A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The...

125
A Genetic Association Study of Glutamate Transporter Genes SLC1A1 and SLC1A3 in Tourette syndrome and Attention Deficit/Hyperactivity Disorder by Alexandra Kentebe A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Alexandra Kentebe 2014

Transcript of A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The...

Page 1: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

A Genetic Association Study of Glutamate Transporter

Genes SLC1A1 and SLC1A3 in Tourette syndrome and

Attention Deficit/Hyperactivity Disorder

by

Alexandra Kentebe

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Institute of Medical Science

University of Toronto

© Copyright by Alexandra Kentebe 2014

Page 2: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

ii

A Genetic Association Study of Glutamate Transporter Genes SLC1A1 and SLC1A3 in Tourette syndrome and Attention

Deficit/Hyperactivity Disorder

Alexandra Kentebe

Master of Science

Institute of Medical Science

University of Toronto

2014

Abstract Tourette syndrome (TS) is a heterogeneous disorder that may share etiological factors with

attention-deficit/ hyperactivity disorder (ADHD) and obsessive-compulsive disorder (OCD).

Genetic factors play a large role in the pathogenesis of these disorders and disruption of the

glutamatergic system has been implicated through multiple lines of study. On this basis, the

current investigation evaluates the role of two glutamate transporter genes: SLC1A1 and

SLC1A3. SLC1A1 was selected for study based on its function and involvement in OCD, and

was tested for association with TS and ADHD, using two independent samples. SLC1A3 was

selected for study based on function and possible involvement in ADHD, and was tested for

association using our TS sample. None of the genotyped markers remained significant

following corrections for multiple testing. Considering that we observed several trends for

association, studies using larger samples are required to determine whether these genes are

associated with TS or ADHD.

Page 3: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

iii

Acknowledgments

I would like to thank my supervisor, Dr. Cathy L. Barr for giving me the opportunity

to learn from her and work in her lab. Without her guidance and patience, this thesis would

not have been possible. I am also grateful for the members of the Barr lab: Kathryn Tzimika,

Karen Wigg, Yu Feng, Lissette Gomez, Ginger Misener, Christopher Tran, Sabah Husain

and Kevin Zhang, for their support and advice during my program. To my Program Advisory

Committee: Dr. Paul Sandor, Dr. Jo Knight and Dr. Margaret Richter, thank you for your

time, your advice and your encouragement. I would also like to thank Dr. Cindi Morshead

for her support throughout my program.

Thank you to the Institute of Medical Science, The Tourette Syndrome Association of

America, the Ontario Mental Health Foundation, The Tourette Syndrome Foundation of

Canada, and the Canadian Institute of Health Research (CIHR) for financially supporting this

project.

Finally, I would like to thank my friends and family for their support during my

program. Thank you to my husband, Ryan, for getting me through the tough days, for being

understanding during the busy days and for your continuous encouragement.

Page 4: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

iv

Table of Contents Abstract(.............................................................................................................................................(ii!

Acknowledgments(........................................................................................................................(iii!

Table(of(Contents(..........................................................................................................................(iv!

List(of(Tables(................................................................................................................................(viii!

List(of(Figures(.................................................................................................................................(ix!

List(of(Abbreviations(.....................................................................................................................(x!

! Introduction(................................................................................................................(1!Chapter!1

1.1! Introduction(to(Tourette(syndrome(.......................................................................................(1!

1.2! Evidence(of(a(genetic(basis(of(TS(.............................................................................................(2!

1.3! Model(of(inheritance(of(TS(.........................................................................................................(3!

1.4! Identification(of(genetic(susceptibility(factors(...................................................................(4!

1.4.1! Linkage!studies!.........................................................................................................................................!4!

1.4.2! Association!studies!.................................................................................................................................!5!

1.4.3! Linkage!Disequilibrium!........................................................................................................................!8!

1.5! The(Behavioral(Spectrum(of(TS(.............................................................................................(10!

1.5.1! Chronic!Multiple!Tics!..........................................................................................................................!11!

1.5.2! TS!comorbidities!and!potential!TS!subgroups!.........................................................................!12!

1.5.3! Comorbid!TS!with!OCD!......................................................................................................................!12!

1.5.4! Evidence!of!genetic!overlap!between!TS!and!OCD!.................................................................!14!

1.5.5! Comorbid!TS!and!ADHD!....................................................................................................................!15!

1.5.6! Relevance!.................................................................................................................................................!17!

Page 5: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

v

1.6! The(Glutamatergic(System(.......................................................................................................(17!

1.7! The(Glutamatergic(Hypothesis(...............................................................................................(19!

1.7.1! Evidence!implicating!the!glutamatergic!system!in!TS!..........................................................!20!

1.7.2! Evidence!implicating!the!glutamatergic!system!in!ADHD!..................................................!21!

1.7.3! Evidence!implicating!the!glutamatergic!system!in!OCD!......................................................!21!

1.8! Genetic(evidence(of(overlap(between(TS,(ADHD(and(OCD(............................................(22!

1.8.1! Common!susceptibility!loci!of!TS,!ADHD!and!OCD!................................................................!22!

1.9! Genetic(studies(in(candidate(region(9p(...............................................................................(23!

1.9.1! Linkage!studies!implicating!9p!in!OCD!.......................................................................................!23!

1.9.2! Case!reports!of!9p!chromosomal!abnormalities!in!TS!.........................................................!23!

1.9.3! Candidate!gene!studies!at!9p24!.....................................................................................................!24!

1.10! SLC1A1(..........................................................................................................................................(24!

1.10.1! SLC1A1!genetic!studies!in!OCD!....................................................................................................!24!

1.10.2! Rationale!for!investigating!SLC1A1!in!TS!and!ADHD!.........................................................!27!

1.11! Genetic(studies(in(candidate(region(5p13(.......................................................................(27!

1.11.1! Linkage!studies!implicating!5p13!in!ADHD!............................................................................!27!

1.11.2! Linkage!studies!implicating!5p13!in!TS!...................................................................................!28!

1.12! SLC1A3(..........................................................................................................................................(29!

1.12.1! SLC1A3!studies!of!ADHD!................................................................................................................!29!

1.12.2! SLC1A3!studies!of!TS!........................................................................................................................!30!

1.12.3! Rationale!for!investigating!SLC1A3!in!TS!and!ADHD!.........................................................!31!

1.13! Chromatin(Signatures(and(Gene(Expression(..................................................................(32!

1.14! Research(Aims(&(Hypotheses(..............................................................................................(38!

Page 6: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

vi

! Materials(and(Methods(........................................................................................(39!Chapter!2

2.1! The(TS(familyUbased(sample(...................................................................................................(39!

2.1.1! Recruitment!and!exclusion!criteria!..............................................................................................!39!

2.1.2! Sample!Composition!...........................................................................................................................!39!

2.1.3! Diagnostic!Assessment!.......................................................................................................................!40!

2.2! The(ADHD(familyUbased(sample(............................................................................................(40!

2.2.1! Recruitment!and!exclusion!criteria!..............................................................................................!41!

2.2.2! Sample!Composition!...........................................................................................................................!41!

2.2.3! Diagnostic!&!Behavioural!Assessment!........................................................................................!41!

2.3! Isolation(&(Extraction(of(DNA(.................................................................................................(42!

2.4! SingleUNucleotide(Polymorphism(Selection(......................................................................(42!

2.4.1! SLC1A1!......................................................................................................................................................!42!

2.4.2! SLC1A3!......................................................................................................................................................!42!

2.5! SingleUNucleotide(Polymorphism(Genotyping(..................................................................(43!

2.6! Statistical(Analysis(.....................................................................................................................(44!

2.6.1! Association!Analyses!...........................................................................................................................!44!

2.6.2! Visualization!and!Interpretation!of!LD!blocks!.........................................................................!46!

2.6.3! Quality!Control!......................................................................................................................................!47!

2.6.4! Power!.........................................................................................................................................................!47!

2.6.5! Correction!for!Multiple!Testing!......................................................................................................!50!

! A(familyUbased(association(study(of(the(putative(ObsessiveUChapter!3

Compulsive(Disorder(gene,(SLC1A1, with(Tourette(Syndrome(and(AttentionU

Deficit/(Hyperactivity(Disorder(..............................................................................................(52!

Page 7: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

vii

3.1 ! Introduction(.................................................................................................................................(52!

3.2! SLC1A1'Results(.............................................................................................................................(58!

3.3! Tables(&(Figures(.........................................................................................................................(60!

! A(familyUbased(association(study(of(a(putative(AttentionUChapter!4

Deficit/Hyperactivity(Disorder(gene,(SLC1A3,(with(Tourette(syndrome(.................(64!

4.1! Introduction(.................................................................................................................................(64!

4.2! SLC1A3'Results(.............................................................................................................................(69!

4.3! Tables(&(Figures(.........................................................................................................................(72!

! Discussion(&(Future(Directions(........................................................................(76!Chapter!5

5.1! Discussion(.....................................................................................................................................(76!

5.1.1! SLC1A1!......................................................................................................................................................!76!

5.1.2! SLC1A3!......................................................................................................................................................!79!

5.1.3! Global!Discussion!of!SLC1A1!and!SLC1A3!.................................................................................!80!

5.2! Global(Limitations(......................................................................................................................(82!

5.3! Future(Direction(.........................................................................................................................(84!

5.3.1! SLC1A1!......................................................................................................................................................!84!

5.3.2! SLC1A3!......................................................................................................................................................!86!

5.3.3! Future!Candidate!Genes!for!Study!................................................................................................!88!

5.4! Conclusion(.....................................................................................................................................(88!

Page 8: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

viii

List of Tables Table 1–1. DSM-V Overview of TS, OCD & ADHD ........................................................... 36!

Table 1–2. Summary of Linkage Studies of TS ..................................................................... 37!

Table 3–1. SLC1A1 Single SNP analysis for TS sample ........................................................ 60!

Table 3–2. SLC1A1 Haplotype analysis of Block 3 for TS sample ........................................ 61!

Table 3–3. SLC1A1 Single SNP analysis for ADHD sample ................................................. 62!

Table 3–4. SLC1A1 Haplotype analysis for ADHD sample ................................................... 62!

Table 4–1. Single-marker TDT analysis for 10 genotyped SLC1A3 SNPs in TS sample ...... 72!

Table 4–2. SLC1A3 Haplotype analysis of rs2562571/rs4869675 for TS sample ................. 72!

Table 4–3. SLC1A3 Haplotype analysis of rs2269272/rs1529461 for TS sample ................. 73!

Page 9: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

ix

List of Figures Figure 3-1. LD plot of 17 genotyped SLC1A1 SNPs .............................................................. 63!

Figure 3-2. Relative positions of 17 genotyped SLC1A1 markers ......................................... 63!

Figure 4-1. LD plot of 10 genotyped SLC1A3 SNPs .............................................................. 74!

Figure 4-2. Relative positions of the 10 genotyped SLC1A3 SNPs. ....................................... 75!

Page 10: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

x

List of Abbreviations 1H-MRS Proton magnetic resonance spectroscopy

ADHD Attention Deficit/Hyperactivity Disorder

AMPA α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid

AMPAR α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid receptor

APA American Psychiatric Association

ASP Affected sib-pair

CMT Chronic Multiple Tics

CSTC Cortico-Striatal-Thalamic-Cortical pathway

DNA Deoxyribonucleic acid

DSM Diagnostic and Statistical Manual of Mental Disorders

DZ Dizygotic

EAAT Excitatory amino-acid transporter

ENCODE Encyclopedia of DNA elements

EO Early-onset

FBAT Family-based association test

GRR Genotype relative risk

Page 11: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

xi

Glx Glutamate + Glutamine

GLAST Glutamate aspartate transporter

GWAS Genome-wide association study

GWLS Genome-wide linkage scan

H3K4Me1 Mono-methylated lysine 4 of histone 3

H3K27Ac Acetylated lysine 27 of histone 3

HAT Histone acetyl-transferase

HWE Hardy-Weinberg equilibrium

K-SADS Kiddie Schedule for Affective Disorders and Schizophrenia

LD Linkage Disequilibrium

LO Late-onset

LOD Logarithm of odds

MAF Minor allele frequency

MLOD Multipoint maximum LOD score

MZ Monozygotic

NMDA N-methyl-D-aspartate

Page 12: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

xii

NMDAR N-methyl-D-aspartate receptor

NPL Nonparametric LOD score

OCB Obsessive-compulsive behaviour

OCD Obsessive-compulsive disorder

PCA Principal components analysis

PCR Polymerase chain reaction

PIC Parent Interview for Child Symptoms

RNA Ribonucleic Acid

SDS Sequence Detection System

SLC1A1 Solute carrier family 1, member 1

SLC1A3 Solute carrier family 1, member 3

SNP Single nucleotide polymorphism

TSAICG The Tourette Syndrome Association International Consortium of Genetics

TDT Transmission disequilibrium test

TF Transcription factor

TFBS Transcription factor binding sites

Page 13: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

xiii

TS Gilles de la Tourette syndrome

TTI Teacher Telephone Interviews

UCSC University of California Santa Cruz

Page 14: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

1

Introduction Chapter 1

1.1 Introduction to Tourette syndrome

The ability to think or behave without interruption is important to quality of life and social

interaction (Cutler, Murphy, Gilmour, & Heyman, 2009; Eddy, Cavanna, Gulisano, Calì,

Robertson, & Rizzo, 2012; Packer, 2005). Generally, interruptions come from external

factors and to some degree can be controlled; however, there are circumstances in which

interruptions originate from the individuals themselves. When unwanted vocalizations or

motor movements result from sudden, recurrent and non-rhythmic movement of a muscle

group it is referred to as a tic (American Psychiatric Association, 2013). It is estimated that

15%-25% of school-aged children worldwide present with tic symptoms that last for less

than a year (Robertson, 2008). An additional 1% of children worldwide experience both

motor and vocal tics (Robertson, 2012). According to the Diagnostic and Statistical Manual

of Mental Disorders 5th ed. (DSM-V) when vocal and motor tic symptoms last for longer

than a year and are unexplained by other medical conditions or substance abuse, a diagnosis

of Gilles de la Tourette syndrome (TS) may be made (APA, 2013; see Table 1-1 for

overview of TS).

Tourette syndrome is a neurobehavioral disorder with an onset generally between 5

and 7 years of age (Robertson, 2012). Initial tic symptoms are often simple and involve a

single muscle group. For instance, common simple tics that result in vocalizations include

grunting and vocal clearing of the throat, and simple motor movements include eye blinking

Page 15: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

2

and shoulder shrugging. Given that these symptoms are mild it is not uncommon for them to

go undiagnosed and therefore untreated (Robertson & Gourdie, 1990). Symptoms of TS

generally become progressively more severe approaching puberty but often improve into

adulthood in approximately 80% of children and adolescence (Bloch & Leckman, 2009).

The course described above suggests that TS may result from neurodevelopmental

delays. This is in contrast to progressive disorders which are typically neurodegenerative and

become more severe with time ( reviewed in Paschou, 2013). Multiple magnetic resonance

imaging studies (Fahim, Yoon, Sandor, Frey, & Evans, 2009; Sowell et al., 2008; Worbe et

al., 2010) provide evidence of support for this hypothesis by demonstrating that the

maturation of brain regions that mediate self-regulation (e.g., basal ganglia and cortex)

coincide with the course of TS (Plessen, 2013; Gogtay et al., 2004). Despite mounting

evidence that alterations to brain regions such as the basal ganglia and cortex are involved in

the pathogenesis of TS, specific neuroanatomical or neurochemical alterations have yet to be

established in the majority of patients (reviewed in McNaught & Mink, 2011).

1.2 Evidence of a genetic basis of TS

To date, multiple family studies have demonstrated that the rate of TS is 10 to 100 times

greater in first-degree relatives of TS patients than in the general population (Kidd, Prusoff,

& Cohen, 1980; Kidd & Pauls, 1982; Pauls, Cohen, Heimbuch, Detlor, & Kidd, 1981; Pauls,

Kruger, Leckman, Cohen, & Kidd, 1984). While substantial clustering of TS within families

does not provide definitive evidence of a genetic basis it can be used along with twin studies

to gain a better understanding of a disorder and related traits.

Page 16: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

3

Given that monozygotic twins (MZ) share identical genetic code and dizygotic twins

(DZ) only share on average 50% of the genetic code, twin studies are commonly used to

provide evidence of a genetic basis of a particular disorder or trait. Assuming environmental

factors are similar for both twins, a higher concordance rate amongst MZ twins when

compared to DZ twins suggests a genetic component for the underlying trait. When the

concordance rate is below 100% for monozygotic twins, environmental factors are also likely

involved in susceptibility.

Price, Kidd, Cohen, Pauls, and Leckman (1985) conducted the first twin study of TS

using 43 pairs of same-sex twins. When using the narrow diagnostic criteria of TS (TS only),

concordance rates for monozygotic and dizygotic twins were 53% and 8%, respectively.

Consistent with these findings, Hyde, Aaronson, Randolph, Rickler, and Weinberger (1992)

also observed a concordance rate of 56% for 16 pairs of MZ twins. These studies provided

evidence that both genetic and environmental factors contribute to TS. It has since been

demonstrated that approximately 60% of phenotypic variability of TS is accounted for by

genetic factors (Davis et al., 2013).

1.3 Model of inheritance of TS

Complex segregation analyses from family studies of TS support a major locus contribution

(Comings et al., 1984; Curtis, Robertson, & Gurling, 1992; Eapen, Pauls, & Robertson,

1993; Hastedt et al., 1995; Pauls, et al., 1986, 1990; Seuchter et al., 2000; Walkup et al.,

1996). However, whether this major locus is inherited in an additive or autosomal dominant

fashion, and whether there is a polygenic contribution of smaller effect remains unclear.

Page 17: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

4

Despite multiple attempts to locate a definitive susceptibility gene of major effect none have

been identified.

1.4 Identification of genetic susceptibility factors

1.4.1 Linkage studies

Genetic linkage studies allow for the identification of broad chromosomal regions that may

harbor susceptibility genes (Teare & Barrett, 2005). In principle this type of study screens for

genetic loci that co-segregate with a particular phenotype amongst relatives (Borecki &

Suarez, 2001). Based on a priori information or lack of information of the trait of interest,

one of two linkage analysis strategies (e.g., parametric or non-parametric methods) can be

used to identify susceptibility loci.

Parametric or model-based linkage analysis is commonly used for Mendelian

disorders with a known disease-allele frequency, and affection status of parents and offspring

in large multigenerational families (Shih & Whittemore, 2001; Teare & Barrett, 2005). This

method is based on the principle that markers proximal to the disease-locus are less likely to

undergo recombination and therefore more likely to segregate within affected relatives than

unaffected relatives (Shih & Whittemore, 2001). The likelihood that the marker and disease-

locus are linked is often measured using the logarithm of odds (LOD) score, with a score of 3

or greater indicating significant evidence of linkage (Teare & Barrett, 2005).

Non-parametric or model-free linkage analysis allows for the identification of

chromosomal susceptibility regions of traits with an unknown mode of inheritance. This is

typically the case for common complex traits with multiple genes and environmental factors

Page 18: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

5

contributing to etiology (Teare & Barrett, 2005; Borecki & Suarez, 2001; Weeks & Lathrop,

1995). Genotypes of affected relatives with common ancestry (e.g., affected siblings) are

analyzed for identical allele or haplotype patterns found at a greater rate than expected by

chance (Teare & Barrett, 2005). These patterns are proposed to be indicative of susceptibility

regions for that particular trait or disorder.

Multiple genome-wide and targeted linkage studies of TS have been conducted using

parametric and non-parametric strategies (see Table 1-2 for summary of TS linkage findings).

Using these methods, over 15 chromosomal risk regions have been implicated. The lack of

definitive TS susceptibility regions may be due to a number of complications including

differing ethnic compositions between studies. Nevertheless, regions implicated by linkage

studies are broad and contain multiple plausible candidate genes. Therefore, linkage studies

alone do not provide good resolution for gene discovery (Teare & Barrett, 2005). Another

challenge of the linkage study is its poor resolution for identifying genetic variants with

small effect sizes (Cirulli & Goldstein, 2010). Therefore, while linkage studies are important

for the discovery of regions harboring potential risk genes, other strategies are also required

to identify genes conferring susceptibility.

1.4.2 Association studies

Association studies are used to determine whether a particular marker is associated with a

trait in a population (Borecki & Suarez, 2001). In contrast to linkage studies, genetic

association studies allow for targeted examination of plausible risk genes within linked

regions, and may be used to identify common risk alleles with small to modest effect sizes

Page 19: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

6

(Cirulli & Goldstein, 2010). Two types of association studies are commonly used i)

candidate gene association studies and ii) genome-wide association studies (GWAS).

Candidate gene association studies are hypothesis-driven, such that gene selection is based

on a priori knowledge. This includes selecting a candidate gene based on biological

relevance to a particular trait or localization within a susceptibility region. In contrast,

GWAS involve genotyping of an array of markers with common alleles and are used to

detect risk alleles with small to moderate effect across the genome without a priori

knowledge (Cirulli & Goldstein, 2010; Hindorff et al., 2009; McCarthy & Hirschhorn, 2008).

Advantages of conducting a GWAS include the identification of novel risk genes or risk

variants located within intergenic regions (Hindorff et al., 2009) that may have originally

been overlooked.

Genetic association studies can be approached as either case-control or family-based.

The case-control design uses two groups: i) cases, which are affected individuals and ii)

controls, individuals who are unaffected with the trait of interest. Case-control studies

determine association by comparing the allele frequencies between these two groups (Clarke

et al., 2011).

A major challenge with the case-control approach is that cases and controls may

differ in ethnicity (Curtis, 1997; Devlin & Roeder, 1999; Nicodemus, Luna, & Shugart,

2007). This is of concern because individuals of differing ethnic groups have different allele

frequencies, which can result in false-positive associations (Nicodemus et al., 2007). To

reduce confounding by population stratification, investigators attempt to select cases and

Page 20: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

7

controls from the same ethnic groups; however, even with careful selection, the effects of

population stratification may not be completely eliminated (Clarke et al., 2011). To address

this limitation, studies involving large number markers, such as GWAS, implement statistical

tools that can make inferences about population structure from genotype data. Individuals

that do not sufficiently match up are removed from the study, improving the ancestral-

homogeneity of the sample. Principal-component analysis (PCA) and multidimensional

scaling (MDS) are two commonly used statistical strategies to describe the data using fewer

variables (Patterson, Price, & Reich, 2006; Price et al., 2008). Population stratification can

also be addressed by using genotypes of relatives (e.g., parents or siblings), rather than

unrelated cases and controls.

Recently the first GWAS of TS was conducted to identify common risk variants

(Scharf et al., 2012). In this study 496,877 SNPs were tested for association in 1285 TS

cases and 4964 European-matched controls (Scharf et al., 2012). Despite using the largest TS

sample to date, no markers reached the genome-wide threshold of significance (p < 5 x 10-8,

Scharf et al., 2012). The strongest signal from the primary analysis of this study was from an

intron of COL27A1 (p = 1.85 x 10-6), which is localized on chromosome 9q32. COL27A1

encodes for a fibrillar collagen and is expressed in multiple brain regions; however,

according to Fox (2008) the involvement of this gene in the development of the central

nervous system remains unclear. Scharf and colleagues (2012) also conducted a secondary

analysis that included 211 TS cases and 285 controls of Latin-American descent. The

expansion of the sample to include affected individuals of Latin-American descent did not

improve efforts to identify significant associations with TS. Given that previous GWAS of

Page 21: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

8

other psychiatric disorders (e.g., bipolar disorder and schizophrenia) required larger sample

sizes to reach the threshold for genome significance (Mühleisen, Leber, Schulze, &

Strohmaier, 2014; Schizophrenia Psychiatric Genome-Wide Association Study Consortium,

2011; Sullivan et al., 2012) it has been proposed that this study was underpowered (Scharf et

al., 2012). Therefore, while no genes met the threshold of genome-wide association, variants

with sub-threshold p values may reflect true associations.

Multiple candidate gene association studies have been conducted, implicating genes

of various pathways. Two of the most commonly studied genes include DAT1 (Yoon et al.,

2007; Comings et al., 1996) and SLITRK1 (Stillman et al., 2009; Abelson et al., 2005;

Miranda et al., 2009; O'Roak et al., 2010; Karagiannidis et al., 2012). Although it is possible

that these genes are involved in TS susceptibility, the risk alleles of these genes alone do not

account for TS susceptibility.

1.4.3 Linkage Disequilibrium

Recombination events over generations produce new combinations of alleles. Depending on

genomic distance, certain chromosomal sites are more likely to undergo recombination than

others. This gives rise to alleles at the same loci being more or less likely to be inherited

together. When the association of alleles at two or more loci is nonrandom it is referred to as

linkage disequilibrium (Pritchard & Przeworski, 2001). Genetic association studies of disease

rely on this property to map genes that cause disease. For this particular investigation LD

was used to systematically select SNPs for association testing with TS and ADHD in the

respective samples.

Page 22: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

9

1.4.3.1 Measurements.of.Linkage.Disequilibrium.

There are multiple statistics used to describe the LD between marker pairs, however, two

frequently used measures are i) the Lewontin D’ statistic and ii) the squared correlation

coefficient, r2 (Mourad, Sinoquet, Dina, & Leray, 2011; Delvin & Risch, 1995).

The Lewontin D’ is a standardized function based on the measure D, the difference

between the observed and expected haplotype frequencies (Mueller, 2004). Lewontin D’ is

derived by dividing D by its maximum value, Dmax and evaluates the independence of two

markers given maximum linkage (Lewontin, 1964). Alleles at two loci with little distance in-

between are more likely to have little to no historical recombination. This relationship is

indicated by a D’ value approaching 1. When D’=1 it is called complete LD. When this is the

case, given the allele frequencies, the correlation between alleles is described as strong as

possible (Carlson et al., 2004). As genetic distance increases, the strength of correlation

between these markers decreases due to recombination throughout generations. Frequent

recombination between markers of two loci on the same chromosome would be represented

by a D’ value approaching 0.

The squared Pearson’s correlation coefficient, r2, similar to D’, may be used to assess

the correlation between two markers. To gain a better understanding of r2 consider two

biallelic loci on the same chromosome. The first locus consists of alleles A and a and the

second locus consists of alleles B and b (common and minor alleles, respectively). Based on

recombination, the possible haplotype combinations include AB, Ab, aB and ab. r2 is

computed with the following equation:

Page 23: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

10

, where p is frequency

Equation (1) shows that although two markers may be correlated, the strength of LD

is reduced if the allele frequencies greatly differ (Edge, Gorroochurn, & Rosenberg, 2013).

