438
Views
8
CrossRef citations to date
0
Altmetric
Review

DRD4 and DAT1 in ADHD: Functional neurobiology to pharmacogenetics

, &
Pages 61-78 | Published online: 21 May 2010

Abstract

Attention deficit/hyperactivity disorder (ADHD) is a common and potentially very impairing neuropsychiatric disorder of childhood. Statistical genetic studies of twins have shown ADHD to be highly heritable, with the combination of genes and gene by environment interactions accounting for around 80% of phenotypic variance. The initial molecular genetic studies where candidates were selected because of the efficacy of dopaminergic compounds in the treatment of ADHD were remarkably successful and provided strong evidence for the role of DRD4 and DAT1 variants in the pathogenesis of ADHD. However, the recent application of non-candidate gene strategies (eg, genome-wide association scans) has failed to identify additional genes with substantial genetic main effects, and the effects for DRD4 and DAT1 have not been replicated. This is the usual pattern observed for most other physical and mental disorders evaluated with current state-of-the-art methods. In this paper we discuss future strategies for genetic studies in ADHD, highlighting both the pitfalls and possible solutions relating to candidate gene studies, genome-wide studies, defining the phenotype, and statistical approaches.

Introduction

Attention deficit/hyperactivity disorder (ADHD) is a common psychiatric disorder of childhood affecting around 5% of the population.Citation1 It is characterized by an early onset and persistent pattern of inattention, impulsivity, and hyperactivity symptoms. The condition is associated with several comorbid disorders (oppositional defiant disorder, anxiety disorders, etc) and conditions (eg, disrupted peer and family relationships), and adverse outcomes emerging with age (eg, educational failure and antisocial behavior). Despite early onset, it is most frequently diagnosed and treated in middle childhood.Citation2

There is an overrepresentation of boys over girls by approximately 3:1.Citation3 ADHD can persist into adulthood, and increases the risk for antisocial personality disorder,Citation4 later criminality,Citation5 as well as drug and alcohol misuse.Citation6 Pharmacologic, neurobiologic, and genetic studies support the notion that ADHD has a neurodevelopmental basis with strong genetic and nongenetic components,Citation7 implicating neurotransmission dysregulation within brain circuits underpinning cognition and motivation.Citation8 Disruption of multiple neurotransmitter systems has been proposed. However, the primary focus has been on the catecholamines, dopamine (DA) and noradrenaline (NA). While other papers have focused on NA,Citation9,Citation10 our focus here is on evidence that variation and disruption of the DA system contributes to the etiology and response to treatment of ADHD.

Dopamine dysregulation in ADHD

Dopamine neurotransmission

DA is a key neurotransmitter in the biology of a wide range of brain processes.Citation11Citation13 It is central to the control of movement,Citation14 cognition,Citation15,Citation16 reward,Citation17 and emotional and motivational responses,Citation18Citation20 including the experience of pleasure and pain in response to positive and negative environmental events.Citation21Citation23 DA is synthesized from the amino acid tyrosine, which is first converted to L-dihydroxyphenylalanine, and then to DA by the enzyme dihydroxyphenylalanine decarboxylase. DA neurons are clustered in several mid brain regions that project to substantial parts of the brain via three major pathways, ie, the nigrostriatal, mesocorticolimbic, and tuberoinfundibular pathways. The nigrostriatal pathway extends from the substantia nigra to the caudate nucleus/putamen, and plays an essential role in voluntary movement.Citation24 The mesocorticolimbic pathway projects from the ventral tegmentum to the mesolimbic and mesocortical regions, and is associated with cognition, reward, and emotion processing.Citation25Citation27 The tuberoinfundibular pathway plays a role in neuronal control of the hypothalamic-pituitary endocrine system.Citation28 DA within these pathways modulates functionally and structurally segregated cortical and basal ganglia loops.Citation29Citation33 These circuits are involved in well-defined brain networks involved in the processes of attention as well as motivation, and disruption of either or both contribute to the etiology of ADHD.Citation17,Citation34 Such parallel organization is now thought to be incomplete,Citation35,Citation36 with thalamic nuclei allowing the passage of signals across different circuits.Citation37

DA is released into the synaptic cleft by action potentials via a calcium-dependent mechanism. Calcium influx triggers fusion of the neurotransmitter vesicles with the presynaptic membrane. DA is then released into the synaptic cleft from where it disperses and binds to postsynaptic receptors. Receptors bind neurotransmitter molecules and open nearby ion channels in the postsynaptic cell membrane. This alters the local transmembrane potential of the cell. DA exerts its effects by binding to DA receptors which are functionally categorized into two families, ie, D1-like and D2-like. The D1-type receptors (D1/D5) couple to the Gs class of G proteins and activate adenylyl cyclase. D2-type receptors (D2/D3/D4) couple to Gi protein which inhibits the production of cAMP.Citation38

Presynaptic receptors (autoreceptors) monitor extracellular DA levels and modulate impulse-dependent release and synthesis of DA.Citation39 Blockade of these receptors leads to increased production and presynaptic release of DA. Stimulation has the opposite effectCitation40 (see later for discussion of the role of presynaptic receptors in the action of methylphenidate). DA clearance from the synaptic cleft is regulated by the products of three genes, ie, DA transporter (SLC6A3/DAT1), monoamine oxidase-A (MAO-A) and catechol-o-methyl transferase (COMT). DAT1 is responsible for the rapid uptake of DA from the synaptic cleft, while MAO-A and COMT are involved in DA catabolism.Citation41

Nongenetic evidence for DA dysregulation

Neurochemical studies support a role for neurotransmitter dysregulation in ADHD pathophysiology.Citation42 Serotonergic, noradrenergic, and glutamatergic pathways have also been implicated.Citation43 Initial interest in DA in ADHD came from the longstanding observation that catecholamine agonists were psychostimulant medications and provided an effective treatment for many ADHD patients.Citation20 Since then, methylphenidate has been shown to inhibit the activity of the DA transporter and increase extrasynaptic levels of DA.Citation44,Citation45 There is evidence that it has little effect on presynaptic DA release,Citation46 but this has been questioned and the possibility of impulse dependency of transmitter release has been highlighted.Citation47 Another psycho-stimulant, amphetamine, has been shown to increase DA levels by modifying its release.Citation48 It interacts with DA transporters to promote DA efflux from the presynaptic neuron into the synaptic cleft.Citation49,Citation50 Other evidence in support of the DA dys-regulation hypothesis of ADHD comes from two main sources (other than the genetic evidence described later).

First, ADHD animal models show dysregulation of DA function.Citation51Citation55 The earliest animal model was developed by administration of 6-hydroxydopamine to neonatal rats that resulted in depletion of DA.Citation56 After treatment with 6-hydroxydopamine, the activity of animals was initially greater than that of controls. This then declined as a result of profound depletion of brain DA. Genetic models also provide evidence. The ADHD-type characteristics of the spontaneously hypertensive rat (SHR)Citation57Citation60 are reduced by DA agents,Citation61,Citation62 while those of the Naples high-excitability/low-excitability strain is associated with larger DA neurons and altered DA functioning in the limbic and cortical areas of the forebrain.Citation63Citation66 In the case of the coloboma mouse, these are associated with altered activity within specific surface proteins that mediate the process of docking and fusion of DA synaptic vesicles to the presynaptic plasma membrane.Citation67 This results from a 2-cM deletion of mouse chromosome 2 containing several genes including SNAP-25. These effects can be reversed by either transgenic insertion or stimulant medication.Citation68

Second, brain imaging studies using positron emission tomography and single photon emission computed tomography suggest altered regulation of striatal DA transporter levels. Studies vary greatly in their methodologic rigor and, perhaps because of this, there are inconsistencies between them.Citation17,Citation69 On the one hand, upregulation of striatal DA transporter densities has been reported in studies with small samples of mostly methylphenidate-treated cases.Citation70Citation73 Other studies with larger sample sizes found no evidence of altered DA transporter activity.Citation74 A recent study with a large sample of treatment-naïve adults with ADHD but without a history of comorbid substance use disorderCitation17 reported downregulation of striatal DAT consistent with higher levels of extracellular DA. This has been confirmed in a recent study in drug-naïve ADHD patientsCitation75 that found decreased striatal DA transporter availability in the basal ganglia. It seems likely that initial reports suggesting DAT upregulation were due to methodologic research limitations. The altered levels of DA transporters are difficult to interpret given the reciprocal and adaptive nature of the relationship between DA transporter densities and DA synthesis and release.Citation17,Citation76

Background genetics

Research has consistently shown a strong genetic component in the etiology of ADHD. Twin studies suggest heritability between 0.7 and 0.8.Citation77Citation79 The effect is similar for boys and girls.Citation80,Citation81 The nonheritable component appears to be attributable almost exclusively to nonshared environmental influences,Citation82,Citation83 but consideration has also been given to “contrast” effects in twin studies.Citation84 Heritability estimatesCitation85 themselves include gene–environment interaction and correlationCitation86 (see later for discussion in relation to DAT1 and DRD4).

There are two commonly used approaches in molecular genetic studies, ie, candidate gene approaches based on theoretical involvement of neurobiologic pathways leading to specific hypotheses, and nonhypothesis-driven genome-wide approaches that consider all genes as equally plausible candidates. Candidate gene approaches use either case-control or family-based association designs. In case-control studies, the frequency of candidate alleles or genotypes is compared in ADHD cases and controls. Family-based approaches such as the transmission disequilibrium test (TDT) examine patterns of genetic transmission disequilibriumCitation87 across generations within affected families to examine whether the probability of transmission of an allele from parents to affected offspring differs from the expected Mendelian pattern of inheritance. There are advantages and disadvantages to these approaches. Family-based studies have an advantage over case-control studies because they are designed to be immune to population stratification.Citation88 By population stratification we mean that in a mixed population, any trait present at a higher frequency in an ethnic group will show a positive association with any allele that also happens to be more common in that group. This can lead to spurious associations and so it is important that the two groups compared are of the same ethnic origin. However, the use of TDT in family-based studies is subject to selection effects due to missing parents and genotyping errors.Citation89,Citation90 Morton and CollinsCitation91 argue that stratification, which reduces the accuracy and power of the case-control design, is a problem only under rare circumstances, while the impact of genotyping errors in family-based approaches may have been underestimated.Citation92 Nonhypothesis-based approachesCitation93 have also used genome-wide association (GWA) studies and linkage design models. In genome-wide linkage studies, related individuals, either siblings or those in extended pedigrees, are studied in an attempt to localize chromosomal regions which may harbor genes influencing a trait by examining the familial cosegregation of the phenotype and genetic markers.Citation94 GWA studies compare markers across a population rather than within families, either for groups with or without a disorder or across the range of a trait in the population. More than a decade ago, it was predicted that GWA study designs are more powerful in detecting common alleles with small effects than are linkage approaches.Citation93 GWA studies require very large numbers of markers (ie, perhaps even millionsCitation95,Citation118) to cover the whole genome.

