Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2014 Mar 20;77(4):654–672. doi: 10.1111/bcp.12312

Pharmacogenomics in psychiatry: the relevance of receptor and transporter polymorphisms

Gavin P Reynolds 1, Olga O McGowan 2, Caroline F Dalton 1
PMCID: PMC3971982  PMID: 24354796

Abstract

The treatment of severe mental illness, and of psychiatric disorders in general, is limited in its efficacy and tolerability. There appear to be substantial interindividual differences in response to psychiatric drug treatments that are generally far greater than the differences between individual drugs; likewise, the occurrence of adverse effects also varies profoundly between individuals. These differences are thought to reflect, at least in part, genetic variability. The action of psychiatric drugs primarily involves effects on synaptic neurotransmission; the genes for neurotransmitter receptors and transporters have provided strong candidates in pharmacogenetic research in psychiatry. This paper reviews some aspects of the pharmacogenetics of neurotransmitter receptors and transporters in the treatment of psychiatric disorders. A focus on serotonin, catecholamines and amino acid transmitter systems reflects the direction of research efforts, while relevant results from some genome-wide association studies are also presented. There are many inconsistencies, particularly between candidate gene and genome-wide association studies. However, some consistency is seen in candidate gene studies supporting established pharmacological mechanisms of antipsychotic and antidepressant response with associations of functional genetic polymorphisms in, respectively, the dopamine D2 receptor and serotonin transporter and receptors. More recently identified effects of genes related to amino acid neurotransmission on the outcome of treatment of schizophrenia, bipolar illness or depression reflect the growing understanding of the roles of glutamate and γ-aminobutyric acid dysfunction in severe mental illness. A complete understanding of psychiatric pharmacogenomics will also need to take into account epigenetic factors, such as DNA methylation, that influence individual responses to drugs.

Keywords: antidepressant drugs, antipsychotic drugs, bipolar illness, depression, gene, schizophrenia

Introduction

Drug action in psychiatric disease

The development and introduction of specific drug treatments for schizophrenia, depression and bipolar disorder in the middle of the last century revolutionized the way in which these disorders are managed. Nevertheless, severe mental illness still represents a huge burden to society in general and healthcare in particular. This reflects the limited efficacy of current treatment; while the past half-century has seen further developments in the drug treatment of psychosis and mood disorders, there has been little improvement in efficacy. Most newly introduced drugs have offered incremental changes, primarily in side-effect profile rather than symptom response. Even now, the pharmacotherapy of depression achieves response rates of little more than 50%, while even lower proportions of patients with schizophrenia achieve adequate response to their antipsychotic drug treatment.

Thus, while there may be small differences between psychiatric drug treatments in terms of their efficacy, there are clearly major differences between individuals in their response to these treatments. Although the psychiatric drugs may differ more in their adverse effect profiles than their efficacies, again the experience of adverse effects varies profoundly between individuals. These unexplained differences in response and side-effects are thought to reflect, at least in part, genetic differences between patients; thus, pharmacogenetics in psychiatry has much to offer in the eventual identification of risk factors for these limitations of drug treatment. Furthermore, it should be born in mind that the information exchange between pharmacology and pharmacogenetics is a two-way process; while our understanding of psychopharmacology provides us with good candidates for pharmacogenetic study, pharmacogenetic findings can also provide clues relating to underlying pharmacological mechanisms.

The pharmacological mechanisms underlying the actions of the major psychiatric drugs are at least partly understood. Thus, the antipsychotic drugs act at dopamine D2 receptors to diminish dopaminergic neurotransmission in the mesolimbic pathway, resulting in relief of some psychotic and manic symptoms. Dopamine D2 receptor blockade in other pathways is also responsible for certain side-effects of these drugs, namely hyperprolactinaemia and the acute extrapyramidal motor symptoms (EPS). The antidepressant action of the selective serotonin reuptake inhibitors (SSRIs) is thought to be the consequence of an initial increase in synaptic serotonin which, via effects including changes in receptor expression, results in alterations in the activity and regulation of serotoninergic neurons.

However, there are still many pharmacological mechanisms that remain elusive. The unique efficacy of clozapine in bringing relief to a good proportion of psychotic patients not responding to other antipsychotic drugs is still not well understood, despite over 40 years of research. The role and importance of the effects of antidepressant drugs on neurogenesis is still debated, and the pharmacological mechanisms behind some adverse effects of psychiatric drug treatments are not fully understood.

Nevertheless, the drug treatment of severe mental illness and other psychiatric disorders primarily involves effects on, and/or regulation of, neurotransmission. Consequently, the genes coding for proteins involved in neurotransmitter action have provided valuable candidates for pharmacogenetic research in psychiatry. It is these studies, the association with drug effects of functional polymorphisms in genes involved in the neurochemical processes of synaptic transmission, on which this review will focus. In addition to these important pharmacokinetic aspects of psychiatric pharmacogenetics, variation in genes associated with pharmacodynamic processes, such as the cytochrome P450 drug-metabolizing enzymes, can also influence the effectiveness of psychiatric drug treatments; however, consideration of these is outside the scope of this review.

Neurochemistry of synaptic transmission

Synaptic chemical neurotransmission is the essential mechanism of communication between neurons; hence, it is central to brain function. Its associated neurochemical processes provide the major point at which we can influence neuronal activity through the effects of drug treatment. Each of the presynaptic processes of transmitter synthesis, storage, release and reuptake, the metabolic removal of transmitter, and the various presynaptic and postsynaptic receptor sites are all potential, if not actual, targets for neuro-and psychoactive drugs.

Transmitters are synthesized by the action of enzymes on readily available precursors; generally, these initial enzymes are fairly specific to the neurons containing the neurotransmitter. Thus, tyrosine hydroxylase and tryptophan hydroxylase are responsible for the initial step in the synthesis of catecholamines and serotonin, glutamic acid decarboxylase for γ-aminobutyric acid (GABA) and choline acetyltransferase for acetylcholine. Storage of transmitter in vesicles is effected by vesicular transporters (e.g. VGluT1 and 2 for glutamate; VMAT2 for monoamines in the central nervous system), while other transporters selective for particular transmitters are involved in transmitter reuptake from the synapse. A wide variety of specific neurotransmitter receptors mediate synaptic neurotransmission or modulate presynaptic neuronal activity and transmitter release. Finally, enzymes such as monoamine oxidase (MAO), catechol-O-methyltransferase and acetylcholinesterase provide a mechanism for metabolic inactivation of neurotransmitter molecules. This is a substantial list of processes, with many candidate proteins to study; this review will focus solely on the two processes involving proteins that recognize the neurotransmitter in the synapse, i.e. receptors and transporters. While receptors mediate the action of neurotransmitters on postsynaptic cells, and occasionally elsewhere, the reuptake transporters are the major mechanism for removal of the transmitter from the synapse, limiting its action.

This restricted focus is inevitably a limitation of our review, undertaken in order to provide a manageable and comprehensible, rather than comprehensive, overview of the current ‘state of the art’. For the same reason, we have not undertaken a systematic approach, but rather provided a critical review with, where possible, some emphasis on genotype functionality. There is a focus on positive findings, again for reasons of space limitation and comprehensibility; it is likely that any attempt to include negative findings systematically will suffer from the fact that many will remain unpublished. Table 1 provides a summary of the findings that we discuss below.

Table 1.

Association studies of single nucleotide polymorphisms in neurotransmitter receptor and transmitter genes with psychiatric treatment response and side-effects