When the allele frequencies are the same and when r2=1, the relationship between markers is

described as perfect LD (Reich et al., 2001). Generally, the LD between two loci is rarely

perfect, as allele frequencies at two loci often differ (Edge, Gorroochurn, & Rosenberg,

2013). Similar to D’, when r2=0 it indicates independence: information regarding one allele

does not provide information for another allele. When alleles are inherited independently

from one another it is referred to as linkage equilibrium.

Ultimately the use of these measurements depends on the context (Pritchard &

Przeworski, 2001). D’ is often used to compare LD between markers in different populations

(Reich et al., 2001), whereas r2 is better suited for association studies (e.g., tagSNP selection).

1.5 The Behavioral Spectrum of TS

Failure to identify many risk alleles is not due to a lack of susceptibility regions. On the

contrary, linkage studies of TS have implicated multiple regions on at least 10 different

chromosomes. While these findings in addition to family and twin studies, support the

involvement of genetic factors they are problematic: many of these linkage findings have yet

to be replicated or have only produced weak evidence of linkage. Furthermore, there are

hundreds of genes within these regions, such that it would be difficult to sufficiently narrow

the search for genes conferring risk.

r2 =pAB − pA pB( )2

pA pB pa pb

Page 24: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

11

The etiologic complexity of TS likely plays a role in the challenge of identifying

specific risk genes. From a clinical perspective, individuals affected with TS vary in

presentation (e.g., severity, age of onset, symptom types). This, in addition to the number of

highlighted linkage regions, suggests that TS may not only be clinically heterogeneous but

also genetically heterogeneous. To clarify current genetic findings of TS, investigators have

searched for more homogeneous subgroups. To date, at least three subgroups have been

identified including: i) TS+ obsessive compulsive behavior, (OCB) the less severe form of

OCD that does not meet the impairment criteria ii) TS+OCD and iii) TS+OCD+ADHD

(Alsobrook & Pauls, 2002; Grados et al., 2008; Mathews et al., 2007; Robertson, et al., 2007,

2008b). One possibility is that there is variable expressivity of TS risk genes, such that these

subgroups represent more severe manifestations of TS, each with common and unique risk

factors (Grados et al., 2008; Mathews & Grados, 2011; Pauls, et al., 1986, 1990, 1992).

Under this assumption of expressivity, additional variables including gene-environment

interactions and non-genetic factors may also extend the behavioral spectrum to also include

milder forms of TS. Therefore, consideration of the behavioral spectrum of TS may improve

efforts to identify additional risk alleles.

1.5.1 Chronic Multiple Tics

Chronic multiple tics (CMT) is a childhood-onset psychiatric disorder characterized by the

presence of either motor or vocal tics for at least a year (APA, 2013). Family studies of TS

demonstrate that the rate of CMT amongst first-degree relatives is 5- to 20- fold greater when

compared to the general population (Pauls, Cohen, Heimbuch, Detlor, & Kidd, 1981; Pauls,

Raymond, Stevenson, & Leckman, 1991). While positive familial aggregation of CMT

Page 25: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

12

within families of TS patients does not provide definitive evidence of a genetic basis,

broadening the diagnostic criteria to include CMT (and other tic disorders) in a twin study of

TS increased partial concordance rates from 53%-56% to 77%-94% in monozygotic twins,

and 8% to 23% in dizygotic twins (Hyde et al., 1992; Price et al., 1985). Based on the

evidence, CMT is believed to represent a less severe manifestation of TS (Pauls, et al., 1981,

1990; Price et al., 1985).

1.5.2 TS comorbidities and potential TS subgroups

Multiple neuropsychiatric disorders have been found at higher rates in relatives of TS

patients when compared to rates in the general population. It is estimated that 90% of

individuals affected with TS also have additional psychiatric or behavioral conditions

(Freeman et al., 2000). TS with comorbid obsessive-compulsive disorder (OCD) and/or

attention-deficit/ hyperactivity disorder (ADHD) are proposed to represent potential subtypes

of the TS spectrum. The relationship between TS and comorbid OCD and ADHD are

discussed in more detail below.

1.5.3 Comorbid TS with OCD

OCD is heritable psychiatric disorder (van Grootheest, Cath, Beekman, & Boomsma, 2005;

Hettema, Neale, & Kendler, 2001) characterized by obsessions and/or compulsions that are

time consuming or result in marked distress, impairment , or are difficult to ignore (APA,

2013; see Table 1-1 for overview of OCD). Obsessions are any persistent, unwanted and

intrusive thoughts, images or urges (APA, 2013). Compulsions are excessive or repetitive

behaviours or mental acts that typically reduce anxiety brought on by obsessions (APA,

Page 26: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

13

2013). Compulsions may also include any behaviors or mental acts that are believed to

prevent or lessen an unrelated event (APA, 2013).

Two distinct onsets have been observed in OCD patients: early-onset and late-onset.

Symptoms of early-onset OCD (also referred to as childhood- or pediatric- OCD) generally

begin around 10 years of age. Clinical and epidemiology studies of OCD have found that

males are four times more likely to be diagnosed with early-onset OCD than females. While

childhood-OCD describes a subgroup of individuals affected with OCD before the age of 18,

late onset (also known as adult-onset) OCD describes individuals with OCD whose

symptoms begin at or after 18 years of age. In addition to differences in age of onset,

evidence from family studies indicate that early-onset OCD is more heritable than late-onset

providing further support that these onsets may have unique and overlapping etiological

factors (van Grootheest et al., 2005; Davis et al., 2013).

The estimated worldwide prevalence of OCD is 2.5% (Ruscio, Stein, Chiu, & Kessler,

The epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey

Replication, 2010) yet clinical-based epidemiology studies show that up to 66% of TS

patients are also affected with OCD (Freeman et al., 2000), and up to 80% of TS patients

present with OCB (Pauls et al., 1986). Considering that these rates are substantially higher in

individuals with TS than the general population (Pauls, Alsobrook, Goodman, Rasmussen, &

Leckman, 1995), it has been proposed that TS and some phenotypes of OCD share

etiological factors (Eichstedt & Arnold, 2001; Mathews & Grados, 2011; Pauls, et al., 1986,

1995). Symptom dimensions of OCD that are related to TS include obsessions involving

Page 27: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

14

symmetry, and compulsions involving touching, order, repetition, arranging and counting

(Bloch & Leckman, 2009; Grados & Mathews, 2009; Leckman et al., 2003; Rizzo, Gulisano,

Calì, & Curatolo, 2012). Other studies have also found that early-onset OCD (Hemmings et

al., 2004), as well as subclinical OCD (OCB) are also frequently associated with TS (Eapen

et al., 1997).

1.5.4 Evidence of genetic overlap between TS and OCD

Evidence of shared genetic susceptibility comes from family and twin studies of large TS

families. These studies show that TS and certain symptom dimensions of OCD co-segregate

within families (Curtis, Robertson, & Gurling, 1992; Grados et al., 2001; McNaught & Mink,

2011; Pauls et al., 1990). Other family studies report increased incidence of OCD (with or

without tics) amongst relatives of TS probands (Pauls et al., 1991). The rate of TS (with or

without OCD) amongst families of OCD probands has also been found to be significantly

greater than expected by chance (Pauls & Leckman, 1986). These rates were observed

regardless of a proband diagnosis of comorbid TS or OCD (Eapen et al. 1993; Pauls, et al.,

1986, 1991; Pitman, Green, Jenike, & Mesulam, 1987).

Within recent years multiple bivariate analyses have been conducted to uncover the

genetic correlation between TS and OCD. These studies have demonstrated heritability

estimate of 0.40, suggesting that genetics account for some of the overlap observed between

TS and OCD (Davis et al., 2013). Given the evidence of genetic overlap between phenotypes

of OCD and TS (Pauls, et al., 1984, 1986, 1991; Price et al., 1985; Walkup, Leckman, Price,

Hardin, Ort, & Cohen, 1988), a recent cross- disorder GWAS consisting of TS and OCD

Page 28: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

15

cases, aimed to gain a better understanding of the genetic etiological architecture between TS

and OCD. Although no genetic variants reached the threshold for genome-wide association

(p= 5x 10-8), additional analysis provided support that OCD associated with TS (TS+OCD) is

likely genetically distinct from OCD only (Davis et al., 2013).

Several overlapping brain regions have also been implicated in both TS and OCD.

For instance, neuroimaging studies of children with TS and OCD demonstrate altered

volume or activation of the striatum (Albin & Mink, 2006; Benedetti et al., 2012; Hoexter et

al., 2012; Peterson, et al., 2000, 2003; Pujol et al., 2004; Zarei et al., 2011) and cerebellum

(Bohlhalter et al., 2006; Pujol et al., 2004; Zarei et al., 2011). Despite this evidence, specific

neuroanatomical alterations involved in TS, with or without OCD, have yet to be identified.

1.5.5 Comorbid TS and ADHD

ADHD is a childhood-onset psychiatric disorder characterized by persistent inattentive or

hyperactive-impulsive behaviour that is inappropriate for one’s developmental age and

results in impaired functioning (APA, 2013; see Table 1-1 for overview of ADHD). The

onset for ADHD is prior to the age of 12 (APA, 2013) with motor hyperactivity generally

preceding inattentiveness or cognitive hyperactivity-impulsivity. As with other

neurodevelopmental psychiatric conditions, clinical presentation of ADHD is dynamic and

commonly changes into adolescence and adulthood (APA, 2013; Turgay et al., 2012).

ADHD affects an estimated 5%-12% children worldwide (Biederman & Faraone,

2005; Polanczyk, et al., 2007a, 2007b). As noted earlier, ADHD is frequently comorbid with

TS, with clinical-based prevalence estimates around 55% (Freeman & Consortium, 2007;

Page 29: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

16

Ludolph, Roessner, Münchau, & Müller-Vahl, 2012) and community-based reports around

38% (Pringsheim & Hammer, 2013). This discrepancy may be due to the fact that comorbid

ADHD is associated with more severe deficits, resulting in increased likelihood of referral to

clinics. Nevertheless, the prevalence of ADHD is substantially greater than expected in both

clinical and population based settings.

Early family studies suggest that ADHD represents an alternative phenotype of TS

risk genes (Comings, et al., 1984, 1987, 2001; Knell & Comings, 1993); however, more

recent findings indicate a more complex relationship (Grados et al., 2008; Pauls, et al., 1986b,

193; Stewart et al., 2006; Mathews & Grados, 2011). It has since been proposed that TS and

ADHD are not manifestations of the same genetic risk factors; rather the purest forms of TS

and ADHD likely represent distinct entities with different genetic factors (Murphy & Muter,

2012; Stewart et al., 2006). Further support for this hypothesis comes from the lack of

evidence of a genetic correlation between TS and ADHD when no other conditions are

considered (Mathews & Grados, 2011). However, there is some evidence indicating that in

some cases the co-occurrence of TS and ADHD may be mediated by OCD. When found in

the same individual TS, ADHD, and OCD (TS+ADHD+OCD) has been described as highly

heritable (Grados et al., 2008) and informative of predicting offspring diagnosis from

parental diagnosis (Mathews & Grados, 2011). It is plausible that TS+ADHD+OCD is a

manifestation with additive overlapping genetic risk factors of TS, OCD and ADHD.

Page 30: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

17

1.5.6 Relevance

In contrast to TS, which infrequently results in impaired functioning (APA, 2013), comorbid

TS with ADHD and/or OCD is often associated with impaired social functioning, self-value

and ability to thrive in a work or academic environment (Conelea et al., 2011; Eddy et al.,

2012; Pringsheim, Lang, Kurlan, Pearce, & Sandor, 2009; Rizzo et al., 2007). Moreover, TS-

affected individuals with additional diagnoses are also more likely to be perceived as being

deliberate disruptive and have a reduced quality of life (Debes, Hjalgrim, & Skov, 2009;

Eddy et al., 2012; Hassan & Cavanna, 2012). Efforts to treat TS only, ADHD only, and OCD

only are currently subpar. It is therefore not unexpected that there are no definitive

treatments for individuals with any combination of these disorders. Due to the lack of

specific treatment strategies, current therapeutic-drug efforts often result in exacerbation of

comorbid symptoms (Lombroso & Scahill, 2008). Refinement of the TS phenotype into

subgroups may not only give researchers a better understanding of the mechanisms

underlying TS, but may also guide the exploration of therapeutics of these comorbid

disorders.

1.6 The Glutamatergic System

Glutamate is the main excitatory neurotransmitter in the mammalian central nervous system

(Kanai et al., 2013). Stored in vesicles, glutamate is released at the presynaptic bouton in

response to influx of calcium ions. At the postsynaptic cleft, glutamate mediates excitatory

signal transduction through binding to one of two classes of glutamate receptors: i) ligand-

gated ion channels (e.g., ionotropic receptors) or ii) G-coupled proteins (metabotropic

receptors). Metabotropic receptors are involved in slow transmission and long- term changes

Page 31: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

18

(reviewed in Riedel, Platt, & Micheau, 2003). In contrast, ionotropic receptors mediate fast

transmission and facilitate the transport of Na2+ and K+ ions. Three ionotropic receptors exist:

N-methyl-D-aspartate (NMDA), α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid

(AMPA) and kainite. Given that AMPA receptors (AMPAR) and kainite receptors can be

activated by the same agonists, these receptors are collectively classified as non-NMDA

receptors (Ozawa, Kamiya, & Tsuzuki, 1998).

The activation of AMPA and NMDA receptors (AMPAR and NMDAR, respectively)

mediate synaptic changes underlying behaviour by a process known as synaptic plasticity

(Ozawa et al., 1998). Glutamate binding to the AMPAR results in excitatory postsynaptic

potentials on the postsynaptic neuron, causing the removal of Mg2+ from the NMDAR ion

channel (Clements & Westbrook, 1991; Singer, Morris, & Grados, 2010). This allows for

extracellular glutamate- and glycine- (or D-serine) binding of the NDMAR, causing further

depolarization and increasing calcium permeability into the postsynaptic neuron. Within the

neuron, calcium ions act as a second messenger, mediating a cascade of intracellular

signaling involved in altering the surface of neurons (Purves et al., 2001). The role of

glutamate in synaptic plasticity is thus important in the maturation of synapses and ultimately

the development of the central nervous system (Ozawa et al., 1998; Riedel, Platt, & Micheau,

2003).

Despite playing a critical role in the development of normal brain function (Zhou &

Danbolt, 2013), when found in excess, glutamate can also act as a neurotoxin via

excitotoxicity (Ozawa, Kamiya, & Tsuzuki, 1998). During this process, excessive activation

Page 32: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

19

of the NMDAR allows for aberrant concentrations of calcium into the postsynaptic neuron

(Purves et al., 2001). As a result calcium-dependent enzymes such as proteasomes become

active and degrade essential cell proteins causing cell death (Brennan-Minnella, Shen, El-

Benna, & Swanson, 2013).

Due to the implications of inappropriate glutamate concentrations in the brain, synaptic

glutamate is regulated by excitatory amino-acid transporters (EAATs), which are localized

on either glia cells or neuron. Alterations to the localization or capacity of these transporters

may in turn disrupt synaptic signaling (Tzingounis & Wadiche, 2007). As a result the

expression of glutamate transporters is critical for maintaining appropriate glutamate levels

within synapses (Portera-Cailliau, Price, & Martin, 1997).

1.7 The Glutamatergic Hypothesis

Although no definitive neuroanatomical localizations of TS have been discovered, multiple

lines of evidence suggest that abnormal synaptic neurotransmission of the cortico-striatal-

thalamic-cortical (CSTC) pathways underlies TS pathophysiology (Singer et al., 2010;

reviewed in Stern, Blair, & Peterson, 2008). These pathways are proposed to influence

behaviors for motor function, impulsivity, attention and executive function (Stahl, 2009;

Udvardi, Nespoli, Rizzo, Hengerer, & Ludolph, 2013). Given the involvement of glutamate

in excitatory neurotransmission, synaptic plasticity and excitotoxicity, aberrant activity in

regions associated with the CSTC circuits may result from dysregulation of the glutamatergic

system. To date, abnormal activity and glutamate concentrations within the CSTC have been

implicated in a variety of neurodevelopment and neurodegenerative disorders including

Page 33: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

20

Huntington’s disease (Behrens, Franz, Woodman, Lindenberg, & Landwehrmeyer, 2002),

Alzheimer’s disease (Francis, 2003), epileptic seizures (Tanaka et al., 1997) and autism

spectrum disorders (Choudhury, Lahiri, & Rajamma, 2012). The following section will

overview findings from rodent models, functional imaging studies, pharmacological studies

and genetic studies that support the hypothesis that dysregulation of the glutamatergic system

in the CSTC may contribute to TS and its related disorders, ADHD and OCD.

1.7.1 Evidence implicating the glutamatergic system in TS

To date, only one proton magnetic resonance spectroscopy (1H-MRS) study of brains of

individuals with TS has been conducted (DeVito et al., 2005). While DeVito and colleagues

were unable to detect abnormal glutamate + glutamine (glx) concentrations in the frontal

cortex, striatum or thalamus there is mounting evidence from other lines of study suggesting

the involvement of abnormal glutamate neurotransmission in TS. For instance, reduced

glutamate concentrations in the globus pallidus have been observed in post-mortem brains of

TS patients (Anderson et al., 1992). Engineered using a transgene encoding for D1 dopamine

receptor neurons with enzymatic cholera toxin, D1CT-7 mice models have also implicated

abnormal glutamatergic transmission in TS (McGrath, Campbell, Parks, & Burton, 2000;

Nordstrom & Burton, 2002). Nordstrom and Burton (2002) observed striatal hyperactivity

from cortical glutamatergic projections in D1CT-7 mice. Exacerbation of tic symptoms (e.g.,

increased number of tics and decreased tic responsiveness to drug therapy) in D1CT-7 mice

has also been observed following administration of glutamatergic drugs (McGrath et al.,

2000). Current pharmaceutical efforts are also investigating anti-glutamatergic drugs (i.e.,

Page 34: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

21

riluzole and N-acetylcysteine) as possible treatment of tics (Singer, 2010; Udvardi et al.,

2013).

1.7.2 Evidence implicating the glutamatergic system in ADHD

Altered glx concentrations in the brains of individuals affected with ADHD have been

reported by multiple studies using 1H-MRS (Perlov et al., 2009). One 1H-MRS study

revealed a significant decrease in the glutamate/glutamine/GABA to

creatine/phosphocreatine ratio in the striatum following treatment with ADHD medication

(Carrey et al., 2003). In another study using 1H-MRS, striatal glx concentrations were also

found to be low in ADHD patients (Maltezos et al., 2014). These studies in addition to others

have suggested that hypoactivation of glutamatergic projections extending to the striatum

may be involved in ADHD pathology; however, opposing findings have also been reported.

For instance, Carrey and colleagues (2007) also conducted a 1H-MRS study, but observed

greater, not lower, striatal glx concentrations in the brains of individuals with ADHD when

compared to controls. Furthermore, hyperactive glutamate terminating in the prefrontal

cortex and the striatum has also been observed in spontaneously hypertensive rats, a rodent

model of ADHD (Miller, Pomerleau, Huettl, Gerhardt, & Glaser, 2014). Finally, genes

involved in the glutamatergic system including GRIN2B (Dorval et al., 2007) and SLC1A3

(Elia et al., 2009; Turic et al., 2005) have also been implicated in ADHD.

1.7.3 Evidence implicating the glutamatergic system in OCD

Abnormal glutamate signaling has been hypothesized to contribute to OCD pathology

(reviewed in Macmaster & Rosenberg, 2010). A neuroimaging study using 1H-MRS

Page 35: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

22

observed higher glx concentration in the striatum of OCD patients when compared to

controls (Rosenberg & Keshavan, 1998). In a subsequent study, Rosenberg and colleagues

found that striatal glx decreased following treatment with paroxetine (Rosenberg et al., 2000),

an accepted drug used to treat OCD symptoms. Considering the evidence, glutamatergic

drugs have been proposed to have therapeutic influence in OCD patients (Coric et al., 2005;

Pasquini & Biondi, 2006; Poyurovsky, Weizman, Weizman, & Koran, 2005). In humans,

Chakrabarty (2005) observed increased glutamate levels in the cerebrospinal fluid of OCD

patients. Multiple genetic studies of OCD have also implicated genes involved in the

glutamatergic system including SLC1A1 (Arnold, Sicard, Burroughs, Richter, & Kennedy,

2006; Dickel et al., 2006; Samuels et al., 2011; Shugart et al., 2009; Stewart et al., 2007;

Porton et al., 2013; Wendland et al., 2009) and GRIN2B (Arnold et al., 2004).

1.8 Genetic evidence of overlap between TS, ADHD and OCD

1.8.1 Common susceptibility loci of TS, ADHD and OCD

Evidence from genome-wide and targeted linkage scans have implicated multiple

chromosomal loci in TS, ADHD and OCD; however, no susceptibility region has been linked

to all three disorders. Reports of chromosomal abnormalities in addition to regions with

positive or suggestive findings from linkage studies may provide clues to chromosomal

regions harboring susceptibility genes involved in all three disorders. Using linkage and case

reports, two regions, chromosome 5p13 and 9p24, have been selected for further study in the

present investigation.

Page 36: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

23

1.9 Genetic studies in candidate region 9p

1.9.1 Linkage studies implicating 9p in OCD

Multiple susceptibility regions have been identified for OCD; however, only linkage to

region 9p has been replicated (Hanna et al., 2002; Willour et al., 2004). Genome-wide

linkage scans of seven multigenerational families affected with early-onset OCD found

suggestive linkage to region 9p using a dominant model of inheritance (LOD=2.25; Hanna et

al., 2002). Fine mapping narrowed this region to marker D9S288, which is located at region

9p24. A subsequent linkage study using an independent sample targeted this region with the

objective of replicating these findings (Willour et al., 2004). The strongest signals came from

markers D9S1792 and D9S1813, two markers in proximity with the region highlighted in the

Hanna et al. study.

1.9.2 Case reports of 9p chromosomal abnormalities in TS

Two case reports have implicated chromosome 9p in TS (Taylor et al., 1991; Singh, Howe,

Jordan, & Hara, 1982). The first case involves a woman with a triple X mosaicism and a

deletion of the 9p region. Clinical features present in this woman included seizure, mental

retardation and TS (Singh et al., 1982). A second case implicating 9p was observed in 1991

by Taylor et al. This report describes an adolescent male with developmental delays who was

also affected with TS and OCD. Further evaluation of this subject’s chromosomal

architecture revealed a partial deletion of 9p of the terminal region.

Page 37: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

24

1.9.3 Candidate gene studies at 9p24

Given the likely role of the glutamatergic pathway in the etiology of TS, ADHD and OCD,

genes involved in the glutamatergic system are of particular interest. Of the 41 genes located

within the 9p24 region, two encode for proteins expressed in the brain (Stewart et al., 2013).

One of these genes encodes for a glutamate transporter, SLC1A1. The following section

reviews relevant scientific publications of this SLC1A1 gene.

1.10 SLC1A1

Solute carrier family 1, member 1 (SLC1A1; also known as EAAT3 or EAAC1) is a

postsynaptic excitatory amino acid transporter that is involved in glutamate clearance from

the extracellular space. In the brain, SLC1A1 is enriched in the cortex, hippocampus,

cerebellum and the striatum (Guillet et al., 2005; Furuta et al., 1997; Maragakis & Rothstein,

2004; Rothstein, 1994), although analyses of rodent brains have shown that eaac1 expression

and distribution vary with CNS development (Furuta, et al., 1997a, 1997b).

1.10.1 SLC1A1 genetic studies in OCD

Arnold and colleagues (2006) conducted a family-based association study using DNA from

157 families with at least one OCD affected child. Two single-nucleotide polymorphisms,

rs301434 and rs301435, were found to be significantly associated in males only (additive

model; p = 0.006 and p = 0.03, respectively). Using a haplotype analysis, two-marker

haplotype rs301434/rs3087879, was also found to be significantly associated with males (p =

0.006).

Page 38: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

25

In another study nine SNPs were tested for association in 71 OCD affected children

(Dickel et al., 2006). Four of the nine polymorphisms had been genotyped by Arnold and

colleagues (rs3780412, rs301430, rs301979, rs301434). Unlike findings from the Arnold et

al. study, the most positive associations were found for markers rs3780412 and rs301430 (p

= 0.04 and p = 0.03, respectively). Additional analysis by sex revealed that marker

rs3780412 association was limited to male probands (p = 0.002). A different two-marker

haplotype rs301430/rs301979 was also found to be nominally associated with this sample (p

= 0.03). This association was limited to males (p = 0.003). In addition to investigating single

locus and haplotype polymorphisms, Dickel and colleagues (2006) also reported a deletion at

the 3’ flanking region of the SLC1A1 gene (chr9:4,594,839-4,594,849, hg19) that segregated

with the OCD trait in a large multigenerational family.

Stewart and colleagues (2007) conducted the next SLC1A1 association study in OCD

using 66 probands (38.6% with comorbid TS); however, the investigation by Stewart et al.

failed to detect significant association for 6 previously OCD associated SNPs (e.g.,

rs3780412, 301430, rs301434, rs301435, rs3087879, rs301979). Only markers rs3780412

and rs2228622 showed a trend towards significance in affected males (p = 0.045) (Stewart el

al., 2007). Furthermore, three-marker haplotype rs12682807/rs2072657/rs301430 spanning

2.7kb, showed a trend with the OCD sample (p = 0.0031). A later study of SLC1A1 reported

association of single marker rs12682807 in OCD-affected males (Stewart et al., 2013).

Many of the variants associated with OCD have no known function. Given that

regulators of a gene may be located outside of the gene, Shugart et al. (2009) tested variants

Page 39: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

26

flanking SLC1A1, in addition to conducting a replication study of previously genotyped

SNPs. In the Shugart study mentioned above, thirteen variants were tested for association

using a sample of 378 affected probands. Only one variant, rs301443 showed significant

association to OCD (p = 0.000067; Bonferroni corrected p = 0.0167); however, this

association was limited to male probands (p = 0.00027). Support of association for this

marker has also been reported by a large meta-analysis, although this finding did not

withstand correction for multiple testing (Stewart et al., 2013). Previous association studies

(Arnold et al., 2006; Dickel et al., 2006) were unable to detect an association with this SNP,

nor was this SNP associated in another study using a large sample consisting of 1576

subjects in 377 OCD affected families (Samuels et al., 2011). Samuel and colleagues were

also unable to identify any common SNPs in LD with rs301443 and were unable to detect an

association of these markers with OCD.

Wendland et al. (2009) used 325 OCD probands to test 4 SNPs that had been

previously genotyped in OCD samples (rs378412, rs301430, rs301434, and rs3087879), in

addition to two tagSNPs that had been yet to be studied in OCD (rs3933331 and rs7858819).