In both candidate gene and genome-wide approaches, the ADHD phenotype can be characterized as a diagnostic category or a quantitative trait. FisherCitation96 developed the theory of quantitative trait loci (QTL) based on the operation of multiple genes of varying effect. Broadly speaking, a continuous trait (rather than a diagnostic category) can be influenced by a few oligogenes with a moderate effect on the phenotype, or by many polygenes each with a very small effect, or by a combination of the two. The polygene example proposed by FisherCitation96 (for the trait of human height) has recently been used to identify multiple loci (at this time, up to 54) associated with height,Citation97 with many more genes predicted to contribute but which remain to be discovered.Citation98 While most studies have defined the ADHD phenotype in terms of diagnostic categories, impulsivity, attention, and activity can be adequately measured in a quantitative way,Citation99,Citation100 and researchers have argued for the use of dimensional approaches in the ADHD field.Citation101,Citation102 Although statistically powerful,Citation103 and despite the fact that they have been successfully applied both in human and animal behavioral studies,Citation104 QTL approaches have so far attracted relatively little interest in the ADHD field. This is probably because the quantification of ADHD when measured using common rating scales focuses only on the severity of psychopathology and does not capture the entire range of the underlying dimensions of attention/inattention and reflectivity/impulsivity.Citation84,Citation102,Citation105

The first ADHD genome scan based on 126 affected sib-pairs identified four regions (5p13, 10q26, 12q23, and 16p13) showing some evidence of linkage with logarithm (base 10) of odds scores >1.5.Citation106 Later genome-wide linkage scans were based on large families in population isolates in ColumbiaCitation107 and the Netherlands,Citation108 which provided a design with much greater statistical power for linkage analysis than the affected-sib pair design. A recent meta-analysisCitation109 of seven ADHD linkage scansCitation107,Citation110Citation115 identified the genomic region on chromosome 16, between 16q21 and 16q24, as the most consistent linkage evidence across the studies. Ten other regions on chromosomes 5, 6, 7, 8, 9, 15, 16, and 17 had nominal significance levels for linkage.Citation109 Two genome-wide linkage studies in humans employing QTL methods have identified linkage to chromosomes 1p36 and 3q13 for ADHD traits.Citation109,Citation116 Interestingly, the chromosomal region 1p36 overlaps with a dyslexia QTL, raising the possibility that pleiotropy (ie, where a single gene may impact on several phenotypes) might play a role in the genetic origins of ADHD and dyslexia.Citation109

Initial GWA studies with hundreds of thousands of markers and thousands of patients have so far failed to identify a significant genome-wide association between ADHD and these markers.Citation2,Citation117 Based on the literature on height, this is not unexpected, because the initial GWA studies of 2000–3000 participants also failed to reveal any associations that reached genome-wide levels of significance, but the strategy of combining samples to achieve increased power did identify loci on chromosome 12 and 20 with strong evidence of associationCitation97 that led to the documentation of an association with genes in these regions (HMAG2 and GDF5). The same approach may be productive for studies of ADHD.

In contrast, candidate gene approaches have been more successful. The first two relevant studies evaluated functional variants of DA genes, and showed an association of ADHD with DAT1Citation118 and DRD4.Citation154 Since then, other candidates within the DA systemCitation119 and other neurotransmitter systemsCitation120,Citation121 have been proposed, but few of these have produced robust and replicable effects. Several meta-analyses for single and multiple loci have been published that review these data.Citation122Citation124

ADHD and the dopamine receptor D4 gene

Distribution and functional polymorphisms

DA receptor D4 (DRD4) is a member of the D2 class of receptors. The D2-like receptors regulate several signaling events, including inhibition of adenylate cyclase, stimulation of arachidonic acid release, and modulation of potassium channels.Citation125Citation128 The human D4 receptor gene maps to chromosome 11p15.5. It consists of four exons and encodes a putative 387-amino acid protein with seven transmembrane domains.Citation129 DRD4 is highly expressed in pyramidal neurons and interneurons in the prefrontal cortex and in the retina. There are lower concentrations in the basal ganglia, hippocampus, and thalamus.Citation128,Citation130Citation133 Genetic variations in the DRD4 sequence have been examined in relation to various neuropsychiatric disorders. These have focused on a variable number of the tandem repeat (VNTR) polymorphism in exon 3, consisting of a 48-base-pair repeat unit. This unit codes for an amino acid sequence located in the third cytoplasmic loop of the receptor, thought to be involved in G-protein coupling.Citation134 In the human population, this VNTR displays a high degree of variability, with multiple nucleotide variation within each repeat.Citation135Citation137 The most common repeat variants are the 4R, 7R, and 2R alleles, respectively. The frequency of these alleles varies widely among different ethnic groupings.Citation136 The 7R allele, for example, has an extremely low prevalence in Asian populations (<2%) but a high frequency in native American populations (∼48%).Citation135 As yet, there is no commonly accepted explanation for this variability at the DRD4 locus. The common and probably ancestral allele has four repeats (4R) originating ∼300,000 years ago, whereas the 7R allele, often associated with psychiatric disorders, is up to 10 times “younger”.Citation138,Citation139 The 7R allele may have arisen as a rare mutational event and then become a high frequency allele by positive selectionCitation136 at a time of the major expansion of human population (the upper Paleolithic).Citation140 In this way, individuals with novelty-seeking personality traits may have driven the expansion of the 7R variant,Citation136 or it may have conferred a reproductive advantage in male-competitive societies.Citation141 In the Americas, an increase in the 7R allele may have been due to a successive founder effect,Citation140 and in China a decrease in the 7R may have been due to selective reproduction of males without the 7R allele.Citation141 At the same time there appears to be selective forces working to balance the alleles in modern societies (balancing selection), and the prevalence of the 7R allele may now be at a stable level or near a fixation point.Citation136

The neurofunctional significance of the DRD4 7R allele is not fully understood. In vitro studies indicate that the sensitivity of the 7R allele to DA is half that of the 2R and 4R variants.Citation134,Citation142 Moreover, DRD4 mRNA is distributed in the prefrontal cortexCitation133,Citation143,Citation144 but also to a lesser extent in the parietal and temporal lobes, cingulate cortex, and cerebellum.Citation132,Citation144 It is found in the basal ganglia, although its density relative to DRD2 is low.Citation143 This suggests it plays a role in cognitive and motivational processes.Citation145,Citation146 DRD4 and DAT1 seem not to be colocalized within brain regions (unlike DRD2 and DAT1), suggesting a different role for these two DA receptors.Citation143 Synthesis and clearance of DA are elevated in mice lacking the DRD4 gene.Citation147 Also, mice lacking a functional DRD4 receptor display cortical hyperexcitabilityCitation148,Citation149 and hypersensitivity to single administrations of alcohol, methamphetamine, and cocaine.Citation147

Categorical diagnoses and quantitative traits

The developing understanding of the neurofunctional significance of DRD4 7R has led to investigation of its association with disorders with a putative DA basis. In relation to ADHD, most studies have focused on the 7R polymorphism. An additional 120-base-pair duplication polymorphism located in the 5′ flanking region of DRD4Citation150 has also been studied recently,Citation151 as well as a single nucleotide polymorphism (−521 C/T; rs1800955) in the same region.Citation152 The association between the 7R allele DRD4 polymorphism and ADHD is well replicated. However, the findings are not completely consistent, and the absolute size of the effects is small, although relative to the maximum size possible if all cases had the allele (which is limited by the allele proportion in the population), in some ethnic groups it may be considered largeCitation153 (ie, if the allele probability is .20 in the population, then the maximum is 1/.2 = 5, and 1.9/5 is about 40%). In a ground-breaking study, LaHoste et alCitation154 first reported the association between DRD4 7R and ADHD. Many studies have followed this lead and the first meta-analysisCitation155 of this association was published in 2001 including both family-based (14 studies, 1665 probands) and case-control studies (eight studies, 1266 children with ADHD and 3068 controls). This gave an odds ratio (OR) of 1.9 for case-control studies (95% confidence interval [CI]: 1.5–2.2, P < 0.001) and 1.4 for family-based studies (95% CI: 1.1–1.6, P = 0.02). Five more meta-/pooled analyses of the 7R allele and ADHD have been published.Citation43,Citation122,Citation156Citation158 All of them have demonstrated a significant association, although the effect has reduced in size as more studies have been conducted and the total sample size has increased.Citation43,Citation122,Citation156Citation158 The most recent meta-analysis showed a fixed effects significance of P < 0.00001 with evidence of significant heterogeneity between studies.Citation43 In contrast with the 7R allele, the 4R allele may confer a protective effect (OR = 0.9, 95% CI: 0.84–0.97).Citation156

Several studies have examined DRD4 in relation to ADHD as a quantitative trait. Curran et alCitation159 first reported an association between the DRD4 7R allele and ADHD trait scores. Lasky-Su et alCitation160 found evidence for an association between two single nucleotide polymorphisms in the promoter region of DRD4 and the quantitative phenotype (mainly inattentive symptoms) generated from the ADHD symptoms. In contrast Mill et alCitation161 and Todd et alCitation62 failed to find evidence for an association between DRD4 and ADHD trait symptoms in the general population. None of these studies used a measure of the full range of attentional abilities in the population, and this could account for these negative results.Citation102

DRD4 and putative ADHD endophenotypes

Endophenotypes are conceptualized as “sitting between” genes and the clinical expression of the disorder.Citation163 To be of value in genetic studies, they should be heritable, cosegregate with a psychiatric illness, be present even when the disease is not (ie, state-independent), and be found in nonaffected family members at a higher rate than in the population.Citation163 Endophenotypes are postulated to be influenced by fewer genes than the clinical phenotype, and consequently the size of the effects of genetic loci contributing to endophenotypes is postulated to be larger than that to disease susceptibility. The fewer the genes that give rise to an endophenotype, the better the chances of revealing their genetic mode of action.Citation163 This concept has been controversial, with the suggestion that genetic effects are no greater in those studies employing endophenotypes than those using standard clinical phenotypes.Citation164 A range of candidate endophenotypes in ADHD has been proposed.Citation165 The best evidence has been found in relation to response inhibition,Citation166,Citation167 temporal processing,Citation168 verbal and visuospatial working memory,Citation166 and delay aversion.Citation169 A number of recent studies have found associations between DRD4 7R and performance on putative endophenotypes of ADHD, although the effects are inconsistent.Citation170 The first study of this sort in ADHD demonstrated the then seemingly paradoxical effect that in a small ADHD sample cases with the 7R-present genotype showed better neuropsychologic performance (faster and less variable reaction time on three tasks) than those with the 7R-absent genotype.Citation171 This direction of findings has been replicated,Citation172,Citation173 although some studies have also shown DRD4 7R is related to worse performance.Citation174 The association between DRD4 7R and neuropsychologic performance is not task-specific. but the strongest and most consistent effects seem to be in relation to high reaction time variability and the absence of 7R.Citation170 There is some evidence for altered speed of processingCitation174 and cognitive impulsiveness on nonreaction tasks in 7R carriers.Citation172 However, there is no effect of genotype on response inhibition.Citation172

DRD4 and gene–environment interactions

Results of behavioral genetic studies are consistent, with a role for environmental factors in ADHD and in personality characteristics in general.Citation175 Gene–environment interaction (GxE) has been an increasing focus of study. Here specific gene variants are shown to exert only a risk effect for a disorder if they are accompanied by exposure to a particular environmental risk factor.Citation176,Citation177 In relation to ADHD, these studies can be divided up into two types, ie, those focusing on the role for pre- and perinatal physical environmental risk factors (eg, maternal smoking and alcohol consumption during pregnancyCitation178) and those focusing on the postnatal social environment (eg, expressed emotion and social deprivation).Citation179 There have been a small number of replicated effects for GxE with DRD4 specifically and the results are currently unconvincing, but this may be due to inadequate statistical power in studies. Neuman et alCitation180 reported an interaction between maternal smoking during pregnancy and the 7R allele but Langley et alCitation181 failed to replicate this. Other DRD4 7R GxE findings include effects of season of birth.Citation182 DRD 7R has also been shown to moderate the effects of parenting on externalizing behavior including ADHD.Citation175,Citation183

ADHD and the SLC6A3/DAT1 gene

Distribution and functional polymorphisms

The DA transporter is a plasma membrane protein that belongs to the large family of NaCl-dependent transporters. It is responsible for terminating neurotransmission by rapid reuptake of DA into presynaptic terminals.Citation184 It has been shown to control the intensity and duration of DA neurotransmission by resetting the DA concentration in the extracellular space.Citation185,Citation186 In situ hybridization and immunochemistry studies have shown that DAT1 mRNA is primarily present in DA-synthesizing neurons of the substantia nigra and ventral tegmentum, and that the corresponding protein coincides with dopaminergic innervation of regions including the ventral mesencephalon, medial forebrain bundle, and dorsal and ventral striatum.Citation187,Citation188 The human DAT1 gene maps to chromosome 5p15.3. Sequence analysis of the 3′UTR of this gene revealed a variable number of tandem repeat (VNTR) polymorphisms with a 40-base-pair unit repeat length, ranging from three to 11 repeats.Citation189 In humans, the 9R and 10R are most common.Citation190 Reporter gene studiesCitation191 and studies of RNA expression in human tissuesCitation192 have shown that expression is significantly higher for the 10R than for other alleles, suggesting this variant may be functional. However, Miller and MadrasCitation193 found greater gene expression for vectors containing the 9R sequence, while othersCitation194 demonstrated that neither the 9R or the 10R allele had an effect on transcription. Furthermore, a brain imaging studyCitation195 showed higher density of striatal DAT1 in 10R homozygotes compared with the 9/10 genotype, but another in vivo experiment yielded conflicting results showing that the 9R carriers (9/9 homozygotes and 9/10 heterozygotes) had significantly higher striatal DAT1 availability.Citation196 However, the density of DAT is not fixed. Turnover of DA transporter protein takes about two days,Citation197 and plasticity has been documented, eg, the effects of drugs on DA transporter density have been established in studies of cocaineCitation198 and methylphenidate.Citation17 In as much as the brain “strives” for biochemical equilibrium, the impact of exposure to high levels of synaptic DA is thought to result in a compensatory increase in DAT to keep DA levels in a narrow range. Thus, exposure to stimulants that block DA transporters and increase synaptic DA is thought to increase the density of DA transporters. However, this must be measured when the drugs are not present in the brain, because occupancy of DA transporters would interfere with estimates of DA transporter density and suggest the opposite.Citation72

Categorical diagnoses and quantitative traits of DAT1

The DAT1 gene was the first DA gene examined in candidate gene association studies.Citation118 Using a family-based association design, the authors reported an association between the 10R allele and ADHD. Since the first publication, a number of studies have also reported an association between the DAT1 10R and ADHD.Citation199,Citation200 However, this association has not always been replicated.Citation201,Citation202 Overall, the evidence from meta-analyses is less supportive for DAT1 than for DRD4. For instance, Curran et alCitation203 reported a small, positive, but nonsignificant OR of 1.16, while Maher et alCitation157 also reported a nonsignificant OR. The most recent study found a significant association (OR = 1.12; P = 0.028), but also significant heterogeneity between studies.Citation43 It has been suggested that specific haplotypes rather than single markers are associated with ADHD.Citation204 Muglia et alCitation205 tested for an association between DAT1 and ADHD, considering the disorder as a category as well as a QTL, finding no association for either measure. Unlike Muglia et al,Citation205 Cornish et alCitation105 and Mill et alCitation161 evaluated ADHD as a continuous trait and found an association between the DAT1 10R allele and ADHD symptom score measure. Most recently Cornish et alCitation206 used a QTL approach to assess the association between the DAT1 high-risk genotype, visual search and vigilance, and ADHD symptoms in a community sample of boys aged 6–11 years. DAT1 genotypes were only related to ADHD symptoms. In contrast, Todd et alCitation207 found that the lower frequency allele (9R), along with the DRD4 7R allele, was overtransmitted in ADHD families.

DAT1 and putative ADHD endophenotypes

The data linking DAT1 to putative endophenotypes of ADHD is less compelling than for DRD4, given the dynamic properties of DA transporter densities. However, once again, high reaction time variability seems to be the most replicated cognitive marker associated with 10R homozygosity.Citation208 It is far from clear what causes such inconsistent results, but it has been suggested that endophenotypes such as delay aversionCitation209 may be better suited when studying DAT1 and ventral striatum-related functions. It is also possible that any association between DAT1 and neuropsychologic performance may be age-specific.Citation210

DAT1 and gene–environment interactions

DAT1 has been implicated in a broader range of GxE effects than has DRD4. In the first study of its kind in ADHD, Kahn et alCitation211 reported that hyperactivity/impulsivity symptom scores in young children were associated with a 10/10 genotype, but only in children exposed to prenatal smoking. It should be noted that the number of cases of children affected by both genetic and environmental risks was small. This was recently replicated in males.Citation212 In contrast, Neuman et alCitation180 reported an association between DAT1 9R and prenatal smoking, while others have found no effect at all.Citation181 Brookes et alCitation178 examined alcohol consumption during pregnancy and found an interaction with a DAT1 haplotype. In terms of psychosocial factors, it has been reported that family adversity moderates the impact of the DAT1 genotype on the expression of ADHD symptoms.Citation213 Sonuga-Barke et alCitation214 reported that DAT1 moderated the effect of parental expressed emotion on the development of conduct problems in ADHD. Stevens et alCitation215 showed that the risk of ADHD was increased only in those children who had experienced severe early institutional deprivation and were either homozygous for the 10R allele or carried a DAT1 haplotype combining a 40-base-pair VNTR in 3’UTR and a 30-base-pair VNTR in intron 8.

Overall the molecular-genetic evidence for DAT1 involvement in the etiology of ADHD is not as strong as for DRD4. The inconsistencies and small ORs may be explained by gene heterogeneity (different mutations at the same locus/gene resulting in an identical phenotype) as suggested in several studies.Citation158 One possibility to overcome this problem might be to examine haplotypes, as has been successfully done in the study by Asherson et al.Citation205

Clinical implications

Pharmacogenetics of DRD4 and DAT1

Individual differences in drug response are well documented in medicine, including psychiatry. A specific drug can be highly beneficial for some patients but can produce little or no effect in others and, for others, the same drug can have serious side effects.

The therapeutic value of medication (stimulants) in ADHD patients was first reported more than 70 years ago.Citation216 Since then, multiple randomized controlled trials have been published confirming without doubt the therapeutic effects of stimulants (eg, methylphenidate and amphetamines).Citation217Citation220 More recently, nonstimulants (eg, atomoxetine) have also been licensed.Citation221 While these treatments are, at least in the short term, very efficacious (eg, response rates of 85% to 90% when titration includes a range of doses for each stimulant and multiple stimulants), and generally well tolerated, there is still a range in the degree of responses.Citation222 The reduction of levels in ADHD to the levels found in healthy controls is relatively uncommon in clinical trials or in normal clinical practice.Citation223 Furthermore, there is likely to be much greater variability in the long-term effect of stimulants, and the optimal clinical dose appears to vary sixfold or more across individuals. These two dimensions of treatment response will be important sources of variance that may be interesting targets for future pharmacogenetic studies (especially given the high “response rates”).

There have been a number of attempts to identify predictors of response with the aim of improved tailoring of treatments to patient characteristics and needs. Factors such as age, gender, comorbidity and clinical have been considered, although evidence of significant effects of these is limited.Citation224,Citation225 In general, pharmacogenetic research in psychiatry studies of gene-drug interactions can help in the validation of therapeutic targets, the detection of factors determining response, and the identification of genetically induced side effects. The long-term goal is to develop more effectively tailored treatment and integrated personalized therapeutics. The therapeutic effects of stimulants at the neuronal level will depend on their ability to alter the release, uptake, and/or enzymatic inactivation of neurotransmitters (see discussion of the effects of methylphenidate and amphetamine earlier.Citation13,Citation52) As we have reviewed, these effects appear to vary as a function of DRD4 and DAT1 variants, and polymorphisms in these genes are important candidates for pharmacogenetic investigation. The working hypothesis is that such polymorphisms alter the impact of stimulant medication on brain systems as well as treatment efficacy.Citation226 Given that methylphenidate is only an “indirect agonist” of DA, via DA transporter blockade, this hypothesis may hold for DAT1 but not DRD4.

A number of pharmacogenetic studies have examined the relationship between methylphenidate response and DA gene polymorphisms in ADHD. The majority of studies have focused on DAT1. The results, so far, are inconclusive for both genes.Citation226,Citation227 The first relevant studyCitation228 reported a better therapeutic response to methylphenidate in ADHD children with the 9/10 genotype compared with children having the 10/10 genotype. While Roman et alCitation229 and Cheon et alCitation230 replicated this finding, othersCitation231,Citation232 found a better treatment response in patients homozygous for 10R. A further two studies demonstrated that the 9/9 genotype was associated with a decreased response to methylphenidate.Citation232,Citation233 In addition, several studies found no effect of DAT1 in terms of medication response.Citation234Citation238 For DRD4, Hamarman et alCitation239 found that patients with the 7R allele required higher doses for symptom improvement, while Cheon et alCitation240 reported that children homozygous for the 4R allele had a better response to methylphenidate. Other studies did not report a significant association between the DRD4 7R.Citation236Citation238 When trying to understand this conflicting and inconsistent set of results it must be acknowledged that studies to date have been in very small samples and therefore papers may be reporting chance findings.

Summary of key findings

  • ADHD is highly heritable (among the highest of all psychiatric disorders and nearly as high as the physical traits such as height) and at the advent of molecular genetic studies of ADHD it was assumed that the discovery of specific genes would be relatively easy.

  • The initial discoveries of associations with candidate genes was remarkably successful (in the context of general psychiatric genetics), with a significant association with first DAT1 and then DRD4 genetic variants that were chosen as candidate genes because of their pattern of distribution and neurofunctionality with regard to DA activity and a presumed role in the response to common pharmacologic treatment of ADHD with stimulant drugs.

  • The subsequent GWA approaches have not discovered additional genes and have not detected the replicated associations with ADHD from the candidate gene studies of DAT and DRD4.Citation117

  • Association studies provide stronger evidence for DRD4 (ie, the 7R allele) than DAT1 (ie, 10/10 genotype) in the pathogenesis of ADHD, probably because of greater between-study heterogeneity in DAT1 findings, with absolute effect sizes quantified as the relative risk for either gene individually have a restricted range. However, due to high allele proportions in the population, these effects may appear to be much larger when this is taken into account and the relative risk is compared with the maximum possible.

  • Evidence relating DRD4 and DAT genotypes to endophenotypes of ADHD is so far weak and inconsistent, but somewhat stronger for DRD4, especially with regard to response time variability.

  • There are also inconsistencies in the evidence implicating these genes in gene–environment interactions, with the strongest findings for DAT1, especially with regard to the impact of maternal smoking during pregnancy, although the role of gene–environment correlations cannot be ruled out.

  • DRD4 and DAT1 polymorphisms are interesting candidates for pharmacogenetic studies. DAT1 has the best evidence but the specific genotype associated with greater efficacy is yet to be determined definitively. This finding has to be treated cautiously given the inconsistency of findings and the small study samples. Recommendations for future pharmacogenetic studies are presented in a recent review.Citation241

Pitfalls and future directions in the ADHD gene search

Despite intense research efforts, progress in understanding the molecular genetic basis of ADHD may seem limited. Over a decade ago, a few candidate genes were found to be associated with ADHD, but their estimated effects were very small. Genome-wide scans have not identified additional loci to be reliably associated with ADHD. So, at the present time, despite high expectations based on heritability of about 0.8, the percentage of variance of the ADHD phenotype that can be explained by specific genetic factors is small. Importantly, this state of affairs is not unique to the ADHD area, and is a generic problem in research on specific genetic polymorphisms associated with other common disorders and traits.Citation242,Citation243 It is generally acknowledged that most of the inherited component of susceptibility to common diseases (including ADHD) remains to be explained.Citation242,Citation243 For ADHD, as for height (which also has high heritability but so far a low amount explained by identified genes), the variance not explained might be best described as “missing” or “dark” heritability.Citation244

What are the next steps? There are a number of options. Should we evaluate further the candidate genes with good documented association using functional genomics? Or should we assume that there are many noncandidate genes with small independent effects that remain to be discovered, and use genome-wide (noncandidate) approaches to continue the search to identify an ever larger set of genes with small effects that may eventually account for the large percentage of phenotypic variance predicted by the high heritability of ADHD? Contrasting candidate gene and genome-wide approaches for the investigation of ADHD, as for other common disorders, raise fundamental questions about what is the best strategy for unraveling the mysteries of the disorder. For example, the reviews and meta-analyses of candidate gene findings suggest evidence of an association for a few genes.Citation43,Citation156 GWA approaches with large samples do not document an association for these replicate candidate genes.Citation117,Citation245 How do we use these findings to suggest directions for future research? If we rely on the findings from the candidate gene approach, do we run the risk of being misdirected by false positives (as has often been suggested) or, if we rely on findings from the GWA approach, do we run the risk of being misdirected by false negatives? Here we address some of the pitfalls of these two general approaches.