Gene Polymorphism Functional effect of polymorphism Type of study/patient cohort Drug(s) Number of patients Length of study Effect observed Reference
SLC6A4 5-HTTLPR s/l S allele is associated with reduced expression 4,7
Primary/MDD Fluvoxamine 102 6 weeks l allele carriers had better response 7
Primary/depression in elderly Paroxetine 34 12 weeks l allele carriers had better response 8
Primary/depression in elderly Nortriptyline 23 12 weeks No effect 8
Meta-analysis/MDD SSRIs 1435 4 weeks Patients with ss alleles took longer to respond and were less likely to achieve remission 9
Meta-analysis/MDD SSRIs/other antidepressants 5408 varied No effect on response to SSRIs 10
Meta-analysis/MDD Caucasian/Asian SSRIs/other antidepressants 5479 2–18 weeks In Caucasians, l allele carriers on SSRIs had better response and remission; weaker effect was seen in an Asian population on mixed antidepressants 13
Primary/MDD (STAR*D study) Citalopram 1762 Varied, with side-effects No association with treatment outcome; l allele was associated with side-effects 17
Primary/MDD Paroxetine 124 2–7 weeks s allele was associated with severe adverse events during study 18
Primary/MDD Mirtazapine 122 2–7 weeks ll genotype was associated with adverse side-effects 18
Primary/MDD/anxiety in adolescents and children Citalopram 83 8 weeks l allele in combination with −703G SNP in tryptophan hydroxylase gene was associated with improved treatment response 21
SLC6A4 rs25531 A/G in l allele of 5-HTTLPR G allele associated with reduced expression 5
Primary/dementia Citalopram Risperidone 92 12 weeks s allele of 5-HTTLPR in combination with G allele of rs25531 predicted greater side-effects 20
SLC6A4 STin2 VNTR Influences gene expression
Systematic review/MDD SSRIs 650 2–18 weeks Greater effect in Asians than seen with 5-HTTLPR 14
Meta-analysis/MDD SSRIs/other antidepressants 1546 Varied No effect on treatment response seen 15
Primary/bipolar Lithium 122 Varied Treatment response in combination with HTTLPR 16
Primary/premature ejaculation Sertraline 246 12 weeks 12/12 genotype was associated with treatment response 19
HTR1A –1019 C/G rs6295 G allele is associated with increased receptor expression and reduced neuronal transmission 22
Review/MDD SSRIs/other antidepressants NA NA G allele was associated with poorer response 24
Primary/panic disorder Sertraline/paroxetine 102 6 weeks C allele was associated with improved initial response 25
Primary/schizophrenia Risperidone 130 8 weeks G allele was associated with poorer response 27
Primary/schizophrenia Mixed antipsychotics 130 4 weeks G allele was associated with poorer response 28
HTR1B rs6298 A/G Functional relevance unknown
Primary/MDD SSRIs/other antidepressants 308 6 weeks AA genotype was associated with poorer response to treatment; interaction with negative life events 29
HTR2A –1438 A/G rs6311 102 T/C rs6313 SNPs are in linkage disquilibrium; 102 C allele is associated with lower expression 30
Primary/schizophrenia Risperidone 100 6 weeks 102 CC genotype was associated with better clinical response 31
Primary/schizophrenia Typical antipsychotics 102 Long term 102 CC genotype was associated with poorer clinical response 32
Meta-analysis/schizophrenia Clozapine 733 Varied 102 CC genotype was associated with poorer clinical response 33
Primary/MDD (STAR*D study) Citalopram 1953 6 weeks AA genotype was more likely to respond to treatment 35
Primary/MDD (Korean cohort) Citalopram 71 4 weeks GG genotype was more likely to respond to treatment 36
Primary/schizophrenia Typical/mixed antipsychotics 635 Varied 102 CC genotype was associated with increased risk of tardive dyskinesia 38
Primary/schizophrenia Olanzapine 164 Varied 102 T allele was associated with weight gain 46
HTR2C –759C/T rs3813929 Influences gene expression 44,45
Primary/schizophrenia Antipsychotics 96 10 weeks C allele was associated with weight gain 42
HTR4 rs1011427 C/T Intronic SNP, functional relevance unknown
Primary/schizophrenia (CATIE study) Antipsychotics 400+ 8 weeks Treatment response – cognitive measures 34
HTR7 rs7916403 G/T Intronic SNP, functional relevance unknown
Primary/alcohol dependence NA 1064 NA G allele is associated with alcohol dependence 47
DRD2 Taq1A rs1800497 1A carriers have reduced striatal dopamine D2 receptor density and reduced dopaminergic activity 51
Meta-analysis/schizophrenia Antipsychotics 748 Varied No association seen with response to treatment 59
Primary/schizophrenia Antipsychotics 206 6 weeks Taq1A polymorphism is associated with weight gain 60
DRD2 –141 ins/del Del allele is associated with lower dopamine D2 receptor expression 53,54
Meta-analysis/schizophrenia Antipsychotics 687 8 weeks Del allele is associated with poorer response to treatment 59
Primary/schizophrenia Risperidone Olanzapine 58 6 weeks Del allele is associated with weight gain 61
DRD3 Gly/Ser rs6280 Gly allele is associated with increased binding of dopamine 67
Schizophrenia Clozapine, risperidone 32; 75; 68; 100 Varied Studies 68 and 69 show that Gly allele is associated with improved treatment; 70 and 71 find no effect 6871
Meta-analyses/schizophrenia Antipsychotics 1610; 2126 Varied Risk of tardive dyskinesia was associated with Gly allele in early studies; effect not seen in later studies 75,76
Primary/schizophrenia (CATIE study) Antipsychotics 710 Varied No effect of polymorphism on tardive dyskinesia risk 77
SLC6A3/DAT1 VNTR 2–11 repeats Conflicting evidence of effect of repeat number on binding 78
Primary/ADHD Methylphenidate 11 8 weeks 10-repeat was associated with poorer treatment outcome 80
Meta-analysis/ADHD Methylphenidate 1572 varied No effect of repeat number on treatment response 81
Primary/schizophrenia Clozapine 160 8 weeks Effect on treatment response 82
Primary/schizophrenia Risperidone 130 8 weeks No effect of repeat number on treatment response 83
Primary/MDD SSRIs 190 3 weeks Poorer response associated with nine-repeat allele 84
ADRA1A –4884 A/G rs922733 May influence gene expression
Primary/schizophrenia Clozapine 289 3 months No effect of polymorphism on treatment response 85
Primary/schizophrenia Risperidone/other antipsychotics 427 11 months AA genotype was associated with weight gain 88
ADRA2A –1291 C/G rs1800544 May influence gene expression
Primary/schizophrenia Clozapine 93 Varied G allele was associated with weight gain 90
Primary/schizophrenia Clozapine 129 6–14 weeks C allele was associated with weight gain 91
Primary/schizophrenia Antipsychotics 139 6–14 weeks SNP only has an effect on weight gain in association with the u-VNTR polymorphism in the MAOA gene 92
Primary/ADHD Methylphenidate 450+ Varied G allele was associated with treatment response in children but not in adults 9398
Primary/MDD Milnacipran 93 4 weeks C allele carriers had improved response to treatment 101
SLC6A2/NET –182 T/C rs2242446 May influence gene expression
Primary/MDD Milnacipran 96 6 weeks T allele was associated with improved response 99
1287 G/A rs5569 May affect transmitter binding or affinity 99
Primary/MDD Milnacipran 96 6 weeks AA genotype was associated with slower response to treatment 99
SLC1A2 (EAAT2) rs4354668 −181 A/C (T/G) C allele is associated with lower transporter activity 104
Primary/bipolar disorder Lithium 110 ≥6 months TT homozygotes had improved response to treatment 105
SLC1A1 (EAAT3) rs2228622 A/G rs3780413 C/G rs 3780412 A/G Synonymous exonic and intronic SNPs, functionality unknown 106
Primary/schizophrenia Mixed antipsychotics 94 ≥24 months A/C/G haplotype is associated with increased risk of development of obsessive–compulsive disorder 107
GRIK4 rs1954787 C/T Intronic SNP may influence gene expression 109
Primary/MDD (STAR*D study) Citalopram 1816 Varied C allele was predictive of improved treatment outcome when combined with HTR2A genotype 109
Primary/MDD Antidepressants 387 5 weeks G allele was associated with remission 110
GRIK4 rs 12800734 A/G Intronic SNP may influence gene expression
Primary/MDD Antidepressants 223 Varied No effect of genotype 111
Primary/MDD Antidepressants 281 Varied (≥6 weeks) G allele and the GG genotype are associated with better response 112
GABRP rs10036156 C/T Synonymous exonic SNP, functionality unknown
Primary/MDD Antidepressants 281 Varied (≥6 weeks) G allele was associated with better response, specifically in the SNRI subgroup 112
GABRQ Phe/Ile rs3810651 May influence binding 112
Primary/MDD Antidepressants 281 Varied (≥6 weeks) A allele was associated with better response 112
GABRA6 rs1992647 C/T (G/A) May influence gene expression
Primary/MDD Antidepressants 281 Varied (≥6 weeks) AA was associated with nonresponse 112
GRIA4 rs2513265 A/T May influence gene expression
Primary/schizophrenia Iloperidone 407 Up to 7 months TT genotype was associated with poorer response to treatment 113
Primary/schizophrenia Ziprasidone 409 4 weeks Treatment response (negative result) 114
Primary/schizophrenia Risperidone 145 12 weeks TT genotype was associated with poorer response to treatment 115
GRM3 rs724226 A/G Intronic SNP, functional relevance unknown
Primary/schizophrenia and related disorders Risperidone 143 2–12 weeks G allele was associated with remission and change in PANSS 117
rs274622 C/T May influence gene expression
Primary/schizophrenia Olanzapine 42 6 weeks C allele was associated with improved response to treatment 119
GR1A2 rs3813296 G/T Intronic SNP, functional relevance unknown
Primary/schizophrenia Antipsychotics 221 Varied T allele was associated with poorer response to treatment 120
SLC6A11 rs4684742 A/G Intronic SNP, functional relevance unknown
Primary/schizophrenia Antipsychotics 272 Long term Association of A allele with tardive dyskinesia 123
SLC6A5 rs2298826 A/G Intronic SNP, functional relevance unknown
Primary/schizophrenia Haloperidol 290 4 weeks Dyskinesia and EPS 124

Abbreviations are as follows: ADHD, attention deficit hyperactivity disorder; EPS, extrapyramidal motor symptoms; NA, not applicable; SNP, single nucleotide polymorphism; SSRI, selective serotonin reuptake inhibitor.

Pharmacogenetic approaches

Given that their effects in influencing synaptic transmitter concentrations and/or synaptic transmission, the genes for these proteins provide strong candidate genes for both psychiatric disorders and, inevitably, individual variability in response to psychiatric pharmacotherapy. In the past, psychiatric pharmacogenetics has concentrated particularly on genes coding for the proteins involved in monoamine neurotransmission, because it is at the dopamine, noradrenaline and serotonin synapses that many of the psychoactive drugs have their effects. Hence, these monoamine-associated genes may provide strong candidates for psychiatric pharmacogenetic associations. However, an increasing focus on the amino acid neurotransmitter genes has reflected a growing interest in the roles that disturbances of GABA and glutamate neurotransmission may play in mental illness.

As the relevant technology has developed, such candidate gene approaches, driven by mechanism-based hypotheses, are increasingly being supplemented by hypothesis-free genome-wide association studies (GWAS). These are not without limitations themselves, because the handling and statistical analysis of typically over 100 million data points has its inherent difficulties; the relative lack of interstudy consistency as well as poor consistency with candidate gene findings are further concerns.

Response to treatment with psychiatric drugs is not dichotomous, although it might be treated as such for purposes of data analysis. The apparently continuous variability generally observed in symptom relief or side-effects cannot be explained by a single genetic variant 1 but must involve multiple factors. These may be multiple genes within a transmitter system or pathway, or different additive or interactive effects across systems. Research is beginning to address this multiplicity of risk factors although, as will be apparent, the majority of studies, even when assessing the effects of multiple genes and systems, look for discrete associations rather than additive and interactive genetic influences.

There remains much inconsistency in the results of pharmacogenetic studies; one contributing factor is thought to be inadequate power due to small sample sizes. However, we would argue that the current focus on quantity rather than quality of the sample is misdirected. Pragmatic investigation of patients who have had a variety of previous drug treatments and treatment periods introduces variance in measures of symptoms and side-effects that can be avoided by, for example, studying first episode, initially drug-naïve, subjects. But the large, systematic and prospective multicentre studies of treatment efficacy, such as CATIE for schizophrenia 2 and STAR*D for depression 3, also provide strong and comprehensive clinical data that have proved useful in pharmacogenetic investigations, and some emphasis will be placed on findings from these and similar studies in our review.

The serotonin system

A large proportion, if not the majority, of pharmacogenetic studies has addressed genes governing or influencing serotonin neurotransmission. Perhaps the most studied of these is the 5-hydroxytryptamine (5-HT) transporter (5-HTT) gene (SLC6A4), which has a common length polymorphism (the HTTLPR) that results in a partial deletion of a tandem repeat sequence within the promoter region. This is found to influence activity of the transporter, in which the deletion (short or s allele) is associated with lower expression 4. There are other promoter polymorphisms in SLC6A4, of which one is found within the deletion sequence (rs25531; A/G), where the G allele in the l allele reduces expression in a similar manner to the HTTLPR deletion s allele 5. Another polymorphism has been described in intron 2 of SLC6A4 consisting of a 17 bp variable number of tandem repeats (termed STin2 VNTR). This also has functional activity with an influence on gene expression 6.

As 5-HTT is the main site of action of SSRIs, its gene has been much studied in association with depression and antidepressant action. The pharmacogenetics of the HTTLPR (l/s) polymorphism has been investigated as a potential marker for symptom response to SSRIs. Thus, Smeraldi et al. 7 showed that in patients with major depression, l allele carriers showed a better response to fluvoxamine than homozygotes for the short variant (s/s). Pollock et al. 8 reported a similar effect on treatment response for paroxetine in major depression, but found no effect on treatment outcome in patients treated with the tricyclic antidepressant nortriptyline. Meta-analyses assessing the remission rate, response rate and response within the first 4 week period of treatment with SSRIs 9 revealed that patients with ss alleles took longer to respond and were less likely to achieve remission. However, results are not always clear cut. A meta-analysis of data obtained from 28 studies with 5408 participants found no significant effect of the HTTLPR on response to SSRI medication 10. This may be because opposite effects in Asians have been reported, with the s allele being associated with better SSRI response 11,12.

Porcelli et al. 13 reviewed 19 studies performed on Caucasians and 11 on Asians. Evaluations were performed separately for SSRIs and mixed/other drugs. There was positive association of the l allele with response and remission in Caucasians within the SSRI group and only a marginal association between the l allele and remission in the mixed antidepressant treatment group. In Asians, however, only a small effect of HTTLPR on remission for mixed antidepressants was detected. Interestingly, an early systematic review suggested that while the HTTLPR influenced antidepressant response in Caucasian subjects, it was the STin2 VNTR that had a greater effect in Asians 14. A recent meta-analysis of candidate gene studies of antidepressant efficacy by Niitsu et al. 15 showed, after exclusion of HTTLPR, no major effect of any single gene variant, but some positive association with STin2 driven by one or two studies. Effects of 5-HTT polymorphisms are seen in response to treatments other than antidepressants; Tharoor et al. 16 reported an association of STin2 polymorphism and its combined effect with 5-HTTLPR variants with lithium response in treatment of bipolar disorder.

Results of the major STAR*D study of citalopram treatment have been summarized by Lin and Chen 17. There was no association observed between treatment outcome and HTTLPR in over 1700 subjects, although presence of the l allele was associated with side-effects. Others have also reported association with SSRI side-effects. Murphy et al. 18 found that among paroxetine-treated subjects, s allele carriers experienced more severe adverse events during the course of the study (gastrointestinal complaints, fatigue, agitation, sweating and dizziness), which caused early discontinuation of treatment. However, in mirtazapine-treated subjects, adverse effects were associated with the ll genotype and included drowsiness, dizziness and anxiety. Interestingly, Safarinejad 19 described an association of STin2 12/12 genotype with response to sertraline treatment of premature ejaculation.

Few studies have included the additional variance associated with rs25531 within the l allele (the ‘trialellic genotype’). One study that did is the 12 week randomized controlled trial 20 which showed that low-expression alleles (s and lG) predicted early appearance and overall greater side-effects of citalopram that led to early treatment discontinuation in patients with dementia and behavioural or psychotic symptoms.

A further limitation common to many pharmacogenetic studies is that they rarely acknowledge that additive and interactive effects of multiple genes are likely to contribute to pharmacogenomic associations with drug effects. One study that did shows additive effects of tryptophan hydroxylase-2 (TPH2) and 5-HTT on clinical response to citalopram 21. Subjects carrying a combination of TPH2 –703G and the HTTLPR l alleles were most likely to respond to citalopram.

The 5-HT1A receptor acts as an autoreceptor on serotonergic neurons, where it imparts an inhibitory control over neuronal activity. It is also found postsynaptically in the hippocampus, cortex and basal ganglia. It is a particularly important site in understanding psychiatric drug action; it is the site of action of some anxiolytic drugs, while several newer antipsychotics have 5-HT1A partial agonist activity, implicated in their action on negative symptoms of schizophrenia. However, its greatest relevance to pharmacotherapeutics is its proposed involvement in the delayed antidepressant mechanism of the SSRIs. Thus, the 5-HT1A receptor gene (HTR1A) is a strong candidate to explain individual differences in response to antidepressants, anxiolytics and perhaps antipsychotics.

