<br />

Ilsley, K., 31<br />

Inada, T., 68, 70, 75<br />

Ingason, A., 16, 129<br />

Inkster, B., 9<br />

Insel, T., 12<br />

Investigators, R., 96, 191<br />

Iossifov, I., 95<br />

Ira, E., 157<br />

Ising, M., 49<br />

Ismail, T., 124, 183<br />

Ivanov, D., 15, 16, 80, 171<br />

Iwase, M., 117, 118<br />

Iwata, N., 68, 70, 75, 118<br />

Iwayama, Y., 81, 124<br />

Jacobi, J., 53, 79, 94<br />

Jacobsen, I., 176<br />

Jain, S., 142<br />

Jain, S., 42, 160, 179<br />

Jakobsen, K., 129<br />

Jamain, S., 59<br />

Janiri, L., 145<br />

Janson, E., 117, 182<br />

Janus, K., 151<br />

Jeffries, A., 21<br />

Jeffries, C., 103<br />

Jenkins, G., 49<br />

Jentsch, J., 34<br />

Jhuang, H., 31<br />

Ji, Y., 49<br />

Jia, P., 24<br />

Jiang, T., 70<br />

Jiang, X., 59, 185<br />

Jimenez Santos, A., 182<br />

Jiménez Z, C., 104<br />

Johansson, S., 127<br />

Johnson, B., 160, 161<br />

Johnson, L., 185<br />

Johnson, M., 47<br />

Jones, I., 47, 59, 89, 100,<br />

138<br />

Jones, L., 42, 47, 59, 73, 89,<br />

138<br />

Jönsson, E., 120, 171<br />

Joo, E., 133<br />

Jovanovic, T., 36<br />

Juárez Rojop, I., 186<br />

Juárez Rojp, I., 182<br />

Juhila, J., 6<br />

I<br />

J<br />

K<br />

Kærgaard, A., 175<br />

Kærlev, L., 175<br />

Kähler, A., 89, 97, 99, 120<br />

Kahn, R., 58, 117, 125, 130,<br />

176, 182<br />

Kaibuchi, K., 75<br />

Kajantie, E., 190<br />

Kallahalli Jayramu , S., 179<br />

Kalu, N., 152<br />

Kamali, M., 5, 174<br />

Kamatani, N., 49<br />

Kamijima, K., 118<br />

Kamino, K., 117<br />

Kam-­‐Thong, T., 9<br />

Kan, K., 29<br />

Kananen, L., 76, 190<br />

Kanazawa, T., 69, 82<br />

Kanba, S., 81<br />

Kaneko, T., 69<br />

Kang, S., 126, 150<br />

Kapitany-­‐Foveny, M., 166<br />

Kaplan, A., 113<br />

Kapoor, M., 150<br />

Kaprio, J., 146, 153<br />

Karagiannidis, J., 63<br />

Karayiorgou, M., 15<br />

Karchin, R., 54, 95<br />

Karege, F., 68<br />

Kasper, S., 197<br />

Kassem, L., 3, 59, 107<br />

Kato, M., 81<br />

Kato, T., 81<br />

Katsanis, N., 12<br />

Kawashige, S., 69<br />

Kay, V., 199<br />

Kazuba, D., 3<br />

Kazui, H., 117, 118<br />

Keddy, P., 50<br />

Kelsoe, J., 15, 39, 127<br />

Kember, R., 187<br />

Kendler, K., 22, 24, 62, 98,<br />

148, 155, 165<br />

Kennedy, J., 40, 48, 102,<br />

111, 113, 139, 140, 154,<br />

158, 163, 200, 201, 202<br />

Kenny, E., 92<br />

Kenny, P., 17<br />

Kent, J., 47<br />

Kent, L., 74<br />

Kepa, A., 184<br />

Khachane, A., 23, 44, 170<br />

Khondoker, M., 75<br />

Kikuchi, T., 68<br />

Kikuyama, H., 82<br />

Kim, S., 78, 177<br />

Kim, Y., 76, 91, 133, 177,<br />

199<br />

Kimura, M., 71<br />

Kiran, H., 160<br />

Kircher, T., 32<br />

Kirov, G., 15, 16, 77, 80,<br />

171, 190<br />

Kishore, B., 160<br />

Kleiber, M., 151<br />

Knight, D., 42<br />

Knowles, J., 23, 31, 100<br />

Kochunov, P., 47


Koefoed, P., 120<br />

Koenen, K., 35<br />

Koh, J., 69, 82<br />

Kohli, M., 9, 52<br />

Kohmura, K., 68<br />

Koide, T., 68, 70<br />

Kolachana, B., 48<br />

Kollins, S., 167<br />

Kolstad, H., 175<br />

Komel, R., 173<br />

Komiyama, N., 16<br />

Kooij, S., 87<br />

Korhonen, ., 153<br />

Kovacsics, C., 31<br />

Kovács-­‐Nagy, R., 134<br />

Kovanen, L., 155<br />

Kovas, Y., 128<br />

Kraft, P., 19<br />

Kramer, J., 126, 148, 150<br />

Kramer, M., 54<br />

Kranz, J., 47, 59, 89<br />

Kraus, L., 123<br />

Kreek, M., 141<br />

Krieger, J., 165<br />

Krishna, N., 179<br />

Kronholm, E., 94, 177<br />

Krug, A., 32<br />

Kubo, M., 49<br />

Kuczenski, R., 66<br />

Kuehner, C., 32<br />