Pitfalls and solutions in candidate gene studies

The pitfalls are different for population-based case-control from family-based approaches. For instance, the question of false-positive effects from the candidate gene approach may be related to methodologic flaws regarding the quality of genotyping and the completeness of samples (especially in family-based studies) and the problems of unbalanced samples in case-control studies. Population-based studies (eg, using case-control approaches) are sometimes easier to do without the need to ascertain parents. However, the methodologic issues associated with unbalanced groups of cases and controls have been a significant stumbling block. The primary “unbalancing” is by ethnicity, and this is particularly relevant for the ADHD area because the 7R allele prevalence of the most replicated candidate gene (DRD4) is known to differ dramatically across ethnicities,Citation137 with extremes from near 0 in Asian ADHD samplesCitation246 to over 30% in some Latin American ADHD samples.Citation247

Family-based studies (eg, the TDT approach with the untransmitted alleles from parents providing a perfectly matched control) can avoid ethnic stratification of cases and controls but have other potential pitfalls. Undetected genotyping errors and missing parents may have a significant impact in TDT analyses.Citation248 Mitchell et alCitation92 addressed genotyping error in a review of the literature on candidate gene studies. They noted an interesting difference in family-based studies and population-based studies; in the family-based studies utilizing TDT, most (87%) indicated that the most common allele was overtransmitted to affected offspring (suggesting a risk factor), but in the population-based studies, the most common allele was enriched in only 32% of cases and 68% of controls (suggesting a protective factor). They pointed out that even if undetected genotyping errors are random, their effect may not be nonrandom and, even if low, they can produce apparent transmission distortion at markers with alleles of unequal frequencies. For associations from TDT analyses between a common allele and risk, or a rare allele and protection, the authors recommend caution because this is in the direction of bias introduced by undetected genotyping error. Curtis and ShamCitation249 showed that computation of the TDT in trios when one parent is missing genotype data increases the false-positive error rate. WeinbergCitation90 and Gordon et alCitation248 proposed methods that allow for missing parents in TDT analyses.

Consideration of genotyping error rate and missing parent genotypes may be particularly relevant to the ADHD area for several reasons. First, the candidate gene approaches (DAT1 and DRD4) have proposed risk alleles with very different population allele frequencies, (ie, the DAT1 10R allele is the most common allele for the 40-base-pair VNTR, with a very high population prevalence that averages about 0.75, while the DRD4 4R allele with a prevalence in most populations of about 0.60 is usually the most common allele for the 48-base-pair VNTR, while the 7R allele has a lower population prevalence that averages about 0.12 in Caucasian populations). Second, the genotyping error rates in ADHD studies have been high for both population-based case-control studies (eg, up to 50% for some genes)Citation250 and family-based GWA studiesCitation117 (eg, 26% of the 500,000 SNPs failed the rigorous quality control implemented). Third, in family-based studies, the fathers are often missing,Citation79 and the use of complete trios may bias the sample.Citation251

What impact might these methodologic problems have on findings in the ADHD literature? For example, consider the observations and cautions outlined by Mitchell et alCitation92 for undetected genotyping errors. If undetected genotyping error rate is assumed and included in the TDT analyses of family-based studies, adjustments would reduce the observed effect for DAT1 and increase the observed effect for DRD4. The proposed allele frequency genotyping error rate effect may account for an observation highlighted in meta-analyses of the DRD4 findings, since the effect size for family-based studies using the TDT have been systematically lower (1.3) than the effect sizes for the population-based studies (1.9), which are not subject to this nonrandom effect. Given these problems, an obvious next step in the ADHD area is to increase rigor in checking for artifacts due to genotyping error and systemically biased samples due to self-selection of cases. Statistical methods have been developed to address these two important methodologic issues. For example, Gordon et alCitation248 developed a variant of the TDT that “allows for error” (ae), and their TDTae is robust to the presence of random genotyping errors and any number of untyped parents.

Pitfalls and solutions in genome-wide studies

In general the GWA approach has been successful in finding genes that were not predicted to be associated with disorders and traits.Citation252 An example of this success is the finding of genes and loci associated with the classic quantitative trait (ie, human height). After almost a century, the predictions provided by FisherCitation96 were finally tested after the 2005 HapMap project provided large sets of SNPs, and GWA methods were developed.Citation97 Initial GWA studies of 2000 to 3000 participants did not identify any loci that reached genome-wide significance levels for association with height, but the combination of samples increased statistical power and identified two genes (HMG2 and GDF5) associated with height. Further use of this strategy identified larger (20 replicated loci)Citation253 and even larger number of SNPs (54 associated loci).Citation254

However, the limitations as well as successes of the GWA approach were highlighted by the studies of height.Citation244 The size of the effects of the genes discovered so far has been very small and account for only 5% of population variance, which contrasted with the prediction by the high estimate of heritability,Citation254 indicating that many other genes will be found. The next step proposed is to conduct GWA studies with even larger sample sizes to identify the many (perhaps hundreds or thousands) genes with small effects that are presumed to contribute to the high heritability of height which have not yet been detected. This approach has been recommended for ADHD research. For example, Neale et alCitation117 did not detect any associated loci with a sample of about 1000 and a set of 500,000 SNPs, which is reminiscent of the initial GWA studies of height. They recommended the use of a larger sample that could be achieved by combining samples, which was a success strategy for identifying loci and genes associated with height.

However, critics of this approach have pointed out potential pitfalls for studies of height that may also be relevant for ADHD. For example, the loci with the largest effects have probably been identified in the initial GWA studies of height, and these account for only 5% of population variance. The contribution of additional SNPs to be identified in the next step with larger samples is expected to be smaller and smaller, so that one estimate of the number of loci required to reach 80% is extraordinarily high (93,000).Citation98 In contrast, other have emphasized that the primary purpose of the GWA approach is not to account completely for the percentage of variance predicted by heritability estimates or to predict the trait itself. Instead, the primary purpose is to identify unexpected biologic pathways involved in a disorder or a trait.Citation97

The current state of affairs has led to a reassessment of the common disease-common variant (CDCV) hypothesis upon which logic the GWA approach is based. The selection of SNP for GWA studies is based on the assumption derived from linkage disequilibrium that common variants within a haplotype block can stand as markers for the common variants, usually defined as having a minor allele frequency of 0.05 or greater. However, the common disease-rare variant (CDRV) hypothesis may be more appropriate.Citation255Citation258 To test the CDRV hypothesis we need a different approach to genotyping (ie, high-depth sequencing) to identify the rare variants, which in absolute numbers (as a set) are expected to be much more frequent than the set of common variants. A next step is to increase the density of SNPs (and eventually to obtain the complete genome sequence of each individual) and this has been proposed to ensure that rare as well as common causal variants could be detected. Several technology developments are currently trying to increase efficiency to a degree that the acquisition of the complete genome sequence for each individual would be feasible. This may be very relevant to the ADHD area. For example, rare variants have been documented for the DRD4 VNTR,Citation153 and these will be detected by complete sequencing.

Pitfalls and solutions for defining the ADHD phenotype

In the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) the ADHD phenotype is based on categoric diagnostic criteria. A different approach would involve reconceptualizing the ADHD phenotype as a trait similar to height. For the application of the FisherCitation96 model of a quantitative trait, the assumption is that it would be normally distributed in the population. More up-to-date approaches employing multiple regression methods of extreme scores on a continuous trait have also been applied.Citation259 However, most dimensional measures of ADHD (eg, derived from the Child Behavior Checklist, Strengths and Difficulties Questionnaire, Score for Neonatal Acute Physiology, Conners, Vanderbilt, DuPaul, or other rating scales) are based on severity of symptoms, so they are fundamentally categoric and produce a highly skewed distribution in the population (ie, for a representative sample that would include ADHD cases and noncases).Citation260

While the use of symptom-severity ratings as a dimension leaves considerable variance unmeasured in the noncases, adaptations at the item level to measure variation across the entire range of behavior in the population can provide trait measures of ADHD that captures this variance.Citation84 The strengths and weaknesses of ADHD symptoms and normal behavior (SWAN) method has not been widely used,Citation261 but it has been used infrequently for the definition of extreme groups for comparison in molecular genetic studiesCitation105 and in population-based twin studies.Citation84,Citation262,Citation263

The approach developed by FisherCitation96 provided the rationale for the evaluation of a quantitative trait considered to be the product of many independent small genetic effects that are additive and produce a normal distribution of the trait in the population. If a trait measure of ADHD that is normally distributed in the population is adopted, then the literature on the molecular genetics of other traits with high heritability may provide clear direction for a next step in the ADHD area that could follow the successes in the studies of the genetic bases of height. As mentioned above, ADHD, like height, has a very high (about 0.8) estimate of heritability. If a normally distributed trait measure of ADHD is used, then the next step in research could follow a general two-stage approach used to identify many genes associated with height.Citation253 In Stage 1, stringent GWA statistical safeguards are applied to protect against false-positive findings in the multiple testing of an extremely large set of SNP markers, and then in Stage 2 the significant set of markers (some assumed to be false positives) are evaluated in an even larger sample at a much reduced genotyping cost. Weedon et alCitation253 described the Stage 1 use of six GWA studies of 13,665 individuals to identify 39 SNPs that exceeded a statistical cut-off to avoid false positives (P < 10 × 10−4), which were investigated for replication in Stage 2 in 16,482 individuals, with replication of association for 20 of the 39 SNPs. This approach has been extended by additional GWA studies of height, which have (so far) identified 54 loci associated with height in a sample of over 63,000 individuals.Citation254,Citation264

A similar approach could be taken for evaluation of normally distributed traits related to ADHD. Associations of SNPs with small but reliable effects might be identified in a similar two-stage approach, with 15,000 to 20,000 individuals included in a Stage 1 GWA scan to identify a set of SNPs with alleles associated with risk (high level of the ADHD trait) and protection (low level of the ADHD trait). Then, in Stage 2 the set of SNPs could be genotyped in an additional set of 15,000 to 20,000 individuals, and for those with a replicated association, the distribution of high-ADHD alleles could be specified. The prediction from the FisherCitation96 quantitative trait model would be a normal distribution of the number of high-ADHD alleles, and a linear relationship between the number of high-ADHD alleles and rating of the ADHD trait. The most rigorous genome-wide linkage study of ADHDCitation111 did not identify any loci associated with ADHD, and the most rigorous GWA study of ADHDCitation117 did not identify any SNP that met the Stage 1 cut-off to carry forward into Stage 2, but this may have been due to the use of a categoric diagnosis of ADHD rather than a normally distributed trait.

Pitfalls and solutions in statistical analyses

The estimate of high heritability (0.80) for ADHD from twin studies includes main effects of genetic factors, as well as interactions of the genetic main effects with environmental effect that have not been measured and included in the model use to generate the estimates of heritability.Citation85 In the next steps of research on ADHD, it may be important to address the violations of assumptions of additivity of main effects, and to measure environmental exposures that affect phenotype so that in statistical analyses, provisions can be made to separate genetic main effects and gene–environment interaction effects. Several approaches for the measurement of environmental exposures that may be involved in gene–environment interactions have been described in a 2008 special issue of the Journal of Child Psychology and Psychiatry.

The strategies to investigate gene–environment interactions will require access to large sample sizes, new technologies, and new analytic methods. Several large samples may be required to take into account differences in the genetic architecture of rare and common alleles that are known to contribute to common disorders and to traits.Citation255 One future sample will be provided by the National Children’s Study which was initiated in 2009 and plans to acquire a representative birth cohort of 100,000 children by 2015, with broad measures of environmental exposures and phenotypic outcomes starting before birth and continuing at birth, in infancy, during childhood and adolescence, and into adulthood. Eventually, the National Children’s Study should have about 5000 cases that would meet the categoric diagnostic criteria for ADHD. Traditionally, these cases would be matched to well-evaluated controls, and a nested case-control study of the disorder. Based on the expected sample of 5000 cases, standard calculations of the statistical power needed to detect association of genetic main effects and gene–environment interaction effectsCitation265 indicated that small association effects should be detectable, and tests of hypotheses of gene–environment interaction would also have adequate power. This would allow for tests of gene–environment interactions that have been proposed based on small samples, such as the interaction of DAT1 genotype and maternal smoking during pregnancy.Citation211 The prospective birth cohort design will allow for evaluation of epigenetic variation related to fetal adaptationsCitation266 which has been proposed as an important etiology of ADHD, based on children born under conditions of stress during pregnancyCitation267 and has been revived by imaging studies during follow-up of that cohort.Citation268

However, if the example of height is used to direct the next steps in research on the genetic basis of ADHD, then a normally distributed trait related to ADHD should be used instead of categorical diagnosis of a disorder. Then the entire sample of 100,000 could be utilized, which would provide a more powerful statistical approach to identify genes associated with ADHD and as yet unknown biologic pathwaysCitation97 that contribute to the etiology, and could be used to develop potential new treatments for this condition.

Disclosures

Dr Sonuga-Barke has been a recent speaker and has done past and present consultancy for Shire and UCB Pharma. He has received past and present research support from Janssen Cilag, Shire, Qbtech, and Flynn Pharma, and is on the advisory boards for Shire, Flynn Pharma, UCB Pharma, and Astra Zeneca, and has had conference support from Shire Dr Swanson has received research support from Alza, Richwood, Shire, Celgene, Novartis, Celltech, Gliatech, Cephalon, Watson, CIBA, Janssen, and McNeil, and has been on the advisory boards of Alza, Richwood, Shire, Celgene, Novartis, Celltech, UCB, Gliatech, Cephalon, McNeil, and Eli Lilly, and has been on the speakers’ bureaus of Alza, Shire, Novartis, Celltech, UCB, Cephalon, CIBA, Janssen, and McNeil. He has also consulted to Alza, Richwood, Shire, Celgene, Novartis, Celltech, UCB, Gliatech, Cephalon, Watson, CIBA, Janssen, McNeil, and Eli Lilly. The authors report no conflict of interest in this research.

References

  • PolanczykGde LimaMSHortaBLBiedermanJRohdeLAThe worldwide prevalence of ADHD: A systematic review and metaregression analysisAm J Psychiatry2007164694294817541055
  • Lasky-SuJAnneyRJNealeBMGenome-wide association scan of the time to onset of attention deficit hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2008147B81355135818937294
  • ReiffMISteinMTAttention-deficit/hyperactivity disorder evaluation and diagnosis: A practical approach in office practicePediatr Clin North Am20035051019104814558680
  • MannuzzaSKleinRGKonigPHGiampinoTLHyperactive boys almost grown up. IV. Criminality and its relationship to psychiatric statusArch Gen Psychiatry19894612107310792589922
  • SatterfieldJHFallerKJCrinellaFMSchellAMSwansonJMHomerLDA 30-year prospective follow-up study of hyperactive boys with conduct problems: Adult criminalityJ Am Acad Child Adolesc Psychiatry200746560161017450051
  • BarkleyRAFischerMSmallishLFletcherKYoung adult follow-up of hyperactive children: Antisocial activities and drug useJ Child Psychol Psychiatry200445219521114982236
  • SwansonJMKinsbourneMNiggJEtiologic subtypes of attention-deficit/hyperactivity disorder: Brain imaging, molecular genetic and environmental factors and the dopamine hypothesisNeuropsychol Rev2007171395917318414
  • Sonuga-BarkeEJElgieSHallMMore to ADHD than meets the eye: Observable abnormalities in search behaviour do not account for performance deficits on a discrimination taskBehav Brain Funct2005111016033644
  • ArnstenAFLiBMNeurobiology of executive functions: Catecholamine influences on prefrontal cortical functionsBiol Psychiatry200557111377138415950011
  • PliszkaSRMcCrackenJTMaasJWCatecholamines in attention-deficit hyperactivity disorder: Current perspectivesJ Am Acad Child Adolesc Psychiatry19963532642728714313
  • SchultzWGetting formal with dopamine and rewardNeuron200236224126312383780
  • SchultzWMultiple dopamine functions at different time coursesAnnu Rev Neurosci20073025928817600522
  • SeemanPMadrasBKAnti-hyperactivity medication: Methylphenidate and amphetamineMol Psychiatry1998353863969774771
  • LeesAJHardyJReveszTParkinson’s diseaseLancet200937396802055206619524782
  • AultmanJMMoghaddamBDistinct contributions of glutamate and dopamine receptors to temporal aspects of rodent working memory using a clinically relevant taskPsychopharmacology (Berl)2001153335336411271408
  • FlorescoSBPhillipsAGDelay-dependent modulation of memory retrieval by infusion of a dopamine D1 agonist into the rat medial prefrontal cortexBehav Neurosci2001115493493911508732
  • VolkowNDWangGJKollinsSHEvaluating dopamine reward pathway in ADHD: Clinical implicationsJAMA2009302101084109119738093
  • HranilovicDBucanMWangYEmotional response in dopamine D2L receptor-deficient miceBehav Brain Res2008195224625018835570
  • PezzeMAFeldonJMesolimbic dopaminergic pathways in fear conditioningProg Neurobiol200474530132015582224
  • WenderPHA possible monoaminergic basis for minimal brain dysfunctionPsychopharmacol Bull197511336371153658
  • GiulianoFAllardJDopamine and sexual functionInt J Impot Res200113Suppl 3S18S2811477488
  • HymanSEMalenkaRCNestlerEJNeural mechanisms of addiction: The role of reward-related learning and memoryAnnu Rev Neurosci20062956559816776597
  • PecinaSBerridgeKCHedonic hot spot in nucleus accumbens shell: Where do mu-opioids cause increased hedonic impact of sweetness?J Neurosci20052550117771178616354936
  • BarbeauAHigh-level levodopa therapy in Parkinson’s disease: Five years laterTrans Am Neurol Assoc1974991601634463526
  • MogensonGJJonesDLYimCYFrom motivation to action: Functional interface between the limbic system and the motor systemProg Neurobiol1980142–369976999537
  • WiseRADopamine, learning and motivationNat Rev Neurosci20045648349415152198
  • WiseRADopamine and food reward: Back to the elementsAm J Physiol Regul Integr Comp Physiol20042861R1314660469
  • Ben-JonathanNHnaskoRDopamine as a prolactin (PRL) inhibitorEndocr Rev200122672476311739329
  • AlexanderGECrutcherMDFunctional architecture of basal ganglia circuits: Neural substrates of parallel processingTrends Neurosci19901372662711695401
  • Bar-GadIBergmanHStepping out of the box: Information processing in the neural networks of the basal gangliaCurr Opin Neurobiol200111668969511741019
  • HaberSNThe primate basal ganglia: Parallel and integrative networksJ Chem Neuroanat200326431733014729134
  • MiddletonFAStrickPLBasal-ganglia ‘projections’ to the prefrontal cortex of the primateCereb Cortex200212992693512183392
  • TakadaMTokunoHNambuAInaseMCorticostriatal projections from the somatic motor areas of the frontal cortex in the macaque monkey: Segregation versus overlap of input zones from the primary motor cortex, the supplementary motor area, and the premotor cortexExp Brain Res199812011141289628410
  • Sonuga-BarkeEJDalenLDaleyDRemingtonBAre planning, working memory, and inhibition associated with individual differences in preschool ADHD symptoms?Dev Neuropsychol200221325527212233938
  • KolomietsBPDeniauJMMaillyPMenetreyAGlowinskiJThierryAMSegregation and convergence of information flow through the cortico-subthalamic pathwaysJ Neurosci200121155764577211466448
  • McFarlandNRHaberSNThalamic relay nuclei of the basal ganglia form both reciprocal and nonreciprocal cortical connections, linking multiple frontal cortical areasJ Neurosci200222188117813212223566
  • HaberSNCalzavaraRThe cortico-basal ganglia integrative network: The role of the thalamusBrain Res Bull2009782–3697418950692
  • CallierSSnapyanMLe CromSProuDVincentJDVernierPEvolution and cell biology of dopamine receptors in vertebratesBiol Cell200395748950214597267
  • AltarCABoyarWCOeiEWoodPLDopamine autoreceptors modulate the in vivo release of dopamine in the frontal, cingulate and entorhinal corticesJ Pharmacol Exp Ther198724211151203612520
  • MarinelliMRudickCNHuXTWhiteFJExcitability of dopamine neurons: modulation and physiological consequencesCNS Neurol Disord Drug Targets200651799716613555
  • HuotariMSanthaMLucasLRKarayiorgouMGogosJAMannistoPTEffect of dopamine uptake inhibition on brain catecholamine levels and locomotion in catechol-O-methyltransferase-disrupted miceJ Pharmacol Exp Ther200230331309131612438556
  • ZametkinAJLiottaWThe neurobiology of attention-deficit/hyperactivity disorderJ Clin Psychiatry199859Suppl 717239680049
  • GizerIRFicksCWaldmanIDCandidate gene studies of ADHD: A meta-analytic reviewHum Genet20091261519019506906
  • VolkowNDWangGJFowlerJSDingYSImaging the effects of methylphenidate on brain dopamine: New model on its therapeutic actions for attention-deficit/hyperactivity disorderBiol Psychiatry200557111410141515950015
  • VolkowNDWangGJFowlerJSEvidence that methylphenidate enhances the saliency of a mathematical task by increasing dopamine in the human brainAm J Psychiatry200416171173118015229048
  • PatrickKSCaldwellRWFerrisRMBreeseGRPharmacology of the enantiomers of threo-methylphenidateJ Pharmacol Exp Ther198724111521583572779
  • SeemanPMadrasBMethylphenidate elevates resting dopamine which lowers the impulse-triggered release of dopamine: A hypothesisBehav Brain Res20021301–2798311864721
  • FanXXuMHessEJD2 dopamine receptor subtype-mediated hyperactivity and amphetamine responses in a model of ADHDNeurobiol Dis20103722823619840852
  • KuczenskiRSegalDSDifferential effects of D- and L-amphetamine and methylphenidate on rat striatal dopamine biosynthesisEur J Pharmacol19753022442511126362
  • KuczenskiRSegalDSEffects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamineJ Neurochem1997685203220379109529
  • AcciliDFishburnCSDragoJA targeted mutation of the D3 dopamine receptor gene is associated with hyperactivity in miceProc Natl Acad Sci U S A1996935194519498700864
  • GirosBJaberMJonesSRWightmanRMCaronMGHyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporterNature199637965666066128628395
  • GranonSPassettiFThomasKLDalleyJWEverittBJRobbinsTWEnhanced and impaired attentional performance after infusion of D1 dopaminergic receptor agents into rat prefrontal cortexJ Neurosci20002031208121510648725
  • LeoDSorrentinoEVolpicelliFAltered midbrain dopaminergic neurotransmission during development in an animal model of ADHDNeurosci Biobehav Rev200327766166914624810
  • XuMMoratallaRGoldLHDopamine D1 receptor mutant mice are deficient in striatal expression of dynorphin and in dopamine-mediated behavioral responsesCell19947947297427954836
  • ShaywitzBAYagerRDKlopperJHSelective brain dopamine depletion in developing rats: An experimental model of minimal brain dysfunctionScience19761914224305308942800
  • MookDMJeffreyJNeuringerASpontaneously hypertensive rats (SHR) readily learn to vary but not repeat instrumental responsesBehav Neural Biol19935921261358476380
  • SagvoldenTBehavioral validation of the spontaneously hypertensive rat (SHR) as an animal model of attention-deficit/hyperactivity disorder (AD/HD)Neurosci Biobehav Rev2000241313910654658
  • WongAHBuckleCEVan TolHHPolymorphisms in dopamine receptors: What do they tell us?Eur J Pharmacol20004102–318320311134669
  • WyssJMFiskGvan GroenTImpaired learning and memory in mature spontaneously hypertensive ratsBrain Res19925921–21351401450905
  • BoixFQiaoSWKolpusTSagvoldenTChronic L-deprenyl treatment alters brain monoamine levels and reduces impulsiveness in an animal model of attention-deficit/hyperactivity disorderBehav Brain Res19989411531629708846
  • MyersMMMustyREHendleyEDAttenuation of hyperactivity in the spontaneously hypertensive rat by amphetamineBehav Neural Biol198234142547073635
  • Gonzalez-LimaFSadileAGNetwork operations revealed by brain metabolic mapping in a genetic model of hyperactivity and attention deficit: The Naples high- and low-excitability ratsNeurosci Biobehav Rev200024115716010654673
  • PapaMSellittiSSadileAGRemodeling of neural networks in the anterior forebrain of an animal model of hyperactivity and attention deficits as monitored by molecular imaging probesNeurosci Biobehav Rev200024114915610654672
  • SadileAGLambertiCSiegfriedBWelzlHCircadian activity, nociceptive thresholds, nigrostriatal and mesolimbic dopaminergic activity in the Naples high- and low-excitability rat linesBehav Brain Res199355117278101085
  • SadileAGPellicanoMPSagvoldenTSergeantJANMDA and non-NMDA sensitive [L-3H] glutamate receptor binding in the brain of the Naples high- and low-excitability rats: An autoradiographic studyBehav Brain Res19967821631748864048
  • HessEJCollinsKAWilsonMCMouse model of hyperkinesis implicates SNAP-25 in behavioral regulationJ Neurosci1996169310431118622140
  • SteffensenSCHenriksenSJWilsonMCTransgenic rescue of SNAP-25 restores dopamine-modulated synaptic transmission in the coloboma mutantBrain Res1999847218619510575087
  • GononFThe dopaminergic hypothesis of attention-deficit/hyperactivity disorder needs re-examiningTrends Neurosci20093212818986716
  • CheonKARyuYHKimYKNamkoongKKimCHLeeJDDopamine transporter density in the basal ganglia assessed with [123I]IPT SPET in children with attention deficit hyperactivity disorderEur J Nucl Med Mol Imaging200330230631112552351
  • DoughertyDDBonabAASpencerTJRauchSLMadrasBKFischmanAJDopamine transporter density in patients with attention deficit hyperactivity disorderLancet199935491962132213310609822
  • KrauseKHDreselSHKrauseJKungHFTatschKIncreased striatal dopamine transporter in adult patients with attention deficit hyperactivity disorder: Effects of methylphenidate as measured by single photon emission computed tomographyNeurosci Lett2000285210711010793238
  • LarischRSitteWAntkeCStriatal dopamine transporter density in drug naive patients with attention-deficit/hyperactivity disorderNucl Med Commun200627326727016479247
  • van DyckCHQuinlanDMCretellaLMUnaltered dopamine transporter availability in adult attention deficit hyperactivity disorderAm J Psychiatry2002159230931211823278
  • HesseSBallaschkeOBarthelHSabriODopamine transporter imaging in adult patients with attention-deficit/hyperactivity disorderPsychiatry Res2009171212012819176281
  • MadrasBKMillerGMFischmanAJThe dopamine transporter: Relevance to attention deficit hyperactivity disorder (ADHD)Behav Brain Res20021301–2576311864718
  • BiedermanJFaraoneSVAttention-deficit hyperactivity disorderLancet2005366948123724816023516
  • StevensonJEvidence for a genetic etiology in hyperactivity in childrenBehav Genet19922233373441616463
  • SwansonJMSergeantJATaylorESonuga-BarkeEJJensenPSCantwellDPAttention-deficit hyperactivity disorder and hyperkinetic disorderLancet199835191004294339482319
  • NadderTSSilbergJLEavesLJMaesHHMeyerJMGenetic effects on ADHD symptomatology in 7- to 13-year-old twins: Results from a telephone surveyBehav Genet199828283999583234
  • SaudinoKJRonaldAPlominRThe etiology of behavior problems in 7-year-old twins: Substantial genetic influence and negligible shared environmental influence for parent ratings and ratings by same and different teachersJ Abnorm Child Psychol200533111313015759595
  • LarssonJOLarssonHLichtensteinPGenetic and environmental contributions to stability and change of ADHD symptoms between 8 and 13 years of age: A longitudinal twin studyJ Am Acad Child Adolesc Psychiatry200443101267127515381894
  • ThaparAHarringtonRMcGuffinPExamining the comorbidity of ADHD-related behaviours and conduct problems using a twin study designBr J Psychiatry200117922422911532799
  • PoldermanTJde GeusEJHoekstraRAAttention problems, inhibitory control, and intelligence index overlapping genetic factors: A study in 9-, 12-, and 18-year-old twinsNeuropsychology200923338139119413451
  • PurcellSStatistical methods in behavioral geneticsPlominRDeFriesJMcClearnGMcGuffinPBehavioral GeneticsNew York, NYWorth2001
  • RutterMGene-environment interdependenceDev Sci2007101121817181693
  • SpielmanRSMcGinnisREEwensWJTransmission test for linkage disequilibrium: The insulin gene region and insulin-dependent diabetes mellitus (IDDM)Am J Hum Genet19935235065168447318
  • SpielmanRSEwensWJThe TDT and other family-based tests for linkage disequilibrium and associationAm J Hum Genet19965959839898900224
  • ClaytonDA generalization of the transmission/disequilibrium test for uncertain-haplotype transmissionAm J Hum Genet19996541170117710486336
  • WeinbergCRAllowing for missing parents in genetic studies of case-parent triadsAm J Hum Genet19996441186119310090904
  • MortonNECollinsATests and estimates of allelic association in complex inheritanceProc Natl Acad Sci U S A1998951911389113939736746
  • MitchellAACutlerDJChakravartiAUndetected genotyping errors cause apparent overtransmission of common alleles in the transmission/disequilibrium testAm J Hum Genet200372359861012587097
  • RischNMerikangasKThe future of genetic studies of complex human diseasesScience19962735281151615178801636
  • LanderESSchorkNJGenetic dissection of complex traitsScience19942655181203720488091226
  • ClarkAGLiJConjuring SNPs to detect associationsNat Genet200739781581617597769
  • FisherRThe correlation between relatives on the supposition of Mendelian inheritanceTrans Roy Soc Edin191852399433
  • HirschhornJNLettreGProgress in genome-wide association studies of human heightHorm Res200971Suppl 251319407491
  • GoldsteinDBCommon genetic variation and human traitsN Engl J Med2009360171696169819369660
  • BurnsGLWalshJAOwenSMSnellJInternal validity of attention deficit hyperactivity disorder, oppositional defiant disorder, and overt conduct disorder symptoms in young children: Implications from teacher ratings for a dimensional approach to symptom validityJ Clin Child Psychol19972632662759292384
  • PintoLPTryonWWActivity measurements support dimensional assessmentBehav Modif19962032432588768507
  • HudziakJJAchenbachTMAlthoffRRPineDSA dimensional approach to developmental psychopathologyInt J Methods Psychiatr Res200716Suppl 1S16S2317623391
  • SwansonJMWigalTLakesKDSM-V and the future diagnosis of attention-deficit/hyperactivity disorderCurr Psychiatry Rep200911539940619785982
  • AshersonPAttention-deficit hyperactivity disorder in the post-genomic eraEur Child Adolesc Psychiatry200413Suppl 1I50I7015322957
  • SpenceJPLiangTLiuLFrom QTL to candidate gene: A genetic approach to alcoholism researchCurr Drug Abuse Rev20092212713419630743
  • CornishKMManlyTSavageRAssociation of the dopamine transporter (DAT1) 10/10-repeat genotype with ADHD symptoms and response inhibition in a general population sampleMol Psychiatry200510768669815809660
  • FisherSEFrancksCMcCrackenJTA genomewide scan for loci involved in attention-deficit/hyperactivity disorderAm J Hum Genet20027051183119611923911
  • Arcos-BurgosMCastellanosFXPinedaDAttention-deficit/hyperactivity disorder in a population isolate: Linkage to loci at 4q13.2, 5q33.3, 11q22, and 17p11Am J Hum Genet2004756998101415497111
  • AminNAulchenkoYSDekkerMCSuggestive linkage of ADHD to chromosome 18q22 in a young genetically isolated Dutch populationEur J Hum Genet200917795896619156173
  • ZhouKDempfleAArcos-BurgosMMeta-analysis of genome-wide linkage scans of attention deficit hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2008147B81392139818988193
  • AshersonPZhouKAnneyRJA high-density SNP linkage scan with 142 combined subtype ADHD sib pairs identifies linkage regions on chromosomes 9 and 16Mol Psychiatry200813551452118180756
  • BakkerSCvan der MeulenEMBuitelaarJKA whole-genome scan in 164 Dutch sib pairs with attention-deficit/hyperactivity disorder: Suggestive evidence for linkage on chromosomes 7p and 15qAm J Hum Genet20037251251126012679898
  • FaraoneSVDoyleAELasky-SuJLinkage analysis of attention deficit hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2008147B81387139118081027
  • HebebrandJDempfleASaarKA genome-wide scan for attention-deficit/hyperactivity disorder in 155 German sib-pairsMol Psychiatry200611219620516222334
  • OgdieMNMacphieILMinassianSLA genomewide scan for attention-deficit/hyperactivity disorder in an extended sample: Suggestive linkage on 17p11Am J Hum Genet20037251268127912687500
  • RomanosMFreitagCJacobCGenome-wide linkage analysis of ADHD using high-density SNP arrays: Novel loci at 5q13.1 and 14q12Mol Psychiatry200813552253018301393
  • DoyleAEFerreiraMASklarPBMultivariate genome-wide linkage scan of neurocognitive traits and ADHD symptoms: Suggestive linkage to 3q13Am J Med Genet B Neuropsychiatr Genet2008147B81399141118973233
  • NealeBMLasky-SuJAnneyRGenome-wide association scan of attention deficit hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2008147B81337134418980221
  • CookEHJrSteinMAKrasowskiMDAssociation of attention-deficit disorder and the dopamine transporter geneAm J Hum Genet19955649939987717410
  • PaytonAHolmesJBarrettJHExamining for association between candidate gene polymorphisms in the dopamine pathway and attention-deficit hyperactivity disorder: A family-based studyAm J Med Genet2001105546447011449400
  • HawiZDringMKirleyASerotonergic system and attention deficit hyperactivity disorder (ADHD): A potential susceptibility locus at the 5-HT(1B) receptor gene in 273 nuclear families from a multi-centre sampleMol Psychiatry20027771872512192616
  • TuricDLangleyKWilliamsHA family based study implicates solute carrier family 1-member 3 (SLC1A3) gene in attention-deficit/hyperactivity disorderBiol Psychiatry200557111461146615950021
  • FaraoneSVPerlisRHDoyleAEMolecular genetics of attention-deficit/hyperactivity disorderBiol Psychiatry200557111313132315950004
  • Purper-OuakilDWohlMMourenMCVerpillatPAdesJGorwoodPMeta-analysis of family-based association studies between the dopamine transporter gene and attention deficit hyperactivity disorderPsychiatr Genet2005151535915722958
  • YangBChanRCJingJLiTShamPChenRYA meta-analysis of association studies between the 10-repeat allele of a VNTR polymorphism in the 3’-UTR of dopamine transporter gene and attention deficit hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2007144B454155017440978
  • JaberMRobinsonSWMissaleCCaronMGDopamine receptors and brain functionNeuropharmacology19963511150315199025098
  • NeveKASeamansJKTrantham-DavidsonHDopamine receptor signalingJ Recept Signal Transduct Res200424316520515521361
  • OakJNOldenhofJVan TolHHThe dopamine D(4) receptor: One decade of researchEur J Pharmacol20004051–330332711033337
  • TaraziFIZhangKBaldessariniRJDopamine D4 receptors: Beyond schizophreniaJ Recept Signal Transduct Res200424313114715521359
  • Van TolHHBunzowJRGuanHCCloning of the gene for a human dopamine D4 receptor with high affinity for the antipsychotic clozapineNature199135063196106141840645
  • ArianoMAWangJNoblettKLLarsonERSibleyDRCellular distribution of the rat D4 dopamine receptor protein in the CNS using anti-receptor antiseraBrain Res19977521–226349106437
  • GanLFalzoneTLZhangKRubinsteinMBaldessariniRJTaraziFIEnhanced expression of dopamine D(1) and glutamate NMDA receptors in dopamine D(4) receptor knockout miceJ Mol Neurosci200422316717814997010
  • MrzljakLBergsonCPappyMHuffRLevensonRGoldman-RakicPSLocalization of dopamine D4 receptors in GABAergic neurons of the primate brainNature199638165792452488622768
  • NoainDAvaleMEWedemeyerCCalvoDPeperMRubinsteinMIdentification of brain neurons expressing the dopamine D4 receptor gene using BAC transgenic miceEur J Neurosci20062492429243817100831
  • Van TolHHWuCMGuanHCMultiple dopamine D4 receptor variants in the human populationNature199235863821491521319557
  • ChangFMKiddJRLivakKJPakstisAJKiddKKThe world-wide distribution of allele frequencies at the human dopamine D4 receptor locusHum Genet1996981911018682515
  • DingYCChiHCGradyDLEvidence of positive selection acting at the human dopamine receptor D4 gene locusProc Natl Acad Sci U S A200299130931411756666
  • LichterJBBarrCLKennedyJLVan TolHHKiddKKLivakKJA hypervariable segment in the human dopamine receptor D4 (DRD4) geneHum Mol Genet1993267677738353495
  • WangEDingYCFlodmanPThe genetic architecture of selection at the human dopamine receptor D4 (DRD4) gene locusAm J Hum Genet200474593194415077199