Of the many identified single nucleotide polymorphisms (SNPs) in HTR1A, only one has been studied to any great extent, namely –1019C/G (rs6295). This SNP, associated with depression and suicidality, occurs at a binding site for transcription factors; the G allele prevents the binding of inhibitory transcription factors, including the repressor Deaf-1, theoretically resulting in an increase in the expression of 5-HT1A autoreceptors with a consequent reduction in 5-HT neuronal transmission 22. In antidepressant pharmacogenetics, Lemonde et al. 23 and subsequently several other studies have, fairly consistently, demonstrated that the –1019G allele is associated with a poorer response to treatment with SSRIs and, in some studies, other antidepressant drugs (reviewed by Drago et al. 24). The effect on SSRI response is not restricted to depression; our study on the initial response of panic disorder to SSRI treatment has also shown a beneficial effect of the C allele 25.

This HTR1A –1019C/G SNP is strongly associated with changes in both negative features and depressive symptoms after 3 months of initial treatment of first-episode psychosis 26. The G allele was again associated with poorer response, a finding replicated in Asian 27 and European samples 28. Thus, the strong functional effect of this SNP has influences that generalize across drug types.

The 5-HT1B subtype is another presynaptic autoreceptor and thus a good candidate for antidepressant function. However, other than a few negative studies, the only notable pharmacogenetic finding is that of Xu et al. 29, who identified an association of two linked HTR1B coding region SNPs with antidepressant response, and an interaction with recent stressful events. While there is little evidence that these synonymous SNPs are functional, the authors mention that they are in linkage disequilibrium with (unstudied) functional promoter polymorphisms, which may therefore mediate an effect on receptor expression.

Perhaps the best studied of 5-HT receptors is 5-HT2A, reflecting the common high affinity of all second-generation antipsychotic drugs for this site, and its postulated role in the clinical effects of these drugs. Antagonism of 5-HT2A with antipsychotics may well contribute to a lower incidence of extrapyramidal motor side-effects. There is also some evidence for their role in antidepressant action; a common consequence of antidepressant drug administration is the downregulation of 5-HT2A receptors. The most studied are the 102T/C, a synonymous coding region SNP, and –1438A/G, a promoter SNP that is in complete linkage with 102T/C and, reportedly, has functional effects on gene expression 30.

Investigation of this genetic factor in relationship to response to treatment has been inconsistent and inconclusive, although from early days of pharmacogenetic studies HTR2A has been considered to be important in the variability in symptom response to antipsychotic drugs. While Lane et al. 31 reported that, in Chinese patients, the 102CC genotype is related to better clinical response to risperidone, particularly of negative symptoms, Joober et al. 32 suggested the 102CC genotype to be more frequent in patients with poor response to typical antipsychotic drugs. Arranz et al. 33 also showed that this genotype is more frequent among European clozapine nonresponders than responders. Perhaps a stronger indication that HTR2A variation may contribute to antipsychotic treatment response comes from the study of the CATIE cohort by Need et al. 34 reporting an intronic SNP in this gene to be one of 22 SNPs, taken from an investigation of 2769 in 118 genes, to associate significantly (P < 0.01) with improvement in Positive and Negative Syndrome Scale (PANSS) scores.

In the STAR*D study, McMahon et al. 35 showed –1438AA homozygotes to have an 18% reduction in absolute risk of having no response to citalopram treatment, compared with those homozygous for the G allele. But, as with the HTTLPR, there may be racial differences in outcomes of antidepressant treatment; Choi et al. 36 reported a better response to citalopram treatment associated with the GG genotype in Korean patients with major depressive disorder.

In addition to treatment response, the 5-HT2A receptor is implicated in side-effects. The reciprocal effects of dopamine and serotonin indicate a role for the latter transmitter in motor function and dysfunction, particularly drug-induced EPS. A small study has shown association of undifferentiated EPS with both HTR2A and HTR2C SNPs 37. Tardive dyskinesia (TD), the severe and chronic EPS which has limited the use of the first generation of antipsychotic drugs, has been found to be associated with the –1438A/G and 102C/T HTR2A polymorphisms, albeit with inconsistent results suggestive of a weak effect. In a large, multicentre study, Lerer et al. 38 found that the 102C/T genotype was significantly associated with TD in older patients and in those with non-orofacial symptoms, illustrating the multiplicity of factors, in addition to ethnicity, that may influence the relationship between genotype and this phenotype.

Genetic variation of the related 5-HT2C receptor may also be weakly related to TD. Here a missense coding region SNP (ser23gly), common in Caucasians but not in Asian populations, is inconsistently reported to associate with occurrence of TD 39. Interestingly, there are additive effects reported between SNPs in the dopamine receptor gene DRD3 and HTR2C 40, illustrating at the level of genetic variation the close interaction between dopamine and serotonin systems in EPS.

Polymorphisms in the 5-HT2C receptor gene are most associated with individual variability in metabolic side-effects, particularly antipsychotic drug-induced weight gain. Antagonism of this receptor is certainly implicated in the pharmacological mechanism of weight gain following treatment with some drugs 41, and it is the site of agonist action of a recently introduced anti-obesity drug, lorcaserin. An early study of a promoter region HTR2C SNP showed an association with weight gain after initial antipsychotic drug treatments 42, which has subsequently been replicated many times over (reviewed by Zhang et al. and Reynolds 39,43). The studied –759C/T polymorphism, along with other promoter region polymorphisms with which it is in linkage disequilibrium, appears to be functional in influencing gene expression 44. Interestingly, the genetic association generalizes to different drugs, including those with both high (clozapine and olanzapine) and relatively low affinity (risperidone) for the 5-HT2C receptor. Thus, the functional effects of the polymorphism may involve mechanisms other than solely mediating the direct effects of drugs at this receptor, perhaps in modifying the downstream physiological control of body weight, which is disrupted by drug action. One clue concerning the mechanism is the association of the –759C/T polymorphism with circulating levels of the hormone leptin, which is important in the control of body weight and also implicated in weight gain pharmacogenetics 45.

Of course, this is only one of many SNPs identified as risk factors for drug-induced weight gain and related metabolic pathology 43. Although one might expect influences from other 5-HT-related genetic factors, there have been few consistent reports of associations with 5-HTT or other 5-HT receptor SNPs, except for some HTR2A findings 46. However, another gene of relevance associated with metabolic side-effects is ADRA2A, as mentioned in the next section. The protein encoded by ADRA2A, the α2A-adrenoceptor, is found as a heteroceptor on serotoninergic neurons; polymorphisms in this gene may thus influence the activity of those neurons in their control of body weight.

Of the other serotonin receptors, 5-HT4, 5-HT6 and 5-HT7 have been particularly implicated in affective behaviour and/or cognitive function. Polymorphisms in each gene have been investigated for association with antidepressant and antipsychotic response, but no consistently reproduced results have been reported. However, a notable finding from the CATIE cohort indicated that SNPs in HTR4 are associated with response to antipsychotic treatment of some symptoms, particularly cognitive measures 34, although the underlying functional relationship between these SNPs and treatment outcome is unclear. Finally, a GWAS identified a significant association of an HTR7 SNP with alcohol dependence, which is an intriguing finding that deserves further investigation 47.

Catecholamine systems

For most of five decades, dopamine has been central to our understanding of the neurotransmitter pathology of schizophrenia, and it is well established that the primary mechanism of the antipsychotic drugs, in relieving the symptoms of both schizophrenia and mania, involves antagonist action at dopamine D2-like receptors in the brain. Dopamine receptor and transporter genes are therefore strong candidates for investigating the association between genetic polymorphisms, antipsychotic drug treatment outcome and the extent of side-effects; however, associations have also been observed between polymorphisms in these genes and susceptibility to major depression, attention deficit hyperactivity disorder (ADHD) and anxiety disorders 4850. Polymorphisms have been reported in all five dopamine receptor genes (DRD1DRD5) and the dopamine transporter gene SLC6A3/DAT1. This has provided a set of potential variants for investigation of genetic associations with drug efficacy and side-effects.

The development of second-generation atypical antipsychotics has had a dramatic influence on the profile of side-effects seen in response to antipsychotic treatment. Extrapyramidal side-effects associated with typical antipsychotics (akathisia, dystonia and parkinsonism) are much less frequent with the newer ‘atypical’ drugs. However, other side-effects, such as hyperprolactinaemia and weight gain, are potential adverse consequences of current drugs, with both drug-to-drug and pharmacogenetic variability.

Inevitably, polymorphisms in the DRD2 gene have been investigated for associations with treatment outcome in schizophrenic patients, with the majority of studies focusing on the Taq1A and –141 Ins/Del polymorphisms. The Taq1A polymorphism, originally believed to be located in the promoter region of the DRD2 gene, but now identified as located in the coding region of the ANKK1 gene, influences the expression of the DRD2 gene; compared with noncarrier homozygotes, carriers of the A1 allele have reduced striatal DRD2 receptor density 51, leading to reduced dopaminergic activity. This reduction in receptor density has been shown in imaging studies to have functional consequences, with the Taq1A polymorphism predicting response to reward 52. The –141 Ins/Del polymorphism is located in the promoter region and represents a deletion or insertion of a cytosine base; the Del allele has been associated with lower expression levels of DRD2 in vivo and in vitro 53,54. Initial studies into associations of these two SNPs with antipsychotic treatment outcome were inconsistent 5558; however, a comprehensive meta-analysis 59 has shown that the Del allele is associated with a poor response to antipsychotic drugs compared with the Ins/Ins genotype, but the Taq1A allele shows no association. The Taq1A polymorphism, along with two others (rs6277 and rs1079598) in the DRD2 gene, has been associated with increased susceptibility to weight gain 60,61.

Hyperprolactinaemia following antagonism of dopamine receptors in the pituitary gland is a serious side-effect of antipsychotics that increases the risk of amenorrhoea, galactorrhoea, sexual dysfunction, breast cancer and osteoporosis 62. Several studies have indicated a dopamine receptor genotype effect on the severity of these side-effects 6365; however, the results are somewhat inconsistent. A study of healthy volunteers given one dose of antipsychotic showed that plasma prolactin increase was modulated by drug, sex and Taq1A polymorphism, with higher levels of prolactin secretion being associated with women, risperidone (in contrast to olanzapine and quetiapine) and the 1A allele 66.

A functional polymorphism in the DRD3 gene (rs6280) results in a coding region substitution of Gly for Ser; in vitro studies have shown increased binding affinity of dopamine for the receptor with the Gly allele 67. The Gly allele has been associated with improved treatment response to clozapine and risperidone 68,69, although other studies have failed to replicate these findings 70,71. The functional effect of this polymorphism may also explain an association with risk of TD. A number of early studies indicated this 7274, but recent meta-analyses are conflicting. Bakker et al. 75 considered 11 studies between 1976 and 2005 covering 1610 patients, and reported an increased risk of TD associated with the Gly allele. Tsai et al. 76 considered 13 studies between 1997 and 2008 and concluded that there was little or no association between the rs6280 polymorphism and TD. The authors of both papers consider publication bias as an explanation for these findings. In each analysis, an effect of publication date is seen, with earlier studies reporting an association, and later studies seeing a weaker or nonsignificant effect. As observed by Tsai et al. 76, this is a common pattern seen in pharmocogenetic studies, where positive findings are reported from early, small studies in discrete groups, followed by larger studies in heterogeneous populations that fail to confirm the initial findings. In the case of TD, there is an additional treatment effect; earlier studies were much more likely to include patients who had been treated with first-generation antipsychotics, with the associated higher risk of TD, whilst treatment protocols used in later studies show a clear bias towards the use of atypical antipsychotics, which are associated with a much lower risk of TD. This theory is supported by results from the CATIE trial in 710 patients 77, which did not find an effect of rs6280; in this trial, the majority of patients were on second-generation antipsychotics. Therefore, it can be concluded that the Gly allele of the rs6280 polymorphism may contribute to increased risk of TD, but this association is predominately seen in patients treated with typical antipsychotics and is less relevant in patients treated with atypical drugs.