Kuntsi, J., 74, 113<br />

Kunugi, H., 118<br />

Kuperman, S., 150<br />

Kuryatov, A., 18<br />

Kushima, I., 70, 75<br />

Kusumawardhani, A., 81<br />

Kutalik, Z., 88<br />

Kutscher, E., 200<br />

Kwagyan, J., 152<br />

Kwan, J., 28<br />

L<br />

Laan, W., 176<br />

Labbe, A., 8<br />

Labonte, B., 38<br />

Lachman, H., 65<br />

Lage, K., 28<br />

Lagus, M., 110<br />

Lahti, J., 155<br />

Lahti, M., 190<br />

Lai, C., 163<br />

Laite, G., 5<br />

Laje, G., 40, 55<br />

Landén, M., 62, 130<br />

Landgraf, R., 5<br />

Langley, K., 74<br />

Laranjeira, R., 165<br />

Largeau, ., 153<br />

Lasalvia, A., 157<br />

Lassen, N., 95<br />

Lavebratt, C., 45, 130, 135<br />

Lawrie, S., 116, 141<br />

Lawson, W., 31, 108, 194<br />

Le Hellard, S., 93<br />

Le, T., 185<br />

Leal, S., 11<br />

Leask, S., 111<br />

Leboyer, M., 59<br />

Leckband, S., 39<br />

Lee, F., 36<br />

Lee, K., 133<br />

Lee, P., 27, 112, 140<br />

Lehner, T., 12<br />

Leibenluft, E., 15<br />

Leistner-­‐Segal, S., 131<br />

Lemery-­‐Chalfant, K., 126<br />

Lencz, T., 44, 54, 96, 202<br />

Le-­‐Niculescu, H., 66<br />

Lennartsson, A., 45<br />

Lenroot, R., 4<br />

Leong, L., 78<br />

Lerch, J., 48<br />

Lerer, B., 88<br />

Lester, K., 134<br />

Lett, T., 40, 48<br />

Leussis, M., 31<br />

Levinson, D., 31, 102<br />

Levitan, R., 113<br />

Levran, O., 141<br />

Levy, D., 15, 112<br />

Levy, F., 4<br />

Lewis, C., 51, 96, 127, 134,<br />

173, 191<br />

Lewis, D., 95<br />

Lewis, G., 7<br />

Lewitzka, U., 3<br />

Li, H., 185<br />

Li, M., 28, 160, 161<br />

Li, X., 71, 198<br />

Liao, D., 163<br />

Liao, H., 163<br />

Lichtenstein, P., 56, 62, 97,<br />

101, 173<br />

Lieberman, J., 40, 48, 76,<br />

201<br />

Liebsch, K., 120<br />

Liew, C., 121<br />

Ligthart, L., 29<br />

Likhodi, O., 40, 202<br />

Lima de Souza, A., 178<br />

Lin, D., 91<br />

Lin, K., 51<br />

Lin, M., 65<br />

Lindell, S., 109<br />

Lindstrom, J., 18<br />

Lino, B., 4<br />

Linotte, S., 197<br />

Littlefield, A., 56<br />

Liu, B., 71, 198<br />

Liu, C., 21, 22<br />

Liu, F., 201<br />

Liu, H., 5<br />

Liu, Q., 164<br />

Liu, X., 84<br />

Liu, Y., 53, 70, 170, 196<br />

Lobina, M., 72<br />

Lobo, D., 163<br />

Lodhi, S., 149<br />

Loerbroks, A., 122<br />

Logan, R., 45<br />

Loh, H., 181, 189<br />

207<br />

Loh<strong>of</strong>f, F., 50<br />

Loken, E., 98<br />

Lönnqvist, J., 76<br />

López N, M., 104<br />

Lopez, J., 8<br />

Lopez, L., 121<br />

Lopéz-­‐Rubalcava , C., 188<br />

Losh, M., 99<br />

Loukola, A., 146, 153<br />

Lourdusamy, A., 61<br />

Lovestone, S., 61, 194<br />

Lóyzaga, C., 125<br />

Lu, L., 149<br />

Lucae , S., 85<br />

Lucae, S., 9, 49, 52<br />

Lucchesi, W., 43<br />

Luna, A., 83<br />

Lundervold, A., 93<br />

Lunnon, K., 61, 194<br />

Luo, L., 193<br />

Luyxk, J., 125<br />

Lyneham, H., 134<br />

Lynskey, M., 56<br />

Lyon, G., 70<br />

M<br />

Macciardi, F., 88<br />

MacGregor, S., 73<br />

Machado-­‐Vieira, R., 114<br />

Maclean, A., 73, 100<br />

Madden, P., 56<br />

Maes, H., 146, 155<br />

Magi, A., 166<br />

Magnusson, P., 44, 97<br />

Maher, B., 88<br />

Mahmood, K., 100<br />

Mahmoodizadeh, A., 172<br />

Mahon, P., 31<br />

Maier, W., 7, 85, 87<br />

Maiti, S., 90<br />

Majumder, S., 161<br />

Malafosse, A., 7, 68<br />

Malhotra, A., 15, 44, 54, 96,<br />

135, 202<br />

Malhotra, D., 15<br />

Malik, M., 194<br />

Malki, K., 186<br />

Mamdani, F., 8<br />

Manchia, M., 50, 172, 201<br />

Mandelli, L., 145<br />

Manduva , V., 179<br />

Manduva, V., 86<br />