This investigation reported a significant association for a single-marker rs3087879 (p =

0.02); however, after correcting for multiple testing this marker was no longer associated (p

= 0.12). Marker rs3933331 was significantly associated with hoarding, and markers

rs7858819 and rs301430 were nominally significantly associated with SLC1A1 expression

levels in the brain. Wendland and colleagues also found that decreased SLC1A1 expression

was correlated with increased number of T alleles at both of these variants. Haplotype

analyses were also conducted: two haplotypes for rs7858819/rs301430/rs3087879 were

Page 40: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

27

significantly associated with OCD, and remained significant after corrections (p < 0.001 and

p = 0.002). These findings support the hypothesis that a causal variant is located within the 3’

end of SLC1A1.

Despite encouraging results by Wendland and colleagues, screening of coding

regions of SLC1A1 have not identified functional variants that may account for previous

association findings in OCD (Veenstra-VanderWeele, et al., 2001, 2012).

1.10.2 Rationale for investigating SLC1A1 in TS and ADHD

As previously indicated, OCD is genetically correlated with both TS and ADHD (Mathews

& Grados, 2011). Putative OCD risk genes that are also strong functional candidates for TS

and ADHD may be used to guide further investigations. SLC1A1 has been associated with

OCD in multiple studies, and is a functional candidate for TS and ADHD. Despite being a

robust candidate, there are no published studies of the association of this gene with TS and

ADHD.

1.11 Genetic studies in candidate region 5p13

1.11.1 Linkage studies implicating 5p13 in ADHD

The first ADHD genome-wide linkage scan (GWLS) consisted of 104 American families

(Fisher et al., 2002). In the GWLS conducted by Fisher and colleagues, modest evidence of

linkage (LOD>2, p = 0.0009) was found for region 5p12. Two follow up GWLS were

conducted, which used the original ADHD sample and additional families. The first study

was conducted by Ogdie et al. (2003) used a sample of 204 nuclear families with at least two

ADHD affected children. The second study used a sample that was expanded to include 308

Page 41: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

28

families (Ogdie et al., 2004). Both studies found weak evidence implicating region 5p13

(MLS>1, p = 0.002 and MLS= 2.55, p = 0.091; Ogdie, et al., 2003, 2004, respectively).

Using a Dutch sample of 164 sib-pairs with ADHD, the results of Bakker et al. (2003) also

yielded modest but supporting evidence of linkage (mMLS= 1.43) to 5p13 under the broad

phenotype. Using a pooled GWLS of the studies mentioned above, chromosome 5p13 was

the only region to be highlighted (Ogdie et al., 2006). Finally, from a GLWS in a German

sample of 155 sib-pairs, the strongest nonparametric multipoint signal (LOD score = 2.59)

also came from chromosome 5p (Hebebrand et al., 2006). Considering the overlap in reports

for chromosome 5p, and more specifically for region 5p13, this region is of interest regarding

ADHD susceptibility.

1.11.2 Linkage studies implicating 5p13 in TS

The chromosome 5p region has also been implicated in TS. Barr and colleagues (1999)

performed a GWLS using seven large multigenerational families. Although no markers

reached statistical significance levels under the autosomal dominant model, multiple markers

yielded LOD scores above 1. Additional fine mapping of regions with LOD > 1 highlighted

two chromosomal regions in some of these large multigenerational families. One of the

highlighted regions spanned chromosome 5p13-q11.2. Although these findings were

suggestive, it provided a basis for future genetic studies in TS. A follow up study by Laurin,

Wigg, Feng, Sandor and Barr (2009) used an extended sample consisting of eleven additional

family members and fine mapping of the 5p region. The results of this study highlighted the

same region of interest as Barr et al. Further, another dependent study that included the

multigenerational families examined by Barr et al. and Laurin et al. (2009), also observed

Page 42: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

29

suggestive evidence of linkage to the 5p13 region (LOD>2) (The Tourette Syndrome

Association International Consortium for Genetics, 2007).

Additional evidence implicating chromosome 5p in TS pathophysiology comes from a

recent investigation that partitioned heritability by chromosomal length (Davis et al., 2013).

This study revealed that chromosome 5 might account for more TS heritability than expected

when compared to other chromosomes (Davis et al., 2013). However, these findings are

recent and this study has yet to be replicated.

1.12 SLC1A3

SLC1A3 (also known as excitatory amino acid transporter 1, EAAT1 or GLAST) is a

glutamate transporter gene located on chromosome 5p13 (Kanai et al., 2013). This gene

encodes for a high affinity sodium-dependent glial glutamate transporter, SLC1A3, that is

predominately involved in synaptic glutamate clearance (Rothstein et al., 1996). Following

the uptake of synaptic glutamate by SLC1A3, glutamate is converted by glutamine-synthase

into its metabolite, glutamine (Udvardi et al., 2013). Glutamine is then shuttled into the

presynaptic neuron where it is converted back into glutamate and repackaged into vesicles

(Udvardi et al., 2013).

1.12.1 SLC1A3 studies of ADHD

Turic et al. (2005) conducted an association study of SLC1A3 using 299 ADHD-

affected families. One marker, rs2269272 (p = 0.007), and two-marker haplotypes,

rs2269272-rs3776581 (p = 0.016) and rs2269272-rs2032893 (p = 0.013) showed weak

evidence of association. A later study in ADHD conducted by Elia and colleagues (2009)

Page 43: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

30

observed associations for other genetic variants spanning SLC1A3: an intronic SNP

(rs3776571) and two downstream variants rs1529461 and rs6863386. A large meta-analysis

of ADHD genome-wide association studies (Neale et al., 2010) observed association for one

of these markers (rs1529461=0.00475), although this association signal did not reach the

threshold for significant genome-wide association.

In 2008, our lab conducted a replication study of findings from Turic et al. using a

sample of 245 nuclear families with at least one ADHD affected child (total of 280 affected

children)(Laurin et al., 2008). Although we were unable to find a significant association

between the three markers of interest (rs2269272, rs3776581 and rs2032893) it would take at

least 27 tagSNPs to cover SLC1A3. Since these studies, no additional genetic studies have

been published regarding the relationship between SLC1A3 and ADHD.

1.12.2 SLC1A3 studies of TS

Based on the overlap in linkage findings highlighting the 5p13 region and the frequency of

comorbid TS with ADHD, Laurin and colleagues (2009) also tested three associated ADHD

variants (rs2269272, rs3776581 and rs2032893) for association with TS. This study failed to

yield evidence of association. In an effort to identify sequence variants that may be involved

in altering SLC1A3 function, another study conducted by Adamczyk and colleagues (2011)

screened the coding region of 256 TS-affected individuals and 224 unaffected controls.

Despite finding a missense variant that could potentially alter the function of this protein,

allele frequencies were not significantly different between individuals with TS and

Page 44: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

31

unaffected controls (Adamczyk et al., 2011). Other than these two studies, no additional

investigations have been conducted assessing the candidacy of SLC1A3 in TS.

1.12.3 Rationale for investigating SLC1A3 in TS and ADHD

As described earlier, the relationship between TS and ADHD is complex and not well

understood. Familial studies of TS patients have identified a subgroup of individuals affected

with TS and comorbid ADHD and OCD (TS+ADHD+OCD). This phenotype may represent

a distinct entity with unique genetic factors. Alternatively, it may represent a more

homogeneous subgroup of TS with additive and overlapping risk alleles from separate

disorders ADHD and OCD. Linkage studies reveal common susceptibility loci for TS and

ADHD, which may guide the exploration for genes conferring susceptibility to the

TS+ADHD+OCD trait.

Our selection of genes involved in the glutamatergic system was based on multiple

lines of evidence from studies of TS, OCD and ADHD (refer to The Glutamatergic

Hypothesis section). Although previous investigation of SLC1A3 by our lab did not find

association of SLC1A3 with TS (Laurin et al., 2009), only three SNPs were tested in a gene

spanning ~82 kilobases.

There is mounting evidence that dysfunction to this gene influences synaptic glutamate

levels (reviewed in Maragakis & Rothstein, 2004). GLAST-null rodents have demonstrated

alterations in motor coordination (Watase et al., 1998) and locomotive hyperactivity

(Karlsson, Tanaka, Heilig, & Holmes, 2008) suggesting that dysfunction of this gene directly

or indirectly influences motor function. Aberrant glutamate uptake has also been observed in

Page 45: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

32

an accepted rat model of ADHD (Miller et al., 2014). Finally, expression patterns of EAAT1

mRNA coincide with postnatal CNS development events including neuronal differentiation

(Sutherland, Delaney, & Noebels, 1996). From a functional and positional standpoint,

SLC1A3 is a strong candidate for study in TS and ADHD.

1.13 Chromatin Signatures and Gene Expression

Identification of robust candidates for study based on gene function or position is merely the

beginning of uncovering the underlying etiology of a disorder. Following the selection of a

candidate gene, genetic association studies can be used to determine whether specific

changes to DNA sequences are associated with a particular trait. One way to begin

addressing this is by identifying variants located within functional regions of the genome.

Historically, variants located within the protein-coding regions of DNA were investigated for

a causative role in pathogenesis; however screening of protein-coding regions of associated

genes have not always clarify the mechanisms underlying complex genetic disorders

(Veenstra-VanderWeele, et al., 2001; Adamczyk et al., 2011). It is now known that only

1.5% of the human genome accounts for protein-coding sequences, with the majority of the

genome (an estimated 80.4%) participating in at least one regulatory event (The ENCODE

Project Consortium, 2012). The results of multiple GWAS studies provide suggestive

evidence that DNA changes to regulatory elements can result in disease (Hindorff et al.,

2009). Given these findings, the regulation of gene expression, or gene regulation, has been

proposed to play a larger role in pathogenesis than previously hypothesized (Ernst & Kellis,

2010; Hindorff et al., 2009).

Page 46: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

33

Gene expression can be regulated at different levels. At the transcriptional level (in

eukaryotes), transcription is initiated by RNA polymerase binding upstream of the gene at

the promoter region. Transcription factors (TF) bind transcription factor binding sites (TFBS)

and form complex interactions with RNA polymerase to influence the expression of the gene.

TFBS can be located within the gene, upstream or downstream, as well as megabases from

the gene they regulate (Bulger & Groudine, 2011; Heintzman et al., 2007; Nobrega,

Ovcharenko, Afzal, & Rubin, 2003). Alterations to DNA sequences where TFBS are located

can increase (enhance) or decrease (repress) the affinity of the existing TF to its binding site

(Wang, Tomso, Liu, & Bell, 2005). Variation may also abolish the original TFBS or produce

a binding site for another TF (Wang et al., 2005).

In its default state DNA is tightly bound to its associated proteins, forming a complex

referred to as chromatin (Phillips & Hoopes, 2008). A unit of chromatin is a nucleosome,

which consists of 146 bp of DNA around four pairs of histone proteins (H2A, H2B, H3, H4).

When tightly bound to the histone proteins, transcription factors assess to DNA is restricted

and chromatin is considered closed; however, chromatin state is dynamic (Phillips & Hoopes,

2008). Specific chemical modifications to particular histone tails alter chromatin state via

chromatin remodeling (Formosa, 2003; reviewed in Kouzarides, 2007; Strahl & Allis, 2000).

The restructuring of nucleosomes using ATP-dependent complexes is what ultimately

influences TF access to DNA.

Considering that the many of GWAS associations map to sequences of non-coding

regions, variants located within regulatory regions are proposed to be involved in the

Page 47: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

34

pathogenesis of many disorders (Hindorff et al., 2009). As combinations of histone

modifications patterns, or histone codes, are useful to detect cis-regulatory elements (Ernst &

Kellis, 2010; Ernst et al., 2011), multiple investigations, including the encyclopedia of DNA

elements (ENCODE), have attempted to map histone codes across the human genome.

Methylation and acetylation of lysine residues of histone tails are believed to play an

important role in chromatin state. For instance, open chromatin with enriched mono-

methylation of lysine 4 of the H3 histone protein (H3K4Me) is suggestive of enhancer

elements, regions where TF bind to facilitate transcription (Spicuglia & Vanhille, 2012; Ernst

et al., 2011). Another modification, acetylation of lysine 27 of the H3 histone protein

(H3K27Ac) is generally an indicator of active regulatory elements (Spicuglia & Vanhille,

2012; Ernst et al., 2011). In contrast, tri-methylation of lysine 4 of the H3 histone protein

(H3K4Me3) is a modification associated with active and poised promoter elements

(Spicuglia & Vanhille, 2012).

Although both histone methylation and acetylation have been associated with

regulatory regions, the mechanisms facilitating these chemical modifications differ. Histone

acetylation of lysine residues is mediated by histone acetyl-transferase (HAT) activity.

Acetylation of lysine residues facilitates transcription by neutralizing the electrostatic

interaction between positively charged histones and negatively charged DNA backbone

(reviewed in Struhl & Moqtaderi, 1998). This improves access of transcription factors to

binding sites. Binding of enzymes with HAT activity is therefore generally indicative of an

open chromatin state (Spicuglia & Vanhille, 2012). In contrast to histone acetylation, the

Page 48: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

35

effect of histone methylation depends on the modified lysine residue, and can either facilitate

or repress transcription (Barski et al., 2007).

Regulatory variation not only plays an important role in genetic susceptibility, but

also phenotypic presentation and may account for the variability of the TS trait.

Investigations of complex genetic disorders such as TS and ADHD can use predictors of

regulatory elements (e.g., histone marks) to highlight noncoding regulatory regions of

candidate genes. This systematic approach to detecting potential causal variants may improve

efforts to clarify the mechanisms underlying disease and is used for this study.

Page 49: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

36

Table 1–1. D

SM-V

Overview

of TS, O

CD

& A

DH

D

Tourette

OC

D

AD

HD

C

haracteristics m

otor and vocal tics obsessions and/or com

pulsions that interferes w

ith academic/occupational,

social, or functional activities

Inattentive and/or hyperactivity/impulsivity

that is inconsistent with developm

ental level and negatively im

pacts development or

functioning in more than one settings

Not due to drugs or other m

edical condition, and not better explained by other medical conditions

Duration

> 1 year > 1 hr/ day or cause

distress/impairm

ent in functioning > 6 m

onths

Onset

<18 years of age Early onset: ≤18 years Late onset: >18 years

< 12 years

Course

Wax and w

ane Increased severity ~10-

12years reduced severity into

adolescence/adulthood

Wax and w

ane Early onset: reduction of sym

ptoms by

adulthood in 40% of affected

individuals

Without treatm

ent improvem

ent of symptom

s is rare

Hyperactivity m

ay diminish in adulthood, but

inattention and impulsivity typically rem

ain

Estim

ated Prevalence 1%

2.5%

1 5%

Gender B

ias 4 m

ale: 1 female

Early onset > in m

ales 2 m

ales: 1 females

Review

ed in APA

(2013)

1Ruscio et al. 2010

Page 50: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

37

Table 1–2. Sum

mary of L

inkage Studies of TS

Im

plicated Region (hg19)

Sample size

Measure

Score Study

Parametric

3q 171 fam

ilies LO

D

2.55 V

erkerk et al. (2006) 5p13.1-q11.2

12 families

LOD

3.05

Laurin et al. (2009) 1p, 3p

1 family

LOD

> 3

Knight et al. (2010)

11q23 1 fam

ily LO

D

3.24 M

érette et al. (2000) 14q31

1 family

LOD

2.4

Breedveld et al. (2010)

N

onparametric

5p15, 7q36, 11p13, 13q34, 14q13, 16p12, 19p13

7 large multigenerational

families

p-value <.00005

Barr (1999)

4q, 8p 110 A

SP M

LOD

> 2

TSAIC

G (1999)

4q, 5q, 17q 77 A

SP p-value

< .0001 Zhang et al. (2002)

5q34 1 fam

ily M

LOD

2.9

Curtis et al. (2004)

13q13 2.5

17q25 4 fam

ilies N

PL 2.61

Paschou et al. (2004) 2p32

304 ASP + 18 fam

ilies -log P

4.42

TSAIC

G (2007)

2p, 3p, 3q, 4p, 6p, 10p, 15p, 21p 304 A

SP -log P

≥ 2

2p, 5p, 6p 18 fam

ilies -log P

> 2 A

SP: Affected sib-pair

MLO

D: m

ultipoint maxim

um LO

D score

N

PL: nonparametric LO

D score

Page 51: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

38

1.14 Research Aims & Hypotheses

For this study, two genes, SLC1A1 and SLC1A3, were investigated.

Study I: A Family-based Association Study of the putative Obsessive-Compulsive

Disorder gene, SLC1A1, with Tourette syndrome and Attention-

Deficit/Hyperactivity Disorder

Objective: The objective of the SLC1A1 study was to determine whether SLC1A1 is

associated with TS and/or ADHD.

Hypothesis: Given its previous association with OCD, as well as the involvement of this

gene in the glutamatergic system, SLC1A1 is a risk gene for TS and ADHD.

Study II: A Family-based Association Study of a Putative Attention-

Deficit/Hyperactivity Disorder gene, SLC1A3, with Tourette syndrome

Objective: The objective of the SLC1A3 study was to determine whether SLC1A3 is

associated with TS.

Hypothesis: Given previous published associations of this gene in ADHD, and the

overlapping linkage findings of ADHD and TS, SLC1A3 is a risk gene for TS.

Page 52: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

39

Materials and Methods Chapter 2

2.1 The TS family-based sample

Approval for this protocol was obtained from the research ethics committee from the

Hospital of Sick Children Toronto and the University Health Network. Parental consent

and child asset were obtained in written form for children below 18 years of age. Signed

consent was obtained for individuals 18 years of age and older.

2.1.1 Recruitment and exclusion criteria

Participants with TS and their families were recruited from Ontario, Canada—with the

majority of the sample coming from the Greater Toronto Area. To qualify for this study,

probands were required to meet the Diagnostic and Statistical Manual of Mental

Disorders (3rd ed.; DSM-III-R; American Psychiatric Association, 1987) for TS or CMT.

Children could not have an additional diagnosis of autism, bipolar disorder or psychosis.

Children with a diagnosis of intellectual disabilities or pervasive developmental disorder

were also excluded from this study. Children with ADHD, OCD and/or OCB were not

excluded from this study.

2.1.2 Sample Composition

Our sample consisted of 303 nuclear families with at least one TS or CMT affected child

and 74 affected siblings (total of 377 affected children). The gender composition of our

sample consisted of 81% male and 19% female. The ethnic composition was

predominantly of European Caucasian ancestry (92%). The remainder of our sample

described their ancestry as non-European (3%) and 5% mixed European and Non-

Page 53: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

40

European. In total 37% of the affected children also met the criteria for comorbid ADHD,

10% for comorbid OCD and 21% comorbid ADHD and OCD.

2.1.3 Diagnostic Assessment

The initial assessment consisted of self- and family- instruments. Information regarding

symptoms of TS and OCD were obtained based on the Yale Global Tic Severity Scale

(Leckman et al., 1989), the symptom checklist and ordinal scales of the Yale-Brown

Obsessive-Compulsive Scale (Goodman et al., 1989). Information regarding other

psychopathologies was also obtained using the Structured Clinical Interview for DSM-

III-R (Spitzer, Williams, Gibbon, & First, 1992) for adults, or the Kiddie Schedule for

Affective Disorders and Schizophrenia (K-SADS, Chambers, et al., 1985; Kaufman,

Birmaher, Brent, Rao, & Ryan, 1995) for individuals below 18 years of age. To insure

accuracy and validity, the collected information was checked by an experienced

neuropsychiatrist and complemented by the direct examination of each subject using the

same scales. Affection status was determined by a diagnosis of TS or CMT. Some of the

additional information regarding other conditions was collected for future analysis of

comorbid conditions, including OCD and ADHD.

2.2 The ADHD family-based sample

Approval for all protocols for this sample was obtained from the research committee at

the Hospital for Sick Children Toronto. Written informed consent and/or assent were

obtained for all subjects and parental consent for individuals below 18 years of age.

Page 54: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

41

2.2.1 Recruitment and exclusion criteria

Participants (ages 7-16) and their families were recruited from the Child Development

and Neuropsychiatry outpatient clinics at the Hospital for Sick Children Toronto. To

qualify for this study, children were required to meet the DSM-IV criteria for one of three

ADHD-subtypes: predominately inattentive, predominantly hyperactive/impulsive, or

combined. Children with a score below 80 on both Performance and Verbal scales of the

Wechsler Intelligence Scale for Children (Wechsler, 1991) were excluded. The exclusion

criteria also included children with a neurological or chronic medical condition,

psychosis, bipolar affective disorder, TS or CMT.

2.2.2 Sample Composition

Our sample consisted of 641 individuals from 207 nuclear families with at least one child

affected with ADHD. Forty-seven siblings with ADHD were also included in our sample

for a total of 254 affected children. The combined subtype diagnosis was made for 61%

of affected children, 28% of children were diagnosed with the inattentive subtype and the

remaining 11% with the hyperactive/impulsive subtype. In regards to gender, the sample

comprised of 77% males and 23% female. The majority of individuals reported their

ethnicity as European or Caucasian (90%), while 10% described their ancestry as African,

Chinese, Indian, Native Canadian, or of mixed descent.

2.2.3 Diagnostic & Behavioural Assessment

For each child, information regarding behaviour and ADHD symptoms was obtained

using semi-structured interviews from both parents (Parent Interview for Child

Symptoms, PIC-IV, Ickowicz et al., 2006) and teachers (Teacher Telephone Interviews,

TTI-IV, Tannock et al., 2002). The PIC-IV provided information regarding additional

Page 55: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

42

psychiatric conditions, behaviour at home, and other psychosocial stressors. While the

TTI-IV provided information regarding functioning at school and the child’s ability to

interact with others. This information was complemented with standardized instruments

including the Conners Parent and Teacher Rating Scales-R (Conners, 1997) and Ontario

Child Health Survey Scales-R (Boyle et al., 1993), which provided additional information

on ADHD symptoms, cognitive deficits and childhood experiences. These assessments

were taken following a medication-free period for at least 24 hours.

2.3 Isolation & Extraction of DNA

The DNA used for this study was isolated from white blood cells using a high-salt

extraction technique (Miller, Dykes, & Polesky, 1988).

2.4 Single-Nucleotide Polymorphism Selection

2.4.1 SLC1A1

SNP selection for the SLC1A1 study was based on two approaches. The first

approach involved selecting six SLC1A1 markers with previous association to OCD. The

second approach used data from the ENCODE project using the University of California,

Santa Cruz genome browser (http://genome.ucsc.edu/). H3K4Me1 and H3K27Ac marks

were used to indicate putative enhancer regions. Eleven SNPs located within putative

enhancer regions were selected and genotyped for study. Only markers with minor allele

frequencies greater than 5% were selected.

2.4.2 SLC1A3

For the SLC1A3 study, a total of ten SNPs were selected using three approaches.

The first approach involved retesting three markers (rs3776581, rs2269272, rs2032893)

Page 56: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

43

previously examined by our lab (Laurin et al. 2009). Since the Laurin et al. (2009)

publications, our TS sample has expanded from 241 TS or CMT affected children to 377

TS or CMT affected children, giving the current study more power. The second approach

involved selecting the four strongest SLC1A3 tagSNP association signals from ADHD

GWAS (Neale, et al., 2010). Finally, the last approach involved the ENCODE project

using the University of California, Santa Cruz genome browser (http://genome.ucsc.edu/).

The latter two tools allowed us to identify enrichment of the H3K4Me1 and H3K27Ac

histone marks, indicators of putative enhancer regions. Enrichment of the H3K4Me3

histone mark was also used to identify the promoter region. Using this approach, three

markers located within these regions were selected and genotyped for study. Only

markers with minor allele frequencies greater than 5% were selected.

2.5 Single-Nucleotide Polymorphism Genotyping

For the global investigation, a total of 27 SNPs were genotyped: 17 SNPs in the SLC1A1

study (refer to Table 3-1 for SLC1A1 SNP list) and 10 SNPs in the SLC1A3 study (refer

to Table 4-1 for SLC1A3 SNP list). All genotyping was conducted using ABI 7900-HT

Sequence Detection System (Applied Biosystems, Foster City, CA) probes, which were

either available by assay-by-demand or requested upon submission of the DNA sequence

flanking the SNP-of-interest, by assay-by-design. TaqMan® 5’nuclease assay was used

for allelic discrimination for all assays.

Each assay consisted of sequence-specific primers that amplified the desired SNP

sequence. In addition, assays also contained two fluorescent-labeled probes used for the

detection of alleles present for the SNP-of-interest. The 3’ end of each probe consisted of

Page 57: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

44

a quencher dye and 5’ fluorescent tag. Each probe is labeled with a different fluorescent

dye: VIC® dye was used to detect allele 1 and FAM® dye for allele 2.

Detection of fluorescence only occurred upon separation of the quencher from the

fluorescent dye. During the Polymerase Chain Reaction (PCR), which is responsible for

amplification of DNA, each probe hybridizes with the complementary DNA sequence.

AmpliTaq Gold® polymerase with nuclease activity separates the probe fluorescents

from the quencher. The fluorescence emitted depends on the allele present at the SNP of

interest. Data was collected for each sample using the ABI 7900HT Sequence Detection

System (SDS) with allelic discrimination mode of the SDS software package, v2.0

(Applied Biosystems, Foster City, CA).

The PCR reaction used for this study consisted of 30 ng of genomic DNA, 2.5 µl

of TaqMan® PCR Mix (Applied Biosystems) and 0.05 µl of allelic discrimination mix

(Applied Biosystems) with 36 µM and 8 µM of each primer and probe, respectively. The

total reaction volume was 5 µl. Ninety-six well optical reaction plates were used for

genotyping with two negative controls.

The PCR thermal cycling stages were as follows: i) 50°C for 2 minutes, ii) 95°C

for 10 minutes, iii) 40 cycles of: 94°C for 15 seconds of denaturing, and 59°C for 1

minute for annealing and extending.

2.6 Statistical Analysis

2.6.1 Association Analyses

Genetic association studies are used to determine whether a genetic marker is

associated with a particular disease or trait within a population. One approach to

Page 58: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

45

determine association is to evaluate the differences of allele frequencies between affected

(cases) and unaffected individuals (controls) (reviewed in Lewis & Knight, 2012).

Employing this method, however, can result in an increase in type I error due to

population stratification, in which the allele frequencies between different ethnic groups

differ. Although the selection of controls from the same ethnic population as cases often

reduces type I error in case-control studies, spurious associations due to population

stratification remains a concern.

Another approach to identify genetic associations is to use a family-based design.

The Transmission Disequilibrium Test (TDT) is a modified χ2 test that controls for

population stratification by evaluating the distribution of allele transmission from

heterozygous parents to affected offspring (Sham & Curtis, 1995). Under the null

hypothesis of independent assortment, transmission of each allele from a heterozygous

parent to offspring should be 50%. Significant deviation in transmission of an allele

suggests that this particular allele is associated with the disorder or trait of interest. Using

an extended TDT for multi-allelic markers, each SNP of this investigation was tested for

single-marker association with TS or ADHD in the respective samples.