The neuronal dopamine transporter (DAT) contributes to the control of synaptic dopamine. A VNTR polymorphism in the 3′ region of the gene that codes for DAT, SLC6A3/DAT1, gives rise to two to 11 tandem repeats of a 40 bp sequence, with nine or 10 repeats being the most common alleles. There is conflicting evidence of differences in DAT binding associated with the number of repeats, with some studies reporting increased binding associated with the 10-repeat allele, and others associating increased binding with the nine-repeat allele 78. Methylphenidate, the most commonly prescribed medication for ADHD, binds to DAT1 and reduces the reuptake of dopamine, thereby increasing synaptic dopamine. A number of studies have demonstrated the association of the 10-repeat allele with poorer treatment outcome 79,80; however, a recent meta-analysis failed to confirm this finding 81.

Other studies have investigated the effect of the DAT 9/10 repeat on treatment response in schizophrenia and depression; in schizophrenics, an association with clozapine response is reported 82, but no association with response to risperidone 83. In major depressive disorder (MDD), Kirchheiner et al. 84 observed a reduced response to SSRIs associated with the nine-repeat allele.

The noradrenergic synapse has long been considered an important target for antidepressant drugs and, although SSRIs have focused attention on serotonin systems, the noradrenaline transporter and certain adrenergic receptors remain important sites of action of some effective antidepressants. Furthermore, several antipsychotics bind to α-adrenergic receptors, and the antagonism of α2-adrenoceptors by clozapine has been suggested to underlie some of the unique properties of this drug.

Despite this, no link has been shown between polymorphisms in these genes and antipsychotic treatment efficacy 85. Associations have been shown between polymorphisms in α-adrenergic receptor genes and metabolic syndrome abnormalities 86,87, but there is limited evidence that variants in these genes confer a risk for antipsychotic drug-induced weight gain. Saiz et al. 88 found a marginal association between the –4884 A/A genotype in the ADRA1A promoter region and higher body mass index scores in patients treated with a range of antipsychotic regimens, while others 89 found a significant association between a bank of SNPs in the regulatory regions of the same gene and body mass index in a similar set of patients. Wang et al. 90 investigated the –1291 C/G promoter polymorphism in the ADRA2A gene and saw an association of the G allele with greater weight gain after clozapine treatment. This finding was contradicted 91 in a study showing that carriers of the C allele had more olanzapine-/clozapine-associated weight gain. Finally, De Luca et al. 92 concluded that –1291 C/G SNP only conferred a risk in a multiplicative model with the u-VNTR promoter polymorphism in the MAOA gene.

In contrast, several ADHD studies have linked methylphenidate response and ADRA2A gene variants 9397; however, this association was not found in adults 98, which may reflect differences in response to amphetamine analogues between adults and children.

The antidepressant serotonin–noradrenaline reuptake inhibitors (SNRIs) in contrast to SSRIs, bind to the noradrenaline transporter (NET), as well as to HTT. Polymorphisms in both ADRA1A and ADRA2A genes and SLC6A2 (the NET gene) are associated with SNRI treatment outcome, with the SLC6A2 SNPs interacting with childhood trauma to influence antidepressant response 29,99,100. Wakeno et al. 101 demonstrated that C allele carriers of the ADRA2A –1291 polymorphism had improved response to the SNRI milnacipran compared with subjects who were GG homozygotes. Polymorphisms in SLC6A2 have been shown to be associated with antidepressant response 99; the –1287 G/A polymorphism was associated with response to milnacipran.

Glutamate and GABA systems

Monoamine systems have classically provided candidate genes for the pharmacogenetic study of treatment response in severe mental illness; typically, dopamine for antipsychotics and serotonin for antidepressants. Naturally, this reflected the major pharmacological mechanisms of these drugs, but also addressed the key theories of neurotransmitter pathology in the respective diseases. However, over the past two decades or so, increasing recognition has been given to the roles of glutamate and GABA neurotransmission in the pathology of these disorders, resulting in these transmitter systems being targets for novel treatments for both schizophrenia and affective disorders 102,103. This, in turn, has encouraged the investigation of glutamate and GABA neuronal transporter and receptor genes in psychiatric pharmacogenetics.

Glutamate and GABA are fast neurotransmitters, with effects that are more rapid and transient than the typical synaptic activity of the monoamine transmitters. Essential to this is a rapid removal mechanism as provided by the reuptake transporters; of the excitatory amino acid transporters (EAATs), EAAT3 is primarily found on neurons while EAAT2 is a glial transporter responsible for most glutamate removal from the synapse. Single nucleotide polymorphisms within the EAAT genes have mainly been studied in the context of neurological disorders. However, given the central role of EAAT2 in the control of synaptic glutamate and the importance of this neurotransmitter in psychiatric pathogenesis, it is perhaps surprising that its gene, SLC1A2, has been so little studied.

A functional SNP (rs4354668) −181 bp from the SLC1A2 transcription start site affects transporter expression, with the variant allele inducing a reduction in promoter activity and resulting in an elevation of plasma glutamate 104. In one of the very few pharmacogenetic studies, this EAAT2 functional polymorphism was shown to have a significant influence on recurrence rate in bipolar disorder, with T homozygotes being associated with fewer episodes as well as improved outcome of lithium treatment 105.

Genetic variation in the EAAT3 gene (SLC1A1) has been associated with obsessive–compulsive disorder in several studies 106, so perhaps it is unsurprising that it is also associated with obsessive–compulsive disorder symptoms occurring as a side-effect of antipsychotic drug treatment 107.

Despite the identification of the neuronal GABA transporter-1 as a potential drug target and the identification of functional polymorphisms in its gene (SLC6A1) 108, there appears to be very little work indicating that they have any association with psychiatric drug treatment.

Glutamate receptors are widely expressed in the central nervous system and are involved in synaptic plasticity, learning and memory processes. The best studied are the ionotropic glutamate receptors, the NMDA, kainate and AMPA receptors, which have GRIN, GRIK and GRIA genes, respectively, encoding their protein subunits.

There is certainly evidence for a fairly consistent, if weak, involvement of a genetic variability in a kainate receptor subunit in antidepressant response. In the STAR*D study of citalopram treatment 109, the effect of rs1954787, a GRIK4 SNP, was found to be predictive of treatment outcome. While this effect alone was fairly modest, the G allele was associated with a greater response to treatment when combined with the HTR2A genotype. A subsequent study of 82 GRIK4 SNPs in another sample identified a polymorphism with a stronger effect, also enhanced in its effect when combined with HTR2A 110. While a multicentre study 111 was unable to replicate an effect of this gene on antidepressant response, the Chinese study of Pu et al. 112 also found a significant effect of rs1954787 in GRIK4 on the outcome of antidepressant treatment.

This study by Pu et al. 112 also identified significant individual effects of several GABA receptor genes (GABRP, GABRQ and GABRA6), as well as some gene–gene interactions between other GABA receptors and glutamic acid decarboxylase, its synthesizing enzyme, on antidepressant response, providing strong support for pharmacogenetic associations of the GABA system with treatment of depression.

Other glutamate receptor genes have been associated with symptom response to antipsychotic drug treatment, although with little consistency and replication. GRIA4 was one of six genes emerging from a GWAS with a significant association with iloperidone response 113. Although the same results were not obtained for ziprasidone 114, GRIA4 was significantly associated with risperidone response 115.

The broad candidate gene analysis of the CATIE sample identified significant associations of seven SNPs in glutamate receptor genes with improvement in total PANSS scores, although it should be noted that glutamate receptors were represented by over 1000 of the 2769 SNPs studied 34. They did not include GRIA4; these were primarily metabotropic glutamate receptor genes, several of which appeared to have fairly specific effects on negative and cognitive symptoms, including GRM7 and GRM3. This latter gene is of particular interest because it codes for a novel target for the treatment of schizophrenia 116, and others have also shown association of functional or potentially functional GRM3 SNPs with antipsychotic response 117,118, particularly of negative symptoms 119.

Another gene associated with antipsychotic-induced improvement of negative symptoms in schizophrenia is GRIA2 120. Furthermore, an association of response to lithium with SNPs spanning this gene was one of several positive findings in a large GWAS of treatment in bipolar disorder 121.

Investigation of the possible role of genetic variability in GABA receptors in lithium response has proved negative (e.g. 122). However, recognition of the importance of GABA systems in motor disorders, particularly dyskinesias, has yielded preliminary positive results for the association of tardive dyskinesia with genes for GABAA receptor subunits and a GABA transporter, SLC6A11 123.

In the absence of strong hypotheses for a primary involvement of glutamatergic dysfunction in adverse effects, few candidate gene studies have investigated amino acid receptor or transporter genes in the side-effects of antipsychotic and antidepressant drugs. One study that did found that the glycine transporter SLC6A5 was associated with dyskinesias and EPS following haloperidol treatment, a finding replicated in further samples 124.

Epigenetics and gene–environment interactions

Variations in the genomic DNA sequence are unlikely to explain fully all the individual differences in the response to, and side-effects of, psychiatric drug treatments. It is now well established that environmental factors can influence the outcome of such treatments. Traumatic events in early life have particularly been studied in this respect; notably, early childhood trauma appears to be a factor predictive of poorer response to the drug treatment of depression in adulthood 125. Such stressful life events can affect response to SSRIs by interacting with functional SLC6A4 polymorphisms 126,127 and with SNPs on other serotoninergic genes, including HTR1B and TPH2 29.

The biological mechanisms underlying these interactions remain obscure. Early life stress can affect synaptic maturation and neuronal development through an influence on the hypothalamic–pituitary–adrenal axis 128. However, epigenetic mechanisms may also be important. There is steadily accumulating evidence that DNA methylation may underlie the influence of early life stress on depression and its response to treatment. For example, expression of the serotonin transporter is related to the extent of DNA methylation in the promoter region of the gene, interacting with the HTTLPR 129. This DNA methylation is greater in subjects reporting child abuse 130, and SLC6A4 methylation relates to the severity of psychopathology 131. Inhibitors of DNA methylation have been reported to have an antidepressant effect 132, although few specific genes associated with antidepressant response have been investigated for their methylation status. The same is true of antipsychotic response, although again DNA methylation appears to be influenced by drug treatment 133.

It is clear that there is still much to learn here. The study of DNA methylation and other epigenetic influences is still very much in its infancy, but it seems that pharmacoepigenetics is likely to be an important future addition to the pharmacogenomic influences on response to treatment of severe mental illness.

Conclusions

As we have indicated, this review has been restricted to consideration of the pharmacogenetics of neurotransmitter receptors and transporters, a fairly arbitrary restriction within the many and various influences on the activity of synaptic neurotransmission, in the treatment of psychiatric disorders. Even within these limitations, it has not been comprehensive; the focus has been primarily on the response to treatment of severe mental illness (depression, bipolar illness and schizophrenia), with some mention of the better-studied side-effects. We have selected, again in a fairly arbitrary manner, mainly positive association studies of known functional polymorphisms; these often underline known pharmacotherapeutic mechanisms in psychiatry, but occasionally can point to new and potential mechanisms of drug action.

There remain many unexplained inconsistencies between studies; perhaps those of greatest concern come from well-replicated candidate gene associations that do not emerge in findings from GWAS. There are always differences between samples in these studies; sample power, ethnicity, sex, extent of prior treatment and drug differences are all likely to contribute, although it is worth noting that despite some suggestions to the contrary, there is so far little evidence for truly drug-specific genetic associations. In addition, the clinical response phenotypes measured may often be complex and multifactorial, comprising several physiological responses under different genetic influences. Furthermore, adherence to drug treatment is notoriously poor in psychiatry and thus any factor that might influence this, such as side-effects or extent of social support, will also be important in determining response.

Nevertheless, we do see some convergence that, where different genes are associated with the same effects, may go unrecognized. Thus, it is notable that functional variants in each of SLC6A4, HTR1A and HTR2A may be associated with antidepressant response, with these gene products mediating synaptic serotonin activity and neurotransmission. There are other examples mentioned above of genetic variability in both a transporter and a postsynaptic receptor having a common influence on psychiatric drug actions; however, more needs to be done to address additive and interactive effects between genes, even in an exploratory manner, if there are sample size constraints that compromise statistical power.

The rapid progress in epigenetics should also help us to explain more of the variance in the individual response to drug treatments. As these new findings converge with more consistent GWAS and candidate gene findings, they will eventually provide us with opportunities for predictive genetic testing, once their scientific validity and clinical utility are established.