  • WangETKodamaGBaldiPMoyzisRKGlobal landscape of recent inferred Darwinian selection for Homo sapiensProc Natl Acad Sci U S A2006103113514016371466
  • ChenCBurtonMGreenbergerEDmitrievaJPopulation migration and the variation of dopamine receptor (DRD4) allele frequencies around the globeEvol Hum Behav199920309324
  • HarpendingHCochranGIn our genesProc Natl Acad Sci U S A2002991101211782544
  • AsghariVSanyalSBuchwaldtSPatersonAJovanovicVVan TolHHModulation of intracellular cyclic AMP levels by different human dopamine D4 receptor variantsJ Neurochem1995653115711657643093
  • De La GarzaR2ndMadrasBK[(3)H]PNU-101958, a D(4) dopamine receptor probe, accumulates in prefrontal cortex and hippocampus of non-human primate brainSynapse200037323224410881045
  • PrimusRJThurkaufAXuJII. Localization and characterization of dopamine D4 binding sites in rat and human brain by use of the novel, D4 receptor-selective ligand [3H]NGD 94-1J Pharmacol Exp Ther19972822102010279262371
  • DolanRJEmotion, cognition, and behaviorScience200229855961191119412424363
  • DurstonSde ZeeuwPStaalWGImaging genetics in ADHD: A focus on cognitive controlNeurosci Biobehav Rev200933567468918789356
  • RubinsteinMPhillipsTJBunzowJRMice lacking dopamine D4 receptors are supersensitive to ethanol, cocaine, and methamphetamineCell199790699110019323127
  • AvaleMEFalzoneTLGelmanDMLowMJGrandyDKRubinsteinMThe dopamine D4 receptor is essential for hyperactivity and impaired behavioral inhibition in a mouse model of attention deficit/hyperactivity disorderMol Psychiatry20049771872614699433
  • RubinsteinMCepedaCHurstRSDopamine D4 receptor-deficient mice display cortical hyperexcitabilityJ Neurosci200121113756376311356863
  • SeamanMIFisherJBChangFKiddKKTandem duplication polymorphism upstream of the dopamine D4 receptor gene (DRD4)Am J Med Genet199988670570910581493
  • KereszturiEKiralyOCsapoZAssociation between the 120-bp duplication of the dopamine D4 receptor gene and attention deficit hyperactivity disorder: Genetic and molecular analysesAm J Med Genet B Neuropsychiatr Genet2007144B223123617171658
  • YangJWJangWSHongSDA case-control association study of the polymorphism at the promoter region of the DRD4 gene in Korean boys with attention deficit-hyperactivity disorder: Evidence of association with the -521 C/T SNPProg Neuropsychopharmacol Biol Psychiatry200832124324817850946
  • GradyDLHarxhiASmithMSequence variants of the DRD4 gene in autism: Further evidence that rare DRD4 7R haplotypes are ADHD specificAm J Med Genet B Neuropsychiatr Genet2005136B1333515892149
  • LaHosteGJSwansonJMWigalSBDopamine D4 receptor gene polymorphism is associated with attention deficit hyperactivity disorderMol Psychiatry1996121211249118321
  • FaraoneSVDoyleAEMickEBiedermanJMeta-analysis of the association between the 7-repeat allele of the dopamine D(4) receptor gene and attention deficit hyperactivity disorderAm J Psychiatry200115871052105711431226
  • LiDShamPCOwenMJHeLMeta-analysis shows significant association between dopamine system genes and attention deficit hyperactivity disorder (ADHD)Hum Mol Genet200615142276228416774975
  • MaherBSMarazitaMLFerrellREVanyukovMMDopamine system genes and attention deficit hyperactivity disorder: A meta-analysisPsychiatr Genet200212420721512454525
  • WohlMPurper-OuakilDMourenMCAdesJGorwoodPMeta-analysis of candidate genes in attention-deficit hyperactivity disorderEncephale2005314 Pt 1437447 French16389711
  • CurranSMillJShamPQTL association analysis of the DRD4 exon 3 VNTR polymorphism in a population sample of children screened with a parent rating scale for ADHD symptomsAm J Med Genet2001105438739311378855
  • Lasky-SuJLangeCBiedermanJFamily-based association analysis of a statistically derived quantitative traits for ADHD reveal an association in DRD4 with inattentive symptoms in ADHD individualsAm J Med Genet B Neuropsychiatr Genet2008147B110010617579349
  • MillJXuXRonaldAQuantitative trait locus analysis of candidate gene alleles associated with attention deficit hyperactivity disorder (ADHD) in five genes: DRD4, DAT1, DRD5, SNAP-25, and 5HT1BAm J Med Genet B Neuropsychiatr Genet2005133B1687315578613
  • ToddRDNeumanRJLobosEAJongYJReichWHeathACLack of association of dopamine D4 receptor gene polymorphisms with ADHD subtypes in a population sample of twinsAm J Med Genet2001105543243811449395
  • GottesmanIIGouldTDThe endophenotype concept in psychiatry: Etymology and strategic intentionsAm J Psychiatry20031604636512668349
  • FlintJMunafoMRThe endophenotype concept in psychiatric geneticsPsychol Med200737216318016978446
  • DoyleAEWillcuttEGSeidmanLJAttention-deficit/hyperactivity disorder endophenotypesBiol Psychiatry200557111324133515950005
  • RommelseNNEndophenotypes in the genetic research of ADHD over the last decade: Have they lived up to their expectations?Expert Rev Neurother20088101425142918928337
  • Slaats-WillemseDSwaab-BarneveldHde SonnevilleLvan der MeulenEBuitelaarJDeficient response inhibition as a cognitive endophenotype of ADHDJ Am Acad Child Adolesc Psychiatry200342101242124814560175
  • BidwellLCWillcuttEGDefriesJCPenningtonBFTesting for neuropsychological endophenotypes in siblings discordant for attention-deficit/hyperactivity disorderBiol Psychiatry200762999199817585884
  • BitsakouPPsychogiouLThompsonMSonuga-BarkeEJDelay aversion in attention deficit/hyperactivity disorder: An empirical investigation of the broader phenotypeNeuropsychologia200947244645618929587
  • KebirOTabbaneKSenguptaSJooberRCandidate genes and neuropsychological phenotypes in children with ADHD: Review of association studiesJ Psychiatry Neurosci20093428810119270759
  • SwansonJOosterlaanJMuriasMAttention deficit/hyperactivity disorder children with a 7-repeat allele of the dopamine receptor D4 gene have extreme behavior but normal performance on critical neuropsychological tests of attentionProc Natl Acad Sci U S A20009794754475910781080
  • LangleyKMarshallLvan den BreeMAssociation of the dopamine D4 receptor gene 7-repeat allele with neuropsychological test performance of children with ADHDAm J Psychiatry2004161113313814702261
  • ManorITyanoSEisenbergJBachner-MelmanRKotlerMEbsteinRPThe short DRD4 repeats confer risk to attention deficit hyperactivity disorder in a family-based design and impair performance on a continuous performance test (TOVA)Mol Psychiatry20027779079412192625
  • WaldmanIDStatistical approaches to complex phenotypes: Evaluating neuropsychological endophenotypes for attention-deficit/hyperactivity disorderBiol Psychiatry200557111347135615950007
  • SheeseBEVoelkerPMRothbartMKPosnerMIParenting quality interacts with genetic variation in dopamine receptor D4 to influence temperament in early childhoodDev Psychopathol20071941039104617931433
  • MoffittTECaspiARutterMStrategy for investigating interactions between measured genes and measured environmentsArch Gen Psychiatry200562547348115867100
  • RutterMSilbergJGene-environment interplay in relation to emotional and behavioral disturbanceAnnu Rev Psychol20025346349011752493
  • BrookesKJMillJGuindaliniCA common haplotype of the dopamine transporter gene associated with attention-deficit/hyperactivity disorder and interacting with maternal use of alcohol during pregnancyArch Gen Psychiatry2006631748116389200
  • WeindrichDLauchtMEsserGSchmidtMHMarital discord and early child developmentActa Paedopsychiatr19925541871921283482
  • NeumanRJLobosEReichWHendersonCASunLWToddRDPrenatal smoking exposure and dopaminergic genotypes interact to cause a severe ADHD subtypeBiol Psychiatry200761121320132817157268
  • LangleyKTuricDRiceFTesting for gene x environment interaction effects in attention deficit hyperactivity disorder and associated antisocial behaviorAm J Med Genet B Neuropsychiatr Genet2008147B1495317579368
  • SeegerGSchlossPSchmidtMHRuter-JungfleischAHennFAGene-environment interaction in hyperkinetic conduct disorder (HD = CD) as indicated by season of birth variations in dopamine receptor (DRD4) gene polymorphismNeurosci Lett2004366328228615288435
  • Bakermans-KranenburgMJVanIMHPijlmanFTMesmanJJufferFExperimental evidence for differential susceptibility: Dopamine D4 receptor polymorphism (DRD4 VNTR) moderates intervention effects on toddlers’ externalizing behavior in a randomized controlled trialDev Psychol200844129330018194028
  • AmaraSGKuharMJNeurotransmitter transporters: Recent progressAnnu Rev Neurosci19931673938096377
  • FrazerAGerhardtGADawsLCNew views of biogenic amine transporter function: Implications for neuropsychopharmacologyInt J Neuropsychopharmacol19992430532011285147
  • GainetdinovRRJonesSRFumagalliFWightmanRMCaronMGRe-evaluation of the role of the dopamine transporter in dopamine system homeostasisBrain Res Brain Res Rev1998262–31481539651511
  • CiliaxBJHeilmanCDemchyshynLLThe dopamine transporter: immunochemical characterization and localization in brainJ Neurosci1995153 Pt 1171417237534339
  • FreedCRevayRVaughanRADopamine transporter immunoreactivity in rat brainJ Comp Neurol199535923403497499533
  • VandenberghDJPersicoAMHawkinsALHuman dopamine transporter gene (DAT1) maps to chromosome 5p15.3 and displays a VNTRGenomics1992144110411061478653
  • MitchellRJHowlettSEarlLDistribution of the 3’ VNTR polymorphism in the human dopamine transporter gene in world populationsHum Biol200072229530410803661
  • FukeSSuoSTakahashiNKoikeHSasagawaNIshiuraSThe VNTR polymorphism of the human dopamine transporter (DAT1) gene affects gene expressionPharmacogenomics J20011215215611911442
  • MillJAshersonPBrowesCD’SouzaUCraigIExpression of the dopamine transporter gene is regulated by the 3’ UTR VNTR: Evidence from brain and lymphocytes using quantitative RT-PCRAm J Med Genet2002114897597912457396
  • MillerGMMadrasBKPolymorphisms in the 3’-untranslated region of human and monkey dopamine transporter genes affect reporter gene expressionMol Psychiatry200271445511803445
  • GreenwoodTAKelsoeJRPromoter and intronic variants affect the transcriptional regulation of the human dopamine transporter geneGenomics200382551152014559208
  • HeinzAGoldmanDJonesDWGenotype influences in vivo dopamine transporter availability in human striatumNeuropsychopharmacology200022213313910649826
  • van DyckCHMalisonRTJacobsenLKIncreased dopamine transporter availability associated with the 9-repeat allele of the SLC6A3 geneJ Nucl Med200546574575115872345
  • KimmelHLCarrollFIKuharMJDopamine transporter synthesis and degradation rate in rat striatum and nucleus accumbens using RTI-76Neuropharmacology200039457858510728879
  • XiaYGoebelDJKapatosGBannonMJQuantitation of rat dopamine transporter mRNA: Effects of cocaine treatment and withdrawalJ Neurochem1992593117911821494906
  • ChenCKChenSLMillJThe dopamine transporter gene is associated with attention deficit hyperactivity disorder in a Taiwanese sampleMol Psychiatry20038439339612740596
  • GillMDalyGHeronSHawiZFitzgeraldMConfirmation of association between attention deficit hyperactivity disorder and a dopamine transporter polymorphismMol Psychiatry1997243113139246671
  • RomanTSchmitzMPolanczykGEizirikMRohdeLAHutzMHAttention-deficit hyperactivity disorder: A study of association with both the dopamine transporter gene and the dopamine D4 receptor geneAm J Med Genet2001105547147811449401
  • ToddRDJongYJLobosEAReichWHeathACNeumanRJNo association of the dopamine transporter gene 3’ VNTR polymorphism with ADHD subtypes in a population sample of twinsAm J Med Genet2001105874574811803523