Alleles that are on the same chromosome that are in proximity to one another are

less likely to undergo recombination, and therefore more likely to be inherited as a set

(reviewed in Clayton, 1999). This set of alleles is referred to as a haplotype. The

TRANSMIT program uses a modified TDT to analyze extended marker haplotypes

(Clayton & Jones, 1999). For this study, the TRANSMIT program v2.5.4 (Clayton &

Jones, 1999) was used to identify any haplotype patterns that were significantly

transmitted to affected offspring. Transmission to affected offspring that is greater than

Page 59: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

46

expect by chance suggested an association of specific haplotypes with TS or ADHD. To

detect a haplotype-association when frequencies were low, haplotypes with frequencies

less than 10% were pooled and only results with frequencies greater than 10% are shown.

2.6.2 Visualization and Interpretation of LD blocks

Neighbouring alleles that are transmitted together reflect haplotypes. Sites were

recombination frequently occurs over generations reflect divisions of haplotypes into

distinct LD blocks (reviewed in Daly, Rioux, Schaffner, Hudson, & Lander, 2001).

Visualization of LD blocks allows for the summarization of marker relationships and

makes it easier to evaluate which alleles belong to the same haplotype.

For this investigation, Haploview v4.2 software was employed to i) derive LD

pair-wise measures and ii) identify LD blocks. Genotype data loaded into the Haploview

software either came from our family-based samples or from the Human Haplotype Map

(HapMap) project, an international collaboration involved in identifying LD patterns and

marker allele frequencies of the human genome (International HapMap Consortium,

2003; www.hapmap.org).

LD blocks were defined using the Solid Spine method. This method partitions

blocks, such that the first and last markers in a block are in strong LD, although strong

LD between intermediate markers within a block is not a requirement.

r2 colour scheme was used to represent the LD relationships between SNPs. The

following characterizes the colour scheme and the LD relationship it highlights: i) r2 = 0

was represented by white, ii) 0 < r2 < 1 by shades of gray, and iii) r2 = 1 by black (Barrett,

Fry, Maller, & Daly, 2005).

Page 60: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

47

2.6.3 Quality Control

The Merlin software v1.1.2 (Abecasis, Cherny, Cookson, & Cardon, 2002) was used to

highlight unlikely genotypes given gene flow patterns and recombination (Abecasis et al.,

2002). The Haploview program (v4.2, Barrett et al., 2005) was also used to check for

Mendelian errors. DNA assays that were highlighted by these programs for genotyping

errors were retyped. DNA samples that often produced inconsistent results were removed

from the study.

To ensure that our genotype frequencies did not significantly deviate from the

expected frequencies, conformance with Hardy-Weinberg equilibrium (HWE) was

checked using the Haploview program (v4.2, Barrett et al., 2005) for each SNP. Common

sources contributing to violations of HWE include: i) nonspecific primer/probe, ii)

genotyping errors iii) inappropriate genotyping calls (Hosking, et al., 2004). Therefore,

deviations were addressed first by ensuring that the primer/probe were specific to the

SNP of interest. This was done by evaluating whether another common SNP was located

within the primer/probe sequences. Nonspecific sequences detect neighbouring alleles,

which can result in deviations to the genotype frequencies. Another possible explanation

for deviations from HWE is due to genotyping errors (Hosking et al., 2004). To address

this, undetermined genotypes and unlikely genotypes were retyped. Finally, we referred

back to the SDS program to ensure that no inappropriate calls were made. Failure to

conform to HWE warranted the removal of that marker from the study.

2.6.4 Power

The primary objective of a genetic association study is to detect associations between

genetic factors and a trait of interest. The likelihood of detecting this association,

Page 61: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

48

assuming that a relationship exists between the variables, can be determined by

conducting a power analysis.

A power analysis is a statistical test of the null hypothesis. Under the null

hypothesis there is no association between the selected marker and phenotype of interest.

Conversely, the alternative hypothesis states there is a relationship between the genetic

factor and phenotype. Rejecting the null hypothesis when it is correct is committing type

I error (α) and is reflected as false positives. Another type of error is failing to reject the

null hypothesis when it is incorrect—this is referred to as type II error (β). Power (1-β) is

the probability of avoiding type II error.

Three factors influence the power of a study: i) sample size (N) ii) threshold of

significance (α), and iii) effect size. Increasing the number of samples in a study is one

method of strengthening the power of a study and reducing the likelihood of committing

type II error. Adjusting the threshold of significance to make it more conservative may

also improve the likelihood that the detected associations reflect true positive findings.

Finally, effect size can also influence the strength of a study.

A measure that describes the effect size of a variable is the genotype relative risk

(GRR). GRR implies that the genotype of an individual is associated with risk of a

particular phenotype. For instance, if the GRR is less than 1, the genotype is considered

protective, as the individual is at a decreased risk affected with the phenotype of interest.

GRR equal to 1 suggests that there is no additional risk, and a GRR greater than 1

suggests that individuals with that particular genotype are at an increased risk. Risk

Page 62: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

49

alleles with greater effect sizes are more likely to be detected by association studies

(Evans & Purcell, 2012).

Conducting a power analysis allows investigators to determine the smallest effect

at a given sample size as well as whether a study is underpowered. For this study, we

employed the Genetic Power Calculator (Purcell et al., 2003) was used to perform the

discrete trait TDT power analysis. Our TS sample consisted of 303 nuclear families with

74 siblings also met the diagnostic criteria for TS or CMT. As a result the power analysis

was calculated for a sample size of 377 parent-child trios. Based on epidemiology studies

of TS and CMT (Robertson, 2008; Scharf, Miller, Mathews, & Ben-Shlomo, 2012), the

disorder prevalence in the general population was specified as 1%.

For the SLC1A1 investigation using our TS/CMT sample, the threshold of

significance and adequate power were defined as 0.003 and 80%, respectively. Using

allele frequencies of 0.10-0.50 our study had adequate power to detect an estimated effect

size from 2.00-2.75 for a marker with a D’ of 1.0.

The power analysis for the SLC1A1 study using the ADHD sample was also

conducted using the Genetic Power Calculator (Purcell, 2003). Our ADHD sample

consisted of 207 nuclear families with 47 ADHD-affected siblings, for a total of 254

parent-child trios. The disorder prevalence used for this analysis was 5% (Polanczyk et al.,

2007) and the threshold of significance was set at 0.013. Using allele frequencies of 0.30-

0.50, our ADHD sample had adequate power (80%) to detect an estimated effect size

from 2.07-3.00 for a marker with a D’ of 1.0.

Page 63: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

50

For the SLC1A3 study adequate power and the threshold of significance were set

to 80% and 0.006, respectively. Using our TS/CMT sample we would have had enough

power to detect an estimated effect size of 1.87-2.10 for allele frequencies of 0.20-0.40.

2.6.5 Correction for Multiple Testing

For this study, the Bonferroni correction was used to adjust the p values to account for

multiple testing. Due to the correlation between some of the SNPs, correcting for all

genotyped makers would have been overly conservative. To address this, the SNPSpD

tool (Nyholt, 2004) was used calculate the number of independent SNPs. SNPSpD uses

principal components analysis (PCA) (also referred to as eigenvalue analysis) to account

for the variance among a set of variables. The first eigenvalue accounts for the greatest

amount of variance between variables; the second eigenvalue, which is uncorrelated to

the first, accounts for the remaining variance (Ringnér, 2008). Each eigenvalue that

follows accounts for the next largest variance that was unaccounted for by prior principal

components (Ringnér, 2008). This method allows for the reduction of highly dimensional

data, so that it can be described using fewer variables (Price et al., 2006).

Eigenvalues may be used to measure the pairwise LD correlation between

markers from a correlation matrix (Cheverud, 2001). This correlation matrix is derived

using the input pedigree and map file (Nyholt, 2004). When the correlation between

markers is high, the first eigenvalue is also high as it accounts for most of the variance.

Further, when all of the SNPs are completely correlated the greatest possible variance of

the eigenvalues is equal to the number of SNPs in the LD matrix (Nyholt, 2004;

Cheverud, 2001). When there is no correlation among a set of SNPs in the matrix, all of

the eigenvalues equal 1 (Nyholt, 2004; Cheverud, 2001). In this case, the SNPs are

Page 64: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

51

independent and the collective variance of eigenvalues is equal to zero (Nyholt, 2004;

Cheverud, 2001). This collective variance is employed to calculate number of

independent SNPs, which was then used to adjust the threshold for statistical significance.

The p values were corrected for multiple testing using the number of independent

SNPs as calculated by SNPSpD (Nyholt, 2004). For the SLC1A1 and SLC1A3 studies

the adjusted threshold of significance was 0.003 (p =0.05/15.029= 0.003) and 0.006 (p =

0.05/8.361= 0.006), respectively.

Page 65: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

52

A family-based association study of the putative Chapter 3

Obsessive-Compulsive Disorder gene, SLC1A1, with

Tourette Syndrome and Attention-Deficit/ Hyperactivity

Disorder

3.1 Introduction

Gilles de la Tourette Syndrome (TS) is a childhood-onset neurobehavioral

disorder characterized by the presence of both motor and phonic tics for at least a year

(American Psychiatric Association, 2013). Family and twin studies provide evidence that

genetics play a substantial role in the pathogenesis of this disorder (Kidd et al., 1980;

Hyde et al., 1992; Pauls et al., 1981; Price et al., 1985; Walkup et al., 1988, 1996);

however, identification of risk genes with major effect remains elusive. Due to the

phenotypic variability of TS, it has been proposed that TS may be genetically

heterogeneous, with different phenotypes having unique and overlapping genetic risk

factors (Hyde et al., 1992; Mathews et al., 2006; Price et al., 1985). Under this

assumption, consideration of clinical phenotypes of TS may clarify genetic signals to date.

Chronic multiple tics (CMT) is a psychiatric disorder characterized by the

presence of either motor or vocal tics for at least a year (APA, 2013). This disorder is

commonly found in relatives of TS patients (Kidd et al., 1980; Pauls et al., 1981, 1984).

Twin studies of TS show that the concordance rate for monozygotic twins increases from

53% to 77%, with the inclusion of CMT subjects, adding to the support that chronic

multiple tics (CMT) represents a less severe manifestation of TS (Price et al., 1985).

Page 66: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

53

Given that approximately 90% of TS-affected individuals have at least one

comorbid disorder (Robertson, 2012) phenotypic variability of TS may not be limited to

CMT, such that other disorders may also share risk genes with TS. Two disorders that are

commonly comorbid with TS are attention-deficit/hyperactivity disorder (ADHD) and

obsessive-compulsive disorder (OCD). ADHD is characterized by inappropriate

(inattentive, hyperactive/ impulsive, or a combination) behavior for a particular

developmental age (APA, 2013). This disorder has an estimated prevalence of 5%

(Polanczyk et al., 2007) and negatively impacts the individual’s quality of social and

academic/occupational functioning (APA, 2013). OCD affects an estimated 2.5% of

individuals (Ruscio et al., 2010) and is characterized by obsessions and/or repetitive

compulsions that result in significant distress or impairs functioning (APA, 2013). Both

ADHD and OCD are heritable (Biederman & Faraone, 2005; Hasler et al., 2007) and are

influenced by environmental factors and multiple genes. Together and individually these

disorders affect over 50% of the TS population (Freeman & Tourette Syndrome

International Database Consortium, 2007). On the basis that the prevalence of these

disorders is substantially higher in TS patients and their relatives than in the general

population, it is hypothesized that at least some subtypes of ADHD and OCD share

common risk factors with TS (Comings 1987, 2001; Pauls, et al., 1986, 1991, 1994;

Sheppard, Bradshaw, Purcell, & Pantelis, 1999)

In addition to high prevalence rates, multiple lines of evidence support an

etiological overlap between TS and OCD (Walkup et al., 1988; Mathews & Grados,

2011). Specifically, tic-related OCD, which describes cases of OCD were tics are also

present, has been observed in an estimated 40% of early-onset OCD patients (Grados et

Page 67: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

54

al., 2001). Moreover, certain obsessive-compulsive symptom involving symmetry, order,

counting and touching are more likely to be found in tic-related OCD patients than those

without tics, suggesting that these OC symptoms may be etiologically related to the tic

disorder spectrum (Leckman et al., 1994-1995). Clinical studies provide further support

of this hypothesis by demonstrating that neuroleptics, a class of medication used to treat

tics, have greater therapeutic effect in treating tic-related OCD than selective-serotonin

reuptake inhibitors alone (reviewed in Singer et al., 2010; Pittenger, Krystal, & Coric,

2006). From a neuroanatomical perspective, many of the same brain regions (e.g.,

striatum) have been implicated in both disorders (Harrison et al., 2013; reviewed in Albin

& Mink, 2006). And from a heritability standpoint, studies have a significant genetic

correlation of around 41% between TS and OCD (Davis et al., 2013).

Due to the clinical heterogeneity among TS patients, it has been proposed that TS

may be separated into homogeneous subgroups, and that these subgroups may have

shared and unique susceptibility factors (Alsobrook & Pauls, 2002; Robertson & Cavanna,

2007; Grados et al., 2008). While comorbid TS and OCD has been identified as a

potential TS subgroup, another subgroup consisting of comorbid TS, OCD and ADHD

has also been identified (Grados et al., 2008). Considering that comorbid TS and ADHD

was not identified as a subgroup of TS from the Grados et al. (2008) study, it was

proposed that OCD may mediate the occurrence of comorbid TS and ADHD. In support

of this hypothesis, subsequent studies of these disorders provide support that OCD may

be mediating this heritable subtype. For instance, relatives of OCD/OCB probands have

been found to be at an increased risk for comorbid TS and ADHD (O'Rourke et al., 2011).

In addition, significant genetic correlations have been observed between TS and OCD

Page 68: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

55

and for OCD and ADHD, but not for TS and ADHD (Mathews & Grados, 2011). The

results of these studies in the least suggest that TS, OCD and ADHD share some common

etiological factors.

One approach to identify genes conferring susceptibility to TS, and some forms of

ADHD and OCD is to investigate genes with biological relevance to all three disorders.

While dopaminergic and serotonergic genes have been the predominant focus in

candidate gene literature across these disorders, and likely contribute to pathogenesis,

additional evidence also suggests a role of the glutamatergic system. For instance,

transgenic mice with altered dopamine receptors of corticostriatal glutamate projections

exhibit tic and OC symptoms (McGrath, 2002; Nordstrom, 2002). In another study,

altered glutamate receptor activation was observed in spontaneous hypertensive rats

(Lehohla, Kellaway, & Russell, 2004), an accepted rodent model of ADHD (reviewed in

Sagvolden & Johansen, 2012). Elevated striatal glx levels have been observed in ADHD

patients through 1H-MRS, when compared to controls (Carrey N. J., MacMaster, Gaudet,

& Schmidt, 2007), and drugs interacting with the glutamatergic system are currently

being studied for the reduction of tic and obsessive-compulsive symptoms (Singer, 2010).

The role of the glutamatergic system in the pathogenesis of these disorders has been

suggested by neuroimaging studies of OCD (reviewed in MacMaster, 2008) and ADHD

(Acros-Burgos et al., 2012; Courvoisie, Hooper, Fine, Kwock, & Castillo, 2004;

Dramsdahl et al., 2011; Ferreira et al., 2009; MacMaster, Carrey, Sparkes, & Kusumakar,

2003; Perlov et al., 2010), as well as analysis of postmortem brain tissue from individuals

with TS (Anderson, 1992). Finally, genetic association studies implicate genes involved

in the glutamatergic system as possible contributors of TS (Crane, et al., 2011), OCD

Page 69: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

56

(Arnold et al., 2004, 2006; Dickel et al., 2006; Stewart et al., 2007; Wendland et al.,

2009) and ADHD (Dorval et al., 2007; Turic et al., 2005; Laurin et al., 2008).

Despite multiple genome-wide linkage scans (e.g., The Tourette Syndrome

Association International Consortium for Genetics, 1999, 2007; Barr et al., 1999;

Breedveld, Fabbrini, Oostra, Berardelli, & Bonifati, 2010; Fisher et al., 2002; Ogdie et al.,

2003, 2004; Hebebrand et al., 2006; Hanna et al., 2002; Shugart et al., 2006; Samuels et

al., 2007) no susceptibility regions have been linked to all three disorders. Recent

genome-wide association studies (GWAS) in TS (Scharf et al., 2012), OCD (Stewart et

al., 2013) and ADHD (Neale et al., 2008, 2010; reviewed in Poelmans et al., 2011), have

also been conducted to identify common genetic variants; however, due to a lack of

power, few variants have met the threshold of genome-wide significance (p = 5 x 10-8).

Nevertheless, association signals from GWAS and targeted association studies may be

used to identify risk genes.

Multiple candidate gene association studies of OCD have reported association

with solute carrier family 1, member 1 (SLC1A1), a gene that encodes for the excitatory

amino acid transporter (EAAT3; Arnold et al., 2006; Dickel et al., 2006; Stewart et al.,

2007; Wendland et al., 2009; Shugart et al., 2009). While a meta-analysis of SLC1A1 did

not provide strong evidence for association of this gene with OCD (Stewart et al., 2013),

this protein is involved in the removal of glutamate from the synaptic clef and is relevant

to the regulation of glutamate in the brain (Tzingounis & Wadiche, 2007). Given the

evidence implicating SLC1A1 in OCD, as well as its biological relevance to TS and

ADHD, it is surprising that there are no published candidate gene studies investigating

the contribution of SLC1A1 to TS and ADHD.

Page 70: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

57

Despite multiple association reports with OCD, SLC1A1 associated variants have

differed between studies and no non-synonymous DNA changes in the coding region

have been found that could explain the potential mechanisms underpinning risk

(Veenstra-VanderWeele et al., 2001, 2012; Wang et al., 2009). Recent evidence suggests

that genetic variation influencing the regulation of gene expression may also contribute to

the susceptibility of complex genetic disorders (Siniscalco, Cirillo, Bradstreet, &

Antonucci, 2013); therefore, we also sought to identify functional DNA changes that may

account for any possible associations.

In this study, we first sought to determine whether SLC1A1 contributes to TS by:

(1) testing associated OCD single-nucleotide polymorphisms (SNPs) and (2) testing

SNPs within putative enhancer regions using our TS sample which consisted of DNA

from 303 nuclear families and 74 affected siblings. We then took our four most positive

markers for study using our ADHD sample, which consisted of DNA from 207 nuclear

families and 47 affected siblings. In total 17 SNPs were genotyped and tested for

association using our TS sample and 4 were tested for association in our ADHD sample.

Page 71: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

58

3.2 SLC1A1 Results

Single-marker analysis

Six polymorphisms that produced association, or trends for association using

OCD samples (rs3933331, rs7858819, rs301430, rs301434, rs301435, rs3087879) were

initially genotyped in our TS sample. Transmission Disequilibrium Test (TDT) analysis

was used to identify biased transmission of alleles from heterozygous parents to affected

children. We observed biased transmission of the rs301430 C allele to affected TS

children (p = 0.014). Although this finding would not withstand correction for multiple

testing biased transmission of this allele has also been reported to individuals with early-

onset OCD (Dickel et al., 2006). Interestingly, the effect of the rs301430 C allele has

been shown increased reporter gene expression when compared to the T allele (Wendland

et al., 2009).

Given the number of previous associations between this gene and OCD, as well as

our trend in TS, we sought to identify functional markers to explain the trends and

improve power. None of the common variants located within this gene are non-

synonymous amino acid changes, therefore we focused on variants located within

putative regulatory elements. Using H3K4Me1 and H3K27Ac marks, which are

indicative of enhancer regions, we selected eleven SNPs with minor allele frequencies

greater than 5%, and tested them for association with our TS sample (see Figure 3-2 for

relative position of genotyped SLC1A1 markers). Subsequent single-marker analysis of

these polymorphisms provided no evidence of association with our TS sample (Table 3-

1).

Page 72: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

59

Four of our most positive markers in TS (based on the single- and haplotype-

analysis results) were also genotyped and tested for association in our ADHD sample. We

observed biased transmission of rs301435 T allele to affected ADHD children (p = 0.006).

This association withstands the correction for multiple testing for the markers genotyped

in the ADHD sample in this study alone.

Haplotype Analysis

The TRANSMIT program (Clayton & Jones, 1999) was used to detect haplotype

association of LD blocks. Using the Solid Spine of LD, in which strongest LD is found

between the first and last markers of a block, 3 blocks were constructed using the TS

sample. While no evidence of association for the first two blocks was found (Blocks 1 &

2 results not shown), we did observe a trend for a three-marker haplotype

(rs301434/rs301435/rs3087879) in block 3 (global χ2 = 20.761, 7 df, global uncorrected p

= 0.004). The TRANSMIT program was also used to detect haplotype association of LD

blocks identified in the ADHD sample. Only one block was constructed using this sample,

which consisted of the same three-marker haplotype (rs301434/rs301435/rs3087879)

found using our TS sample. Despite observing a trend in TS, this haplotype did not reach

the threshold of significance in ADHD (Table 3-4 for results).

Page 73: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

60

3.3 Tables & Figures

Table 3–1. SLC1A1 Single SNP analysis for TS sample

Marker Position MAF

(Allele)

HW p-value Transmission Ratio

(Allele)

χ2 p-value

rs3933331 4389941 0.186 (C) 0.653 87:69 (G:C) 2.077 0.150

rs10491731 4542803 0.140 (C) 0.862 54:70 (A:C) 2.065 0.151

rs4641119 4544907 0.290 (C) 0.264 92:101 (A:C) 0.420 0.517

rs7858819 4559892 0.230 (T) 0.983 93:108 (C:T) 1.119 0.290

rs7858877 4560569 0.465 (C) 0.925 123:126 (T:C) 0.036 0.849

rs3780413 4567353 0.314 (C) 0.460 110:98 (G:C) 0.692 0.405

rs4740790 4568342 0.310 (A) 0.313 106:90 (G:A) 1.306 0.253

rs7871243 4573218 0.461 (A) 0.732 123:125 (G:A) 0.016 0.899

rs12682807 4574022 0.089 (C) 0.014 49:54 (A:C) 0.243 0.622

rs10117931 4575120 0.464 (A) 0.882 129:134 (G:A) 0.095 0.758

rs301430 4576680 0.280 (C) 0.909 90:126 (T:C) 6.000 0.014

rs301434 4582082 0.473 (T) 0.896 138:125 (C:T) 0.643 0.423

rs301435 4582843 0.468 (C) 0.734 144:139 (T:C) 0.088 0.766

rs3087879 4586808 0.343 (C) 0.933 136:113 (G:C) 2.124 0.145

rs301443 4594919 0.293 (G) 0.091 114:114 (C:G) 0.000 1.000

rs10974636 4596213 0.285 (T) 0.448 99:103 (G:T) 0.079 0.778

rs34345944 4596315 0.173 (G) 0.192 66:81 (A:G) 1.531 0.216

Page 74: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

61

Table 3–2. SLC1A1 Haplotype analysis of Block 3 for TS sample

rs301434 rs301435 rs3087879 Frequency Observed Expected Var

(O-E)

χ2 (1df) p-value

1 (C) 2 (T) 2 (G) 0.522 381.230 370.700 80.254 1.382 0.240

2 (T) 1 (C) 1 (C) 0.332 226.260 237.480 68.927 1.828 0.176

2 (T) 1 (C) 2 (G) 0.124 98.398 90.671 35.205 1.696 0.193

Global χ2 test (7 df) with frequencies > 10% = 20.761, uncorrected p = 0.004

Page 75: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

62

Table 3–3. SLC1A1 Single SNP analysis for ADHD sample

Marker Position MAF

(Allele)

HW p-value Transmission Ratio

(Allele)

χ2 p-value

rs301430 4576680 0.302 (C) 0.380 70:67 (T:C) 0.066 0.798

rs301434 4582082 0.473 (T) 0.248 107:83 (T:C) 3.032 0.082

rs301435 4582843 0.476 (C) 0.096 115:77 (C:T) 7.521 0.006

rs3087879 4586808 0.333 (C) 0.659 100:79 (C:G) 2.464 0.117

Table 3–4. SLC1A1 Haplotype analysis for ADHD sample

rs301434 rs301435 rs3087879 Frequency Observed Expected Var

(O-E)

χ2

(1df)

p-value

1 (C) 2 (T) 2 (G) 0.505 306.930 314.200 61.278 0.862 0.353

2 (T) 1 (C) 1 (C) 0.308 195.470 191.870 56.830 0.229 0.633

2 (T) 1 (C) 2 (G) 0.146 99.960 91.091 32.101 2.450 0.118

Global χ2 test (7 df) = 13.165 with frequencies > 10%, p = 0.068

Page 76: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

63

Figure 3-1. LD plot of 17 genotyped SLC1A1 SNPs. The numbers within each block represents r2 and are indicative of LD between SNPs in the TS sample. LD blocks defined using the solid spine of LD are outlined in bold.

Figure 3-2. Relative positions of 17 genotyped SLC1A1 markers

Page 77: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

64

A family-based association study of a putative Chapter 4

Attention-Deficit/Hyperactivity Disorder gene, SLC1A3,

with Tourette syndrome

4.1 Introduction

Gilles de la Tourette syndrome (TS) is a childhood-onset neurobehavioral

disorder characterized by the presence of both motor and phonic tics for at least a year

(American Psychiatry Association, 2013). Family studies of TS demonstrate that this

disorder is heritable, with both environmental and genetic factors playing a role in its

etiology (Kidd et al., 1980; Pauls et al., 1981; Walkup et al., 1988, 1996; Price et al.,

1985; Hyde et al., 1992). Despite this evidence, multiple association studies and the only

published genome-wide association study (GWAS) of TS have been unable to identify

many genes contributing to susceptibility (Scharf et al., 2012). One explanation for the

difficulty identifying risk genes is due to the heterogeneity of the TS phenotype. For

instance, multiple lines of evidence suggests that chronic multiple tics (CMT) is a less

severe phenotype of TS (Pauls et al., 1986, 1990, 1991; Walkup et al., 1996; Grados et al.,

2008). The presence of additional psychiatric disorders in the majority of TS probands,

including combinations of comorbid attention-deficit/ hyperactivity disorder (ADHD) or

obsessive-compulsive disorder (OCD), and subclinical OCD (OCB) suggest that TS is

heterogeneous and of complex etiology. Consideration of homogeneous TS subgroups by

genetic studies may improve efforts in identifying risk genes by clarifying whether these

manifestations represent TS subgroups with shared and additional risk factors.

Page 78: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

65

ADHD is a psychiatric disorder characterized by inappropriate inattentive,

hyperactive/impulsive behaviour for a particular developmental age (American

Psychiatric Association, 2013). This disorder interferes with functioning or development

(APA, 2013) and affects approximately 5% of children worldwide (Biederman & Faraone,

2005). Clinical and epidemiological studies reveal that although TS affects approximately

1% of children worldwide (Robertson, 2012), an estimated 50% of these children also

meet the diagnostic criteria for ADHD (Freeman et al,. 2000, 2007).

Despite the high prevalence of ADHD with TS (TS+ADHD), the relationship

between these disorders is unclear (Comings & Comings, 1984; Pauls et al., 1986, 1991,

1993; Knell et al., 1993; Stewart et al., 2006; Grados et al., 2008; O’Rourke et al., 2011;

Mathews & Grados, 2011). For instance, Knell et al. (1993) observed a significantly

greater rate of ADHD among relatives of TS only and TS+ADHD probands, when

compared to relatives of unaffected individuals; however, the rates of ADHD between TS

only and TS+ADHD probands also significantly differed (Knell et al., 1993). In another

study the rate of TSonly was not significantly elevated amongst relatives of ADHDonly

probands, while the rate of TS+ADHD in relatives was significantly greater across all

probands (TS-only, ADHD-only, TS+ADHD) (Stewart et al., 2006). A plausible

explanation for these findings is that ADHD that is comorbid with TS may be a subtype

of TS, with unique and shared risk factors as ADHD without TS (Knell et al., 1993;

Rizzo et al., 2007; O’Rourke et al., 2011).

Multiple reports of linkage to several chromosomal regions support a genetic

overlap between these disorders, including: 4q, 5p, 8p, 10q, 11q, and 17p (Arcos-Burgos

et al., 2004; Barr et al. 1999; Curtis et al., 2004; Fisher et al., 2002; Hebebrand et al.,

Page 79: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

66

2006; Laurin et al., 2008; Ogdie et al., 2006; Simonic, Gericke, Ott, & Weber, 1998;

TSAICG, 1999, 2007). Amongst the overlapping susceptibility loci, chromosomal region

5p12-13 has been consistently linked to ADHD through several genome-wide linkage

scans (Fisher et al., 2002; Ogdie, et al., 2003, 2004, 2006; Bakker et al., 2003; Hebebrand

et al., 2006) and suggestive evidence from two genome-wide linkage scans have also

implicated part of this region in TS (Barr et al., 1999; TSAICG, 2007). Although none of

the genetic variants mapping to 5p13 reached the threshold of genome-wide significance

in the recent GWAS of TS (Scharf et al., 2012) a more recent investigation of the

heritability of TS by chromosome demonstrated greater heritability loading for

chromosome 5 (Davis et al., 2013). Based on these findings, genes mapping to 5p13 with

biological relevance to both disorders are good candidates for study in the search for

shared risk genes between TS and ADHD.

Altered glutamate neurotransmission has been implicated in the pathogenesis of

TS and ADHD from multiple lines of study. For instance, proton magnetic resonance

studies have observed altered glx (glutamate and glutamine) levels in the frontal-striatal

regions of ADHD patients (Dramsdahl, 2011; MacMaster et al., 2003; Perlov, 2007,

2009) and altered glutamate levels have been reported using postmortem analysis of

brains of TS affected patients (Anderson et al., 1992). Proton magnetic resonance

spectroscopy studies have reported abnormal glx levels in the basal ganglia of ADHD

patients (Carrey et al., 2007; Maltezos et al., 2014) and new treatment options that

modulate glutamate release are being studied for a role in tic management (Singer, 2010).

Finally, disrupted glutamate transmission has also been observed in spontaneous

hypertensive rats (Warton, Howells, & Russell, 2009), an accepted model of ADHD

Page 80: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

67

(Lehoha et al., 2004) as well as tic-like movements in rodent models of TS (McGrath et

al., 2000; Nordstrom, 2002). On the basis of this evidence, dysfunction to the

glutamatergic system may contribute to TS and ADHD. Therefore, genes involved the in

the glutamatergic system are biologically relevant to the pathogenesis of TS and ADHD.

Solute carrier family 1, member 3 (SLC1A3) maps to chromosome 5p13 and

encodes for a glial excitatory amino acid transporter (EAAT1). This transporter is

involved in the clearance of glutamate from the synaptic cleft and prevents glutamate-

induced exictoxicity (Kanai et al., 2013). To date, SLC1A3 has been implicated in two

family-based association studies of ADHD (Turic et al., 2005; Elia et al., 2009) and

produced association signals in a GWAS of ADHD (Neale et al., 2010).

Given the overlap in linkage findings, genes found within the 5p13 region that

have been implicated in ADHD are relevant to the genetics of TS. In 2009, our lab

conducted the first association study of SLC1A3 in TS using a sample consisting of 241

affected children (Laurin et al. 2009). Three ADHD associated single nucleotide

polymorphisms (SNPs) were genotyped in the TS sample. While no positive findings

with TS were observed, it would take 27 tagSNPs (r2 > 0.80) to adequately test whether

genetic variation to this gene is associated with TS. Since the Laurin et al. (2009) study, a

subsequent genetic study of SLC1A3 tested the only non-synonymous mutation, marker

rs2032892, for association with an independent TS sample of 256 affected individuals

and 224 unaffected controls; however, the result of this study did not reach the threshold

of significance (Adamczyk et al., 2011). Other than these investigations, no other studies

have been published exploring whether other SLC1A3 markers are associated with TS.

Page 81: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

68

Since the publication by Laurin et al. (2009), an additional candidate gene study

has associated SLC1A3 with ADHD (Elia et al., 2009). In addition, GWAS have since

been conducted for both ADHD (Neale, et al., 2008, 2010; Polemans et al., 2011) and TS

(Scharf et al., 2012). Although no significant evidence of association for SLC1A3 has

been identified by these GWAS, few variants reached genome-wide significance (p = 5 x

10-8). Given that top signals from these studies may reflect true associations, follow-up

studies of these signals of SLC1A3 merit further investigation.

Prior to the GWAS era, studies of complex genetic disorders focused on the

impact of non-synonymous mutations of protein-coding region. Recent evidence suggests

that variation in non-coding sequences responsible for the regulation of gene expression,

may also play a role in the pathogenesis of complex genetic disorders (Hindorff et al.,

2009; Siniscalco et al., 2013) At the transcriptional level, the regulation of gene

expression is mediated through binding of transcription factors to transcription factor

binding sites located within regulatory regions (e.g., enhancer or promoter regions).

Given that many disease-associated SNPs identified by GWAS are located in or near non-

coding putative regulatory regions of the genome (Manolio, Brooks, Collins, 2008; The

ENCODE project consortium, 2012) alleles located within regulatory regions may have

functional implications on complex genetic disorders and merit further study.

To date, there is little evidence demonstrating that non-synonymous variants in

the coding region of SLC1A3 account for previous associations with ADHD. Despite this,

variants located within non-coding putative regulatory regions of SLC1A3 have not been

well studied. Similar to results from other GWAS (reviewed in Hindorff et al., 2009), the

strongest GWAS findings of ADHD and TS predominately consist of variants located

Page 82: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

69

outside of protein-coding regions (e.g., intronic or intergenic SNPs) that have no known

function (Scharf et al., 2012; Neale, et al., 2008, 2010, reviewed in Poelmans et al., 2011).

It has been proposed that many of the top-signaling SNPs may localize to putative

regulatory regions (reviewed in Manolio, 2013; The ENCODE project consortium, 2012).

For this reason, predictions of regulatory regions may be used to interpret strong signals

from GWAS and improve the power of studies attempting to identify causal variants of

disease (Ernst et al., 2011; The ENCODE project consortium, 2012).

Based on the candidacy strength of SLC1A3, we conducted a family-based

association study using a TS sample of 303 nuclear families with at least one TS affected

child (for a total of 377 affected children). Our objective was to determine if SLC1A3, a

putative ADHD gene, was also associated to TS. SLC1A3 SNPs were selected using three

approaches: (1) re-evaluating three previously investigated SNPs for association using

our extended sample (from 241 affected children to 377 affected children), (2) testing the

strongest four SLC1A3 tagSNP signals from the ADHD GWAS (Neale, et al., 2008,

2010), and (3) testing three markers localized to putative regulatory regions that may help

interpret previous association findings. In total ten SLC1A3 markers were genotyped and

tested for association in our TS sample.

4.2 SLC1A3 Results

Single-marker analysis

To determine whether SLC1A3, an associated ADHD gene, is also associated with

TS we selected seven associated ADHD SLC1A3 markers based on previous reports.

Three of the seven markers (rs2269272, rs3776581 and rs2032893) were selected from a

Page 83: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

70

candidate gene association study (Turic et al., 2005) and four tagSNPs (rs4354072,

rs2562571, rs2303716, rs1529461) were selected from GWAS of ADHD (Neale et al.,

2010). Each marker was tested for association in our TS sample, which consisted of 303

nuclear families with at least one TS or CMT affected child (a total of 377 affected

children). Transmission Disequilibrium Test (TDT) analysis, a test for biased

transmission of alleles from heterozygous parents to affected offspring, was used to

determine whether the marker alleles were associated with TS.

Similar to published results from our lab (Laurin et al., 2009), we were unable to

find an association between three ADHD associated SNPs (rs2269272, rs3776581,

rs2032893; Turic et al., 2005) and TS using our sample. Biased transmission of the

rs2562571 T allele to affected TS children (p = 0.025) was observed, although this

finding would not withstand correction for multiple testing. This marker was originally

selected from a meta-analysis of ADHD GWAS (Neale et al., 2010) and is located

upstream of SLC1A3 in the intergenic region. To determine the potential mechanism of

this variant, we used histone markers that were informative of enhancer regions (see

Methods for further detail). We found no evidence that marker rs2562571 maps to a

putative regulatory region nor were we able to identify any common SNPs in LD (r2

> .80) with this marker that localize within a regulatory region, using HapMap data.

Although no SNPs tagged marker rs2562571 (r2 > 0.80), using HapMap data

marker rs2731901 showed the strongest correlation (r2 > 0.50) and mapped to a predicted

enhancer region. Based on the proximity of the rs2562571 association signal to the

promoter region, we also selected two additional SNPs (rs4443439 and rs2869675) for

association testing that overlapped the promoter region of SLC1A3. Testing of these

Page 84: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

71

additional SNPs using the TDT analysis failed to meet the threshold of significance or

demonstrate any trends (see Table 4-1).

Haplotype analysis

In addition to single-marker analysis, we also conducted haplotype analysis for

two LD blocks using the TRANSMIT program (Clayton & Jones, 1999). LD blocks were

defined using Solid Spine of LD, in which strongest LD can be found between first and

last markers of a block. While no significant evidence of haplotype association was found,

two haplotypes rs2562571/rs4869675 and rs2269272/rs1529461 showed trends were

observed with TS (p-values shown in Table 4-2 and 4-3, respectively; LD structure

shown in Figure 4-1). For the first haplotype, rs2562571/rs4869675, we observed an

overtransmission of the rs2562571 T and rs4869675 G haplotype (χ2 = 3.924, 1 df, p =

0.048; see Table 4-2), and an undertransmission of the rs2562571 C and rs4869675 T

haplotype (χ2 = 4.223, 1 df, p = 0.040; see Table 4-2) to affected offspring. For the

second haplotype, we found an undertransmission of the rs2269272 C and rs1529461 A

haplotype to affected TS offspring (χ2 = 4.379, 1 df, p = 0.036; see Table 4-3). Although

haplotypes rs2562571/ rs4869675 and rs2269272/ rs1529461 showed trends in TS,

neither global association tests were significant (see Table 4-2 and Table 4-3, respectively

for global analysis results). Furthermore, these findings did not reach the threshold of

significance in TS, and therefore should be interpreted with caution.

Page 85: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

72

4.3 Tables & Figures

Table 4–1. Single-marker TDT analysis for 10 genotyped SLC1A3 SNPs in TS sample

Marker Position MAF

(Allele)

HWp Transmission

Ratio (Allele)

χ2 p-value

rs4354072 36367860 0.333 (T) 0.250 110:126 (C:T) 0.949 0.330

rs2731901 36574507 0.190 (G) 0.962 97:82 (A:G) 1.257 0.262

rs2562571 36597841 0.211 (C) 0.849 105:75 (T:C) 5.000 0.025

rs4869675 36600919 0.309 (G) 0.341 129:112 (G:T) 1.199 0.274

rs4443439

(rs4020423)

36607791 0.316 (T) 0.163 136:125 (C:T) 0.464 0.496

rs3776581 36661944 0.336 (A) 0.900 92:84 (G:A) 0.364 0.547

rs2303716 36680125 0.302 (G) 0.175 110:97 (A:G) 0.816 0.366

rs2269272 36687856 0.178 (T) 0.720 84:90 (C:T) 0.207 0.649

rs1529461 36689363 0.219 (A) 0.995 86:105 (A:G) 1.890 0.169

rs2032893 36698622 0.411 (T) 1.000 93:92 (C:T) 0.005 0.941

Table 4–2. SLC1A3 Haplotype analysis of rs2562571/rs4869675 for TS sample

rs2562571 rs4869675 Frequency Observed Expected Var

(O-E)

χ2

(1df)

p value

T T 0.488 336.600 337.830 78.812 0.019 0.890

T G 0.297 223.350 206.990 68.222 3.924 0.048

C T 0.203 131.740 146.450 51.293 4.223 0.040

Global χ2 (3df) with frequencies > 10% = 6.022, p = 0.111

Page 86: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

73

Table 4–3. SLC1A3 Haplotype analysis of rs2269272/rs1529461 for TS sample

rs2269272 rs1529461 Frequency Observed Expected Var

(O-E)

χ2

(1df)

p value

C G 0.606 452.090 442.340 80.603 1.179 0.278

C A 0.218 143.680 158.900 52.862 4.379 0.036

T G 0.175 130.170 124.180 47.853 0.750 0.387

Global χ2 (3df) with frequencies > 10% = 5.668, p = 0.129

Page 87: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

74

Figure 4-1. LD plot of 10 genotyped SLC1A3 SNPs. The numbers within each block represent r2 and are indicative of LD between SNPs. LD blocks were defined using the solid spine of LD are outlined in bold.

Page 88: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

75

Figure 4-2. Relative positions of the 10 genotyped SLC1A3 SNPs.

Page 89: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

76

Discussion & Future Directions Chapter 5

5.1 Discussion

In this investigation, we presented the first association study of SLC1A1 in TS and

ADHD, as well as a follow-up study of SLC1A3 in TS. Our TS sample consisted of DNA

from 377 TS or CMT affected children, and our ADHD sample consisted of DNA from

254 ADHD affected children. While we hypothesized that SLC1A1 and SLC1A3 confer

susceptibility to TS and ADHD, we were unable to detect significant associations using

our samples. However, we did observe trends, which may be indicative of true findings.

Given the role of these genes in the glutamatergic system, these loci merit further study in

larger samples.

5.1.1 SLC1A1

SLC1A1 is a gene that has been associated with OCD, a disorder that is commonly

comorbid with TS. Despite evidence implicating SLC1A1 as a risk gene of OCD, the

proposed genetic relationship of OCD, TS and ADHD, and its involvement in neuronal

glutamate clearance, there are no studies assessing SLC1A1 as a risk gene for TS or

ADHD. For this reason, we sought to determine whether SLC1A1 is also a common risk

gene for TS and ADHD through a family-based association study. Considering that the

genetic relationship between TS and ADHD may be mediated by OCD, we tested OCD

associated variants of SLC1A1 using our TS and ADHD samples.

For the single-marker analysis of SLC1A1 in TS, our strongest association signal

came from marker rs301430, a synonymous mutation within exon 10. This marker was

selected based on its association with early-onset OCD as a single-marker (Arnold et al.,

Page 90: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

77

2006) and due to its inclusion of several OCD-associated haplotypes (Dickel et al., 2006;

Stewart et al., 2009; Wendland et al., 2009). In the current investigation we also observed

weak evidence of association for a three-marker haplotype rs301434/rs301435/rs3087879

with the TS sample (global uncorrected p = 0.004). Interestingly, this corresponds to a

haplotype associated in an OCD study reported by Arnold and colleagues (2006).

However, our findings did not withstand correction for multiple tests.

In a study using tissue from postmortem brains, Wendland and colleagues (2009)

investigated whether SLC1A1 SNPs were associated with SLC1A1 mRNA expression

levels in the prefrontal cortex. The association signal of marker rs301430 was nominally

significant with SLC1A1 mRNA levels, with an increase in expression coinciding with

the number of C alleles (Wendland et al., 2009). The results of a follow-up in-vitro

analysis of SLC1A1 expression using a luciferase reporter gene assay suggested that

rs301430 might be a causal variant (Wendland et al., 2009). We were unable to find any

replication studies to refute or support these findings. However, inconsistent reports of

which rs301430 allele is associated with OCD brings doubt as to whether this variant is a

causal polymorphism. For instance, Dickel et al. (2006) observed overtransmission of the

rs301430 C allele to subjects with early-onset OCD. However, using an independent

OCD sample, Stewart et al. (2007) observed an overtransmission of the rs301430 T allele

to OCD probands. In the present study we observed an overtransmission of the rs301430

C allele to TS probands. One possible explanation for the association of opposite alleles

is that rs301430 is in LD with the causal variant (Lin, Vance, Pericak-Vance & Martin,

2007); however, other association studies of OCD and SLC1A1 were unable to detect an

association signal for this single-marker (Shugart et al., 2009; Samuels et al., 2011;

Page 91: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

78

Taylor et al., 2012; Wu et al., 2013), including a recent meta-analysis of SLC1A1 using

OCD subjects (Stewart et al., 2013). Alternatively, the lack of replicated associations for

this marker may be the result of sampling from differing ancestral populations or

phenotypes examined across studies (Lin et al., 2007; reviewed in Hemmings & Stein,

2006).

In light of our trends in TS for both single- and haplotype- analysis, we tested

these four markers (rs301430, rs301434, rs301435, rs3087879) for association with our

ADHD sample. In contrast to our TS results, we were unable to identify a significant

association between single-marker rs301430 and haplotype

rs301434/rs301435/rs3087879 with the ADHD sample. However, we did observe a weak

association for single-marker rs301435 with our ADHD sample (p = 0.006), but this

finding barely remained significant after multiple-testing corrections.

Despite observing trends for two markers relatively proximal to each other, the

two implicated SNPs were poorly correlated. Considering that the majority of SLC1A1

associations with OCD localize to non-coding regions of the human genome, changes to

DNA sequences of regulatory elements may underlie susceptibility to OCD and its

related disorders (e.g., TS and ADHD). In addition to testing OCD associated SNPs with

TS we also selected SNPs that localized to predicted enhancer elements. The premise

behind this methodology was based on recent findings of large association studies,

including GWAS, which indicated enrichment for SNPs within regulatory elements

(Edwards, Beesley, French, & Dunning, 2013; Schorck et al., 2013). SNPs that localize to

enhancer regions are more likely to be functionally relevant and provide a possible

explanation of the mechanisms underlying associated variants. Despite our efforts to

Page 92: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

79

narrow the search for functionally relevant alleles none of the additionally genotyped

SNPs reached the threshold of significance.

5.1.2 SLC1A3

The second part of this investigation was based on a common TS- and ADHD-

susceptibility region on chromosome 5p13. To date, several genetic association studies

have implicated this gene in ADHD. Yet comprehensive investigations of this gene in TS

are lacking. On the basis that SLC1A3 is a strong positional and functional candidate risk

gene for both ADHD and TS, we sought to determine whether previously reported

association findings for ADHD and three SLC1A3 SNPs (Turic et al., 2005) would be

associated with TS. While the results of our initial investigation (Laurin et al., 2009)

failed to detect a significant association with our sample of 241 TS-affected probands,

our TS sample has since expanded to include 377 TS-affected children. For this reason

we conducted a follow-up association study, re-evaluating the original three markers as

well as more recent markers identified with trends for association with ADHD near

SLC1A3, for association with TS using our larger sample.

The results of our investigation revealed a trend for marker rs2562571 with TS

(p = 0.025): a marker that was selected based on an ADHD GWAS association signal

(Neale et al., 2010). This SNP is located in the intergenic region, approximately 8.6

kilobases upstream of SLC1A3 and has no known function. Using the publically available

HapMap database, we were unable to identify any markers in high LD with marker

rs2562571. Therefore, it is difficult to determine the significance of this finding. Since

our finding did not withstand correction for multiple tests it may not reflect a true

association. Haplotype analysis failed to identify significant global haplotypes, while two

Page 93: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

80

individual haplotypes consisting of markers rs2562571/rs4869675 and

rs2269279/rs1529461 showed trends of association with the TS sample.

A published study of SLC1A3 using 299 ADHD subjects reported a significant

association of a two-marker haplotype (rs2269272/rs2032893) spanning approximately

11 kilobases (Turic et al., 2005). Although we were unable to find a significant

association of this particular haplotype with TS using our sample (not shown), one of the

haplotypes that demonstrated a trend in our study, rs2269272/rs1529461, lies within an

ADHD associated region reported by Turic and colleagues. Considering that this region

has been implicated in ADHD, and was highlighted using our TS sample, it is plausible

that it harbors risk alleles relevant to both TS and ADHD. Using a subset of our TS

sample that was included in the GWAS of TS we were unable to find any positive

findings that would point to further studies. The lack of positive association signals from

this region may have been the result of an underpowered study; therefore this region

remains of interest.

5.1.3 Global Discussion of SLC1A1 and SLC1A3

For both investigations our strongest signals came from SNPs with no known function.

This is not uncommon in genetic association studies of complex traits, were associated

SNPs are commonly found outside of protein-coding regions (Hindorff et al., 2009).

Considering that the purpose of genetic association studies is to identify genetic variants

that can clarify the mechanisms underlying pathophysiology of a particular trait or

disorder, it is important to identify associated variants that can result in functional

changes.

Page 94: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

81

Mutations of the coding regions of SLC1A1 and SLC1A3 do not account for

reported association signals, or the trends observed in the present study. Associated

variants that are found within regulatory elements may influence susceptibility by altering

transcription factor binding. Under the assumption that association signals from other

studies and the trends of this study reflect true associations, we hypothesized that variants

of SLC1A1 and SLC1A3 were influencing gene regulation by altering transcription factor

binding sites. Integration of the ENCODE regulatory track and the UCSC browser

allowed for the identification of regulatory elements. Specifically, we used enrichment of

histone marks H3K4Me1, H3K4Me3 and H3K27Ac to indicate promoter or enhancer

regions.

In the present study, four of the five markers that produced trends (either through

single-marker or haplotype) did not map to putative regulatory regions. For this reason,

potentially functional variants (i.e., alleles that tag putative regulatory regions) that are in

LD with associated markers were also tested for association; however, none of the

additionally genotyped SNPs produced any evidence of association with TS using our

sample.

While the co-occurrence of TS, ADHD, and OCD in the same individual is likely

influenced by genetic factors, identifying a single susceptibility locus that has been

implicated in all three disorders has been challenging. For instance, despite conducting a

PubMed search using the words “Tourette syndrome” “ADHD” “OCD” “GWAS” and

“linkage” it was difficult to identify candidate genes based on strong positional evidence

for all three disorders. This is not entirely unexpected given that most linkage studies of

these disorders have only produced weak evidence of linkage, and sub-threshold findings

Page 95: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

82

are less likely to be published. Despite the number of linked regions that have been

reported for these disorders, evidence of overlapping risk loci is lacking. Recent efforts to

identify genetic variants conferring susceptibility through GWAS studies have also failed

to identify shared susceptibility loci for TS, ADHD and OCD. This may in part be due to

the relatively small sample sizes of the first GWAS of TS and OCD, which included 1285

cases and 1465 cases with 400 trios, respectively. While these GWAS collaborations

have resulted in the largest TS- and OCD- samples to date, it is now clear that more

substantial sample sizes will be required to detect susceptibility loci (Schizophrenia

Working Group of the Psychiatric Genomics Consortium, 2014). For example, a recently

published schizophrenia GWAS of 36,989 cases and 113,075 controls identified 83

additional susceptibility loci, a large increase from the 25 risk loci that had been

previously found using much smaller sample sizes. Collaborative efforts of consortiums

of TS, ADHD and OCD are currently underway to acquire much larger sample sizes to

improve the power of GWAS studies. Therefore, while common susceptibility loci fro all

three disorders remains elusive, future GWAS collaborations will likely highlight

possible genetic candidates based on position.

5.2 Global Limitations

The purpose of the SLC1A1 and SLC1A3 studies were to identify common genetic risk

variants across TS and ADHD. To improve the probability of detecting a functionally

relevant polymorphism, we systematically selected SNPs that located to predicted

promoter or enhancer regions derived from ENCODE histone marks H3K4Me1,

H3K4Me3 and H4K27Ac; however, the use of ENCODE histone marks to predict

regulatory elements are not without limitations. The human body is composed of a wide

Page 96: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

83

variety of cell-types shaped by differential gene expression (Vaquerizas, Kummerfeld,

Teichmann & Luscombe, 2009; Lee et al., 2012). Neural cell-types are no exception to

this and have cell-type specific expression of transcription factors (Ernst et al., 2011).

One of the limitation of using histone mark signals from ENCODE, is that they are

derived from non-neural cell lines. Therefore, histone mark signals should be interpreted

with caution when drawing assumptions of regulatory elements in the brain.

Complex genetic traits often have multiple causal variants with small effects

contributing to susceptibility (reviewed in Glazier, Nadeau, & Aitman, 2002; Manolio et

al., 2008; Hindorff et al., 2009). Association studies of diseases of polygenic inheritance

generally identify risk variants with effect sizes greater or less than 1.2 (Schanze et al.,

2011; Wiste et al., 2014). For the SLC1A1 study we would only have been able to detect

an association for a genetic variant with a relative risk of 1.75-1.96 for TS and 1.65-2.01

for ADHD using our respective samples. For the SLC1A3 study, we only had adequate

power to detect relatively large effect sizes for TS. Both TS and ADHD are proposed to

be due to polygenic inheritance with multiple common risk variants of small but additive

effects. It is plausible that the actual genetic effect sizes were too low to detect given our

relatively modest samples sizes. Under these assumptions non-significant trends observed

in our studies may be due to insufficient power to detect variants of small effect, and may

reflect true associations. However, to confirm these assumptions, meta-analysis or larger

samples in respective samples would be required.

A methodological limitation of this study derives from the use of the family-based

association test, TDT. While this test controls for population stratification by using

parental genotypes of affected offspring, only transmissions of heterozygous parents are

Page 97: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

84

informative. Therefore, although our entire respective samples consisted of 377 TS-

affected probands and 247 affected-ADHD probands, only a proportion of the parental-

offspring transmissions were informative. This may in part account for our lack of

positive findings, but can be addressed by using larger sample sizes.