Competing Interests

All authors have completed the Unified Competing Interest form at http://www.icmje.org/coi_disclosure.pdf (available on request from the corresponding author) and declare: no support from any organization for the submitted work; no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years; and no other relationships or activities that could appear to have influenced the submitted work, except for GPR who acts as advisor to Optimal Medicine Ltd.

References

  • 1.Pickar D, Rubinow K. Pharmacogenomics of psychiatric disorders. Trends Pharmacol Sci. 2001;22:75–83. doi: 10.1016/s0165-6147(00)01603-5. [DOI] [PubMed] [Google Scholar]
  • 2.Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD, Severe J, Hsiao JK Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353:1209–1223. doi: 10.1056/NEJMoa051688. [DOI] [PubMed] [Google Scholar]
  • 3.Rush AJ, Fava M, Wisniewski SR, Lavori PW, Trivedi MH, Sackeim HA, Thase ME, Nierenberg AA, Quitkin FM, Kashner TM, Kupfer DJ, Rosenbaum JF, Alpert J, Stewart JW, McGrath PJ, Biggs MM, Shores-Wilson K, Lebowitz BD, Ritz L, Niederehe G STAR*D Investigators Group. Sequenced treatment alternatives to relieve depression (STAR*D): rationale and design. Control Clin Trials. 2004;25:119–142. doi: 10.1016/s0197-2456(03)00112-0. [DOI] [PubMed] [Google Scholar]
  • 4.Heils A, Teufel A, Petri S, Stöber G, Riederer P, Bengel D, Lesch KP. Allelic variation of human serotonin transporter gene expression. J Neurochem. 1996;66:2621–2624. doi: 10.1046/j.1471-4159.1996.66062621.x. [DOI] [PubMed] [Google Scholar]
  • 5.Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, Xu K, Arnold PD, Richter MA, Kennedy JL, Murphy DL, Goldman D. Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet. 2006;78:815–826. doi: 10.1086/503850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Battersby S, Ogilvie AD, Smith CA, Blackwood DH, Muir WJ, Quinn JP, Fink G, Goodwin GM, Harmar AJ. Structure of a variable number tandem repeat of the serotonin transporter gene and association with affective disorder. Psychiatr Genet. 1996;6:177–181. doi: 10.1097/00041444-199624000-00001. [DOI] [PubMed] [Google Scholar]
  • 7.Smeraldi E, Zanardi R, Benedetti F, Di Bella D, Perez J, Catalano M. Polymorphism within the promoter of the serotonin transporter gene and antidepressant efficacy of fluvoxamine. Mol Psychiatry. 1998;3:508–511. doi: 10.1038/sj.mp.4000425. [DOI] [PubMed] [Google Scholar]
  • 8.Pollock BG, Ferrell RE, Mulsant BH, Mazumdar S, Miller M, Sweet RA, Davis S, Kirshner MA, Houck PR, Stack JA, Reynolds CF, Kupfer DJ. Allelic variation in the serotonin transporter promoter affects onset of paroxetine treatment response in late-life depression. Neuropsychopharmacology. 2000;23:587–590. doi: 10.1016/S0893-133X(00)00132-9. [DOI] [PubMed] [Google Scholar]
  • 9.Serretti A, Kato M, De Ronchi D, Kinoshita T. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients. Mol Psychiatry. 2007;12:247–257. doi: 10.1038/sj.mp.4001926. [DOI] [PubMed] [Google Scholar]
  • 10.Taylor MJ, Sen S, Bhagwagar Z. Antidepressant response and the serotonin transporter gene-linked polymorphic region. Biol Psychiatry. 2010;68:536–543. doi: 10.1016/j.biopsych.2010.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kim DK, Lim SW, Lee S, Sohn SE, Kim S, Hahn CG, Carroll BJ. Serotonin transporter gene polymorphism and antidepressant response. Neuroreport. 2000;11:215–219. doi: 10.1097/00001756-200001170-00042. [DOI] [PubMed] [Google Scholar]
  • 12.Myung W, Lim SW, Kim S, Kim H, Chung JW, Seo MY, Kim JW, Carroll BJ, Kim DK. Serotonin transporter genotype and function in relation to antidepressant response in Koreans. Psychopharmacology (Berl) 2013;225:283–290. doi: 10.1007/s00213-012-2813-y. [DOI] [PubMed] [Google Scholar]
  • 13.Porcelli S, Fabbri C, Serretti A. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with antidepressant efficacy. Eur Neuropsychopharmacol. 2012;22:239–258. doi: 10.1016/j.euroneuro.2011.10.003. [DOI] [PubMed] [Google Scholar]
  • 14.Smits KM, Smits LJ, Schouten JS, Stelma FF, Nelemans P, Prins MH. Influence of SERTPR and STin2 in the serotonin transporter gene on the effect of selective serotonin reuptake inhibitors in depression: a systematic review. Mol Psychiatry. 2004;9:433–441. doi: 10.1038/sj.mp.4001488. [DOI] [PubMed] [Google Scholar]
  • 15.Niitsu T, Fabbri C, Bentini F, Serretti A. Pharmacogenetics in major depression: a comprehensive meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2013;35:183–194. doi: 10.1016/j.pnpbp.2013.05.011. [DOI] [PubMed] [Google Scholar]
  • 16.Tharoor H, Kotambail A, Jain S, Sharma PS, Satyamoorthy K. Study of the association of serotonin transporter triallelic 5-HTTLPR and STin2 VNTR polymorphisms with lithium prophylaxis response in bipolar disorder. Psychiatr Genet. 2013;23:77–81. doi: 10.1097/YPG.0b013e32835d6fad. [DOI] [PubMed] [Google Scholar]
  • 17.Lin E, Chen PS. Pharmacogenomics with antidepressants in the STAR*D study. Pharmacogenomics. 2008;9:935–946. doi: 10.2217/14622416.9.7.935. [DOI] [PubMed] [Google Scholar]
  • 18.Murphy GM, Jr, Hollander SB, Rodrigues HE, Kremer C, Schatzberg AF. Effects of the serotonin transporter gene promoter polymorphism on mirtazapine and paroxetine efficacy and adverse events in geriatric major depression. Arch Gen Psychiatry. 2004;61:1163–1169. doi: 10.1001/archpsyc.61.11.1163. [DOI] [PubMed] [Google Scholar]
  • 19.Safarinejad MR. Analysis of association between the 5-HTTLPR and STin2 polymorphisms in the serotonin-transporter gene and clinical response to a selective serotonin reuptake inhibitor (sertraline) in patients with premature ejaculation. BJU Int. 2010;105:73–78. doi: 10.1111/j.1464-410X.2009.08714.x. [DOI] [PubMed] [Google Scholar]
  • 20.Dombrovski AY, Mulsant BH, Ferrell RE, Lotrich FE, Rosen JI, Wallace M, Houck PR, Mazumdar S, Pollock BG. Serotonin transporter triallelic genotype and response to citalopram and risperidone in dementia with behavioral symptoms. Int Clin Psychopharmacol. 2010;25:37–45. doi: 10.1097/YIC.0b013e328333ee10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rotberg B, Kronenberg S, Carmel M, Frisch A, Brent D, Zalsman G, Apter A, Weizman A. Additive effects of 5-HTTLPR (serotonin transporter) and tryptophan hydroxylase 2 G-703T gene polymorphisms on the clinical response to citalopram among children and Adolescents with depression and anxiety disorders. J Child Adolesc Psychopharmacol. 2013;23:117–122. doi: 10.1089/cap.2012.0020. [DOI] [PubMed] [Google Scholar]
  • 22.Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD, Bown CD, Sequeira A, Kushwaha N, Morris SJ, Basak A, Ou XM, Albert PR. Impaired repression at a 5-hydroxytryptamine 1A receptor gene polymorphism associated with major depression and suicide. J Neurosci. 2003;23:8788–8799. doi: 10.1523/JNEUROSCI.23-25-08788.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lemonde S, Du L, Bakish D, Hrdina P, Albert PR. Association of the C (-1019)G 5-HT1A functional promoter polymorphism with antidepressant response. Int J Neuropsychopharmacol. 2004;7:501–506. doi: 10.1017/S1461145704004699. [DOI] [PubMed] [Google Scholar]
  • 24.Drago A, Ronchi DD, Serretti A. 5-HT1A gene variants and psychiatric disorders: a review of current literature and selection of SNPs for future studies. Int J Neuropsychopharmacol. 2008;11:701–721. doi: 10.1017/S1461145707008218. [DOI] [PubMed] [Google Scholar]
  • 25.Yevtushenko OO, Oros MM, Reynolds GP. Early response to selective serotonin reuptake inhibitors in panic disorder is associated with a functional 5-HT1A receptor gene polymorphism. J Affect Disord. 2010;123:308–311. doi: 10.1016/j.jad.2009.09.007. [DOI] [PubMed] [Google Scholar]
  • 26.Reynolds GP, Arranz B, Templeman LA, Fertuzinhos S, San L. Effect of 5-HT1A receptor gene polymorphism on negative and depressive symptom response to antipsychotic treatment of drug-naive psychotic patients. Am J Psychiatry. 2006;163:1826–1829. doi: 10.1176/ajp.2006.163.10.1826. [DOI] [PubMed] [Google Scholar]
  • 27.Wang L, Fang C, Zhang A, Du J, Yu L, Ma J, Feng G, Xing Q, He L. The –1019 C/G polymorphism of the 5-HT1A receptor gene is associated with negative symptom response to risperidone treatment in schizophrenia patients. J Psychopharmacol. 2008;22:904–909. doi: 10.1177/0269881107081522. [DOI] [PubMed] [Google Scholar]
  • 28.Mössner R, Schuhmacher A, Kühn KU, Cvetanovska G, Rujescu D, Zill P, Quednow BB, Rietschel M, Wölwer W, Gaebel W, Wagner M, Maier W. Functional serotonin 1A receptor variant influences treatment response to atypical antipsychotics in schizophrenia. Pharmacogenet Genomics. 2009;19:91–94. doi: 10.1097/FPC.0b013e328311a917. [DOI] [PubMed] [Google Scholar]
  • 29.Xu Z, Zhang Z, Shi Y, Pu M, Yuan Y, Zhang X, Li L, Reynolds GP. Influence and interaction of genetic polymorphisms in the serotonin system and life stress on antidepressant drug response. J Psychopharmacol. 2012;26:349–359. doi: 10.1177/0269881111414452. [DOI] [PubMed] [Google Scholar]
  • 30.Parsons MJ, D'Souza UM, Arranz MJ, Kerwin RW, Makoff AJ. The-1438A/G polymorphism in the 5-hydroxytryptamine type 2A receptor gene affects promoter activity. Biol Psychiatry. 2004;56:406–410. doi: 10.1016/j.biopsych.2004.06.020. [DOI] [PubMed] [Google Scholar]
  • 31.Lane HY, Chang YC, Chiu CC, Chen ML, Hsieh MH, Chang WH. Association of risperidone treatment response with a polymorphism in the 5-HT2A receptor gene. Am J Psychiatry. 2002;159:1593–1595. doi: 10.1176/appi.ajp.159.9.1593. [DOI] [PubMed] [Google Scholar]
  • 32.Joober R, Benkelfat C, Brisebois K, Toulouse A, Turecki G, Lal S, Bloom D, Labelle A, Lalonde P, Fortin D, Alda M, Palmour R, Rouleau GA. T102C polymorphism in the 5HT2A gene and schizophrenia: relation to phenotype and drug response variability. J Psychiatry Neurosci. 1999;24:141–146. [PMC free article] [PubMed] [Google Scholar]
  • 33.Arranz MJ, Munro J, Sham P, Kirov G, Murray RM, Collier DA, Kerwin RW. Meta-analysis of studies on genetic variation in 5-HT2A receptors and clozapine response. Schizophr Res. 1998;32:93–99. doi: 10.1016/s0920-9964(98)00032-2. [DOI] [PubMed] [Google Scholar]