  • CurranSMillJTahirEAssociation study of a dopamine transporter polymorphism and attention deficit hyperactivity disorder in UK and Turkish samplesMol Psychiatry20016442542811443527
  • AshersonPBrookesKFrankeBConfirmation that a specific haplotype of the dopamine transporter gene is associated with combined-type ADHDAm J Psychiatry2007164467467717403983
  • MugliaPJainUInksterBKennedyJLA quantitative trait locus analysis of the dopamine transporter gene in adults with ADHDNeuropsychopharmacology200227465566212377402
  • CornishKMWildingJMHollisCVisual search performance in children rated as good or poor attenders: The differential impact of DAT1 genotype, IQ, and chronological ageNeuropsychology200822221722518331164
  • ToddRDHuangHSmalleySLCollaborative analysis of DRD4 and DAT genotypes in population-defined ADHD subtypesJ Child Psychol Psychiatry200546101067107316178930
  • LooSKSpecterESmolenAHopferCTealePDReiteMLFunctional effects of the DAT1 polymorphism on EEG measures in ADHDJ Am Acad Child Adolesc Psychiatry200342898699312874502
  • Sonuga-BarkeEJSergeantJANiggJWillcuttEExecutive dysfunction and delay aversion in attention deficit hyperactivity disorder: Nosologic and diagnostic implicationsChild Adolesc Psychiatr Clin N Am200817236738418295151
  • RommelseNNAltinkMEArias-VasquezAA review and analysis of the relationship between neuropsychological measures and DAT1 in ADHDAm J Med Genet B Neuropsychiatr Genet2008147B81536154618729135
  • KahnRSKhouryJNicholsWCLanphearBPRole of dopamine transporter genotype and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and oppositional behaviorsJ Pediatr2003143110411012915833
  • BeckerKEl-FaddaghMSchmidtMHEsserGLauchtMInteraction of dopamine transporter genotype with prenatal smoke exposure on ADHD symptomsJ Pediatr2008152226326918206700
  • LauchtMSkowronekMHBeckerKInteracting effects of the dopamine transporter gene and psychosocial adversity on attention-defiit/hyperactivity disorder symptoms among 15-year-olds from a high-risk community sampleArch Gen Psychiatry200764558559017485610
  • Sonuga-BarkeEJOadesRDPsychogiouLDopamine and serotonin transporter genotypes moderate sensitivity to maternal expressed emotion: The case of conduct and emotional problems in attention deficit/hyperactivity disorderJ Child Psychol Psychiatry20095091052106319490304
  • StevensSEKumstaRKreppnerJMBrookesKJRutterMSonuga-BarkeEJDopamine transporter gene polymorphism moderates the effects of severe deprivation on ADHD symptoms: Developmental continuities in gene-environment interplayAm J Med Genet B Neuropsychiatr Genet2009150B675376119655343
  • BradleyCThe behavior of children receiving benzedrineAm J Psychiatry193794577585
  • ChavezBSopkoMAJrEhretMJAn update on central nervous system stimulant formulations in children and adolescents with attention-deficit/hyperactivity disorderAnn Pharmacother20094361084109519470858
  • MaloneMASwansonJMEffects of methylphenidate on impulsive responding in children with attention-deficit hyperactivity disorderJ Child Neurol1993821571638505479
  • PatrickKSStraughnABPerkinsJSGonzalezMAEvolution of stimulants to treat ADHD: Transdermal methylphenidateHum Psychopharmacol200924111719051222
  • WilensTEBiedermanJPrinceJSix-week, double-blind, placebo-controlled study of desipramine for adult attention deficit hyperactivity disorderAm J Psychiatry19961539114711538780417
  • SpencerTBiedermanJHeiligensteinJAn open-label, dose-ranging study of atomoxetine in children with attention deficit hyperactivity disorderJ Child Adolesc Psychopharmacol200111325126511642475
  • Sonuga-BarkeEJVan LierPSwansonJMHeterogeneity in the pharmacodynamics of two long-acting methylphenidate formulations for children with attention deficit/hyperactivity disorder. A growth mixture modelling analysisEur Child Adolesc Psychiatry200817424525418071840
  • CoghillDRRhodesSMMatthewsKThe neuropsychological effects of chronic methylphenidate on drug-naive boys with attention-deficit/hyperactivity disorderBiol Psychiatry200762995496217543895
  • CornforthCCoghillDSonuga-BarkeESex and age effects in MPH response in ADHDPsychopharmacology (Berl)2010In press
  • Sonuga-BarkeEJCoghillDMarkowitzJSSwansonJMVandenbergheMHatchSJSex differences in the response of children with ADHD to once-daily formulations of methylphenidateJ Am Acad Child Adolesc Psychiatry200746670171017513982
  • SteinMAMcGoughJJThe pharmacogenomic era: Promise for personalizing attention deficit hyperactivity disorder therapyChild Adolesc Psychiatr Clin N Am200817247549018295157
  • LevyFWhat do dopamine transporter and catechol-o-methyltransferase tell us about attention deficit-hyperactivity disorder? Pharmacogenomic implicationsAust N Z J Psychiatry2007411101617464676
  • WinsbergBGComingsDEAssociation of the dopamine transporter gene (DAT1) with poor methylphenidate responseJ Am Acad Child Adolesc Psychiatry199938121474147710596245
  • RomanTSzobotCMartinsSBiedermanJRohdeLAHutzMHDopamine transporter gene and response to methylphenidate in attention-deficit/hyperactivity disorderPharmacogenetics200212649749912172219
  • CheonKARyuYHKimJWChoDYThe homozygosity for 10-repeat allele at dopamine transporter gene and dopamine transporter density in Korean children with attention deficit hyperactivity disorder: Relating to treatment response to methylphenidateEur Neuropsychopharmacol20051519510115572278
  • KirleyALoweNHawiZAssociation of the 480 bp DAT1 allele with methylphenidate response in a sample of Irish children with ADHDAm J Med Genet B Neuropsychiatr Genet2003121B1505412898575
  • SteinMAWaldmanIDSarampoteCSDopamine transporter genotype and methylphenidate dose response in children with ADHDNeuropsychopharmacology20053071374138215827573
  • JooberRGrizenkoNSenguptaSDopamine transporter 3’-UTR VNTR genotype and ADHD: A pharmaco-behavioural genetic study with methylphenidateNeuropsychopharmacology20073261370137617063150
  • LangleyKTuricDPeirceTRNo support for association between the dopamine transporter (DAT1) gene and ADHDAm J Med Genet B Neuropsychiatr Genet2005139B171016082688
  • McGoughJMcCrackenJSwansonJPharmacogenetics of methylphenidate response in preschoolers with ADHDJ Am Acad Child Adolesc Psychiatry200645111314132217023870
  • TharoorHLobosEAToddRDReiersenAMAssociation of dopamine, serotonin, and nicotinic gene polymorphisms with methylphenidate response in ADHDAm J Med Genet B Neuropsychiatr Genet2008147B452753017948872
  • van der MeulenEMBakkerSCPaulsDLHigh sibling correlation on methylphenidate response but no association with DAT1-10R homozygosity in Dutch sibpairs with ADHDJ Child Psychol Psychiatry200546101074108016178931
  • ZeniCPGuimaraesAPPolanczykGVNo significant association between response to methylphenidate and genes of the dopaminergic and serotonergic systems in a sample of Brazilian children with attention-deficit/hyperactivity disorderAm J Med Genet B Neuropsychiatr Genet2007144B339139417171656
  • HamarmanSFossellaJUlgerCBrimacombeMDermodyJDopamine receptor 4 (DRD4) 7-repeat allele predicts methylphenidate dose response in children with attention deficit hyperactivity disorder: A pharmacogenetic studyJ Child Adolesc Psychopharmacol200414456457415662148
  • CheonKAKimBNChoSCAssociation of 4-repeat allele of the dopamine D4 receptor gene exon III polymorphism and response to methylphenidate treatment in Korean ADHD childrenNeuropsychopharmacology20073261377138317077808
  • FroehlichTEMcGoughJJSteinMAProgress and promise of attention-deficit hyperactivity disorder pharmacogeneticsCNS Drugs20102429911720088618
  • McCarthyMIAbecasisGRCardonLRGenome-wide association studies for complex traits: Consensus, uncertainty and challengesNat Rev Genet20089535636918398418
  • McCarthyMIHirschhornJNGenome-wide association studies: Potential next steps on a genetic journeyHum Mol Genet200817R2R156R16518852205
  • MaherBPersonal genomes: The case of the missing heritabilityNature20084567218182118987709
  • FrankeBNealeBMFaraoneSVGenome-wide association studies in ADHDHum Genet20091261135019384554
  • LeungPWLeeCCHungSFDopamine receptor D4 (DRD4) gene in Han Chinese children with attention-deficit/hyperactivity disorder (ADHD): Increased prevalence of the 2-repeat alleleAm J Med Genet B Neuropsychiatr Genet2005133B1545615578612
  • GabrielaMLJohnDGMagadelaBVGenetic interaction analysis for DRD4 and DAT1 genes in a group of Mexican ADHD patientsNeurosci Lett2009451325726019146920
  • GordonDHaynesCJohnnidisCPatelSBBowcockAMOttJA transmission disequilibrium test for general pedigrees that is robust to the presence of random genotyping errors and any number of untyped parentsEur J Hum Genet200412975276115162128
  • CurtisDShamPCA note on the application of the transmission disequilibrium test when a parent is missingAm J Hum Genet19955638118127887437
  • SwansonJMMoyzisRKMcGoughJJEffects of source of DNA on genotyping success rates and allele percentages in the Preschoolers with Attention-Deficit/Hyperactivity Disorder Treatment Study (PATS)J Child Adolesc Psychopharmacol200717563564617979583
  • WestALangleyKHamshereMLEvidence to suggest biased phenotypes in children with attention deficit hyperactivity disorder from completely ascertained triosMol Psychiatry20027996296612399949
  • PearsonTAManolioTAHow to interpret a genome-wide association studyJAMA2008299111335134418349094
  • WeedonMNLangoHLindgrenCMGenome-wide association analysis identifies 20 loci that influence adult heightNat Genet200840557558318391952
  • VisscherPMSizing up human height variationNat Genet200840548949018443579
  • PritchardJKCoxNJThe allelic architecture of human disease genes: common disease-common variant...or not?Hum Mol Genet200211202417242312351577
  • SchorkNJMurraySSFrazerKATopolEJCommon vs rare allele hypotheses for complex diseasesCurr Opin Genet Dev200919321221919481926
  • RobinsonRCommon disease, multiple rare (and distant) variantsPLoS Biol201081e100029320126253
  • DicksonSPWangKKrantzIHakonarsonHGoldsteinDBRare variants create synthetic genome-wide associationsPLoS Biol201081e100029420126254
  • DeFriesJCFulkerDWMultiple regression analysis of twin dataBehav Genet19851554674734074272
  • SwansonJDeutschCCantwellDGenes and attention-deficit hyperactivity disorderClin Neurosci Res20011217216
  • ZametkinADopamine reward pathway in adult ADHD (Letter to editor)JAMA2010303323223420085952
  • HayDABennettKSLevyFSergeantJSwansonJA twin study of attention-deficit/hyperactivity disorder dimensions rated by the strengths and weaknesses of ADHD-symptoms and normal-behavior (SWAN) scaleBiol Psychiatry200761570070516962074
  • YoungDJLevyFMartinNCHayDAAttention deficit hyperactivity disorder: A Rasch analysis of the SWAN Rating ScaleChild Psychiatry Hum Dev200940454355919455417
  • AulchenkoYSStruchalinMVBelonogovaNMPredicting human height by Victorian and genomic methodsEur J Hum Genet20091781070107519223933
  • GaudermanWJSample size requirements for matched case-control studies of gene-environment interactionStat Med2002211355011782049
  • GluckmanPDHansonMACooperCThornburgKLEffect of in utero and early-life conditions on adult health and diseaseN Engl J Med20083591617318596274
  • LouHCEtiology and pathogenesis of attention-deficit hyperactivity disorder (ADHD): Significance of prematurity and perinatal hypoxic-haemodynamic encephalopathyActa Paediatr19968511126612718955450
  • Rosa NetoPLouHCummingPPrydsOGjeddeAMethylphenidate-evoked potentiation of extracellular dopamine in the brain of adolescents with premature birth: Correlation with attentional deficitAnn N Y Acad Sci200296543443912105118