Manfro, G., 131<br />

Mann, J., 37<br />

Mannila, H., 137<br />

Männistö, S., 94<br />

Manor, I., 113<br />

Mansour, H., 178<br />

Manz, N., 126, 150<br />

Marangon, V., 157<br />

Maria, S., 191<br />

Markt, P., 5<br />

Márquez, L., 125<br />

Marshall, V., 152<br />

Martin, E., 141<br />

Martin, N., 56, 124<br />

Martínez G, M., 104<br />

Martinotti, G., 145<br />

Martorell , L., 132<br />

Maschieto, M., 189<br />

Mash, D., 71<br />

Massat, I., 197<br />

Mathé, A., 45<br />

Matsuzaki, H., 183<br />

Mattay, V., 48, 142<br />

Matthäus, F., 120<br />

Mattheisen, M., 72, 78, 85,<br />

87, 180<br />

Matthews, P., 9<br />

Mattingsdal, M., 85, 120<br />

Maussion, G., 38<br />

Mavruk, S., 95<br />

May, C., 161<br />

May, V., 36<br />

Mayfield , R., 151<br />

Mazza, M., 145<br />

McCarley, R., 118<br />

McCarthy, M., 39<br />

Mccarthy, S., 15, 90<br />

McClay JL, J., 53<br />

McClay, J., 23, 44, 170<br />

McClernon, F., 167<br />

McCombie, W., 54, 90, 95<br />

McCormack, C., 4<br />

McCue, C., 4<br />

McDonald, J., 184<br />

McGuffin, P., 59, 96, 127,<br />

173, 186, 191<br />

McGuire, J., 185<br />

McInnis, M., 4, 5, 174<br />

McIntosh, A., 116, 141<br />

McKechanie, A., 73<br />

Mckenzie, B., 154<br />

McLeod, H., 199<br />

Mcmahon, F., 15, 40, 55, 59,<br />

64, 107, 185, 201<br />

McMahon, W., 70<br />

McMichael, O., 62<br />

McQuillin, A., 176<br />

McRae, A., 73<br />

Meaney, M., 38<br />

Meary, A., 68<br />

Medeiros, H., 23<br />

Meera , P., 152<br />

Mehta, D., 123<br />

Meichle, S., 78<br />

Meier, S., 72, 85, 120, 122<br />

Meirelles, O., 72<br />

Melas, P., 45<br />

Melle, I., 85, 120<br />

Mello, B., 189<br />

Meltzer, H., 40, 48, 201, 202<br />

Meltzer-­‐Brody, S., 100<br />

Menchón, J., 132<br />

Mendlewicz, J., 197<br />

Mendoza, P., 188<br />

Mercer, K., 36<br />

Merwood, A., 74<br />

Messas, G., 165<br />

Meta-­‐Analysis Group, C., 17


Meyer-­‐Lindenberg, A., 19,<br />

132<br />

Meyers, J., 146<br />

Michael, G., 136<br />

Michael, O., 175<br />

Mick, E., 97<br />

Middeldorp, C., 29<br />

Middleton, F., 65, 69<br />

Mikkelsen, S., 175<br />

Miliar G, A., 104<br />

Mill, J., 158, 187<br />

Millard, S., 78<br />

Miller, G., 148<br />

Miller, M., 196<br />

Mills, N., 124<br />

Mir, A., 100<br />

Miranda, A., 74, 113<br />

Missaglia, S., 145<br />

Mitchell, P., 4, 5<br />

Miyake, A., 156<br />

Monnier, P., 45<br />

Montgomery, G., 56<br />

Moore, B., 70<br />

Moore, M., 126<br />

Moran, J., 15, 16, 47, 55, 56,<br />

59, 62, 80, 89, 101, 129<br />

Morell, C., 44<br />

Moreno, R., 114<br />

Morgan, M., 76<br />

Mori, N., 124, 183<br />

Morihara, T., 117<br />

Morris, B., 77<br />

Morris, D., 20, 92<br />

Morris, N., 82<br />

Mors, O., 7, 78, 95, 120, 169,<br />

175, 176, 180<br />

Mortensen, P., 78, 95, 169,<br />

180<br />

Moses, E., 47<br />

Moskvina, V., 58<br />

Mostafavi Abdolmalek, H.,<br />

109<br />

Mostafavi, S., 109<br />

Mould, G., 113<br />

Moumita, M., 152<br />

Moy, W., 53, 79, 94<br />

Mozhui, K., 149<br />

Mrazek, D., 49<br />

Muck Seler, D., 173<br />

Mueller, K., 193<br />

Mueller-­‐Myhsok, B., 49<br />

Muglia, P., 59, 191<br />

Mühleisen , T., 85<br />

Mühleisen, T., 72, 87, 180<br />

Mukherjee, O., 142<br />

Mulas, A., 72<br />

Mulas, F., 113<br />

Muller, C., 43<br />

Müller, D., 40, 48, 200, 201,<br />

202<br />

Müller-­‐Myhsok, B., 9, 52, 85<br />

Mulsant, B., 48<br />

Multani, P., 50<br />

Munro, J., 199<br />

Murdoch, J., 25<br />

Murphy, E., 3<br />

Murray, J., 193<br />

Murray, R., 51, 112<br />