  • 34.Need AC, Keefe RS, Ge D, Grossman I, Dickson S, McEvoy JP, Goldstein DB. Pharmacogenetics of antipsychotic response in the CATIE trial: a candidate gene analysis. Eur J Hum Genet. 2009;17:946–957. doi: 10.1038/ejhg.2008.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.McMahon FJ, Buervenich S, Charney D, Lipsky R, Rush AJ, Wilson AF, Sorant AJ, Papanicolaou GJ, Laje G, Fava M, Trivedi MH, Wisniewski SR, Manji H. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antidepressant treatment. Am J Hum Genet. 2006;78:804–814. doi: 10.1086/503820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Choi MJ, Kang RH, Ham BJ, Jeong HY, Lee MS. Serotonin receptor 2A gene polymorphism (–1438A/G) and short-term treatment response to citalopram. Neuropsychobiology. 2005;52:155–162. doi: 10.1159/000087847. [DOI] [PubMed] [Google Scholar]
  • 37.Gunes A, Scordo MG, Jaanson P, Dahl ML. Serotonin and dopamine receptor gene polymorphisms and the risk of extrapyramidal side effects in perphenazine-treated schizophrenic patients. Psychopharmacology (Berl) 2007;190:479–484. doi: 10.1007/s00213-006-0622-x. [DOI] [PubMed] [Google Scholar]
  • 38.Lerer B, Segman RH, Tan EC, Basile VS, Cavallaro R, Aschauer HN, Strous R, Chong SA, Heresco-Levy U, Verga M, Scharfetter J, Meltzer HY, Kennedy JL, Macciardi F. Combined analysis of 635 patients confirms an age-related association of the serotonin 2A receptor gene with tardive dyskinesia and specificity for the non-orofacial subtype. Int J Neuropsychopharmacol. 2005;8:411–425. doi: 10.1017/S1461145705005389. [DOI] [PubMed] [Google Scholar]
  • 39.Zhang JP, Malhotra AK. Pharmacogenetics and antipsychotics: therapeutic efficacy and side effects prediction. Expert Opin Drug Metab Toxicol. 2011;7:9–37. doi: 10.1517/17425255.2011.532787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Segman RH, Heresco-Levy U, Finkel B, Inbar R, Neeman T, Schlafman M, Dorevitch A, Yakir A, Lerner A, Goltser T, Shelevoy A, Lerer B. Association between the serotonin 2C receptor gene and tardive dyskinesia in chronic schizophrenia: additive contribution of 5-HT2Cser and DRD3gly alleles to susceptibility. Psychopharmacology (Berl) 2000;152:408–413. doi: 10.1007/s002130000521. [DOI] [PubMed] [Google Scholar]
  • 41.Reynolds GP, Kirk SL. Metabolic side effects of antipsychotic drug treatment–pharmacological mechanisms. Pharmacol Ther. 2010;125:169–179. doi: 10.1016/j.pharmthera.2009.10.010. [DOI] [PubMed] [Google Scholar]
  • 42.Reynolds GP, Zhang ZJ, Zhang XB. Association of antipsychotic drug-induced weight gain with a 5-HT2C receptor gene polymorphism. Lancet. 2002;359:2086–2087. doi: 10.1016/S0140-6736(02)08913-4. [DOI] [PubMed] [Google Scholar]
  • 43.Reynolds GP. Pharmacogenetic aspects of antipsychotic drug-induced weight gain – a critical review. Clin Psychopharmacol Neurosci. 2012;10:71–77. doi: 10.9758/cpn.2012.10.2.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hill MJ, Reynolds GP. Functional consequences of two HTR2C polymorphisms associated with antipsychotic-induced weight gain. Pharmacogenomics. 2011;12:727–734. doi: 10.2217/pgs.11.16. [DOI] [PubMed] [Google Scholar]
  • 45.Templeman LA, Reynolds GP, Arranz B, San L. Polymorphisms of the 5-HT2C receptor and leptin genes are associated with antipsychotic drug-induced weight gain in Caucasian subjects with a first-episode psychosis. Pharmacogenetics. 2005;15:195–200. doi: 10.1097/01213011-200504000-00002. [DOI] [PubMed] [Google Scholar]
  • 46.Ujike H, Nomura A, Morita Y, Morio A, Okahisa Y, Kotaka T, Kodama M, Ishihara T, Kuroda S. Multiple genetic factors in olanzapine-induced weight gain in schizophrenia patients: a cohort study. J Clin Psychiatry. 2008;69:1416–1422. doi: 10.4088/jcp.v69n0909. [DOI] [PubMed] [Google Scholar]
  • 47.Zlojutro M, Manz N, Rangaswamy M, Xuei X, Flury-Wetherill L, Koller D, Bierut LJ, Goate A, Hesselbrock V, Kuperman S, Nurnberger J, Jr, Rice JP, Schuckit MA, Foroud T, Edenberg HJ, Porjesz B, Almasy L. Genome-wide association study of theta band event-related oscillations identifies serotonin receptor gene HTR7 influencing risk of alcohol dependence. Am J Med Genet B Neuropsychiatr Genet. 2011;156B:44–58. doi: 10.1002/ajmg.b.31136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Opmeer EM, Kortekaas R, Aleman A. Depression and the role of genes involved in dopamine metabolism and signalling. Prog Neurobiol. 2010;92:112–133. doi: 10.1016/j.pneurobio.2010.06.003. [DOI] [PubMed] [Google Scholar]
  • 49.Wu J, Xiao H, Sun H, Zou L, Zhu LQ. Role of dopamine receptors in ADHD: a systematic meta-analysis. Mol Neurobiol. 2012;45:605–620. doi: 10.1007/s12035-012-8278-5. [DOI] [PubMed] [Google Scholar]
  • 50.Faludi G, Gonda X, Bagdy G, Dome P. Pharmaco-and therapygenetic aspects in the treatment of anxiety disorders beyond the serotonergic system: a brief review. Neuropsychopharmacol Hung. 2012;14:221–229. [PubMed] [Google Scholar]
  • 51.Thompson J, Thomas N, Singleton A, Piggott M, Lloyd S, Perry EK, Morris CM, Perry RH, Ferrier IN, Court JA. D2 dopamine receptor gene (DRD2) Taq1 A polymorphism: reduced dopamine D2 receptor binding in the human striatum associated with the A1 allele. Pharmacogenetics. 1997;7:479–484. doi: 10.1097/00008571-199712000-00006. [DOI] [PubMed] [Google Scholar]
  • 52.Cohen MX, Young J, Baek JM, Kessler C, Ranganath C. Individual differences in extraversion and dopamine genetics predict neural reward responses. Brain Res Cogn Brain Res. 2005;25:851–861. doi: 10.1016/j.cogbrainres.2005.09.018. [DOI] [PubMed] [Google Scholar]
  • 53.Jönsson EG, Nöthen MM, Grünhage F, Farde L, Nakashima Y, Propping P, Sedvall GC. Polymorphisms in the dopamine D2 receptor gene and their relationships to striatal dopamine receptor density of healthy volunteers. Mol Psychiatry. 1999;4:290–296. doi: 10.1038/sj.mp.4000532. [DOI] [PubMed] [Google Scholar]
  • 54.Arinami T, Gao M, Hamaguchi H, Toru M. A functional polymorphism in the promoter region of the dopamine D2 receptor gene is associated with schizophrenia. Hum Mol Genet. 1997;6:577–582. doi: 10.1093/hmg/6.4.577. [DOI] [PubMed] [Google Scholar]
  • 55.Suzuki A, Kondo T, Mihara K, Yasui-Furukori N, Ishida M, Furukori H, Kaneko S, Inoue Y, Otani K. The-141C Ins/Del polymorphism in the dopamine D2 receptor gene promoter region is associated with anxiolytic and antidepressive effects during treatment with dopamine antagonists in schizophrenic patients. Pharmacogenetics. 2001;11:545–550. doi: 10.1097/00008571-200108000-00009. [DOI] [PubMed] [Google Scholar]
  • 56.Hwang R, Shinkai T, De Luca V, Müller DJ, Ni X, Macciardi F, Potkin S, Lieberman JA, Meltzer HY, Kennedy JL. Association study of 12 polymorphisms spanning the dopamine D(2) receptor gene and clozapine treatment response in two treatment refractory/intolerant populations. Psychopharmacology (Berl) 2005;181:179–187. doi: 10.1007/s00213-005-2223-5. [DOI] [PubMed] [Google Scholar]
  • 57.Hwang R, Shinkai T, Deluca V, Macciardi F, Potkin S, Meltzer HY, Kennedy JL. Dopamine D2 receptor gene variants and quantitative measures of positive and negative symptom response following clozapine treatment. Eur Neuropsychopharmacol. 2006;16:248–259. doi: 10.1016/j.euroneuro.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • 58.Lencz T, Robinson DG, Xu K, Ekholm J, Sevy S, Gunduz-Bruce H, Woerner MG, Kane JM, Goldman D, Malhotra AK. DRD2 promoter region variation as a predictor of sustained response to antipsychotic medication in first-episode schizophrenia patients. Am J Psychiatry. 2006;163:529–531. doi: 10.1176/appi.ajp.163.3.529. [DOI] [PubMed] [Google Scholar]
  • 59.Zhang JP, Lencz T, Malhotra AK. D2 receptor genetic variation and clinical response to antipsychotic drug treatment: a meta-analysis. Am J Psychiatry. 2010;167:763–772. doi: 10.1176/appi.ajp.2009.09040598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Müller DJ, Zai CC, Sicard M, Remington E, Souza RP, Tiwari AK, Hwang R, Likhodi O, Shaikh S, Freeman N, Arenovich T, Heinz A, Meltzer HY, Lieberman JA, Kennedy JL. Systematic analysis of dopamine receptor genes (DRD1-DRD5) in antipsychotic-induced weight gain. Pharmacogenomics J. 2012;12:156–164. doi: 10.1038/tpj.2010.65. [DOI] [PubMed] [Google Scholar]
  • 61.Lencz T, Robinson DG, Napolitano B, Sevy S, Kane JM, Goldman D, Malhotra AK. DRD2 promoter region variation predicts antipsychotic-induced weight gain in first episode schizophrenia. Pharmacogenet Genomics. 2010;20:569–572. doi: 10.1097/FPC.0b013e32833ca24b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cookson J, Hodgson R, Wildgust HJ. Prolactin, hyperprolactinaemia and antipsychotic treatment: a review and lessons for treatment of early psychosis. J Psychopharmacol. 2012;26(5 Suppl):42–51. doi: 10.1177/0269881112442016. [DOI] [PubMed] [Google Scholar]
  • 63.Young RM, Lawford BR, Barnes M, Burton SC, Ritchie T, Ward WK, Noble EP. Prolactin levels in antipsychotic treatment of patients with schizophrenia carrying the DRD2*A1 allele. Br J Psychiatry. 2004;185:147–151. doi: 10.1192/bjp.185.2.147. [DOI] [PubMed] [Google Scholar]
  • 64.Calarge CA, Ellingrod VL, Acion L, Miller DD, Moline J, Tansey MJ, Schlechte JA. Variants of the dopamine D2 receptor gene and risperidone-induced hyperprolactinemia in children and adolescents. Pharmacogenet Genomics. 2009;19:373–382. doi: 10.1097/FPC.0b013e328329a60f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhang XR, Zhang ZJ, Zhu RX, Yuan YG, Jenkins TA, Reynolds GP. Sexual dysfunction in male schizophrenia: influence of antipsychotic drugs, prolactin and polymorphisms of the dopamine D2 receptor genes. Pharmacogenomics. 2011;12:1127–1136. doi: 10.2217/pgs.11.46. [DOI] [PubMed] [Google Scholar]
  • 66.Lopez-Rodriguez R, Roman M, Novalbos J, Pelegrina ML, Ochoa D, Abad-Santos F. DRD2 Taq1A polymorphism modulates prolactin secretion induced by atypical antipsychotics in healthy volunteers. J Clin Psychopharmacol. 2011;31:555–562. doi: 10.1097/JCP.0b013e31822cfff2. [DOI] [PubMed] [Google Scholar]
  • 67.Lundstrom K, Turpin MP. Proposed schizophrenia-related gene polymorphism: expression of the Ser9Gly mutant human dopamine D3 receptor with the Semliki Forest virus system. Biochem Biophys Res Commun. 1996;225:1068–1072. doi: 10.1006/bbrc.1996.1296. [DOI] [PubMed] [Google Scholar]
  • 68.Scharfetter J, Chaudhry HR, Hornik K, Fuchs K, Sieghart W, Kasper S. Aschauer HN Dopamine D3 receptor gene polymorphism and response to clozapine in schizophrenic Pakastani patients. Eur Neuropsychopharmacol. 1999;10:17–20. doi: 10.1016/s0924-977x(99)00044-9. [DOI] [PubMed] [Google Scholar]