The composition of our TS sample was also a challenge of this study. Many of

our TS probands had also been diagnosed with comorbid ADHD or OCD, with only a

small proportion (32%) of probands having a diagnosis of TS only. This was not entirely

unexpected as TS is rarely found without comorbid conditions. However, our TS sample

was more reflective of a TS+ADHD sample. Once again this was not entirely unexpected

considering that the prevalence of comorbid ADHD with TS is substantially higher than

that of TS without ADHD in both clinical and population based studies. The presence of

subgroups could have been beneficial given a larger sample size. Stratification into more

homogeneous groups (TS only, TS + ADHD, TS + ADHD + OCD, TS + OCD) would

have allowed us to compare the association signals of the genotyped variants across

subgroups; however, considering that none of our findings remained significant after

corrections for multiple tests using our entire sample, analysis of these subgroups would

require a much larger sample to ensure adequate power.

5.3 Future Direction

5.3.1 SLC1A1

While we were unable to detect a significant association between SNPs within predicted

enhancer regions and our TS and ADHD samples, it is plausible that DNA changes to

other cis-regulatory elements of SLC1A1 may account for reported associations or trends.

Page 98: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

85

For instance, while alternatively spliced transcripts do not always result in dysfunctional

proteins (reviewed in Tazi, Bakkour & Stamm, 2009), a recent study identified an

isoform transcript of eaat3 that lacks exon 11 in the brain (Porton et al., 2013). The

resulting protein from this transcript has a truncated carboxylic end—an alteration that

has been proposed to interfere with the assembly of the glutamate transporter trimer

(Porton et al., 2013) and reduce the transporter’s affinity for glutamate (Maragakis &

Rothstein, 2004).

The haplotype detected in our study, rs301434/rs301435/rs3087879, spans 10kb

overlapping the 11th exon and 3’ untranslated region. Based on association signals from

this region in studies of OCD, and the trends observed using our TS and ADHD samples,

it is plausible that DNA sequence changes involving these variants play a role in pre-

mRNA splicing. Another possibility is that the causal variants lie within this region and

influence the regulation of SLC1A1 through post-transcriptional effects.

Alternatively splice transcripts are the result of a process called splicing. Splicing

involves the removal of introns from pre-mRNA transcripts and is regulated by binding

of spliceosomes to specific regions of the DNA (splice regulatory elements). Mutations to

regulatory elements including intronic splice enhancer have been associated with disease

and exon skipping (Wei, Lin, Modafferi et al., 1997; McCarthy and Phillips, 1998).

Therefore, changes to DNA sequences where splice regulatory proteins bind may account

for the alternatively spliced transcript reported by Porton et al.

Publically available databases including the Human Splice Finder

(http://www.umd.be/HSF/) may be used to evaluate this region for splice regulatory

Page 99: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

86

elements. Future studies using larger samples are also required to determine whether

signals from this study reflect true findings.

5.3.2 SLC1A3

Future investigations should evaluate marker rs2562571 using an independent

sample of TS. Assuming this finding is replicated, future studies should attempt to clarify

the underlying mechanism of this SNP. One possibility is that it is located within an

enhancer region specific to neural tissue. Enrichment of histone marks sequenced from

neural tissue can be used to determine whether enhancer activity is present at or near this

locus. Further, luciferase gene assays may be used to determine whether marker

rs2562571 coincides with SLC1A3 expression levels.

Haplotype analysis using our TS sample highlighted an ADHD associated region.

Assuming that our trend represents a true finding, it is plausible that markers within this

haplotype region can account for the observed association signal. Using the genome

browser and ENCODE transcription factor binding track we were unable to find a

putative enhancer element within the haplotype region; however, we did identify a

putative insulator element. Insulator elements are also involved in the regulation of gene

transcription. Unlike enhancer elements, which are involved in facilitating transcription

upon binding of transcription factors, insulator elements may block enhancer activity of

distal genes (reviewed in Burgess-Beusse, et al., 2002). We were able to identify a single

marker, rs1122900, located within this putative insulator element that has a MAF greater

than 30%. Using HapMap data we were able to determine that this marker is more

strongly correlated with single-markers rs2269272 and rs1529461 than those markers are

with each other. Considering the correlation of marker rs1122900 with markers

Page 100: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

87

rs1529461 and rs2269272, as well as its functional relevance, it is a robust candidate to

explain the haplotype signal detected in this investigation. Further study is required to test

this marker for association using TS and ADHD samples. Under the assumption that a

significant association signal is detected, subsequent investigations should also test this

polymorphism for regulatory activity in neural tissue.

As noted earlier it would take 27 tagSNPs (r2 > 0.80) to adequately capture the

SLC1A3 gene. In this study only 10 markers were tested for association. It is plausible

that other polymorphisms of this gene contribute to TS, and that the GWAS results from

the subset of TS samples lacked adequate power to detect association signals. Therefore,

markers that tag the result of this gene should also be tested for association with TS

samples.

The present investigation examined SLC1A3 in TS and ADHD. However, TS and

ADHD are likely mediated by OCD. To our knowledge, the 5p13 region has not been

implicated in OCD. Nevertheless, SLC1A3 may contribute to OCD susceptibility, as

multiple candidate genes are assessed based on function without positional evidence.

Furthermore, variants associated with disease do not always produce an association signal

in every study or reach the threshold for genome-wide significance (Xu & Taylor, 2009;

Yeager et al., 2007; Thomas et al., 2008). It is plausible that studies of OCD to date have

not had adequate power to detect an association signal at the 5p13 region because its

effect size is very small. Based on the trends observed in the TS sample of this study and

other investigations of ADHD, as well as its involvement in the glutamatergic system,

association testing of SLC1A3 is required using OCD samples.

Page 101: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

88

5.3.3 Future Candidate Genes for Study

As mentioned earlier, it is plausible that the trends observed in this study reflect positive

findings that are limited by the lack of power (discussed further above in Global

Limitations). Alternatively, the lack of significant findings from this study may suggest

that SLC1A1 and SLC1A3 are not involved in TS or ADHD.

A fairly novel gene of study is protein tyrosine phosphatase receptor type D,

PTPRD. PTPRD is encoded on chromosome 9p24, in the same region as SLC1A1. In

addition to being involved in the differentiation of glutamatergic synapses, and

interacting with other highly studied genes (e.g., SLITRK3), PTPRD has been implicated

in OCD (Matthesien et al., 2014) and ADHD (Elia, Gai, Xie et al., 2010), but as not been

examined in TS. Therefore, PTPRD is an attractive gene for study in the search for

common susceptibility loci for TS, ADHD and OCD.

5.4 Conclusion

SLC1A1 and SLC1A3 play important roles in glutamate regulation in the brain. While

we were unable to produce concrete evidence that the genes of these glutamate

transporters are associated with TS and ADHD, we cannot rule out that they contribute to

these disorders. Alterations to gene expression provide a plausible explanation of the

mechanisms underlying the observed trends and reported association signals. Specifically,

identification of regulatory elements using neural tissue and studies using larger samples

will allow for further insight as to whether SLC1A1 and SLC1A3 are risk genes for TS

and ADHD.

Page 102: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

89

References Abecasis, G. R., Cherny, S. S., Cookson, W. O., & Cardon, L. R. (2002). Merlin--rapid analysis

of dense genetic maps using sparse gene flow trees. Nat Genet , 30 (1), 97-101.

Abelson, J. F., Kwan, K. Y., O'Roak, B. J., Baek, D. Y., Stillman, A. A., Morgan, T. M., et al. (2005). Sequence variants in SLITRK1 are associated with Tourettes syndrome. Science , 10 (5746), 317-320.

Adamczyk, A., Gause, C. D., Sattler, R., Vidensky, S., Rothstein, J. D., Singer, H., et al. (2011). Genetic and functional studies of a missense variant in a glutamate transporter, SLC1A3, in Tourette syndrome. Psychiatr Genet , 21 (2), 90-97.

Albin, R. L., & Mink, J. W. (2006). Recent advances in Tourette syndrome research. Trends Neurosci , 29 (3), 175-182.

Alsobrook, J. P., & Pauls, D. L. (2002). A factor analysis of tic symptoms in Gilles de la Tourette's syndrome. Am J Psychiatry , 159 (2), 291-296.

American Psychiatric Association. (1987). Diagnostic and statistical manual of mental disorders (3rd ed., text rev.). Washington, DC: American Psychiatric Association.

American Psychiatric Association. (2013). Diagnostic and statistical manual of mental disorders, fifth edition. Washington, DC: American Psychiatric Association.

Anderson, G. M., Pollak, E. S., Chatterjee, D., Leckman, J. F., Riddle, M. A., & Cohen, D. J. (1992). Postmortem analysis of subcortical monoamines and amino acids in Tourette syndrome. Adv Neurol , 58, 123-133.

Aoyama, K., Suh, S. W., Hamby, A. M., Liu, J., Chan, W. Y., Chen, Y., et al. (2006). Neuronal glutathione deficiency and age-dependent neurodegeneration in the EAAC1 deficient mouse. Nat Neurosci , 9 (1), 119-126.

Arcos-Burgos, M., Castellanos, F. X., Pineda, D., Lopera, F., Palacio, J. D., Palacio, L. G., et al. (2004). Attention-deficit/hyperactivity disorder in a population isolate: linkage to loci at 4q13.2, 5q33.3, 11q22, and 17p11. Am J Hum Genet , 75 (6), 998-1014.

Arcos-Burgos, M., Londoño, A. C., Pineda, D. A., Lopera, F., Palacio, J. D., Arbelaez, A., et al. (2012). Analysis of brain metabolism by proton magnetic resonance spectroscopy (1H-MRS) in attention-deficit/hyperactivity disorder suggests a generalized differential ontogenic pattern from controls. Atten Defic Hyperact Disord , 4 (4), 205-212.

Arnold, P. D., Rosenberg, D. R., Mundo, E., Tharmalingam, S., Kennedy, J. L., & Richter, M. A. (2004). Association of a glutamate (NMDA) subunit receptor gene (GRIN2B) with obsessive-compulsive disorder: a preliminary study. Psychopharmacology , 174 (4), 530-538.

Arnold, P. D., Sicard, T., Burroughs, E., Richter, M. A., & Kennedy, J. L. (2006). Glutamate transporter gene SLC1A1 associated with obsessive-compulsive disorder. Arch Gen Psychiatry , 63 (7), 769-776.

Page 103: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

90

Bakker, S. C., van der Meulen, E. M., Buitelaar, J. K., Sandkuijl, L. A., Pauls, D. L., Monsuur, A. J., et al. (2003). A whole-genome scan in 164 Dutch sib pairs with attention-deficit/hyperactivity disorder: suggestive evidence for linkage on chromosomes 7p and 15q. Am J Hum Genet , 75 (5), 1251-1260.

Banaschewski, T., Neale, B. M., Rothenberger, A., & Roessner, V. (2007). Comorbidity of tic disorders & ADHD: conceptual and methodological considerations. Eur Child Adolesc Psychiatry , 16 (Suppl 1), 5-14.

Barr, C. L., Wigg, K. G., Pakstis, A. J., Kurlan, R., Pauls, D., Kidd, K. K., et al. (1999). Genome scan for linkage to Gilles de la Tourette syndrome. Am J Med Genet , 88 (4), 437-445.

Barrett, J. C., Fry, B., Maller, J., & Daly, M. J. (2005). Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics , 21 (2), 263-265.

Barski, A., Cuddapah, S., Cui, K., Roh, T. Y., Schones, D. E., Wang, Z., et al. (2007). High-resolution profiling of histone methylations in the human genome. Cell , 129 (4), 823-837.

Behrens, P. F., Franz, P., Woodman, B., Lindenberg, K. S., & Landwehrmeyer, G. B. (2002). Impaired glutamate transport and glutamate-glutamine cycling: downstream effects of the Huntington mutation. Brain , 125 (Pt 8), 1908-1922.

Benedetti, F., Poletti, S., Radaelli, D., Pozzi, E., Giacosa, C., Ruffini, C., et al. (2012). Caudate gray matter volume in obsessive-compulsive disorder is influenced by adverse childhood experiences and ongoing drug treatment. J Clin Psychopharmacol , 32 (4), 544-547.

Biederman, J., & Faraone, S. V. (2005). Attention-deficit hyperactivity disorder. Lancet , 366 (9481), 237-248.

Bienvenu, O. J., et al. (2009). Sapap3 and pathological grooming in humans: Results from the OCD collaborative genetics study. Am J med Genet B Neuropsychiatr Genet , 150B (5), 710-720.

Bloch, M. H., & Leckman, J. F. (2009). Clinical course of Tourette syndrome. J Psychosom Res , 67 (6), 497-501.

Bloch, M. H., Leckman, J. F., Zhu, H., & Peterson, B. S. (2005). Caudate volumes in childhood predict symptom severity in adults with Tourette syndrome. Neurology , 65 (8), 1253-1258.

Bloch, M. H., Peterson, B. S., Scahill, L., Otka, J., Katsovich, L., Zhang, H., et al. (2006). Adulthood outcome of tic and obsessive-compulsive symptom severity in children with Tourette syndrome. Arch Pediatr Adolesc Med , 160 (1), 65-69.

Bohlhalter, S., Goldfine, A., Matteson, S., Garraux, G., Hanakawa, T., Kansaku, K., et al. (2006). Neural correlates of tic generation in Tourette syndrome: an event-related functional MRI study. Brain , 129 (Pt 8), 2029-2037.

Borecki, I. B., & Suarez, B. K. (2001). Linkage and association: basic concepts. Adv Genet , 42, 45-66.

Page 104: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

91

Boyle, M. H., Offord, D. R., Racine, Y., Fleming, J. E., Szatmari, P., & Sanford, M. (1993). valuation of the revised Ontario Child Health Study scales. J Child Psychol Psychiatry , 34, 189-213.

Breedveld, G. J., Fabbrini, G., Oostra, B. A., Berardelli, A., & Bonifati, V. (2010). Tourette disorder spectrum maps to chromosome 14q31.1 in an Italian kindred. Neurogenetics , 11 (4), 417-423.

Breiter, H. C., Rauch, S. L., Kwong, K. K., Baker, J. R., Weisskoff, R. M., Kennedy, D. N., et al. (1996). Functional magnetic resonance imaging of symptom provocation in obsessive-compulsive disorder. Arch Gen Psychiatry , 53 (7), 595-606.

Brennan-Minnella, A. M., Shen, Y., El-Benna, J., & Swanson, R. A. (2013). Phosphoinositide 3-kinase couples NMDA receptors to superoxide release in excitotoxic neuronal death. Cell Death Dis , 4.

Bulger, M., & Groudine, M. (2011). Functional and mechanistic diversity of distal transcription enhancers. Cell , 144 (3), 327-339.

Burgess-Beusse, B., Farrell, C., Gaszner, M., Litt, M., Mutskov, V., Recillas-Targa, F., et al. (2002). The insulation of genes from external enhancers and silencing chromatin. Proc Natl Acad Sci USA , 99 (Suppl 4), 16433-16437.

Carlson, C. S., Eberle, M. A., Rieder, M. J., Yi, Q., Kruglyak, L., & Nickerson, D. A. (2004). Selecting a maximally informative set of single-nucleotide polymorphisms for association analyses using linkage disequilibrium. Am J Hum Genet , 74 (1), 106-120.

Carrey, N., MacMaster, F. P., Fogel, J., Sparkes, S., Waschbusch, D., Sullivan, S., et al. (2003). Metabolite changes resulting from treatment in children with ADHD: a 1H-MRS study. Clin Neuropharmacol , 26 (4), 218-221.

Carrey, N., MacMaster, F. P., Sparkes, S. J., Khan, S. C., & Kusumakar, V. (2002). Glutamatergic changes with treatment in attention deficit hyperactivity disorder: a preliminary case series. J Child Adolesc Psychopharmacol , 12 (4), 331-336.

Chambers, W. J., Puig-Antich, J., Hirsch, M., Paez, P., Ambrosini, P. J., Tabrizi, M. A., et al. (1985). The assessment of affective disorders in children and adolescents by semistructured interview. Test-retest reliability of the schedule for affective disorders and schizophrenia for school-age children, present episode version. Arch Gen Psychiatry , 42 (7), 696-702.

Cheverud, J. M. (2001). A simple correction for multiple comparisons in interval mapping genome scans. Heredity , 87 (Pt 1), 52-58.

Choudhury, P. R., Lahiri, S., & Rajamma, U. (2012). Glutamate mediated signaling in the pathophysiology of autism spectrum disorders. Pharmacol Biochem Behav , 100 (4), 841-849.

Cirulli, E. T., & Goldstein, D. B. (2010). Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nat Rev Genet , 11 (6), 415-425.

Page 105: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

92

Clarke, G. M., Anderson, C. A., Pettersson, F. H., Cardon, L. R., Morris, A. P., & Zondervan, K. T. (2011). Basic stastical analysis in genetic case-control studies. Nat Protoc , 6 (2), 121-133.

Clayton, D., & Jones, H. (1999). Transmission/disequilibrium tests for extended marker haplotypes. Am J Hum Genet , 65 (4), 1161-1169.

Clements, J. D., & Westbrook, G. L. (1991). Activation kinetics reveal the number of glutamate and glycine binding sites on the N-methyl-D-aspartate receptor. Neuron , 7 (4), 605-613.

Comings, D. E. (2001). Clinical and molecular genetics of ADHD and Tourette syndrome. Two related polygenic disorders. Ann N Y Acad Sci , 931, 50-83.

Comings, D. E., & Comings, B. G. (1987). Tourette's syndrome and attention deficit disorder with hyperactivity. Arch Gen Psychiatry , 44 (11), 1023-1026.

Comings, D. E., & Comings, B. G. (1984). Tourette's syndrome and attention deficit disorder with hyperactivity: are they genetically related? J Am Acad Child Psychiatry , 23 (2), 138-146.

Comings, D. E., Wu, S., Chiu, C., Ring, R. H., Gade, R., Ahn, C., et al. (1996). Polygenic inheritance of Tourette syndrome, stuttering, attention deficit hyperactivity, conduct, and oppositional defiant disorder: the additive and subtractive effect of three dopaminergic genes-- DRD2, D beta H, and DAT1. Am J Med Genet , 67 (3), 264-288.

Conelea, C. A., Woods, D. W., Zinner, S. H., Budman, C., Murphy, T., Scahill, L. D., et al. (2011). Exploring the impact of tic disorders on youth: results from the Tourette Syndrome Impact Survey. Child Psychiatry Hum Dev , 42 (2), 219-242.

Conners, C. K. (1997). Conners' Rating Scales- Revised. Toronto, Canada: Multi-Health Systems.

Coric, V., Taskiran, S., Pittenger, C., Wasylink, S., Mathalon, D. H., Valentine, G., et al. (2005). Riluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. Biol Psychiatry , 58 (5), 424-428.

Courvoisie, H., Hooper, S. R., Fine, C., Kwock, L., & Castillo, M. (2004). Neurometabolic functioning and neuropsychological correlates in children with ADHD-H: preliminary findings. J Neuropsychiatry Clin Neurosci , 16 (1), 63-69.

Curtis, D. (1997). Use of siblings as controls in case-control assocation studies. Ann Hum Genet , 61 (Pt 4), 319-333.

Curtis, D., Brett, P., Dearlove, A. M., McQuillin, A., Kalsi, G., Robertson, M. M., et al. (2004). Genome scan of Tourette syndrome in a single large pedigree shows some support for linkage to regions of chromosomes 5, 10 and 13. Psychiatr Genet , 14 (2), 83-87.

Curtis, D., Robertson, M. M., & Gurling, H. M. (1992). Autosomal dominant gene transmission in a large kindred with Gilles de la Tourette syndrome. Br J Psychiatry , 160, 845-849.

Page 106: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

93

Cutler, D., Murphy, T., Gilmour, J., & Heyman, I. (2009). The quality of life of young people with Tourette syndrome. Child Care Health Dev , 35 (4), 496-504.

Daly, M. J., Rioux, J. D., Schaffner, S. F., Hudson, T. J., & Lander, E. S. (2001). High-resolution haplotype structure in the human genome. Nat Genet , 29 (2), 229-232.

Davis, L. K., Yu, D., Keenan, C. L., Gamazon, E. R., Konkashbaev, A. I., & et al. (2013). Partitioning the heritability of Tourette syndrome and obsessive compulsive disorder reveals differences in genetic architecture. PLoS Genet , 9 (10), 1-14.

Debes, N. M., Hjalgrim, H., & Skov, L. (2009). The presence of comorbidity in Tourette syndrome increases the need for pharmacological treatment. J Child Neurol , 24 (12), 1504-1512.

Delorme, R., Krebs, M. O., Chabane, N., Roy, I., Millet, B., Mouren-Simeoni, M. C., et al. (2004). Frequency and transmission of glutamate receptors GRIK2 and GRIK3 polymorphisms in patients with obsessive compulsive disorder. Neuroreport , 15 (4), 699-702.

Delvin, B., & Risch, N. (1995). A comparison of linkage disequilibrium measures for fine-scale mapping. Genomics , 29 (2), 311-322.

DeVito, T. J., Drost, D. J., Pavlosky, W., Neufeld, R. W., Rajakumar, N., McKinlay, B. D., et al. (2005). Brain magnetic resonance spectroscopy in Tourette's disorder. J Am Acad Child Adolesc Psychiatry , 44 (12), 1301-1308.

Devlin, B., & Roeder, K. (1999). Genomic control for association studies. Biometrics , 55 (4), 997-1004.

Diamond, J. S. (2001). Neuronal glutamate transporters limit activation of NMDA receptors by neurotransmitter spillover on CA1 pyramidal cells. J Neurosci , 21 (21), 8328-8338.

Dickel, D. E., Veenstra-VanderWeele, J., Cox, N. J., Wu, X., Fischer, D. J., Van Etten-Lee, M., et al. (2006). Association testing of the positional and functional candidate gene SLC1A1/EAAC1 in early-onset obsessive-compulsive disorder. Arch Gen Psychiatry , 63 (7), 778-785.

Dorval, K. M., Wigg, K. G., Crosbie, J., Tannock, R., Kennedy, J. L., Ickowicz, A., et al. (2007). Association of the glutamate receptor subunit gene GRIN2B with attention-deficit/hyperactivity disorder. Genes Brain Behav , 6 (5), 444-452.

Dramsdahl, M., Ersland, L., Plessen, K. J., Haavik, J., Hugdahl, K., & Specht, K. (2011). Adults with attention-deficit/hyperactivity disorder - a brain magnetic resonance spectroscopy study. Front Psychiatry , 2 (65), 1-8.

Duan, Q. L., & et al. (2014). A genome-wide association study of bronchodilator response in asthmatics. Pharmacogenomics J , 14 (1), 41-47.

Page 107: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

94

Eapen, V., Pauls, D. L., & Robertson, M. M. (1993). Evidence for autosomal dominant transmission in Tourette's syndrome. United Kingdom cohort study. Br J Psychiatry , 162, 593-596.

Eapen, V., Robertson, M. M., Alsobrook, J. P., & Pauls, D. L. (1997). Obsessive compulsive symptoms in Gilles de la Tourette syndrome and obsessive compulsive disorder: differences by diagnosis and family history. Am J Med Genet , 74 (4), 432-438.

Eddy, C. M., Cavanna, A. E., Gulisano, M., Calì, P., Robertson, M. M., & Rizzo, R. (2012). The effects of comorbid obsessive-compulsive disorder and attention-deficit hyperactivity disorder on quality of life in Tourette syndrome. Journal of Neuropsychiatry and Clinical Neurosciences , 24, 458-462.

Edge, M. D., Gorroochurn, P., & Rosenberg, N. A. (2013). Windfalls and pitfalls: Applications of population genetics to the search for disease genes. Evol Med Public Health , 2013 (1), 254-272.

Edwards, S. L., Beesley, J., French, J. D., & Dunning, A. M. (2013). Beyond GWASs: illuminating the dark road from association to function. Am J Hum Genet , 93 (5), 779-797.

Eichstedt, J. A., & Arnold, S. L. (2001). Childhood-onset obsessive-compulsive disorder: a tic-related subtype of OCD? Clin Psychol Rev , 21 (1), 137-157.

Elia, J., Capasso, M., Zaheer, Z., Lantieri, F., Ambrosini, P., Berrettini, W., et al. (2009). Candidate gene analysis in an on-going genome-wide association study of attention-deficit hyperactivity disorder: suggestive association signals in ADRA1A. Psychiatr Genet , 19 (3), 134-141.

Ernst, J., & Kellis, M. (2010). Discovery and characterization of chromatin states for systematic annotation of the human genome. Nat Biotechnol , 28 (8), 817-825.

Ernst, J., Kheradpour, P., Mikkelsen, T. S., Shoresh, N., Ward, L. D., Epstein, C. B., et al. (2011). Mapping and analysis of chromatin state dynamics in nine cell types. Nature , 473 (7345), 45-49.

Evans, D. M., & Purcell, S. (2012). Power calculations in genetic studies. Cold Spring Harb Protoc , 2012 (6), 664-674.

Fahim, C., Yoon, U., Sandor, P., Frey, K., & Evans, A. C. (2009). Thinning of the motor-cingulate-insular cortices in siblings concordant for Tourette syndrome. Brain Topogr , 22 (3), 176-184.

Faye, L. L., Machiela, M. J., Kraft, P., Bull, S. B., & Sun, L. (2013). Re-ranking sequencing variants in the post-GWAS era for accurate causal variant identification. PLoS Genet , 9 (8).

Ferreira, P. E., Palmini, A., Bau, C. H., Grevet, E. H., Hoefel, J. R., Rohde, L. A., et al. (2009). Differentiating attention-deficit/hyperactivity disorder inattentive and combined types: a

Page 108: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

95

(1)H-magnetic resonance spectroscopy study of fronto-striato-thalamic regions. J Neural Transm , 116 (5), 623-629.

Fisher, S. E., Francks, C., McCracken, J. T., McGough, J. J., Marlow, A. J., MacPhie, I. L., et al. (2002). A genomewide scan for loci involved in attention-deficit/hyperactivity disorder. Am J Hum Genet , 70 (5), 1183-1196.

Formosa, T. (2003). Changing the DNA landscape: putting a SNP on chromatin. Curr Top Microbiol Immunol , 274, 171-201.

Fox, M. A. (2008). Novel roles for collagens in wiring the vertebrate nervous system. Curr Opin Cell Biol , 20, 508-513.

Francis, P. T. (2003). Glutamatergic systems in Alzheimer's disease. Int J Geriatr Psychiatry , 18 (Suppl 1), S15-S21.

Freeman, R. D., & Consortium, T. S. (2007). Tic disorders and ADHD: anwers from a world-wide clinical dataset on Tourette syndrome. Eur Child Adolesc Psychiatry , 16 (Supplement 1), 1/15-1/23.

Freeman, R. D., Fast, D. K., Burd, L., Kerbeshian, J., Robertson, M. M., & Sandor, P. (2000). An international perspective on Tourette syndrome: selected findings from 3500 individuals in 22 countries. Dev Med Child Neurol , 42, 436-447.

Furuta, A., Martin, L. J., Lin, C. L., Dykes-Hoberg, M., & Rothstein, J. D. (1997). Cellular and synaptic localization of the neuronal glutamate transporters excitatory amino acid transporter 3 and 4. Neuroscience , 81 (4), 1031-1042.

Furuta, A., Rothstein, J. D., & Martin, L. J. (1997). Glutamate transporter protein subtypes are expressed differentially during rat CNS development. J Neurosci , 17 (21), 8363-8375.

Geller, D., Petty, C., Vivas, F., Johnson, J., Pauls, D., & Biederman, J. (2007). Examining the relationship between obsessive-compulsive disorder and attention-deficit/hyperactivity disorder in children and adolescents: a familial risk analysis. Biol Psychiatry , 61 (3), 316-321.

Geller, D., Petty, C., Vivas, F., Johnson, J., Pauls, D., & Biederman, J. (2007). Further evidence for co-segregation between pediatric obsessive compulsive disorder and attention deficit hyperactivity disorder: a familial risk analysis. Biol Psychiatry , 61 (12), 1388-1394.

Genetics, T. T. (2007). Genome Scan for Tourette Disorder in Affected-Sibling-Pair and Multigenerational Families. Am J Hum Genet , 80, 265-272.

Glazier, A. M., Nadeau, J. H., & Aitman, T. J. (2002). Finding genes that underlie complex traits. Science , 298 (5602), 2345-2349.

Gogtay, N., Giedd, J. N., Lusk, L., Hayashi, K. M., Greenstein, D., Vaituzis, A. C., et al. (2004). Dynamic mapping of human cortical development during childhood through early adulthood. Proc Natl Acad Sci USA , 101 (21), 8174-8179.

Page 109: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

96

Goodman, W. K., Price, L. H., Rasmussen, S. A., Mazure, C., Fleischmann, R. L., Hill, C. L., et al. (1989). The Yale-Brown Obsessive-Compulsive Scale: development, use, and reliability. Arch Gen Psychiat , 46, 1006-1011.

Grados, M. A., & Mathews, C. A. (2009). Clinical phenomenology and phenotype variability in Tourette syndrome. J Psychosom Res , 67 (6), 491-496.

Grados, M. A., Mathews, C. A., & TSAICG. (2008). Latent class analysis of Gilles de la Tourette syndrome using comorbidities: clinical and genetic implications. Biol Psychiatry , 64 (3), 219-225.

Grados, M. A., Riddle, M. A., Samuels, J. F., Liang, K. Y., Hoehn-Saric, R., Bienvenu, O. J., et al. (2001). The familial phenotype of obsessive-compulsive disorder in relation to tic disorders: the Hopkins OCD family study. Biol Psychiatry , 50 (8), 559-565.

Grados, M. A., Specht, M. W., Sung, H. M., & Fortune, D. (2013). Glutamate drugs and pharmacogenetics of OCD: a pathway-based exploratory approach. Expert Opin Drug Discov , 8 (12), 1515-1527.

Green, E. K., & et al. (2013). Replication of bipolar disorder susceptibility alleles and identification of two novel genome-wide significant associations in a new bipolar disorder case-control sample. Mol Psychiatry , 18 (12), 1302-1307.

Greenberg, B. D., Ziemann, U., Corá-Locatelli, G., Harmon, A., Murphy, D. L., Keel, J. C., et al. (2000). Altered cortical excitability in obsessive-compulsive disorder. Neurology , 54 (1), 142-147.

Hanna, G. L., Veenstra-VanderWeele, J., Cox, N. J., Boehnke, M., Himle, J. A., Curtis, G. C., et al. (2002). Genome-wide linkage analysis of families with obsessive-compulsive disorder ascertained through pediatric probands. Am J Med Genet , 114 (5), 541-552.

Harrison, B. J., Pujol, J., Cardoner, N., Deus, J., Alonso, P., & et al. (2013). Brain corticostriatal systems and the major clinical symptom dimensions of obsessive-compulsive disorder. Biol Psychiatry , 73 (4), 321-328.

Hasler, G., Pinto, A., Greenberg, B. D., Samuels, J., Fyer, A. J., Pauls, D., et al. (2007). Familiality of factor analysis-derived YBOCS dimensions in OCD-affected sibling pairs from the OCD Collaborative Genetics Study. Biol Psychiatry , 61 (5), 617-625.

Hassan, N., & Cavanna, A. E. (2012). The prognosis of Tourette syndrome: implications for clinical practice. Funct Neurol , 27 (1), 23-27.

Hasstedt, S. J., Leppert, M., Filloux, F., van de Wetering, B. J., & McMahon, W. M. (1995). Intermediate inheritance of Tourette syndrome, assuming assortative mating. Am J Hum Genet , 57 (3), 682-689.

Hebebrand, J., Dempfle, A., Saar, K., Thiele, H., Herpertz-Dahlmann, B., Linder, M., et al. (2006). A genome-wide scan for attention-deficit/hyperactivity disorder in 155 German sib-pairs. Mol Psychiatry , 11 (2), 196-205.

Page 110: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

97

Heintzman, N. D., Stuart, R. K., Hon, G., Fu, Y., Ching, C. W., Hawkins, R. D., et al. (2007). Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet , 39 (3), 311-318.

Hemmings, S. M., & Stein, D. J. (2006). The current status of association studies in obsessive-compulsive disorder. Psychiatr Clin North Am , 29 (2), 411-444.

Hemmings, S. M., Kinnear, C. J., Lochner, C., Niehaus, D. J., Knowles, J. A., Moolman-Smook, J. C., et al. (2004). Early- versus late-onset obsessive-compulsive disorder: investigating genetic and colinical correlates. Psychiatry Res , 128 (2), 175-182.

Hettema, J. M., Neale, M. C., & Kendler, K. S. (2001). A review and meta-analysis of the genetic epidemiology of anxiety disorders. Am J Psychiatry , 158 (10), 1568-1578.

Hindorff, L. A., Sethupathy, P., Junkins, H. A., Ramos, E. M., Mehta, J. P., Collins, F. S., et al. (2009). Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci USA , 106 (23), 9362-9367.

Hoexter, M. Q., de Souza Duran, F. L., D'Alcante, C. C., Dougherty, D. D., Shavitt, R. G., Lopes, A. C., et al. (2012). Gray matter volumes in obsessive-compulsive disorder before and after fluoxetine or cognitive-behavior therapy: a randomized clinical trail. Neuropsychopharmacology , 37 (3), 734-745.

Holmseth, S., Dehnes, Y., Huang, Y. H., Follin-Arbelet, V. V., Grutle, N. J., Mylonakou, M. N., et al. (2012). The density of EAAC1 (EAAT3) glutamate transporters expressed by neurons in the mammalian CNS. J Neurosci , 32 (17), 6000-6013.

Hosking, L., Lumsden, S., Lewis, K., Yeo, A., McCarthy, L., Bansal, A., et al. (2004). Detection of genotyping errors by Hardy–Weinberg equilibrium testing. Eur J Hum Genet , 12, 395-399.

Hughes, J. R., & et al. (2014). Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment. Nat Genet , 46 (2), 205-212.

Hyde, G. W., Hern, K. L., Novey, E. S., Elipoulos, D., Marshall, R., Gonzalez, J. J., et al. (1993). Attention deficit-hyperactivity disorder and asymmetry of the caudate nucleus. J Child Neurol , 8 (4), 339-347.

Hyde, T. M., Aaronson, B. A., Randolph, C., Rickler, K. C., & Weinberger, D. R. (1992). Relationship of birth weight to the phenotypic expression of Gilles de la Tourette's syndrome in monozygotic twins. Neurology , 42 (3 Pt 1), 652-658.

Ickowicz, A., Schachar, R. J., Sugarman, R., Chen, S. X., Millette, C., & Cook, L. (2006). The parent interview for child symptoms: a situation-specific clinical research interview for attention-deficit hyperactivity and related disorders. Can J Psychiatry , 51 (5), 325-328.

Jin, Z., Zang, Y. F., Zeng, Y. W., Zhang, L., & Wang, Y. F. (2001). Striatal neuronal loss or dysfunction and choline rise in children with attention-deficit hyperactivity disorder: a 1H-magnetic resonance spectroscopy study. Neurosci Lett , 215 (1-2), 45-48.

Page 111: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

98

Kanai, Y., Clémençon, B., Simonin, A., Leuenberger, M., Lochner, M., Weisstanner, M., et al. (2013). The SLC1 high-affinity glutamate and neutral amino acid transporter family. Mol Aspects Med , 34 (2-3), 108-120.

Karagiannidis, I., Rizzo, R., Tarnok, Z., Wolanczyk, T., Hebebrand, J., Nöthen, M. M., et al. (2012). Replication of association between a SLITRK1 haplotype and Tourette Syndrome in a large sample of families. Mol Psychiatry , 17 (7), 665-668.

Karlsson, R. M., Tanaka, K., Heilig, M., & Holmes, A. (2008). Loss of glial glutamate and aspartate transporter (excitatory amino acid transporter 1) causes locomotor hyperactivity and exaggerated responses to psychotomimetics: rescue by haloperidol and metabotropic glutamate 2/3 agonist. Biol Psychiatry , 64 (9), 810-814.

Karlsson, R. M., Tanaka, K., Heilig, M., & Holmes, A. (2008). Loss of glial glutamate and aspartate transporter (excitatory amino acid transporter 1) causes locomotor hyperactivity and exaggerated responses to psychotomimetics: rescue by haloperidol and metabotropic glutamate 2/3 agonist. Biol Psychiatry , 64 (9), 810-814.

Karno, M., Golding, J. M., Sorenson, S. B., & Burnham, M. A. (1988). The epidemiology of obsessive-compulsive disorder in five U.S. communities. Arch Gen Psychiatry , 45, 1084-1099.

Kaufman, J., Birmaher, B., Brent, D., Rao, U., & Ryan, N. (1995). Kiddie- SADS-Lifetime Version (K-SADS-PL) . Pittsburgh: University of Pittsburgh.

Kidd, K. K., & Pauls, D. L. (1982). Genetic hypotheses for Tourette syndrome. Adv Neurol , 35, 243-249.

Kidd, K. K., Prusoff, B. A., & Cohen, D. J. (1980). Familial pattern of Gilles de la Tourette syndrome. Arch Gen Psychiatry , 37, 1336-1339.

Knell, E. R., & Comings, D. E. (1993). Tourette's syndrome and attention-deficit hyperactivity disorder: evidence for a genetic relationship. J Clin Psychiatry , 54 (9), 331-337.

Knight, S., & et al. (2010). Linkage analysis of Tourette syndrome in a large Utah pedigree. Am J Med Genet B Neuropsychiatr Genet , 153B (2), 656-662.

Kouzarides, T. (2007). Chromatin modifications and their function. Cell , 128 (4), 693-705.

Laurin, N., Lee, J., Ickowicz, A., Pathare, T., Malone, M., Tannock, R., et al. (2008). Association Study for Genes at Chromosome 5p13-q11 in Attention Deficit Hyperactivity Disorder. Am J Med Genet B Neuropsychiatr Genet , 147B, 600-605.

Laurin, N., Wigg, K. G., Feng, Y., Sandor, P., & Barr, C. L. (2009). Chromosome 5 and Gilles de la Tourette syndrome: Linkage in a large pedigree and association study of six candidates in the region. Am J Med Genet B Neuropsychiatr Genet , 150B (1), 95-103.

Leckman, J. F., Bloch, M. H., Smith, M. E., Larabi, D., & Hampson, M. (2010). Neurobiological substrates of Tourette's disorder. J Child Adolesc Psychopharmacol , 20 (4), 237-247.

Page 112: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

99

Leckman, J. F., Grice, D. E., Barr, L. C., de Vries, A. L., Martin, C., Cohen, D. J., et al. (1994-1995). Tic-related vs. non-tic-related obsessive compulsive disorder. Anxiety , 1 (5), 208-215.

Leckman, J. F., Pauls, D. L., Zhang, H., Rosario-Campos, M. C., Katsovich, L., Kidd, K. K., et al. (2003). Obsessive-compulsive symptom dimensions in affected sibling pairs diagnosed with Gilles de la Tourette syndrome. Am J Med Genet B Neuropsychiatr Genet , 116B (1), 60-68.

Leckman, J. F., Riddle, M. A., Hardin, M. T., Ort, S. I., Swartz, K. L., Stevenson, J., et al. (1989). The Yale Global Tic Severity Scale: initial testing of a clinican-rated scale of tic severity. J Am Acad Child Adolesc Psychiatry , 28, 566-573.

Lee, B. K., & et al. (2012). Cell-type specific and combinatorial usage of diverse transcription factors revealed by genome-wide binding studies in multiple human cells. Genome Res , 22 (1), 9-24.

Lehohla, M., Kellaway, L., & Russell, V. A. (2004). NMDA receptor function in the prefrontal cortex of a rat model for attention-deficit hyperactivity disorder. Metab Brain Dis , 19 (1-2), 35-42.

Lencz, T., & et al. (2013). Genome-wide association study implicates NDST3 in schizophrenia and bipolar disorder. Nat Commun , 4 (2739), 1-10.

Lewontin, R. C. (1964). The interaction of selection and linkage. I. General consideration; Heterotic models. Genetics , 49 (1), 49-67.

Lin, C. I., Orlov, I., Ruggiero, A. M., Dykes-Hoberg, M., Lee, A., Jackson, M., et al. (2001). Modulation of the neuronal glutamate transporter EAAC1 by the interacting protein GTRAP3-18. Nature , 410 (6824), 84-88.

Lin, P., Vance, J. M., Pericak-Vance, M. A., & Martin, E. R. (2007). No Gene Is an island: The Flip-Flop Phenomenon. Am J Hum Genet , 80, 531-538.

Lombroso, P. J., & Scahill, L. (2008). Tourette syndrome and obsessive-compulsive disorder. Brain Dev , 30 (4), 231-237.

Ludolph, A. G., Roessner, V., Münchau, A., & Müller-Vahl, K. (2012). Tourette syndrome and other tic disorders in childhood, adolescents and adulthood. Dtsch Arztebl Int , 109 (48), 821-828.

Mühleisen, T. W., Leber, M., Schulze, T. G., & Strohmaier, J. (2014). Genome-wide association study reveals two new risk loci for bipolar disorder. Nat Commun , 5 (3339).

Macmaster, F. P., & Rosenberg, D. R. (2010). Glutamate and the Treatment of Obsessive-Compulsive Disorder. Psychopharm Rev , 45 (5), 33-40.

MacMaster, F. P., Carrey, N., Sparkes, S., & Kusumakar, V. (2003). Proton spectroscopy in medication-free pediatric attention-deficit/hyperactivity disorder. Biol Psychiatry , 53 (2), 184-187.

Page 113: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

100

MacMaster, F. P., O'Neill, J., & Rosenberg, D. R. (2008). Brain imaging in pediatric obsessive compulsive disorder. J Am Aacd Child Adolesc Psychiatry , 47 (11), 1262-1272.

Maltezos, S., Horder, J., Coghlan, S., Skirrow, C., O'Gorman, R., Lavender, T. J., et al. (2014). Glutamate/glutamine and neuronal integrity in adults with ADHD: a proton MRS study. Transl Psychiatry , 4 (e373), 1-8.

Manolio, T. A. (2013). Bringing genome-wide association findings into clinical use. Nat Rev Genet , 14 (8), 549-558.

Manolio, T. A., Brooks, L. D., & Collins, F. S. (2008). A HapMap harvest of insights into the genetics of common disease. J Clin Invest , 118 (5), 1590-1605.

Maragakis, N. J., & Rothstein, J. D. (2004). Glutamate transporters: animal models to neurologic disease. Neurobiol Dis , 15 (3), 461-473.

Mathews, C. A., & Grados, M. A. (2011). Familiality of Tourette syndrome, obsessive-compulsive disorder, attention deficit/hyperactivity disorder: heritability analysis in a large sib-pair sample. J Am Acad Child Adolesc Psychiatry , 50 (1), 46-54.

Mathews, C. A., Jan, K. L., Herrera, L. D., Lowe, T. L., Budman, C. L., Erenberg, G., et al. (2007). Tic symptom profiles in subjects with Tourette syndrome from two genetically isolated populations. Biol Psychiatry , 61 (3), 292-300.

McCarthy, M. I., & Hirschhorn, J. N. (2008). Genome-wide association studies: past, present and future. Hum Mol Genet , 15 (17R(2)), R100-101.

McDonald, M. L., & et al. (2014, May 13). Common Genetic Variants Associated with Resting Oxygenation in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol .

McGrath, M. J., Campbell, K. M., Parks, C. R., & Burton, F. H. (2000). Glutamatergic drugs exacerbate symptomatic behavior in a transgenic model of comorbid Tourette's syndrome and obsessive-compulsive disorder. Brain , 877 (1), 23-30.

McNaught, K. S., & Mink, J. W. (2011). Advances in understanding and treatment of Tourette syndrome. Nat Rev Neurol , 7, 667-676.

Mérette, C., & et al. (2000). Significant linkage for Tourette syndrome in a large French Canadian family. Am J Hum Genet , 67 (4), 1008-1013.

Miller, E. M., Pomerleau, F., Huettl, P., Gerhardt, G. A., & Glaser, P. E. (2014). Aberrant glutamate signaling in the prefrontal cortex and striatum of the spontaneously hypertensive rat model of attention-deficit/hyperactivity disorder. Psychopharmacology .

Miller, S. A., Dykes, D. D., & Polesky, H. F. (1988). A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res , 16, 1215.

Miranda, D. M., Wigg, K., Kabia, E. M., Feng, Y., Sandor, P., & Barr, C. L. (2009). Association of SLITRK1 to Gilles de la Tourette syndrome. Am J Med Genet B Neuropsychiatr Genet , 150B (4), 483-486.

Page 114: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

101

Mourad, R., Sinoquet, C., Dina, C., & Leray, P. (2011). Visualization of pairwise and multilocus linkage disequilibrium structure using latent forests. PLoS One , 6 (12).

Mourad, R., Sinoquet, C., Dina, C., & Leray, P. (2011). Visualization of Pairwise and Multilocus Linkage Disequilibrium Structures using Latent Forests. PLoS ONE , 6 (12), e27320.

Mueller, J. C. (2004). Linkage diseqilibrium for different scales and applications. Brief Bioinform , 5 (4), 355-364.

Murphy, T., & Muter, V. (2012). Risk factors for comorbidity in ADHD and GTS: looking beyond a single-deficit model. Appl Neuropsychol Child , 1 (2), 129-136.

Neale, B. M., et al. (2008). Genome-wide Association Scan of Attention Deficit Hyperactivity Disorder. Am J Med Genet B Neuropsychiatr Genet , 147B (8), 1337-1344.

Neale, B. M., Medland, S. E., Ripke, S., Asherson, P., Franke, B., Lesch, K.-P., et al. (2010). Meta-analysis of genome-wide association studies of attention deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry , 49 (9), 884-897.

Nicodemus, K. K., Luna, A., & Shugart, Y. Y. (2007). An evaluation of power and type 1 error of single nucleotide transmission/disequilibrium-based statistical methods under different family structures, missing parental data, and population stratification. Am J Hum Genet , 80 (1), 178-185.

Nicodemus, K. K., Luna, A., & Shugart, Y. Y. (2007). An evaluation of power and type I error of single-nucleotide polymorphism transmission/disequilibrium-based statistical methods under different family structures, missing parental data, and population stratification. Am J Hum Genet , 80 (1), 178-185.

Nobrega, M. A., Ovcharenko, I., Afzal, V., & Rubin, E. M. (2003). Scanning human gene deserts for long-range enhancers. Science , 302 (5644), 413.

Nordstrom, E. J. (2002). A transgenic model of comorbid Tourette's syndrome and obsessive-compulsive disorder circuitry. Mol Psychiatry , 7 (6), 617-625.

Nyholt, D. R. (2004). A simple correction for multiple testing for single-nucleotide polymorphisms in linkage disequilibrium with each other. Am J Hum Genet , 74 (4), 765-769.

Ogdie, M. N., Bakker, S. C., Fisher, S. E., Francks, C., Yang, M. H., Cantor, R. M., et al. (2006). Pooled genome-wide linkage data on 424 ADHD ASPs suggests genetic heterogeneity and a common risk locus at 5p13. Mol Psychiatry , 11 (1), 5-8.

Ogdie, M. N., Fisher, S. E., Yang, M., Ishii, J., Francks, C., Loo, S. K., et al. (2004). Attention deficit hyperactivity disorder: fine mapping supports linkage to 5p13, 6q12, 16p13, and 17p11. Am J Hum Genet , 75 (4), 661-668.

Ogdie, M. N., Macphie, I. L., Minassian, S. L., Yang, M., Fisher, S. E., Francks, C., et al. (2003). A genomewide scan for attention-deficit/hyperactivity disorder in an extended sample: suggestive linkage on 17p11. Am J Hum Genet , 72 (5), 1268-1279.

Page 115: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

102

O'Roak, B. J., Morgan, T. M., Fishman, D. O., Saus, E., Alonso, P., Gratacòs, M., et al. (2010). Additional support for the association of SLITRK1 var321 and Tourette syndrome. Mol Psychiatry , 15 (5), 447-450.

O'Rourke, J. A., Scharf, J. M., Platko, J., Stewart, S. E., Illmann, C., Geller, D. A., et al. (2011). The familial association of tourette's disorder and ADHD: the impact of OCD symptoms. Am J Med Genet B Neuropsychiatr Genet , 156B (5), 553-560.

Ozawa, S., Kamiya, H., & Tsuzuki, K. (1998). Glutamate receptors in the mammalian central nervous system. Prog Neurobiol , 54 (5), 581-618.

Packer, L. E. (2005). Tic-related school problems: impact on functioning, accommadations, and interventions. Behav Modif , 29 (6), 876-899.

Paschou, P. (2013). The genetic basis of Gilles de la Tourette syndrome. Neurosci Biobehav Rev , 1-14.

Paschou, P., & et al. (2004). Indications of linkage and association of Gilles de la Tourette syndrome in two independent family samples: 17q25 is a putative susceptibility region. Am J Hum Genet , 75 (4), 545-560.

Pasquini, M., & Biondi, M. (2006). Memantine augmentation for refractory obsessive-compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry , 30 (6), 1173-1175.

Patterson, N., Price, A. L., & Reich, D. (2006). Population structure and eigenanalysis. PLoS Genet , 2 (12), 2074-2093.

Pauls, D. L. (2010). The genetics of obsessive-compulsive disorder: a review. Dialogues Clin Neurosci , 12 (2), 149-163.

Pauls, D. L., & Leckman, J. F. (1986). The inheritance of Gilles de la Tourette's syndrome and associated behaviors. Evidence for autosomal dominant transmission. N Engl J Med , 315 (16), 993-997.

Pauls, D. L., Alsobrook, J. P., Goodman, W., Rasmussen, S., & Leckman, J. F. (1995). A family study of obsessive-compulsive disorder. Am J Psychiatry , 152 (1), 76-84.

Pauls, D. L., Cohen, D. J., Heimbuch, R., Detlor, J., & Kidd, K. K. (1981). Familial pattern and transmission of Gilles de la Tourette syndrome and multiple tics. Arch Gen Psychiatry , 38 (10), 1091-1093.

Pauls, D. L., Kruger, S. D., Leckman, J. F., Cohen, D. J., & Kidd, K. K. (1984). The risk of Tourette's syndrome and chronic multiple tics among relatives of Tourette syndrome patients obtained by direct interview. J Am Acad Child Psychiatry , 23 (2), 134-137.

Pauls, D. L., Leckman, J. F., & Cohen, D. J. (1994). Evidence against a genetic relationship between Tourette's syndrome and anxiety, depression, panic and phobic disorders. Br J Psychiatry , 164 (2), 215-221.

Page 116: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

103

Pauls, D. L., Leckman, J. F., & Cohen, D. J. (1993). Familial relationship between Gilles de la Tourette's syndrome, attention deficit disorder, learning disabilities, speech disorders, and stuttering. J Am Acad Child Adolesc Psychiatry , 32 (5), 1044-1050.

Pauls, D. L., Pakstis, A. J., Kurlan, R., Kidd, K. K., Leckman, J. F., Cohen, D. J., et al. (1990). Segregation and linkage analyses of Tourette's syndrome and related disorders. Acad Child Adolesc Psychiatry , 29 (2), 195-203.

Pauls, D. L., Raymond, C. L., Stevenson, J. M., & Leckman, J. F. (1991). A family study of Gilles de la Tourette syndrome. Am J Hum Genet , 48 (1), 154-163.

Pauls, D. L., Towbin, K. E., Leckman, J. F., Zahner, G. E., & Cohen, D. J. (1986). Gilles de la Tourette's syndrome and obsessive-compulsive disorder. Evidence supporting a genetic relationship. Arch Gen Psychiatry , 43 (12), 1180-1182.

Peghini, P., Janzen, J., & Stoffel, W. (1997). Glutamate transporter EAAC-1-deficient mice develop dicarboxylic aminoaciduria and behavioral abnormalities but no neurodegeneration. EMBO J , 16 (13), 3822-3832.

Perlov, E., Philipsen, A., Hesslinger, B., Buechert, M., Ahrendts, J., Feige, B., et al. (2007). Reduced cingulate glutamate/glutamine-to-creatine ratios in adult patients with attention deficit/hyperactivity disorder -- a magnet resonance spectroscopy study. J Psychiatr Res , 41 (11), 934-941.

Perlov, E., Philipsen, A., Matthies, S., Drieling, T., Maier, S., Bubl, E., et al. (2009). Spectroscopic findings in attention-deficit/hyperactivity disorder: review and meta-analysis. World J Biol Psychiatry , 10 (4 Pt 2), 355-365.

Perlov, E., Tebarzt van Elst, L., Buechert, M., Maier, S., Matthies, S., Ebert, D., et al. (2010). H¹-MR-spectroscopy of cerebellum in adult attention deficit/hyperactivity disorder. J Psychiatr Res , 44 (14), 938-943.

Peterson, B. S., Leckman, J. F., Tucker, D., Scahill, L., Staib, L., Zhang, H., et al. (2000). Preliminary findings of antistreptococcal antibody titers and basal ganglia volumes in tic, obsessive-compulsive, and attention deficit/hyperactivity disorders. Arch Gen Psychiatry , 57 (4), 364-372.

Peterson, B. S., Thomas, P., Kane, M. J., Scahill, L., Zhang, H., Bronen, R., et al. (2003). Basal Ganglia volumes in patients with Gilles de la Tourette syndrome. Arch Gen Psychiatry , 60 (4), 415-424.

Phillips, T., & Hoopes, L. (2008). Transcription factors and transcriptional control in eukaryotic cells. Nat Edu , 1 (1), 119.