Murtha, M., 25<br />

Murthy A, R., 86<br />

Murthy, P., 160<br />

Mushiroda, T., 49<br />

Myers, A., 36<br />

Myers, J., 155<br />

Myers, P., 46, 164<br />

N<br />

Naeem, F., 100<br />

Naik, R., 5<br />

Nair, C., 170<br />

Nakamura, J., 145, 192<br />

Nakamura, K., 124, 183<br />

Nakamura, M., 81<br />

Nakamura, Y., 49, 70<br />

Nanko, S., 188<br />

Narasimhan, S., 50<br />

Neale, B., 26, 28, 60, 129<br />

Neary, J., 46, 103<br />

Nedic, G., 173<br />

Nees, F., 137<br />

Negrão, A., 165<br />

Negri, G., 145<br />

Nelson, C., 112<br />

Nelson, E., 56<br />

Nelson, K., 29, 196<br />

Németh, N., 136, 166<br />

Nemoda, Z., 123, 195<br />

Nerella, S., 23, 44<br />

Newport, J., 123<br />

Nicodemus, K., 83<br />

Nicolae, D., 22<br />

Nicolini, H., 125, 186, 188<br />

Niculescu, A., 66, 108<br />

Nieratschker, V., 120, 122,<br />

137<br />

Nigg, J., 93<br />

Nikolac, M., 173<br />

Nikolas, M., 93<br />

Nimgaonkar, V., 64, 178<br />

Nishiguchi, M., 82<br />

Nivard, M., 29<br />

Nohesara, S., 109<br />

Nordent<strong>of</strong>t, M., 78<br />

Nossova, N., 121<br />

Nöthen, M., 72, 85, 87, 91,<br />

180<br />

Numans, M., 125<br />

Nummy, K., 93<br />

Nurnberger , J., 5, 150<br />

Nurnberger Jr, J., 108, 148,<br />

194<br />

Nwulia, E., 108, 147, 152,<br />

194<br />

Nyman, E., 146<br />

O<br />

O'Donnell, L., 174<br />

O'Donovan, M., 7, 15, 16, 42,<br />

47, 58, 59, 73, 74, 77, 80,<br />

84, 86, 89, 136, 171, 190<br />

208<br />

O'Dushlaine, C., 27, 56, 62,<br />

129<br />

O'Neill, F., 98<br />

O'Reilly, R., 90<br />

Oades, R., 74, 113<br />

Oedegaard, K., 127<br />

Ohi, K., 70, 117, 118<br />

Ohmori, O., 145, 192<br />

Okochi, T., 118<br />

Olason, P., 15<br />

Olgiati, P., 198<br />

Ollila, H., 94, 138, 177<br />

Olsson, S., 130<br />

Olvera, R., 47<br />

Oph<strong>of</strong>f, R., 16, 117, 125,<br />

130, 176, 182<br />

Opmeer, E., 115<br />

Oquendo, M., 37<br />

Ørnt<strong>of</strong>t, T., 95, 169, 180<br />

Ösby, U., 62, 130, 135<br />

Oscar-­‐Berman, M., 196<br />

Osimo, E., 88<br />

Ossowski, S., 80<br />

Ostrosky, P., 188<br />

Otowa, T., 88<br />

Owen, M., 15, 16, 42, 47, 58,<br />

59, 60, 74, 77, 80, 84, 86,<br />

89, 136, 171, 190<br />

Ozaki, N., 68, 70, 75, 118<br />

P<br />

Pae, C., 197<br />

Palha, J., 189<br />

Pallesen, J., 78, 95, 176<br />

Palotie, A., 16, 25, 155<br />

Papaleo, F., 83<br />

Papasotiriou, S., 63<br />

Papeleo, F., 33<br />

Park, J., 59<br />

Parla, J., 54, 95<br />

Partonen, T., 94, 138, 155<br />

Paschou, P., 63<br />

Pasparakis, M., 32<br />

Patel, S., 66<br />

Pato, C., 23<br />

Pato, M., 23<br />

Patterson, D., 165<br />

Pattwell, S., 36<br />

Pauline, M., 141<br />

Pauls, D., 52<br />

Paunio T, T., 153<br />

Paunio, T., 94, 110, 137,<br />

138, 177<br />

Payo-­‐Cano, J., 186<br />

Pedersen, C., 78, 95, 180<br />

Pedrosa, E., 65<br />

Pendleton, N., 93<br />

Peng, Z., 22<br />

Penninx, B., 30, 87, 100, 115<br />

Pereira, A., 165<br />

Pereira, C., 189<br />

Pergadia, M., 56, 153<br />

Perkins, D., 103<br />

Perlis, R., 38, 79<br />

Perola, M., 94, 138<br />

Perrine, K., 196<br />

Perroud, N., 7, 68<br />

Perry, J., 191<br />

Peterson, R., 146<br />

Petryshen, T., 31, 118<br />

Pibiri, F., 21, 22<br />

Pickard, B., 73<br />

Pickens, C., 164<br />

Pierce, K., 121<br />

Piras, C., 50<br />

Piras, M., 72<br />

Pirkola, S., 76<br />

Pirooznia, M., 54, 95, 104<br />

Pitkäniemi, J., 153<br />

Pivac, N., 173<br />

Piven, J., 99<br />

Platt, D., 148<br />

Plomin, R., 128, 174<br />

Pocklington, A., 16, 27, 74<br />

Poelmans, G., 52<br />

Poggio, T., 31<br />

Pohlack, S., 137<br />

Ponomarev, I., 151<br />

Pool García, S., 182<br />

Popoli, M., 8<br />

Porcelli, S., 156<br />

Porjesz, B., 126, 150<br />

Potash, J., 15, 31, 54, 95,<br />

104<br />

Potkin, S., 40, 48<br />

Poulsen, P., 175<br />

Powell, J., 51, 61<br />

Powell, T., 187<br />

Power, R., 127, 173<br />

Prabhat, C., 152<br />

Prabhu, S., 42, 142<br />

Pradip, P., 152<br />

Pratima, M., 152<br />

Pratt, A., 199<br />

Pratt, J., 77<br />

Pregelj, P., 173<br />

Preisig, M., 88<br />

Prescott, C., 62, 165<br />

Price, J., 21<br />

Price, T., 74, 113<br />

Priebe, L., 72, 85<br />

Proitsi, P., 61, 194<br />

Propping, P., 87<br />

Puga, R., 189<br />

Pulver, A., 122<br />

Purcell, S., 15, 16, 27, 47,<br />

55, 56, 59, 62, 73, 76, 80,<br />

89, 97, 101, 118, 129,<br />

140<br />

Purushottam, M., 42, 142,<br />

160, 179<br />

Puttonen, S., 110<br />

Q<br />

Qi, H., 45<br />

Qin, W., 81<br />

Quackenbush, C., 199<br />

Quinn, J., 46, 164


R<br />

Radant, A., 78<br />

Räikkönen, K., 155, 190<br />

Raimondi, G., 64<br />

Rajji, T., 48<br />

Rajkumar, A., 176<br />

Ramanathan, S., 42<br />

Randesi, M., 141<br />

Rangaswamy, M., 126, 150<br />

Rapee, R., 134<br />

Rasetti, R., 48, 142<br />

Rauch, A., 48<br />

Ravindran, A., 111<br />

Raychaudhuri, S., 28<br />

Reddy, J., 86<br />

Rees, E., 15, 80, 171<br />

Reich-­‐Erkelenz, D., 116<br />

Reimers, M., 22<br />

Reinvang, I., 93<br />

Reitschel, M., 15<br />

Ren, B., 45<br />

Ressler, K., 36<br />

Rex-­‐Haffner, M., 123<br />

Rice, J., 126<br />

Richards, A., 73<br />

Richardson, T., 50<br />

Richter, M., 139<br />

Rickels, K., 50<br />

Riemers, M., 165<br />

Rietschel , M., 85<br />

Rietschel, M., 7, 16, 32, 59,<br />

72, 87, 91, 120, 122, 137,<br />

180<br />

Rijpkema, M., 175<br />

Rijsdijk, F., 112<br />

Riley, B., 62, 98, 165<br />

Rimol, L., 120<br />

Riolo, R., 157<br />

Ripatti, S., 76, 153<br />

Ripke, S., 28, 52, 60<br />

Rivera Angles, M., 186<br />

Rivera, M., 96, 191<br />

Roberts, G., 4<br />

Robertson, G., 50<br />

Robison, R., 70<br />

Roeyers, H., 74<br />

R<strong>of</strong>fman, J., 140<br />

Rogdaki, M., 45<br />

Rónai, Z., 134, 136<br />

Ronald, A., 174<br />

Roncada, P., 50<br />

Rose, E., 20<br />

Rose, R., 146<br />

Rosenberg, D., 139, 140<br />

Rosenfeld, J., 54, 96<br />

Ross, C., 30<br />

Rossin, E., 28<br />

Rostami, M., 172<br />

Rothenberger, A., 74, 113<br />

Roubeson, M., 25<br />

Roussos, P., 32, 114<br />

Rouvinen-­‐Lagerström, N.,<br />

155<br />

Royers, H., 113<br />

Rubin, L., 141<br />

Rudd, D., 101<br />

Ruderfer, D., 15, 16, 27, 56,<br />

62, 80, 101, 129<br />

Rudolf, G., 23, 44<br />

Ruggeri, B., 158<br />

Ruggeri, M., 157<br />

Rujescu, D., 16, 37, 136<br />

Russell, E., 138<br />

Russo, G., 136, 175<br />

Ruttorf, M., 137<br />

S<br />

Saccone, N., 147<br />

Sadee, W., 18<br />

Saemann, P., 52<br />

Saetre, P., 171<br />

Sagvolden, T., 65<br />

Saito, M., 31<br />

Saitoh, O., 118<br />

Sajeev, G., 200<br />

Sakata, S., 145, 192<br />

Salah, H., 178<br />

Salisbury, D., 112<br />

Salk, R., 119<br />

Salminen, O., 153<br />

Salomaa, V., 94, 138<br />

Salum, G., 131<br />

Samaan, Z., 96<br />

Sämann, P., 9<br />

Sambataro, F., 142<br />

Samocha, K., 28<br />

Sanchez, M., 182<br />

Sanders, A., 53, 79, 94<br />

Sanders, S., 25<br />

Sandman, N., 94<br />

Sanjeev, J., 152<br />

Sano, A., 81<br />

Santangelo, S., 172<br />

Sarin, S., 153<br />

Sarkozy, P., 123, 166<br />

Sasaki, T., 84<br />

Sasanfar, R., 172<br />

Sasikala, K., 107<br />

Sasvári-­‐Székely, M., 123,<br />

134, 136, 166, 195<br />

Sathyan, P., 161<br />

Sattlecker, M., 194<br />

Savage, A., 164<br />

Saviouk, V., 87<br />

Savolainen, K., 190<br />

Sawa, A., 122<br />

Schaid, D., 49<br />

Schalkwyk, L., 186, 187<br />

Schalling, M., 62, 130, 135<br />

Scheet, P., 29<br />

Scheftner, W., 31<br />

Schellenberg, G., 26<br />

Scherer, S., 24<br />

Schlessinger, D., 72<br />

Schmäl, C., 72<br />

Schmidt, K., 58<br />

Schmidt, M., 52<br />

Schoenauer, S., 137<br />

Schork, N., 66, 121<br />

Schubart, C., 182<br />

209<br />

Schubert, M., 180<br />

Schuch, I., 131<br />

Schuckit, M., 126, 148, 150<br />

Schulz, A., 169<br />

Schulze, K., 112<br />

Schulze, T., 15, 32, 59, 72,<br />

116, 120, 122, 180, 201<br />

Schumann, G., 43, 143, 158<br />

Schur<strong>of</strong>f, F., 68<br />

Schwab, S., 81, 82<br />

Schwahn, C., 169<br />

Schweitzer, R., 5<br />

Scolnick, E., 62<br />

Scott , D., 152<br />

Scott, A., 82<br />

Sebat, J., 15<br />

Sedvall, G., 171<br />

Seidman, L., 118, 121<br />

Seneviratne, C., 160, 161<br />

Sergeant, J., 74<br />

Serre, T., 31<br />

Serretti, A., 145, 156, 162,<br />

187, 197, 198<br />

Severino, G., 50<br />

Shah, A., 65<br />

Shaham, Y., 164<br />

Shaikh, S., 201<br />

Sham, P., 28<br />

Sharma, B., 147<br />

Sharma, R., 149<br />

Shekhar, A., 66<br />

Shekhtman, T., 39<br />

Shen, Q., 71<br />

Shen, Y., 131<br />

Shenton, M., 118<br />

Sher, K., 56<br />

Shi, J., 31, 53<br />

Shinkai, T., 145, 192<br />

Shirtcliff, E., 126<br />

Short, S., 19<br />

Shugart, Y., 99<br />

Siburian, R., 172<br />

Sidoti, A., 187<br />

Siegel, S., 34<br />

Siever, L., 78<br />

Sigurdsson, E., 16<br />

Silander, K., 155<br />

Silva, A., 189<br />

Silveira, P., 131<br />

Silverman, J., 78<br />

Sims, R., 60<br />

Sinclair, D., 155<br />

Singh, S., 90, 151<br />

Sinopoli, V., 140<br />

Sipilä, T., 6, 76<br />

Skjødt, C., 129<br />

Sklar, P., 15, 16, 31, 47, 55,<br />

56, 59, 62, 73, 76, 80, 89,<br />

97, 101, 129<br />

Skrobot, O., 199<br />

Smeets, H., 176<br />

Smit, J., 87<br />

Smith, C., 110<br />

Smith, R., 18, 84<br />

Smoller, J., 112, 129, 140<br />

Snyder, K., 49<br />

Snyder, L., 197<br />

Soeiro-­‐de-­‐Souza, M., 114<br />

Soggiu, A., 50<br />

Soliman, F., 36<br />

Somers, M., 117<br />

Sommer, I., 117, 182<br />

Somogyi, A., 195<br />

Song, J., 133, 177<br />

Soria, V., 132<br />

Souery, D., 197<br />

Soundy, T., 196, 200<br />

Souza, R., 201<br />

Sprooten, E., 116, 141<br />

Squassina, A., 50<br />

Srinivas, L., 170<br />

St Clair, D., 16, 73<br />

Stallings, M., 156<br />

State, M., 25<br />

Stathias, V., 63<br />

Steele, C., 59<br />

Steen, V., 85, 93<br />

Stefanson, H., 16<br />

Stefansson, H., 15, 78<br />

Stefansson, K., 15, 16<br />

Steffens, M., 87<br />

Steinberg, S., 16, 78<br />

Steinhausen, H., 74, 113<br />

Stephens, S., 147<br />

Stergiakouli, E., 74<br />

Stewart, A., 61, 194<br />

Stone, W., 121<br />

Stowe, Z., 123<br />

Straub, R., 33<br />

Strauss, J., 102, 191<br />

Strengman, E., 125<br />

Stringer, S., 58<br />

Strohmaier, J., 32, 72, 85,<br />

87, 122<br />

Stürmer, T., 122<br />

Subhashree, D., 160<br />

Suda, S., 124, 183<br />

Suderman, M., 38<br />

Sulkava, S., 137<br />

Sullivan, P., 44, 53, 55, 56,<br />

62, 76, 89, 91, 97, 98, 99,<br />

100, 101, 170, 199<br />

Sun, B., 193<br />

Sun, J., 22, 23, 24<br />

Sun, W., 91<br />

Sundermann, E., 141<br />

Suomi, S., 109<br />

Sussmann, J., 116, 141<br />

Sutcliffe, J., 26<br />

Sutin, A., 72<br />

Suzuki, M., 70, 75<br />

Svenningsson, P., 45<br />

Svensson, A., 173<br />

Sweeney, C., 148<br />

Syed, Z., 174<br />

Szántai, E., 134<br />

Szatkiewicz, J., 91, 99<br />

Szekely, A., 123<br />

Szyf, M., 38


T<br />

Taj, R., 100<br />

Takahashi, H., 118<br />

Takata, A., 81<br />

Takeda, M., 70, 75, 117, 118<br />

Takei, N., 124<br />

Talati, A., 166<br />

Talbot, K., 34<br />

Tang, P., 181<br />

Tansey, K., 7<br />

Tatsumi, M., 118<br />

Taub, M., 122<br />

Tavian, D., 145<br />

Taylor, R., 152<br />

Taylor, A., 99<br />

Taylor, G., 112<br />

Tee, S., 181<br />

Telles Ribeiro, H., 178<br />

Tenesa, A., 93<br />

Teo, C., 102, 111, 154<br />

Terenius, L., 171<br />

Termorshuizen, F., 176<br />

Terracciano, A., 72<br />

Terwisscha van Schel, A.,<br />

130<br />

Tesli, M., 120<br />

Teumer, A., 169<br />

Thakkar, M., 149<br />

Thapar, A., 74<br />

Theall, K., 112<br />

Theberge, F., 164<br />

Thennarasu, K., 160<br />

Thiagalingam, S., 109<br />

Thimm, M., 32<br />

Thiselton, D., 62<br />

Thomas, G., 104<br />

Thompson, S., 31<br />

Thomsen, J., 175<br />

Thurin, K., 142<br />

Thygesen, J., 129<br />

Tian, Q., 83<br />

Tinsley, C., 20<br />

Tischfield, J., 126, 148, 150<br />

Tiwari, A., 40, 48, 158, 200,<br />

201, 202<br />

Tobar, S., 178<br />

Tokunaga, K., 84<br />

Tolouei, A., 172<br />

Tomblin, B., 193<br />

Tommerup, N., 176<br />

Toner, D., 129<br />

Tooney, P., 92<br />

Torma, N., 166<br />

Tornador, C., 80<br />

Torri, F., 88<br />

Tosato, S., 16, 157<br />

Tosto, M., 128<br />

Tovilla-­‐Zarate, C., 182, 186<br />

Toyota, T., 81<br />

Tozzi, F., 191, 200<br />

Treutlein, J., 72<br />

Tropeano, M., 134<br />

Trujillano, D., 80, 184<br />

Tsivtsivadze, E., 26<br />

Tsuang, D., 78<br />

Tsuang, M., 69, 121<br />

Tsutsumi, A., 69, 82<br />

Tümer, Z., 176<br />

Tunc-­‐Skarka, N., 120<br />

Turecki, G., 8, 37, 38<br />

Tzvetkov, M., 116<br />

U<br />

Ueno, M., 188<br />

Uher, R., 7, 127, 186<br />

Ujike, H., 75<br />

Urban, A., 102<br />

Urban, R., 166<br />

Urretavizcaya, M., 132<br />

Ursano, R., 185<br />

Utge, S., 138, 177<br />

Utsunomiya, K., 145, 192<br />

V<br />

V., N., 170<br />

Vakkalanka, R., 83<br />

Valero , J., 132<br />

Vallada, H., 165<br />

Valle, D., 104, 122<br />

Vallender, E., 148<br />

Valli, K., 94<br />

van 't Slot, R., 117<br />

van Beek, J., 29<br />

van Beijsterveldt, T., 29<br />

van de Lagemaat, L., 16<br />

van den Oord, E., 23, 44, 53,<br />

88, 170, 199<br />

van der Meere, J., 113<br />

van der Wee, N., 115<br />

van Duijn, C., 87<br />

van Eijk, K., 117<br />

van Gastel, W., 182<br />

van Haren, N., 130<br />

Van Hulle, C., 126<br />

van Tol, M., 115<br />

Vandever, J., 156<br />

VanWijk, R., 70<br />

Varga, G., 123, 136<br />

Vargas, I., 125<br />

Vargas, L., 125<br />

Varghese, M., 142<br />

Vasiliou, S., 164<br />

Vasquez, A., 175<br />

Vassos, E., 72<br />

Vazquez, V., 139<br />

Vedernikov, A., 58<br />

Veltman, D., 115<br />

Ventura, S., 44<br />

Vereczkei, A., 195<br />

Veres-­‐Székely, A., 136<br />

Verhage, M., 26<br />

Videtič Paska, A., 173<br />

Vieten, C., 143<br />

Vilella , E., 132<br />

Villar Soto, M., 182<br />

Vincent, J., 191<br />

Vink, J., 29<br />

Visscher, P., 73<br />

Vivek, B., 152<br />

210<br />

Vladimirov, V., 62<br />

Voineskos, A., 48<br />

Vorstman, J., 128<br />

Vreeburg, S., 115<br />

Vuokko, K., 153<br />

W<br />

Waldman, I., 93, 179<br />

Walsh, D., 98, 165<br />

Walter, H., 32<br />

Walters, J., 136, 138<br />

Wang, J., 126, 150<br />

Wang, K., 70<br />

Wang, L., 24, 198<br />

Wang, S., 151<br />

Wang, T., 104<br />

Wang, W., 91<br />

Wang, X., 92, 161<br />

Wang, Y., 71, 198<br />

Warburton, A., 164<br />

Wassink, T., 101<br />

Watson, A., 64, 178<br />

Webb, B., 98, 146, 148, 165<br />

Weber, K., 141<br />

Webster, M., 55, 185<br />

Wedenoja , J., 153<br />

Wegener, G., 45, 175<br />

Weinberger, D., 33, 48, 83,<br />

142<br />

Weinfeld, M., 121<br />

Weinshilboum, R., 49<br />

Weissman, M., 31, 166<br />

Weissman, S., 102<br />

Wendland, J., 55, 59<br />

Werge, T., 16, 120, 129, 171<br />

Werme, M., 45<br />

Wessa, M., 32<br />

Whalley, H., 141<br />

White, M., 142<br />

Whitfield, J., 124<br />

Wickramaratne, P., 166<br />

Wigg, K., 45<br />

Wijsman, E., 78<br />

Wilcox, H., 5<br />

Wildenauer, D., 81, 82<br />

Wildenauer, M., 81<br />

Wilhelmsen, K., 143<br />

Wilkinson, L., 42<br />

Willemsen, G., 29<br />

Williams, C., 152<br />

Williams, H., 84, 86, 136<br />

Williams, J., 60<br />

Williams, N., 74, 97<br />

Williams, R., 149, 167<br />

Williams, S., 61, 194<br />

Winchester, C., 77<br />

Winkler, A., 47<br />

Winn, M., 121<br />

Wirgenes, K., 120<br />

Witasp, A., 45<br />

Witt, S., 32, 120, 122, 137<br />

Wiuf, C., 95, 169, 180<br />

Wolf , C., 85<br />

Wolf, I., 120<br />

Wong, C., 158<br />

Woo, T., 118<br />

Wood, J., 178<br />

Woudstra, S., 115<br />

Wray, N., 58, 73, 124<br />

Wright, A., 4<br />

Wright, E., 151<br />

Wright, M., 124<br />

Wu, J., 92<br />

Wu, K., 140<br />

Wu, R., 70<br />

Wu, Y., 193<br />

Wüst, S., 122<br />

Xavier, R., 28<br />

Xiao, X., 29<br />

Xie, W., 45<br />

Xing, G., 185<br />

Xing, J., 70<br />

Xiong, M., 99<br />

Xu, F., 22<br />

Xu, J., 22, 24<br />

Xu, Q., 131<br />

Xu, X., 193<br />

X<br />

Y<br />

Yamada, K., 81, 124, 145,<br />

192<br />

Yamamori, H., 117, 118<br />

Yan, J., 148<br />

Yandell, M., 70<br />

Yang, L., 70<br />

Yang, X., 70<br />

Yassin, A., 178<br />

Yasuda, Y., 70, 117, 118<br />

Yemmiganur Chandrash, J.,<br />

179<br />

Yeo, A., 200<br />

Yilmaz, Z., 113<br />

Yolken, R., 180<br />

Yoneda, H., 69, 82<br />

Yoshikawa, T., 81, 124<br />

Yoshimi, A., 70, 75<br />

Young, S., 156<br />

Young-­‐Wolff, K., 153<br />

Yu, C., 78<br />

Yu, H., 133<br />

Yuan, Q., 71, 109<br />

Z<br />

Zaharieva, I., 74<br />

Zai, C., 102, 113, 154, 158,<br />

200, 201<br />

Zai, G., 139, 158<br />

Zandi, P., 31, 54, 95, 104<br />

Zanoni, M., 157<br />

Zeanah, C., 112<br />

Zeggini, E., 98<br />

Zeledon, M., 122<br />

Zemojtel, T., 88<br />

Zhang, C., 21, 22<br />

Zhang, F., 48<br />

Zhang, J., 131, 198, 202


Zhang, L., 151, 185<br />

Zhang, P., 70<br />

Zhang, Y., 22, 102<br />

Zhang-­‐James, Y., 65<br />

Zhao, Q., 71, 198<br />

Zhao, Z., 22, 23, 24<br />

Zheng, D., 65<br />

Zhou, Z., 71, 109<br />

211<br />

Zhu, S., 30<br />

Zhu, X., 102<br />

Zitman, F., 115<br />

Zlojutro, M., 46, 103<br />

Zohar, J., 197<br />

Zsanett, T., 191<br />

Zs<strong>of</strong>ia, N., 191<br />

Zupanc, T., 173


Please join us for these<br />

Future Events:<br />

XX th <strong>World</strong> <strong>Congress</strong> <strong>of</strong> <strong>Psychiatric</strong> <strong>Genetics</strong><br />

October 14-18, 2012<br />

Hamburg, Germany<br />

XXI st <strong>World</strong> <strong>Congress</strong> <strong>of</strong> <strong>Psychiatric</strong> <strong>Genetics</strong><br />

October 17-21, 2013<br />

Boston, Massachusetts, USA<br />

http://www.ispg.net/

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!