  • 69.Szekeres G, Kéri S, Juhász A, Rimanóczy A, Szendi I, Czimmer C, Janka Z. Role of dopamine D3 receptor (DRD3) and dopamine transporter (DAT) polymorphism in cognitive dysfunctions and therapeutic response to atypical antipsychotics in patients with schizophrenia. Am J Med Genet B Neuropsychiatr Genet. 2004;124B:1–5. doi: 10.1002/ajmg.b.20045. [DOI] [PubMed] [Google Scholar]
  • 70.Malhotra AK, Goldman D, Buchanan RW, Rooney W, Clifton A, Kosmidis MH, Breier A, Pickar D. The dopamine D3 receptor (DRD3) Ser9Gly polymorphism and schizophrenia: a haplotype relative risk study and association with clozapine response. Mol Psychiatry. 1998;3:72–75. doi: 10.1038/sj.mp.4000288. [DOI] [PubMed] [Google Scholar]
  • 71.Kim B, Choi EY, Kim CY, Song K, Joo YH. Could HTR2A T102C and DRD3 Ser9Gly predict clinical improvement in patients with acutely exacerbated schizophrenia? Results from treatment responses to risperidone in a naturalistic setting. Hum Psychopharmacol. 2008;23:61–67. doi: 10.1002/hup.897. [DOI] [PubMed] [Google Scholar]
  • 72.Steen VM, Lovlie R, MacEwan T, McCreadie RG. Dopamine D3-receptor gene variant and susceptibility to tardive dyskinesia in schizophrenic patients. Mol Psychiatry. 1997;2:139–145. doi: 10.1038/sj.mp.4000249. [DOI] [PubMed] [Google Scholar]
  • 73.Liao DL, Yeh YC, Chen HM, Chen H, Hong CJ, Tsai SJ. Association between the Ser9Gly polymorphism of the dopamine D3 receptor gene and tardive dyskinesia in Chinese schizophrenic patients. Neuropsychobiology. 2001;44:95–98. doi: 10.1159/000054924. [DOI] [PubMed] [Google Scholar]
  • 74.Lerer B, Segman RH, Fangerau H, Daly AK, Basile VS, Cavallaro R, Aschauer HN, McCreadie RG, Ohlraun S, Ferrier N, Masellis M, Verga M, Scharfetter J, Rietschel M, Lovlie R, Levy UH, Meltzer HY, Kennedy JL, Steen VM, Macciardi F. Pharmacogenetics of tardive dyskinesia: combined analysis of 780 patients supports association with dopamine D3 receptor gene Ser9Gly polymorphism. Neuropsychopharmacology. 2002;27:105–119. doi: 10.1016/S0893-133X(02)00293-2. [DOI] [PubMed] [Google Scholar]
  • 75.Bakker PR, van Harten PN, van Os J. Antipsychotic-induced tardive dyskinesia and the Ser9Gly polymorphism in the DRD3 gene: a meta analysis. Schizophr Res. 2006;83:185–192. doi: 10.1016/j.schres.2006.01.010. [DOI] [PubMed] [Google Scholar]
  • 76.Tsai HT, North KE, West SL, Poole C. The DRD3 rs6280 polymorphism and prevalence of tardive dyskinesia: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:57–66. doi: 10.1002/ajmg.b.30946. [DOI] [PubMed] [Google Scholar]
  • 77.Tsai HT, Caroff SN, Miller DD, McEvoy J, Lieberman JA, North KE, Stroup TS, Sullivan PF. A candidate gene study of Tardive dyskinesia in the CATIE schizophrenia trial. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:336–340. doi: 10.1002/ajmg.b.30981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Willeit M, Praschak-Rieder N. Imaging the effects of genetic polymorphisms on radioligand binding in the living human brain: a review on genetic neuroreceptor imaging of monoaminergic systems in psychiatry. Neuroimage. 2010;53:878–892. doi: 10.1016/j.neuroimage.2010.04.030. [DOI] [PubMed] [Google Scholar]
  • 79.Roman T, Rohde LA, Hutz MH. Polymorphisms of the dopamine transporter gene: influence on response to methylphenidate in attention deficit-hyperactivity disorder. Am J Pharmacogenomics. 2004;4:83–92. doi: 10.2165/00129785-200404020-00003. [DOI] [PubMed] [Google Scholar]
  • 80.Cheon KA, Ryu YH, Kim JW, Cho DY. The 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 methylphenidate. Eur Neuropsychopharmacol. 2005;15:95–101. doi: 10.1016/j.euroneuro.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 81.Kambeitz J, Romanos M, Ettinger U. Meta-analysis of the association between dopamine transporter genotype and response to methylphenidate treatment in ADHD. Pharmacogenomics J. 2013 doi: 10.1038/tpj.2013.9. Apr 16. doi: 10.1038/tpj.2013.9. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 82.Xu M, Xing Q, Li S, Zheng Y, Wu S, Gao R, Yu L, Guo T, Yang Y, Liu J, Zhang A, Zhao X, He G, Zhou J, Wang L, Xuan J, Du J, Li X, Feng G, Lin Z, Xu Y, St Clair D, Lin Z, He L. Pharmacogenetic effects of dopamine transporter gene polymorphisms on response to chlorpromazine and clozapine and on extrapyramidal syndrome in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:1026–1032. doi: 10.1016/j.pnpbp.2010.05.017. [DOI] [PubMed] [Google Scholar]
  • 83.Zhang A, Xing Q, Wang L, Du J, Yu L, Lin Z, Li X, Feng G, He L. Dopamine transporter polymorphisms and risperidone response in Chinese schizophrenia patients: an association study. Pharmacogenomics. 2007;8:1337–1345. doi: 10.2217/14622416.8.10.1337. [DOI] [PubMed] [Google Scholar]
  • 84.Kirchheiner J, Nickchen K, Sasse J, Bauer M, Roots I, Brockmoller J. A 40-basepair VNTR polymorphism in the dopamine transporter (DAT1) gene and the rapid response to antidepressant treatment. Pharmacogenomics J. 2007;7:48–55. doi: 10.1038/sj.tpj.6500398. [DOI] [PubMed] [Google Scholar]
  • 85.Bolonna AA, Arranz MJ, Munro J, Osborne S, Petouni M, Martinez M, Kerwin RW. No influence of adrenergic receptor polymorphisms on schizophrenia and antipsychotic response. Neurosci Lett. 2000;280:65–68. doi: 10.1016/s0304-3940(99)01000-9. [DOI] [PubMed] [Google Scholar]
  • 86.Cheng C, Chiu HJ, Loh W, Chan CH, Hwu TM, Liu YR, Lan TH. Association of the ADRA1A gene and the severity of metabolic abnormalities in patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2012;36:205–210. doi: 10.1016/j.pnpbp.2011.10.011. [DOI] [PubMed] [Google Scholar]
  • 87.Talmud PJ, Cooper JA, Gaunt T, Holmes MV, Shah S, Palmen J, Drenos F, Shah T, Kumari M, Kivimaki M, Whittaker J, Lawlor DA, Day IN, Hingorani AD, Casas JP, Humphries SE. Variants of ADRA2A are associated with fasting glucose, blood pressure, body mass index and type 2 diabetes risk: meta-analysis of four prospective studies. Diabetologia. 2011;54:1710–1719. doi: 10.1007/s00125-011-2108-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Saiz PA, Susce MT, Clark DA, Kerwin RW, Molero P, Arranz MJ, de Leon J. An investigation of the alpha1A-adrenergic receptor gene and antipsychotic-induced side-effects. Hum Psychopharmacol. 2008;23:107–114. doi: 10.1002/hup.903. [DOI] [PubMed] [Google Scholar]
  • 89.Liu YR, Loh EW, Lan TH, Chen SF, Yu YH, Chang YH, Huang CJ, Hu TM, Lin KM, Yao YT, Chiu HJ. ADRA1A gene is associated with BMI in chronic schizophrenia patients exposed to antipsychotics. Pharmacogenomics J. 2010;10:30–39. doi: 10.1038/tpj.2009.55. [DOI] [PubMed] [Google Scholar]
  • 90.Wang YC, Bai YM, Chen JY, Lin CC, Lai IC, Liou YJ. Polymorphism of the adrenergic receptor alpha 2a-1291C>G genetic variation and clozapine-induced weight gain. J Neural Transm. 2005;112:1463–1468. doi: 10.1007/s00702-005-0291-7. [DOI] [PubMed] [Google Scholar]
  • 91.Sickert L, Müller DJ, Tiwari AK, Shaikh S, Zai C, De Souza R, De Luca V, Meltzer HY, Lieberman JA, Kennedy JL. Association of the alpha 2A adrenergic receptor-1291C/G polymorphism and antipsychotic-induced weight gain in European-Americans. Pharmacogenomics. 2009;10:1169–1176. doi: 10.2217/pgs.09.43. [DOI] [PubMed] [Google Scholar]
  • 92.De Luca V, Souza RP, Viggiano E, Sickert L, Teo C, Zai C, Tiwari AK, Müller DJ, Lieberman JA, Volavka J, Meltzer HY, Kennedy JL. Genetic interactions in the adrenergic system genes: analysis of antipsychotic-induced weight gain. Hum Psychopharmacol. 2011;26:386–391. doi: 10.1002/hup.1219. [DOI] [PubMed] [Google Scholar]
  • 93.Polanczyk G, Zeni C, Genro JP, Guimarães AP, Roman T, Hutz MH, Rohde LA. Association of the adrenergic alpha2A receptor gene with methylphenidate improvement of inattentive symptoms in children and adolescents with attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2007;64:218–224. doi: 10.1001/archpsyc.64.2.218. [DOI] [PubMed] [Google Scholar]
  • 94.da Silva TL, Pianca TG, Roman T, Hutz MH, Faraone SV, Schmitz M, Rohde LA. Adrenergic alpha2A receptor gene and response to methylphenidate in attention-deficit/hyperactivity disorder-predominantly inattentive type. J Neural Transm. 2008;115:341–345. doi: 10.1007/s00702-007-0835-0. [DOI] [PubMed] [Google Scholar]
  • 95.Cheon KA, Cho DY, Koo MS, Song DH, Namkoong K. Association between homozygosity of a G allele of the alpha-2a-adrenergic receptor gene and methylphenidate response in Korean children and adolescents with attention-deficit/hyperactivity disorder. Biol Psychiatry. 2009;65:564–570. doi: 10.1016/j.biopsych.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 96.Hong SB, Kim JW, Cho SC, Shin MS, Kim BN, Yoo HJ. Dopaminergic and noradrenergic gene polymorphisms and response to methylphenidate in korean children with attention-deficit/hyperactivity disorder: is there an interaction? J Child Adolesc Psychopharmacol. 2012;22:343–352. doi: 10.1089/cap.2011.0076. [DOI] [PubMed] [Google Scholar]
  • 97.Kim BN, Kim JW, Cummins TD, Bellgrove MA, Hawi Z, Hong SB, Yang YH, Kim HJ, Shin MS, Cho SC, Kim JH, Son JW, Shin YM, Chung US, Han DH. Norepinephrine genes predict response time variability and methylphenidate-induced changes in neuropsychological function in attention deficit hyperactivity disorder. J Clin Psychopharmacol. 2013;33:356–362. doi: 10.1097/JCP.0b013e31828f9fc3. [DOI] [PubMed] [Google Scholar]
  • 98.Contini V, Victor MM, Cerqueira CC, Polina ER, Grevet EH, Salgado CA, Karam RG, Vitola ES, Belmonte-de-Abreu P, Bau CH. Adrenergic alpha2A receptor gene is not associated with methylphenidate response in adults with ADHD. Eur Arch Psychiatry Clin Neurosci. 2011;261:205–211. doi: 10.1007/s00406-010-0172-4. [DOI] [PubMed] [Google Scholar]
  • 99.Yoshida K, Takahashi H, Higuchi H, Kamata M, Ito K, Sato K, Naito S, Shimizu T, Itoh K, Inoue K, Suzuki T, Nemeroff CB. Prediction of antidepressant response to milnacipran by norepinephrine transporter gene polymorphisms. Am J Psychiatry. 2004;161:1575–1580. doi: 10.1176/appi.ajp.161.9.1575. [DOI] [PubMed] [Google Scholar]
  • 100.Houston JP, Zou W, Aris V, Fijal B, Chen P, Heinloth AN, Martinez J. Evaluation of genetic models for response in a randomized clinical trial of duloxetine in major depressive disorder. Psychiatry Res. 2012;200:63–65. doi: 10.1016/j.psychres.2012.06.002. [DOI] [PubMed] [Google Scholar]
  • 101.Wakeno M, Kato M, Okugawa G, Fukuda T, Hosoi Y, Takekita Y, Yamashita M, Nonen S, Azuma J, Kinoshita T. The alpha 2A-adrenergic receptor gene polymorphism modifies antidepressant responses to milnacipran. J Clin Psychopharmacol. 2008;28:518–524. doi: 10.1097/JCP.0b013e31818455fc. [DOI] [PubMed] [Google Scholar]