Pitman, R. K., Green, R. C., Jenike, M. A., & Mesulam, M. M. (1987). Clinical comparison of Tourette's disorder and obsessive-compulsive disorder. Am J Psychiatry , 144 (9), 1166-1171.

Page 117: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

104

Pittenger, C., Krystal, J. H., & Coric, V. (2006). Glutamate-modulating drugs as novel pharmacotherapeutic agents in the treatment of obsessive-compulsive disorder. NeuroRx , 3 (1), 69-81.

Plessen, K. J. (2013). Tic disorders and Tourette's syndrome. Eur Child Adolesc Psychiatry , 22 Suppl, S55-60.

Poelmans, G., Pauls, D. L., Buitelaar, J. K., & Franke, B. (2011). Integrated genome-wide association study findings: identification of a neurodevelopmental network for attention deficit hyperactivity disorder. Am J Psychiatry , 168 (4), 365-377.

Polanczyk, G., & Rohde, L. A. (2007). Epidemiology of attention-deficit/hyperactivity disorder across the lifespan. Curr Opin Psychiatry , 20 (4), 386-392.

Polanczyk, G., de Lima, M. S., Horta, B. L., Biederman, J., & Rohde, L. A. (2007). The worldwide prevalence of ADHD: a systematic review and metaregression analysis. Am J Psychiatry , 164 (6), 942-948.

Portera-Cailliau, C., Price, D. L., & Martin, L. J. (1997). Excitotoxic neuronal death in the immature brain is an apoptosis-necrosis morphological continuum. J Comp Neurol , 378 (1), 70-87.

Porton, B., Greenberg, B. D., Askland, K., Serra, L. M., Gesmonde, J., Rudnick, G., et al. (2013). Isoforms of the neuronal glutamate transporter gene, SLC1A1/EAAC1, negatively modulate glutamate uptake: relevance to obsessive-compulsive disorder. Transl Psychiatry , 3, 1-10.

Poyurovsky, M., Weizman, R., Weizman, A., & Koran, L. (2005). Memantine for treatment-resistant OCD. Am J Psychiatry , 162 (11), 2191-2192.

Price, A. L., Patterson, N. J., Plenge, R. M., Weinblatt, M. E., Shadick, N. A., & Reich, D. (2006). Principal components analysis corrects for stratification in genome-wide association studies. Nature Genetics , 38 (8), 904-909.

Price, R. A., Kidd, K. K., Cohen, D. J., Pauls, D. L., & Leckman, J. F. (1985). A twin study of Tourette syndrome. Arch Gen Psychiatry , 42 (8), 815-820.

Pringsheim, T., & Hammer, T. (2013). Social behavior and comorbidity in children with tics. Pediatr Neurol , 49 (6), 406-410.

Pringsheim, T., Lang, A., Kurlan, R., Pearce, M., & Sandor, P. (2009). Understanding disability in Tourette syndrome. Dev Med Child Neurol , 51 (6), 468-472.

Pritchard, J. K., & Przeworski, M. (2001). Linkage disequilibrium in humans: models and data. Am J Hum Genet , 69 (1), 1-14.

Pujol, J., Soriano-Mas, C., Alonso, P., Cardoner, N., Menchón, J. M., Deus, J., et al. (2004). Mapping structural brain alterations in obsessive-compulsive disorder. Arch Gen Psychiatry , 61 (7), 720-730.

Page 118: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

105

Purcell, S., Cherny, S. S., & Sham, P. C. (2003). Genetic Power Calculator: design of linkage and association genetic mapping studies of complex traits. Bioinformatics , 19 (1), 149-150.

Purves, D., Augustine, G. J., Fitzpatrick, D., & et al. editors. (2001). Neuroscience. 2nd edition. Sunderland (MA): Sinauer Associates.

Rauen, T., Taylor, W. R., Kuhlbrodt, K., & Wiessner, M. (1998). High-affinity glutamate transporters in the rat retina: a major role of the glial glutamate transporter GLAST-1 in transmitter clearance. Cell Tissue Res , 291 (1), 19-31.

Reich, D. E., Cargill, M., Bolk, S., Ireland, J., Sabeti, P. C., Richter, D. J., et al. (2001). Linkage Disequilibrium in the human genome. Nature , 411 (6834), 199-204.

Riedel, G., Platt, B., & Micheau, J. (2003). Glutamate receptor function in learning and memory. Behav Brain Res , 140 (1-2).

Ringnér, M. (2008). What is principal component analysis? Nature Biotechnology , 26, 303-304.

Rizzo, R., Curatolo, P., Gulisano, M., Virzì, M., Arpino, C., & Robertson, M. M. (2007). Disentangling the effects of Tourette syndrome and attention deficit hyperactivity disorder on cognitive and behavioral phenotypes. Brain Dev , 29 (7), 413-420.

Rizzo, R., Gulisano, M., Calì, P. V., & Curatolo, P. (2012). Long term clinical course of Tourette syndrome. Brain Dev , 34 (8), 667-673.

Rizzo, R., Gulisano, M., Calì, P. V., & Curatolo, P. (2013). Tourette syndrome and comorbid ADHD: Current pharmacological treatment options. Eur J Paediatr Neurol , 17 (5), 421-428.

Robertson, M. M. (2012). The Gille de la Tourette syndrome: the current status. Arch Dis Child Educ Pract Ed , 97 (5), 166-175.

Robertson, M. M. (2008). The prevalence and epidemiology of Gilles de la Tourette syndrome. Part 1: the epidemiology and prevalence studies. J Psychosom Res , 65 (5), 461-472.

Robertson, M. M., & Cavanna, A. E. (2007). The Gilles de la Tourette syndrome: a principal component factor analysis study of a large pedigree. Psychiatr Genet , 17 (3), 143-152.

Robertson, M. M., & Gourdie, A. (1990). Familial Tourette's syndrome in a large British pedigree. Associated psychopathology, severity, and potential for linkage analysis. Br J Psychiatry , 156, 515-521.

Robertson, M. M., Althoff, R. R., Hafez, A., & Pauls, D. L. (2008). Principal components analysis of a large cohort with Tourette syndrome. Br J Psychiatry , 193 (1), 31-36.

Rosenberg, D. R., & Keshavan, M. S. (1998). A.E. Bennett Research Award. Toward a neurodevelopmental model of of obsessive--compulsive disorder. Biol Psychiatry , 43 (9), 623-640.

Page 119: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

106

Rosenberg, D. R., MacMaster, F. P., Keshavan, M. S., Fitzgerald, K. D., Stewart, C. M., & Moore, G. J. (2000). Decrease in caudate glutamatergic concentrations in pediatric obsessive-compulsive disorder patients taking paroxetine. J Am Acad Child Adolesc Psychiatry , 39 (9), 1096-1103.

Rothstein, J. D., & et al. (1994). Localization of neuronal and glial glutamate transporters. Neuron , 13 (3), 713-725.

Rothstein, J. D., Dykes-Hoberg, M., Pardo, C. A., Bristol, L. A., Jin, L., Kuncl, R. W., et al. (1996). Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron , 16 (3), 675-686. (Ruscio, Stein, Chiu, & Kessler, The epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey Replication, 2010) (Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2014)

Ruscio, A. M., Stein, D. J., Chiu, W. T. & Kessler, R. C. (2010). The epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey Replication. Mol Psychiatry, 15(1), 53-63.

Sagvolden, T., Russell, V. A., Aase, H., Johansen, E. B., & Farshbaf, M. (2005). Rodent models of attention-deficit/hyperactivity disorder. Biol Psychiatry , 57 (11), 1239-1247.

Samuels, J., Shugart, Y. Y., Grados, M. A., Willour, V. L., Bienvenu, O. J., & Greenberg, B. D. (2007). Significant linkage to compulsive hoarding on chromosome 14 in families with obsessive-compulsive disorder: results from the OCD Collaborative Genetics Study. Am J Psychiatry , 164 (3), 493-499.

Samuels, J., Wang, Y., Riddle, M. A., Greenberg, B. D., Fyer, A. J., McCracken, J. T., et al. (2011). Comprehensive family-based association study of the glutamate transporter gene SLC1A1 in obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet , 156B (4), 472-477.

Saxena, S., & Rauch, S. L. (2000). Functional neuroimaging and the neuroanatomy of obsessive-compulsive disorder. Psychiatr Clin North Am , 23 (3), 563-586.

Schanze, D., Ekici, A. B., Gawlik, M., Pfuhlmann, B., Reis, A., Stober, G. (2011). Evaluation of risk loci for schizophrenia derived from genome-wide association studies in a German population. Am J Med Genet B Neuropsychiatr Genet, 156(2), 198-203.

Scharf, J. M. (2012). Genome-wide association study of Tourette's syndrome. Molecular Psychiatry , 1-8.

Scharf, J. M., & Pauls, D. L. (2007). Principles and Practices of Medical Genetics (5th ed.). New York: Elsevier.

Scharf, J. M., Miller, L. L., Mathews, C. A., & Ben-Shlomo, Y. (2012). Prevalence of Tourette syndrome and chronic tics in the population-based Avon longitudinal study of parents and children cohort. J Am Acad Child Adolesc Psychiatry , 51 (2), 192-201.

Page 120: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

107

Schaub, M. A., Boyle, A. P., Kundaje, A., Batzoglou, S., & Snyder, M. (2012). Linking disease associations with regulatory information in the human genome. Genome Res , 22 (9), 1748-1759.

Schizophrenia Psychiatric Genome-Wide Association Study Consortium. (2011). Genome-wide association study identifies five new schizophrenia loci. Nat Genet , 43 (10), 969-976.

Schizophrenia Working Group of the Psychiatric Consortium. (2014). Biological insights from 108 schizophrenia- associated genetic loci. Nature, 511, 421-426.

Schorck, A. J., Thompson, W. K., Pham, P., et al. (2013). All SNPs are not created equal: genome-wide association studies reveal a consistent pattern of enrichment among functionally annotated SNPs. PLoS Genet , 9 (4), e1003449.

Scimemi, A., Tian, H., & Diamond, J. S. (2009). Neuronal transporters regulate glutamate clearance, NMDA receptor activation, and synaptic plasticity in the hippocampus. J Neurosci , 29 (46), 14581-14595.

Seuchter, S. A., Hebebrand, J., Klug, B., Knapp, M., Lehmkuhl, G., Poustka, F., et al. (2000). Complex segregation analysis of families ascertained through Gilles de la Tourette syndrome. Genet Epidemiol , 18 (1), 33-47.

Sham, P. C., & Curtis, D. (1995). An extended transmission/disequilibrium test (TDT) for multi-allele marker loci. Ann Hum Genet , 59 (Pt 3), 323-336.

Sham, P. C., & Purcell, S. M. (2014). Statistical power and significance testing in large-scale genetic studies. Nat Rev Genet , 15 (5), 335-346.

Sheppard, D. M., Bradshaw, J. L., Purcell, R., & Pantelis, C. (1999). Tourette's and comorbid syndromes: obsessive compulsive and attention deficit hyperactivity disorder. A common etiology? Clin Psychol Rev , 19 (5), 531-552.

Shih, M. C., & Whittemore, A. S. (2001). Allele-sharing among affected relatives: non-parametric methods for identifying genes. Stat Methods Med Res , 10 (1), 27-55.

Shugart, Y. Y., Samuels, J., Willour, V. L., Grados, M. A., Greenberg, B. D., Knowles, J. A., et al. (2006). Genomewide linkage scan for obsessive-compulsive disorder: evidence for susceptibility loci on chromosomes 3q, 7p, 1q, 15q, and 6q. Mol Psychiatry , 11 (8), 763-770.

Shugart, Y. Y., Wang, Y., Samuels, J. F., Grados, M. A., Greenberg, B. D., Knowles, J. A., et al. (2009). A family-based association study of the glutamate transporter gene SLC1A1 in obsessive-compulsive disorder in 378 families. Am J Med Genet B Neuropsychiatr Genet , 150B (6), 886-892.

Simonic, I., Gericke, G. S., Ott, J., & Weber, J. L. (1998). Identification of genetic markers associated with Gilles de la Tourette syndrome in an Afrikaner population. Am J Hum Genet , 63 (3), 839-846.

Page 121: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

108

Singer, H. S., Morris, C., & Grados, M. (2010). Glutamatergic modulatory therapty for Tourette syndrome. Med Hypotheses , 74 (5), 862-867.

Singh, D. N., Howe, G. L., Jordan, H. W., & Hara, S. (1982). Tourette's syndrome in a black woman with associated triple X and 9p mosaicism. J Natl Med Assoc , 74 (7), 675-682.

Siniscalco, D., Cirillo, A., Bradstreet, J. J., & Antonucci, N. (2013). Epigenetic findings in autism: new perspectives for therapy. Int J Environ Res Public Health , 10 (9), 4261-4273.

Sowell, E. R., Kan, E., Yoshii, J., Thompson, P. M., Bansal, R., Xu, D., et al. (2008, Jun). Thinning of sensorimotor cortices in children with Tourette syndrome. Nat Neurosci , 637-639.

Spencer, T., Biederman, J., Harding, M., O'Donnell, D., Wilens, T., Faraone, S., et al. (1998). Disentangling the overlap between Tourette's disorder and ADHD. J Child Psychol Psychiatry , 39 (7), 1037-1044.

Spicuglia, S., & Vanhille, L. (2012). Chromatin signatures of active enhancers. Nucleus , 3 (2), 126-131.

Spielman, R. S., McGinnis, R. E., & Ewens, W. J. (1993). Transmission test for linkage disequilibrium: the insulin gene region and insulin-dependent diabetes mellitus (IDDM). Am J Hum Genet , 52 (3), 506-516.

Spitzer, R., Williams, J., Gibbon, M., & First, M. (1992). The Structured Clinical Interview for DSM-III-R (SCID). I. History, rationale, and description. Arch Gen Psychiat , 49, 624-629.

Stahl, S. M. (2009). The prefrontal cottex is out of tune in attention-deficit/hyperactivity disorder. J Clin Psychiatry , 70 (7), 950-951.

Starck, G., Ljungberg, M., Nilsson, M., et al. (2008). A 1H magnetic resonance spectroscopy study in adults with obsessive compulsive disorder: relationship between metabolite concentrations and symptom severity. J Neural Transm , 115 (7), 1051-1062.

Stern, E. R., Blair, C., & Peterson, B. S. (2008). Inhibitory deficits in Tourette's syndrome. Dev Psychobiol , 50 (1), 9-18.

Stewart, S. E., Fagerness, J. A., Platko, J., Smoller, J. W., Scharf, J. M., Illmann, C., et al. (2007). Association of the SLC1A1 glutamate transporter gene and obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet , 144B (8), 1027-1033.

Stewart, S. E., Illmann, C., Geller, D. A., Leckman, J. F., King, R., & Pauls, D. L. (2006). A controlled family study of attention-deficit/hyperactivity disorder and Tourette's disorder. J Am Acad Child Adolesc Psychiatry , 45 (11), 1354-1362.

Stewart, S. E., Mayerfeld, C., Arnold, P. D., Crane, J. R., O'Dushlaine, C., & et al. (2013). Meta-analysis of association between obsessive compulsive disorder and the 3' region of neuronal glutamate transporter gene SLC1A1. Am J Med Genet B Neuropsychiatr Genet , 162B (4), 367-379.

Page 122: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

109

Stewart, S. E., Yu, D., Scharf, J. M., Neale, B. M., & et al. (2013). Genome-wide association study of obsessive-compulsive disorder. Molecular Psychiatry , 18 (7), 788-798.

Stillman, A. A., Krsnik, Z., Sun, J., Rasin, M. R., State, M. W., Sestan, N., et al. (2009). Developmentally regulated and evoltionarily conserved expression of SLITRK1 in brain circuits implicated in Tourette syndrome. J Comp Neurol , 513 (1), 21-37.

Strahl, B. D., & Allis, C. D. (2000). The language of covalent histone modifications. Nature , 403 (6765), 41-45.

Struhl, K., & Moqtaderi, Z. (1998). The TAFs in the HAT. Cell , 94 (1), 1-4.

Sullivan, P. (2012). Don't give up on GWAS. Mol Psychiatry , 17 (1), 2-3.

Sutherland, M. L., Delaney, T. A., & Noebels, J. L. (1996). Glutamate transporter mRNA expression in proliferative zones of the developing and adult murine CNS. J Neurosci , 16 (7), 2191-2207.

Swerdlow, N. R., & Sutherland, A. N. (2005). Using animal models to develop therapeutics for Tourette syndrome. Pharmacol Ther , 108 (3), 281-293.

Tanaka, K., Watase, K., Manabe, T., Yamada, K., Watanabe, M., Takahashi, K., et al. (1997). Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter GLT-1. Science , 276 (5319), 1699-1702.

Tannock, R., Hum, M., Masellis, M., Humphries, T., & Schachar, R. (2002). Interview for Children's Academic Performance, Attention, Behaviour and Learning: DSM-IV Version (TTI-IV). Unpublished document .

Taylor, L. D., Krizman, D. B., Jankovic, J., Hayani, A., Steuber, P. C., Greenberg, F., et al. (1991). 9p monosomy in a patient with Gilles de la Tourette's syndrome. Neurology , 41 (9), 1513-1515.

Teare, M. D., & Barrett, J. H. (2005). Genetic linkage studies. Lancet , 366 (9490), 1036-1044.

The ENCODE Project Consortium. (2012). An integrated encyclopedia of DNA elements in the human genome. Nature , 489, 57-74.

The Tourette Syndrome Association International Consortium for Genetics. (1999). A Complete Genome Screen in Sib Pairs Affected by Gilles de la Tourette Syndrome . Am J Hum Genet , 65, 1428-1436.

The Tourette Syndrome Association International Consortium for Genetics. (2007). Genome Scan for Tourette Disorder in Affected-Sibling-Pair and Multigenerational Families. Am J Hum Genet , 80, 265-272.

Thomas, G., et al. (2008). Multiple loci identified in a genome-wide association study of prostate cancer. Nat Genet , 40 (3), 310-315.

Page 123: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

110

Turgay, A., Goodman, D. W., Asherson, P., Lasser, R. A., Babcock, T. F., Pucci, M. L., et al. (2012). Lifespan persistence of ADHD: the life transition model and its application. J Clin Psychiatry , 73 (2), 192-201.

Turic, D., Langley, K., Williams, H., Norton, N., Williams, N. M., Moskvina, V., et al. (2005). A family based study implicates solute carrier family 1-member 3 (SLC1A3) gene in attention-deficit/hyperactivity disorder. Biol Psychiatry , 57 (11), 1461-1466.

Tzingounis, A. V., & Wadiche, J. I. (2007). Glutamate transporters: confining runaway excitation by shaping synaptic transmission. Nat Rev Neurosci , 8 (12), 935-947.

Udvardi, P. T., Nespoli, E., Rizzo, F., Hengerer, B., & Ludolph, A. G. (2013). Nondopaminergic neurotranmission in the pathophysiology of Tourette syndrome. Int Rev Neurobiol , 112, 95-130.

van Grootheest, D. S., Cath, D. C., Beekman, A. T., & Boomsma, D. I. (2005). Twin studies on obsessive-compulsive disorder: a review. Twin Res Hum Genet , 8 (5), 450-458.

Vaquerizas, J. M., Kummerfeld, S. K., Teichmann, S. A., & Luscombe, N. M. (2009). A census of human transcription factors: function, expression and evolution. Nat Rev Genet , 10 (4), 252-263.

Veenstra-VanderWeele, J., Kim, S. J., Gonen, D., Hanna, G. L., Leventhal, B. L., & Cook, E. H. (2001). Genomic organization of the SLC1A1/EAAC1 gene and mutation screening in early-onset obsessive-compulsive disorder. Mol Psychiatry , 6 (2), 160-167.

Veenstra-VanderWeele, J., Kim, S. J., Gonen, D., Hanna, G. L., Leventhal, B. L., & Cook, E. H. (2001). Genomic organization of the SLC1A1/EAAC1 gene and mutation screening in early-onset obsessive-compulsive disorder. Mol Psychiatry , 6 (2), 160-167.

Veenstra-VanderWeele, J., Xu, T., Ruggiero, A. M., Anderson, L. R., Jones, S. T., Himle, J. A., et al. (2012). Functional studies and rare variant screening of SLC1A1/EAAC1 in males with obsessive-compulsive disorder. Psychiatr Genet , 22 (5), 256-260.

Vennstra-VanderWeele, J., Xu, T., Ruggiero, A. M., Anderson, L. R., Jones, S. T., & et al. (2012). Functional studies and rare variant screening of SLC1A1/EAAC1 in males with obsessive-compulsive disorder. Psychiatr Genet , 22 (5), 256-260.

Verkerk, A. J., & et al. (2006). Genetic and clinical analysis of a large Dutch Gilles de la Tourette family. Mol Psychiatry , 11 (10), 954-964.

Walkup, J. T., LaBuda, M. C., Singer, H. S., Brown, J., Riddle, M. A., & Hurko, O. (1996). Family study and segregation analysis of Tourette syndrome: evidence for a mixed model of inheritance. Am J Hum Genet , 59 (3), 684-693.

Walkup, J. T., Leckman, J. F., Price, R. A., Hardin, M., Ort, S. I., & Cohen, D. J. (1988). The relationship between obsessive-compulsive disorder and Tourette's syndrome: a twin study. Psychopharmacol Bull , 24 (3), 375-379.

Page 124: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

111

Wang, X., Cui, D., Wang, Z., Fan, Q., Xu, H., Qiu, J., et al. (2012). Cross-sectional comparison of the clinical characteristics of adults with early-onset and late-onset obsessive compulsive disorder. J Affect Disord , 136, 498-504.

Wang, X., Tomso, D. J., Liu, X., & Bell, D. A. (2005). Single nucleotide polymorphism in transcriptional regulatory regions and expression of environmentally responsive genes. Toxicol Appl Pharmacol , 207 (2 Suppl), 84-90.

Wang, Y., Adamczyk, A., Shugart, Y. Y., Samuels, J. F., Grados, M. A., & et al. (2010). A screen of SLC1A1 for OCD-related alleles. Am J Med Genet B Neuropsychiat Genet , 153B (2), 675-679.

Wang, Y., Adamczyk, A., Shugart, Y. Y., Samuels, J. F., Grados, M. A., Greenberg, B. D., et al. (2010). A screen of SLC1A1 for OCD-related alleles. Am J Med Genet B Neuropsychiatr Genet , 153B (2), 675-679.

Warton, F. L., Howells, F. M., & Russell, V. A. (2009). Increased glutamate-stimulated release of dopamine in substantia nigra of a rat model for attention-deficit/hyperactivity disorder--lack of effect of methylphenidate. Metab Brain Dis , 24 (4), 599-613.

Watase, K., Hashimoto, K., Kano, M., Yamada, K., Watanabe, M., Inoue, Y., et al. (1998). Motor discoordination and increased susceptibility to cerebellar injury in GLAST mutant mice. Eur J Neurosci , 10 (3), 976-988.

Wechsler, D. Wechsler Intelligence Scale for Children - Third Edition. San Antonio, TX: Harcourt Brace & Co.

Weeks, D. E., & Lathrop, G. M. (1995). Polygenic disease: methods for mapping complex disease traits. Trends Genet , 11 (12), 513-519.

Weissman, M. M., Bland, R. C., & Canino, G. J. (1994). The cross national epidemiology of obsessive compulsive disorder. J Clin Psychiatry , 55, 5-10.

Welch, J. M., Lu, J., Rodriguiez, R. M., et al. (2007). Cortico-striatal synaptic defects and OCD-like behaviours in Sapap3-mutant mice. Nature , 448 (7156), 894-900.

Wendland, J. R., Moya, P. R., Timpano, K. R., Anavitarte, A. P., Kruse, M. R., Wheaton, M. G., et al. (2009). A haplotype containing quantitative trait loci for SLC1A1 gene expression and its association with obsessive-compulsive disorder. Arch Gen Psychiatry , 66 (4), 408-416.

Willour, V. L., Yao Shugart, Y., Samuels, J., Grados, M., Cullen, B., Bienvenu, O. J., et al. (2004). Replication study supports evidence for linkage to 9p24 in obsessive-compulsive disorder. Am J Hum Genet , 75 (3), 508-513.

Wiste, A., et al. (2014). Bipolar polygenic loading and bipolar spectrum features in major depressive disorder. Bipolar Disord, 16(6), 608-616.

Page 125: A Genetic Association Study of Glutamate Transporter ......1.1 Introduction to Tourette syndrome The ability to think or behave without interruption is important to quality of life

112

Worbe, Y., Gerardin, E., Hartmann, A., Valabrégue, R., Chupin, M., Tremblay, L., et al. (2010). Distinct structural changes underpin clinical phenotypes in patients with Gilles de la Tourette syndrome. Brain , 133 (12), 3649-3660.

Wu, H., Wang, X., Yu, S., Wang, D., Chen, J., Jiang, K. Z., et al. (2013). Association of the candidate gene SLC1A1 and obsessive-compulsive disorder in Han Chinese population. Psychiatry Res , 209 (3), 737-739.

Wu, K., Hanna, G. L., Easter, P., Kennedy, J. L., Rosenberg, D. R., & Arnold, P. D. (2013). Glutamate system genes and brain volume alterations in pediatric obsessive-compulsive disorder: a preliminary study. Psychiatry Res , 211 (3), 214-220.

Xu, Z., & Taylor, J. A. (2009). SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies. Nucleic Acids Res , 37, 600-605.

Yeager, M., et al. (2007). Genome-wide association study of prostate cancer identifies a second risk locus at 8q24. Nat Genet , 39, 645-649.

Yoon, D. Y., Rippel, C. A., Kobets, A. J., Morris, C. M., Lee, J. E., Williams, P. N., et al. (2007). Dopaminergic polymorphisms in Tourette syndrome: association with the DAT1 gene (SLC6A3). Am J Med Genet B Neuropsychiatr Genet , 144B (5), 605-610.

Zarei, M., Mataix-Cols, D., Heyman, I., Hough, M., Doherty, J., Burge, L., et al. (2011). Changes in gray matter volume and white matter microstructure in adolescents with obsessive-compulsive disorder. Biol Psychiatry , 70 (11), 1083-1090.

Zhang, H., Leckman, J. F., Pauls, D. L., Tsai, C. P., Kidd, K. K., Campos, M. R., et al. (2002). Genomewide scan of hoarding in sib pairs in which both sibs have Gilles de la Tourette syndrome. Am J Hum Genet , 70 (4), 896-904.

Zhou, Y., & Danbolt, N. C. (2013). GABA and Glutamate Transporters in Brain. Front Endocrinol , 4 (165).

(Wiste, et al., 2014) (Schanze, Ekici, Gawlik, Pfuhlmann, Reis, & Stober, 2011)