  • 102.Kantrowitz J, Javitt DC. Glutamatergic transmission in schizophrenia: from basic research to clinical practice. Curr Opin Psychiatry. 2012;25:96–102. doi: 10.1097/YCO.0b013e32835035b2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Krystal JH, Sanacora G, Blumberg H, Anand A, Charney DS, Marek G, Epperson CN, Goddard A, Mason GF. Glutamate and GABA systems as targets for novel antidepressant and mood-stabilizing treatments. Mol Psychiatry. 2002;7(Suppl. 1):S71–80. doi: 10.1038/sj.mp.4001021. [DOI] [PubMed] [Google Scholar]
  • 104.Mallolas J, Hurtado O, Castellanos M, Blanco M, Sobrino T, Serena J, Vivancos J, Castillo J, Lizasoain I, Moro MA, Dávalos A. A polymorphism in the EAAT2 promoter is associated with higher glutamate concentrations and higher frequency of progressing stroke. J Exp Med. 2006;203:711–717. doi: 10.1084/jem.20051979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Dallaspezia S, Poletti S, Lorenzi C, Pirovano A, Colombo C, Benedetti F. Influence of an interaction between lithium salts and a functional polymorphism in SLC1A2 on the history of illness in bipolar disorder. Mol Diagn Ther. 2012;16:303–309. doi: 10.1007/s40291-012-0004-5. [DOI] [PubMed] [Google Scholar]
  • 106.Walitza S, Wendland JR, Gruenblatt E, Warnke A, Sontag TA, Tucha O, Lange KW. Genetics of early-onset obsessive-compulsive disorder. Eur Child Adolesc Psychiatry. 2010;19:227–235. doi: 10.1007/s00787-010-0087-7. [DOI] [PubMed] [Google Scholar]
  • 107.Kwon JS, Joo YH, Nam HJ, Lim M, Cho EY, Jung MH, Choi JS, Kim B, Kang DH, Oh S, Park T, Hong KS. Association of the glutamate transporter gene SLC1A1 with atypical antipsychotics-induced obsessive-compulsive symptoms. Arch Gen Psychiatry. 2009;66:1233–1241. doi: 10.1001/archgenpsychiatry.2009.155. [DOI] [PubMed] [Google Scholar]
  • 108.Hirunsatit R, George ED, Lipska BK, Elwafi HM, Sander L, Yrigollen CM, Gelernter J, Grigorenko EL, Lappalainen J, Mane S, Nairn AC, Kleinman JE, Simen AA. Twenty-one-base-pair insertion polymorphism creates an enhancer element and potentiates SLC6A1 GABA transporter promoter activity. Pharmacogenet Genomics. 2009;19:53–65. doi: 10.1097/FPC.0b013e328318b21a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Paddock S, Laje G, Charney D, Rush AJ, Wilson AF, Sorant AJ, Lipsky R, Wisniewski SR, Manji H, McMahon FJ. Association of GRIK4 with outcome of antidepressant treatment in the STAR*D cohort. Am J Psychiatry. 2007;164:1181–1188. doi: 10.1176/appi.ajp.2007.06111790. [DOI] [PubMed] [Google Scholar]
  • 110.Horstmann S, Lucae S, Menke A, Hennings JM, Ising M, Roeske D, Müller-Myhsok B, Holsboer F, Binder EB. Polymorphisms in GRIK4, HTR2A, and FKBP5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology. 2010;35:727–740. doi: 10.1038/npp.2009.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Serretti A, Chiesa A, Crisafulli C, Massat I, Linotte S, Calati R, Kasper S, Bailer U, Lecrubier Y, Fink M, Antonijevic I, Forray C, Snyder L, Bollen J, Zohar J, De Ronchi D, Souery D, Mendlewicz J. Failure to replicate influence of GRIK4 and GNB3 polymorphisms on treatment outcome in major depression. Neuropsychobiology. 2012;65:70–75. doi: 10.1159/000329553. [DOI] [PubMed] [Google Scholar]
  • 112.Pu M, Zhang Z, Xu Z, Shi Y, Geng L, Yuan Y, Zhang X, Reynolds GP. Influence of genetic polymorphisms in the glutamatergic and GABAergic systems and their interactions with environmental stressors on antidepressant response. Pharmacogenomics. 2013;14:277–288. doi: 10.2217/pgs.13.1. [DOI] [PubMed] [Google Scholar]
  • 113.Lavedan C, Licamele L, Volpi S, Hamilton J, Heaton C, Mack K, Lannan R, Thompson A, Wolfgang CD, Polymeropoulos MH. Association of the NPAS3 gene and five other loci with response to the antipsychotic iloperidone identified in a whole genome association study. Mol Psychiatry. 2009;14:804–819. doi: 10.1038/mp.2008.56. [DOI] [PubMed] [Google Scholar]
  • 114.Volpi S, Potkin SG, Malhotra AK, Licamele L, Lavedan C. Applicability of a genetic signature for enhanced iloperidone efficacy in the treatment of schizophrenia. J Clin Psychiatry. 2009;70:801–809. doi: 10.4088/jcp.08m04391. [DOI] [PubMed] [Google Scholar]
  • 115.Fijal BA, Stauffer VL, Kinon BJ, Conley RR, Hoffmann VP, Witte MM, Zhao F, Houston JP. Analysis of gene variants previously associated with iloperidone response in patients with schizophrenia who are treated with risperidone. J Clin Psychiatry. 2012;73:367–371. doi: 10.4088/JCP.10m06507. [DOI] [PubMed] [Google Scholar]
  • 116.Chaki S. Group II metabotropic glutamate receptor agonists as a potential drug for schizophrenia. Eur J Pharmacol. 2010;639:59–66. doi: 10.1016/j.ejphar.2009.12.041. [DOI] [PubMed] [Google Scholar]
  • 117.Fijal BA, Kinon BJ, Kapur S, Stauffer VL, Conley RR, Jamal HH, Kane JM, Witte MM, Houston JP. Candidate-gene association analysis of response to risperidone in African-American and white patients with schizophrenia. Pharmacogenomics J. 2009;9:311–318. doi: 10.1038/tpj.2009.24. [DOI] [PubMed] [Google Scholar]
  • 118.Bishop JR, Miller D, Ellingrod VL, Holman T. Association between type-three metabotropic glutamate receptor gene (GRM3) variants and symptom presentation in treatment refractory schizophrenia. Hum Psychopharmacol. 2011;26:28–34. doi: 10.1002/hup.1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bishop JR, Ellingrod VL, Moline J, Miller D. Association between the polymorphic GRM3 gene and negative symptom improvement during olanzapine treatment. Schizophr Res. 2005;77:253–260. doi: 10.1016/j.schres.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 120.Crisafulli C, Chiesa A, De Ronchi D, Han C, Lee SJ, Park MH, Patkar AA, Pae CU, Serretti A. Influence of GRIA1, GRIA2 and GRIA4 polymorphisms on diagnosis and response to antipsychotic treatment in patients with schizophrenia. Neurosci Lett. 2012;506:170–174. doi: 10.1016/j.neulet.2011.10.074. [DOI] [PubMed] [Google Scholar]
  • 121.Perlis RH, Smoller JW, Ferreira MA, McQuillin A, Bass N, Lawrence J, Sachs GS, Nimgaonkar V, Scolnick EM, Gurling H, Sklar P, Purcell S. A genomewide association study of response to lithium for prevention of recurrence in bipolar disorder. Am J Psychiatry. 2009;166:718–725. doi: 10.1176/appi.ajp.2009.08111633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Duffy A, Turecki G, Grof P, Cavazzoni P, Grof E, Joober R, Ahrens B, Berghöfer A, Müller-Oerlinghausen B, Dvoráková M, Libigerová E, Vojtěchovský M, Zvolský P, Nilsson A, Licht RW, Rasmussen NA, Schou M, Vestergaard P, Holzinger A, Schumann C, Thau K, Robertson C, Rouleau GA, Alda M. Association and linkage studies of candidate genes involved in GABAergic neurotransmission in lithium-responsive bipolar disorder. J Psychiatry Neurosci. 2000;25:353–358. [PMC free article] [PubMed] [Google Scholar]
  • 123.Inada T, Koga M, Ishiguro H, Horiuchi Y, Syu A, Yoshio T, Takahashi N, Ozaki N, Arinami T. Pathway-based association analysis of genome-wide screening data suggest that genes associated with the gamma-aminobutyric acid receptor signaling pathway are involved in neuroleptic-induced, treatment-resistant tardive dyskinesia. Pharmacogenet Genomics. 2008;18:317–323. doi: 10.1097/FPC.0b013e3282f70492. [DOI] [PubMed] [Google Scholar]
  • 124.Giegling I, Drago A, Dolžan V, Plesničar BK, Schäfer M, Hartmann AM, Sander T, Toliat MR, Möller HJ, Stassen HH, Rujescu D, Serretti A. Glutamatergic gene variants impact the clinical profile of efficacy and side effects of haloperidol. Pharmacogenet Genomics. 2011;21:206–216. doi: 10.1097/FPC.0b013e32833efb18. [DOI] [PubMed] [Google Scholar]
  • 125.Nemeroff CB, Vale WW. The neurobiology of depression: inroads to treatment and new drug discovery. J Clin Psychiatry. 2005;66(Suppl. 7):5–13. [PubMed] [Google Scholar]
  • 126.Mandelli L, Marino E, Pirovano A, Calati R, Zanardi R, Colombo C, Serretti A. Interaction between SERTPR and stressful life events on response to antidepressant treatment. Eur Neuropsychopharmacol. 2009;19:64–67. doi: 10.1016/j.euroneuro.2008.08.005. [DOI] [PubMed] [Google Scholar]
  • 127.Keers R, Uher R, Huezo-Diaz P, Smith R, Jaffee S, Rietschel M, Henigsberg N, Kozel D, Mors O, Maier W, Zobel A, Hauser J, Souery D, Placentino A, Larsen ER, Dmitrzak-Weglarz M, Gupta B, Hoda F, Craig I, McGuffin P, Farmer AE, Aitchison KJ. Interaction between serotonin transporter gene variants and life events predicts response to antidepressants in the GENDEP project. Pharmacogenomics J. 2011;11:138–145. doi: 10.1038/tpj.2010.14. [DOI] [PubMed] [Google Scholar]
  • 128.McCormick CM, Mathews IZ. Development, hypothalamic-pituitary-adrenal function, and programming of adult learning and memory. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:756–765. doi: 10.1016/j.pnpbp.2009.09.019. [DOI] [PubMed] [Google Scholar]
  • 129.Vijayendran M, Beach SR, Plume JM, Brody GH, Philibert RA. Effects of genotype and child abuse on DNA methylation and gene expression at the serotonin transporter. Front Psychiatry. 2012;3:55. doi: 10.3389/fpsyt.2012.00055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Beach SR, Brody GH, Todorov AA, Gunter TD, Philibert RA. Methylation at SLC6A4 is linked to family history of child abuse: an examination of the Iowa Adoptee sample. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:710–713. doi: 10.1002/ajmg.b.31028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kang HJ, Kim JM, Stewart R, Kim SY, Bae KY, Kim SW, Shin IS, Shin MG, Yoon JS. Association of SLC6A4 methylation with early adversity, characteristics and outcomes in depression. Prog Neuropsychopharmacol Biol Psychiatry. 2013;44:23–28. doi: 10.1016/j.pnpbp.2013.01.006. [DOI] [PubMed] [Google Scholar]
  • 132.Sales AJ, Biojone C, Terceti MS, Guimarães FS, Gomes MV, Joca SR. intra-hippocampal administration of DNA methylation inhibitors. Br J Pharmacol. 2011;164:1711–1721. doi: 10.1111/j.1476-5381.2011.01489.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Melas PA, Rogdaki M, Ösby U, Schalling M, Lavebratt C, Ekström TJ. Epigenetic aberrations in leukocytes of patients with schizophrenia: association of global DNA methylation with antipsychotic drug treatment and disease onset. FASEB J. 2012;26:2712–2718. doi: 10.1096/fj.11-202069. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES