Supplementary Information (docx 473K)

advertisement
Supplemental File
Evaluating Historical Candidate Genes For Schizophrenia
Farrell et al.
Table of Contents
Supplemental text from informed investigators ........................................................................................... 2
AKT1.............................................................................................................................................................................................................................. 2
CHRNA7 ....................................................................................................................................................................................................................... 2
COMT............................................................................................................................................................................................................................. 2
DAO and DAOA .......................................................................................................................................................................................................... 3
DISC1............................................................................................................................................................................................................................. 4
DRD2 ............................................................................................................................................................................................................................. 5
DRD3 ............................................................................................................................................................................................................................. 5
DTNPB1........................................................................................................................................................................................................................ 6
HTR2A........................................................................................................................................................................................................................... 6
NRG1 ............................................................................................................................................................................................................................. 7
RGS4 .............................................................................................................................................................................................................................. 8
Supplementary Figures .............................................................................................................................. 10
Figure S1. PGC SCZ mega-analysis vs. SZGene candidate gene meta-analysis .............................................................................10
Figure S2. Gene ratings from 24 schizophrenia geneticists .................................................................................................................11
Supplemental Tables ................................................................................................................................. 12
Table S1. Results for 59 markers in/near 25 schizophrenia candidate genes.............................................................................12
Table S2. Independent meta-analyses for historical genes published 2008-2014 ....................................................................14
Table S3. Candidate gene studies of DAO ....................................................................................................................................................15
Table S4. Candidate gene studies of DAOA .................................................................................................................................................19
References ................................................................................................................................................ 31
1
Supplemental text from informed investigators
AKT1
Personal communication from Drs Joseph Gogos and Maria Karayiorgou (Columbia University, New
York, NY, USA).
Authoritative investigator rating: 5
PGC schizophrenia PI mean rating: 2.5
The rating for AKT1 was 5 (very likely). We base this rating on the original integrative biological data,
subsequent work in various genetic animal models (including unpublished work from our labs) as well as
gene network analysis that implicates AKT1/GSK3beta signaling as a major node connected to many SCZ
candidate genes. 1,2
CHRNA7
Personal communication from Drs Robert Freedman and Sherry Leonard (University of Colorado,
Denver, CO, USA).
Authoritative investigator rating: 5
PGC schizophrenia PI mean rating: 2.9
Linkage to schizophrenia of the CHRNA7 gene, which codes for the α7-nicotinic receptor subunit, was first
observed using a physiological endophenotype, auditory P50 evoked potential inhibition, that had been
previously associated with schizophrenia and α-7 nicotinic receptor function (LOD 5.2) 3. Subsequent
study showed association of both schizophrenia and the P50 endophenotype with SNPs in the proximal
CHRNA7 promoter that decrease expression in vitro 4. This finding is consonant with several reports of
decreased expression of α7-nicotinic receptor subunits in post mortem schizophrenia samples 5. A SNP in
the more distal promoter has the most significant reported association, P=0.0009, African-American;
P=0.013, Caucasian 6. Perhaps the most persuasive evidence for the CHRNA7 gene’s role in schizophrenia
is the well-replicated finding of 15q13.3 CNV CHRNA7 deletions 7,8. CHRFAM7A, a partial duplication
containing CHRNA7 exons 5-10, which is only found in humans, also maps nearby on 15q 9, and
polymorphisms in CHRFAM7A are associated with schizophrenia (Sinkus 2009). CHRFAM7A product
subunits can associate with CHRNA7 subunits as part of the α7-nicotinic acetylcholine receptor, which is
a pentameric ring. Compared to a receptor composed entirely of CHRNA7 subunits, those incorporating
CHRFAM7A subunits have significantly decreased ion flux, which suggests that CHRFAM7A may limit
CHRNA7 function and thereby have an additional pathological role 10.
COMT
Personal communication from Dr Daniel R Weinberger (Lieber Institute, Johns Hopkins University
School of Medicine).
Authoritative investigator rating: 1 (main effect), 4 (epistatic effect)
PGC schizophrenia PI mean rating: 2.4
Catechol-o-methyl transferase (COMT) has been perhaps the most extensively studied candidate gene in
behavioral genetics since the initial report of an association with schizophrenia in 1996 11. Its popularity
is based on many factors, including that it is mapped to an early schizophrenia linkage region on
chromosome 22, it is contained within the 22q11 hemi-deletion critical region of the VCFS syndrome, and
it is involved in catecholamine metabolism, a virtually sacrosanct biochemistry of neurotransmitters
related to psychiatric disorders and their treatment. Since the initial report, over twenty independent
case control studies found significant association with common COMT variants and psychosis, with
another ten family based studies also showing significant association 12. Even an early genome-wide
association study found a nominally significant signal in COMT 13. COMT shows significant association
signals in the current SZGene meta-analysis 12 and has been linked with psychosis in cases with
velocardiofacial syndrome (VCFS) 14,15. COMT also has been shown in several studies to interact with
environmental risk factors associated with psychosis (e.g. cannabis) 16,17. Most of the early studies
2
focused on a common coding variant with established effect on enzyme activity, the Val158Met variation,
but recent reports have emphasized signals in other polymorphisms. In addition to these associations
with clinical diagnosis, beginning with the first study of association of COMT with cognition and cortical
physiology 18 and with evidence that COMT is primarily involved in cortical dopamine flux 19, over 50
reports have documented a role for COMT in cortical information processing, establishing a biologic basis
for understanding a potential mechanism of association of COMT with behavioral syndromes. Virtually all
of the clinical associations of COMT with aspects of cortical function related to cognition and emotion
have been confirmed in studies of genetically engineered mice with analogous COMT variation 20-22.
Given these multiple positive reports of clinical association with albeit relatively small samples and the
elaboration of a biological mechanism by which COMT could influence brain function linked to psychosis
and other psychiatric disorders, why has association been so decidedly negative at the single locus level
in the current incarnation of GWAS data? And is the prior evidence of a role of COMT in brain function
related to psychiatric disorders a “red herring”? A reasoned response to these questions is that the jury is
still out on both counts. The failure to find association in the current GWAS data may reflect interactions
with modifying factors and the biologic heterogeneity of functional variants within the COMT gene that
are not accounted for in the current GWAS single locus association analyses. Several functional
polymorphisms within the COMT gene have been implicated 23-25, each acting via different molecular
mechanisms, so their cumulative effects are complex. A recent study of molecular haplotypes in
individuals with VCFS showed that alleles at one variant have paradoxical effects on COMT expression
and enzyme activity in the context of alleles at other variants 14. There are considerable population
differences in allele frequencies at the various functional polymorphisms within COMT, adding
insuperable heterogeneity to single locus analyses. Sex effects on COMT associations also have been
identified in clinical studies and in mice 20,22. Another complication with association with COMT involves
the biology of cortical dopamine function. The relationship between cortical function and dopamine
activity is nonlinear 26 and thus, predictable relationships between single SNP allele load and clinical
biology may require multilocus and nonlinear (e.g. epistatic) models. A recent study in mice and in
humans demonstrated that the COMT Val158Met polymorphism shows opposite associations with
cortical function depending on haplotype at another gene (DTNBP1) that also influences cortical
dopamine signaling 27. Interestingly, one meta-analysis of COMT associations with psychosis suggested
that the strongest signal was a protective association with heterozygosity at the Val158Met variant,
implying that in a given context (i.e. varying genomes and environments), too much or too little COMT
mediated cortical dopamine could have similar associations 28. Such effects would literally obliterate
single locus association in heterogeneous samples such as current GWAS. It is reasonable to assume that
COMT is a genetic risk factor for psychiatric syndromes linked to cortical dopamine signaling, but
statistical confirmation of this association will require multifactorial and probably nonlinear genetic
models that address the critical role of context, both genomic and environmental.
DAO and DAOA
Personal communication from Drs Marcella Rietschel and Jens Treutlein (Central Institute of Mental
Health, University of Heidelberg, Mannheim, Germany).
Authoritative investigator rating: 3, 3
PGC schizophrenia PI mean rating: 2.2, 2.0
See Tables S2 and S3. The first report of an association between schizophrenia and variants in the genes
d-amino acid oxidase (DAO) and d-amino acid oxidase activator (DAOA, alias G72) was published in 2002.
In a sample of 213 schizophrenia patients and 241 controls from Canada, Chumakov et al. performed a
systematic investigation of SNPs in a 5-Mb segment of the 13q34 chromosomal region. Two of the six
nominally significant variants from this study were subsequently replicated in an independent
schizophrenia sample 29, and in a sample of patients with bipolar disorder 30. These two variants were
located in the DAOA gene, whose product interacts with DAO. DAO metabolizes D-serine, which is an
3
activator of NMDA glutamate receptors, and glutamatergic signaling has been implicated in the etiology
of psychotic symptoms and schizophrenia.
Since these genes were plausible candidates for psychiatric disorders, numerous studies attempted to
corroborate their involvement in schizophrenia, bipolar disorder, and related traits. To our knowledge, a
total of ~76 studies for DAOA and ~25 studies for DAO have tested markers in schizophrenia and related
phenotypes. However, the findings have been inconsistent. Furthermore, few findings remained
significant following correction for the number of SNPs tested.
Several meta-analyses of the role of DAOA in schizophrenia and bipolar disorder have been published.
The meta-analysis by Detera-Wadleigh & McMahon in 2006 31 included seven studies of schizophrenia
and three studies of bipolar disorder, and found association between schizophrenia and the markers
M12, M15, M22, M23, and M24. The meta-analysis by Shi et al. in 2008 32 included 23 studies, and
detected association between schizophrenia and the markers M18, M22, and M24. The meta-analysis by
Müller et al. in 2011 33 included 24 studies of schizophrenia and 11 studies of bipolar disorder, and
revealed association between schizophrenia and the marker M15. The most recent meta-analysis by Tan
et al. in 2014 34 included 46 studies (17,515 cases and 25,189 controls), and tested the markers M15,
M18, and M23. However, no significant finding was identified.
In summary, the available data provide no support for the hypothesis that variants in DAO or DAOA
confer risk for schizophrenia. This view is supported by the observation that the number of positive
findings in meta-analyses is inversely proportional to the number of studies examined. It could be argued,
however, that since the anticipated effect sizes are small, and that the effects of genetic variants will be
blurred by interaction with other factors, this inverse relationship is unsurprising when increasing
heterogeneity of the samples. Interestingly, research has indicated possible association between specific
markers in DAO and DAOA and more homogenous patient groups, e.g. association between markers M23
and M24 in Europeans. Given their function, and the fact that genetic variation in these genes is unlikely
to occur without any effect, DAO and DAOA remain plausible candidate genes for schizophrenia. However,
confirmation of this hypothesis can not be made on the basis on the available data but will require further
analyses.
DISC1
Personal communication from Dr David Porteous (Centre for Genomic and Experimental Medicine,
Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital,
Crewe Road, Edinburgh, UK).
Authoritative investigator rating: 4.5
PGC schizophrenia PI mean rating: 2.7
Disrupted in Schizophrenia 1 (DISC1) is an archetypal positional candidate gene, disrupted by a balanced
t(1;11) translocation that is linked, with genome-wide significance, to schizophrenia alone (LOD = 3.6)
and to schizophrenia plus related mental illness (MLOD = 7.1) in a large Scottish family 35,36. Thus, the
DISC1 locus meets the primary genetic criterion for candidature and merits biological validation. DISC1
encodes a scaffold protein regulating neuronal proliferation, migration, integration and signaling, spine
formation and synapse maintenance-neurobiology that aligns well with current concepts in
schizophrenia 37. Moreover, DISC1 protein binding partners, including PDE4B, PCM1 and NDE1, are
independently implicated as genetic risk factors for schizophrenia and related major mental illness
through cytogenetics, copy number variation and linkage (reviewed in 38). Rare DISC1 amino acid
substitution variants have been reported in schizophrenia 39, recurrent major depressive disorder,
segregating with psychiatric disorder 40 and autism 41. In contrast, meta-analysis of common DISC1 SNPs
has found no evidence of genome-wide significant association with schizophrenia 42 and DISC1 does not
appear on the GWAS list of significant associations for schizophrenia 43. The absence of evidence from
4
GWAS is, of course, not evidence of absence of effect; while GWAS can identify ancient variants common
to all populations, variants that are moderate to rare and/or population-specific and/or disorder
subtype-specific for illness will fall below the accepted level of genome wide significance. A further
limitation of GWAS and many CNV studies is that they do not identify functional variants directly. In this
regard, structure-function analysis of DISC1 has proved fruitful, demonstrating biological pleiotropy and
epistasis that is hidden from current GWAS analysis 44. There is good evidence from human 45,46 and
mouse 47 studies that modulation of DISC1 expression affects systems level changes in biologically
relevant pathways and clinical endophenotypes. Amino acid substitution polymorphisms and rare
variants have been shown experimentally to modulate structural and oligomerisation properties of DISC1
and to modulate or abrogate relevant biological traits, including -catenin signaling, ATF4-dependent
stress response and mitochondrial trafficking 48-51. Thus, DISC1 is analogous to the examples of APP and
PSEN1 and 2 in Alzheimer’s disease 52; risk genes that may score low on GWAS criteria, but high on rare
variant criteria do offer valuable and direct etiological insight into psychiatric disorders. Score = 4.5/5
(genome wide significant by linkage; functional genetic variants and biological evidence for relevance).
DRD2
Personal communication from Drs James Kennedy and Clement Zia (Centre for Addiction and Mental
Health, University of Toronto, Toronto, Ontario, Canada).
Authoritative investigator rating: 4
PGC schizophrenia PI mean rating: 4.1
One of the top hits in the most recent Psychiatric Genomics Consortium GWAS meta-analysis of
schizophrenia is rs2514218, located approximately 50kb upstream of DRD2. There is substantial
neurobiological evidence supporting a role of the dopamine D2 receptor in schizophrenia, most notably
the fact that all efficacious antipsychotic medications have significant D2 antagonism (Seeman et al,
1975). Furthermore, schizophrenia patients show elevated striatal D2 levels (e.g., SPECT 53) as well as D2
receptor super-sensitivity 54,55. These findings collectively suggest that rs2514218 and/or correlated
variants in regulatory sites may create elevated risk for schizophrenia by giving rise to higher D2
receptor expression. While rs2514218 does not appear to have a strong functional role based on ENCODE
data 56-58, molecular studies using tissues from specific brain regions will clarify the molecular
mechanisms of this genome-wide significant association. Interrogation of DRD2 regulatory sites for
possible association with the increased striatal D2 levels in schizophrenia patients may be useful.
D2 receptor occupancy is correlated with therapeutic efficacy for antipsychotics 59. Putative functional
single-nucleotide polymorphisms in DRD2 have been implicated in antipsychotic response (rs1799732/141C insertion/deletion 60) and side effects including rs1800497/TaqIA in tardive dyskinesia 61,62 and
rs6277/C957T 63 in antipsychotic-induced weight gain. However, association findings of these and other
DRD2 markers in the diagnosis of schizophrenia have been mixed 64, suggesting that the role of D2 in the
etiopathophysiology of schizophrenia may be modulatory rather than directly causative. Interestingly,
ZNF804A, another one of the top hits in the PGC2 GWAS meta-analysis, may regulate the expression of
DRD2 in addition to other schizophrenia candidate genes 65. Given this observation as well as the
complexity of D2-mediated signaling, further DRD2 gene-based studies as well as gene-gene interaction
and pathway analyses are required to decipher the role of DRD2 in schizophrenia. Based on its welldocumented therapeutic role, we can be cautiously optimistic that DRD2 may become one of the more
meaningful GWAS hits in terms of translational impact.
DRD3
Personal communication from Dr Michael Owen (Cardiff University, Cardiff, Wales, UK).
Authoritative investigator rating: 2
PGC schizophrenia PI mean rating: 2.3
5
This was an excellent functional candidate gene for schizophrenia given its role in pharmacology and its
location in the brain. In the light of what we know now about the genetic architecture of schizophrenia,
the initial studies and even the several meta-analyses that we, and others, conducted were
underpowered. The updated results of meta-analysis of candidate gene studies in SZGene and the
findings from PGC2 provide no support for the hypothesis that common genetic variants in this gene
confer risk of schizophrenia.
DTNPB1
Personal communication from Dr Brien Riley (Virginia Institute of Psychiatric and Behavioral
Genetics, Virginia Commonwealth University, Richmond, VA, USA).
Authoritative investigator rating: 2
PGC schizophrenia PI mean rating: 2.4
The score is based on the following rationale. If a “1” signifies a locus for which there has never been any
credible support (i.e., null expectation), there is some evidence for a locus in this region of chr 6 in family
studies and it does not seem to me, although this is intuitive rather than empirical, that the chr 6p signal
is being driven by the HLA region signal, although it is certainly possible. However, the pattern of
different haplotypes associated in almost every sample examined strongly suggests that there is no
stable association signal detectable at this locus in European samples. I think this is also the answer to
the publication pattern question: the focus in the early days of association studies was on any signal from
a locus of interest with very little attention paid to whether the specific signal observed was consistent
(i.e., really represented replication).
HTR2A
Personal communication from Drs Jiang Li and Herbert Y. Meltzer (Northwestern University,
Feinberg School of Medicine).
Authoritative investigator rating: 4
PGC schizophrenia PI mean rating: 2.3
Three HTR2A polymorphisms, rs6311, rs6313, rs6314, have been widely investigated in association
studies with schizophrenia. rs6311 (-1438G>A), located in the promoter region of HTR2A, is in strong
linkage disequilibrium with rs6313 (102T>C), a synonymous mutation. rs6314 (His452Tyr), the most
common missense variant, is located at the C-terminal region. It has been implicated in binding of
accessory/scaffolding proteins as well as G protein-coupled receptor desensitization and internalization
66. rs6313, a silent substitution, has been controversially associated with schizophrenia 67-75. It has been
associated with a subgroup of schizophrenia patients characterized by poor long-term outcome and
poor response to typical antipsychotic drugs 76. Prepulse inhibition and habituation has been reported to
be modulated by rs6311/rs6313 in schizophrenia, suggesting alterations in brain HTR2A may
contributes to the prepulse inhibition deficits present in some patients with schizophrenia. It conferred
an increased risk for psychosis in a meta-analysis of Alzheimer dementia subjects 77. These studies
support the conclusion that rs6311/rs6313 may contribute to an endophenotype characterized by poor
treatment response to clozapine, Only Arranz et al. reported positive finding with regard to clozapine
response in association with rs6311/rs6313 78,79. Others cannot replicate their findings 80-85. A metaanalysis of the association between HTR2A polymorphisms and clozapine response showed a deviation
from theoretically expected pattern in the funnel plots where studies should converge with increasing
sample size, suggesting a possibility of publication bias. The weighted mean of the odds ratios of all
studies on HTR2A genotypes including rs6311/rs6313/rs6314 was 1.7, thus, predicting only a minor
influence of this gene on clozapine response86.
rs6314 has been associated with hippocampal volume 87 and novelty processing in humans 88. It has been
shown to predict poor response to the atypical antipsychotic drug, clozapine, in some 78-80,89, but not all
studies 82-84. A recent study examined evidence from functional imaging and clinical data and reported
6
that rs6314 is associated with prefrontal function and response of negative symptoms to olanzapine, a
clozapine-like atypical antipsychotic drug 90. Functional studies have suggested that rs6314 may impact
on calcium signaling and mobilization as well as on reduced activation of phospholipases C and D,
resulting in markedly reduced intracellular signaling capacity 91,92. An in silico-based bioinformatics
analysis predict that rs6314 alters patterns of splicing with possible effects on HTR2A expression 90.
Based on an early study of allelic expression imbalance (AEI) in 23 adult brain tissue heterozygous for
rs6313, Bray et al. asserted that the HTR2A locus does not contain common variants affecting mRNA
levels in adult brain 93. However, Smith et al. recently combined in vitro gene-reporter assay and AEI
analysis with next-generation sequencing technology to show that significant AEI can be observed only in
the extended form of HTR2A harboring rs6311, but not in any mature mRNA transcripts. The
translational efficiency of long UTR is higher than short UTR, suggesting that rs6311 decreases usage of
an upstream transcription start site encoding a longer 5’UTR with greater translational efficiency 94.
According to postmortem prefrontal cortex samples from BrainCloud 95, promoter SNP rs6311(LD with
rs6313) is also a methylation QTL for HTR2A as it is highly correlated with CpG methylation at the HTR2A
across ethnicity 94, further suggesting potential epigenetic effect of this polymorphism on gene
expression. In conclusion, the common functional variants including rs6311, rs6313 and rs6314 of
HTR2A make a limited but definite contribution to the risk of schizophrenia and treatment response to
clozapine. Controversial results warrant more extensive investigation. Further study should focus on the
association with endophenotypes instead of a complex disease trait alone, or treatment response in
general, as the biological and pharmacological background influencing those traits could be largely
heterogeneous. Further study should also focus on the interaction between the genetic variants and
environmental factors in the context of the association with schizophrenia or drug response. With
increased sample size, not only common variants, but also rare mutations should also be included in
analysis to determine the accumulated genetic burden in conferring the risk for the disease or
endophenotypes.
NRG1
Personal communication from Dr Amanda Law (University of Colorado, Denver, CO, USA).
Authoritative investigator rating: 3
PGC schizophrenia PI mean rating: 2.9
Neuregulin 1 (NRG1) is an essential neurotrophin, a critical mediator of neurodevelopment, synaptic
plasticity and neocortical function and one of the most researched candidate susceptibility genes for
schizophrenia. NRG1 maps to 8p21-p12, a chromosomal region with prior evidence of genome-wide
linkage to schizophrenia 96-98. Association to common genetic variation in NRG1 and schizophrenia was
first reported in the landmark study of Stefansson and colleagues, in 2002 99, whereby genome-wide
linkage scans, extensive fine mapping and association analyses of extended Icelandic pedigrees identified
a 290kb risk haplotype in the 5’ region of NRG1 (HAPICE); a finding confirmed in Scottish 100 and Irish 101
populations. Subsequent association has been validated in three separate meta-analyses 102-104. Since the
original report, 30 independent studies have reported significant association to common NRG1 variants
(most notably complex haplotypes) and schizophrenia in European, Asian and African populations 12.
While NRG1 did not show significant association in the current SZGene meta-analysis 12, more recent
updates list NRG1 as positive 12 and recent analyses of three European genome-wide data sets provide
support for association of NRG1 HAPICE and additional novel haplotypes with schizophrenia 105.
Nevertheless, it is important to note that the associated haplotype or variants are not always HAPICE, (a
finding likely related to population heterogeneity), no obviously pathogenic mutations or copy number
variations have been identified in NRG1 and several negative studies exist12,106.
In addition to statistical associations, the evidence in favor of NRG1 as a schizophrenia risk gene is
strengthened by biological findings. First, risk polymorphisms in NRG1 are associated in humans with
multiple neurobiological phenotypes relevant to schizophrenia, including cognitive function, prefrontal
7
cortical physiology, and neocortical development 107-109. Second, neurobiological roles of NRG1 are
relevant to the disease process of schizophrenia, including key roles in neuronal migration,
neurotransmitter function, cortical inhibitory circuit development and myelination 106. Third, NRG1 gene
expression is altered in the brain of patients with schizophrenia and related to risk polymorphisms 110112; and fourth, mutation of NRG1 in rodents produces abnormalities in several behavioral domains
relevant to schizophrenia, including learning, memory, cognitive function and sociability 99,113-115.
Interestingly, genetic manipulations in mice 99,113-115 demonstrate that ‘overexpression’ or ‘knockdown’ of
the NRG1 gene leads to similar schizophrenia-related behaviors, suggesting a non-linear, inverted-U
model of NRG1 signaling and schizophrenia, an observation which may have important implications for
genetic association studies.
Even so, despite multiple positive reports of clinical association, the distinct neurobiological relevance of
NRG1’s functions to neurodevelopment and cortical function, and identification of potential biological
mechanisms of risk, large genome-wide association studies of schizophrenia have failed to provide
corroborative evidence of association to NRG1. Lack of support from this approach raises several
important questions regarding the validity of pre-GWAS genetic associations to NRG1; challenges
whether NRG1 is a reliable, disease-relevant genetic factor for schizophrenia, and raises the issue of
whether current GWAS single-marker association approaches are capable of identifying association to
complex genetic loci. It would be only pragmatic to conclude that the current data are inconclusive as to
whether NRG1 is a significant genetic risk factor for schizophrenia. However, failure to find association to
NRG1 in current schizophrenia GWAS may relate to several factors, including: limitations of singlemarker analysis; noting that the majority of previous associations to NRG1 are to complex and diverse
haplotypes, not single nucleotide variants 99-104,106; heterogeneity; noting that original isolation of the
gene was in small, relatively homogenous populations 99-101 and heterogeneity within the 1.2Mb, vastly
complex gene 106 has been shown to be pervasive within and across larger populations 104,106; geneenvironment interactions; noting the significant interaction of NRG1 with environmental risk variables,
including obstetric complications, psychosocial stress and cannabis 116,117, and finally gene-gene
interactions; noting variant interactions between NRG1 and genes in the neureuglin signaling pathway to
increase risk 108,118. Despite the failure of current GWAS to confirm association to NRG1, previous data
support critical roles for NRG1 in neurodevelopmental and neurobehavioral functions relevant to
schizophrenia. The ‘pre’- ‘post-GWAS’ validity conundrum will only likely resolve for NRG1 with the
application of multifactorial methods for GWAS data analysis which account for association in the context
of genetic background, genetic interplay and environmental modification.
RGS4
Personal communication from Dr Karoly Mirnics (Vanderbilt University, Nashville, KY, USA).
Authoritative investigator rating: 2
PGC schizophrenia PI mean rating: 2.1
Based on the current golden standards, the evidence that RGS4 sequence variants predispose to
schizophrenia is weak. However, this does not mean that RGS4 is not important for the disease
pathophysiology. These two categories should be always considered separately as a gene that has no
sequence variant predisposing toward a disease can be still critical for the overall progression of
pathology and emergence of symptoms. RGS4, just like most of the initial schizophrenia candidate genes,
did not show positive association in GWA studies (including BDNF, COMT, RELN and others), but the
evidence for their involvement in the disease progression and schizophrenia-associated endophenotypes
can be considered very strong. The following genetic studies support RGS4 as a schizophrenia
susceptibility gene 119-129. The following candidate gene studies (in addition to GWAS) do not support the
notion that sequence variants of RGS4 predispose to schizophrenia 130-139. The pathophysiological
evidence supporting a role for RGS4 in schizophrenia include:
1) Decreased RGS4 brain transcript expression in schizophrenia 140-143
8
2)
3)
4)
5)
6)
7)
8)
9)
10)
Decreased RGS4 brain protein expression in schizophrenia 144
Plausibility of biological effects – regulation of GPCR signaling 145-150
Genetic association/DNA methylation with suicide attempts in schizophrenia 119
Converging “hub” gene and interaction with other schizophrenia susceptibility genes 121,151-156
Cortical thickness associated with genotype 125
Brain connectivity associated with genotype 157
Working memory/cognitive performance associated with genotype 120,158-160
Medication efficacy depending on genotype 161,162
Antipsychotic treatment increases RGS4 membrane expression 163
9
Supplementary Figures
Figure S1. PGC SCZ mega-analysis vs. SZGene candidate gene meta-analysis
For each gene, the x-axis is the –log10(P) from the SZGene meta-analysis and the y-axis is the –log10(P)
from the PGC mega-analysis for a 25 kb window encompassing the gene. The data points are sized and
colored by the number of pre-GWAS candidate gene studies as reported in Table 1. The horizontal line
indicates the genome-wide significance threshold (5x10-8).
10
Figure S2. Gene ratings from 24 schizophrenia geneticists
The x-axis labels indicate the gene being rated as a risk factor for schizophrenia and the y-axis indicates
the distribution of ratings. The vertical length of a colored bar represents the proportion of investigators
giving a particular rating to that gene. The rating color scheme is in the legend on the right. The ratings
were a summary judgment of the current status of a gene as a genetic risk factor for schizophrenia
(1=very unlikely and 5=very likely). On the far left, “equal distribution reference” provides a reference
bar size for equal distribution amongst raters for a given gene. The absence of a particular colored bar
indicates that none of the investigators gave that rating for a gene.
11
Supplemental Tables
Table S1. Results for 59 markers in/near 25 schizophrenia candidate genes
Gene
Marker
Studies
Ncase
AKT1
APOE
BDNF
rs3730358
.2/3/4
270C/T
rs6265
rs28531779
rs737865
rs165599
rs4680
rs3918346
rs947267
rs778294
rs3916967
rs3916965
rs778293
rs1421292
rs3918342
rs2391191
rs1322784
rs1000731
rs999710
rs6675281
rs3738401
rs821616
rs1800497
rs1799732
rs1801028
rs6280
rs4646983
rs2619528
rs909706
rs2619522
rs2619538
rs1018381
rs2005976
rs760761
rs2619539
rs1011313
rs3213207
rs2228595
2
15
7
9
2
6
8
28
5
6
6
8
9
9
9
10
11
6
6
7
8
9
9
10
10
16
44
3
6
7
7
7
9
9
10
10
11
13
3
2543
1549
1242
2397
861
4552
5359
7622
1364
1524
3334
3574
4058
4348
4712
3968
5083
3297
3412
3608
3896
4705
4705
1228
3457
4160
6031
406
3370
4081
4101
4332
4227
4761
4560
5451
5385
6305
1227
CHRNA7
COMT
DAO
DAOA
DISC1
DRD2
DRD3
DRD4
DTNBP1
GRM3
SZG OR
(95% CI)
1.01 (0.91-1.13)
0.99 (0.82-1.20)
0.68 (0.52-0.87)
0.95 (0.87-1.04)
0.97 (0.72-1.30)
1.00 (0.95-1.06)
1.00 (0.95-1.05)
1.00 (0.96-1.05)
1.00 (0.89-1.12)
1.05 (0.95-1.16)
0.96 (0.89-1.03)
1.00 (0.94-1.07)
0.95 (0.90-1.01)
1.00 (0.94-1.06)
1.02 (0.97-1.08)
0.99 (0.94-1.05)
0.97 (0.92-1.02)
0.99 (0.92-1.07)
1.06 (0.98-1.14)
1.07 (1.00-1.14)
1.03 (0.95-1.12)
1.05 (0.99-1.12)
1.00 (0.94-1.06)
0.90 (0.79-1.03)
0.92 (0.82-1.02)
0.85 (0.71-1.03)
1.03 (0.97-1.08)
1.13 (0.76-1.67)
1.01 (0.93-1.09)
1.04 (0.97-1.10)
1.01 (0.93-1.08)
1.01 (0.95-1.07)
1.04 (0.93-1.15)
0.95 (0.89-1.02)
0.97 (0.90-1.04)
1.04 (0.99-1.10)
1.12 (1.02-1.23)
1.10 (1.02-1.19)
1.21 (0.96-1.52)
SZG P
0.82
0.95
0.0028
0.29
0.82
0.96
0.99
0.92
0.94
0.33
0.22
0.97
0.11
0.93
0.37
0.80
0.26
0.81
0.12
0.045
0.48
0.095
0.93
0.14
0.12
0.10
0.33
0.56
0.89
0.29
0.88
0.80
0.51
0.17
0.37
0.12
0.015
0.015
0.099
PGC OR
(95% CI)
1.02 (0.99-1.06)
0.99 (0.96-1.02) †
1.01 (0.97-1.06) †
0.95 (0.92-0.97)
1.01 (0.96-1.05)
0.98 (0.96-1.01)
1.00 (0.99-1.03)
0.99 (0.97-1.01)
1.03 (1.00-1.05)
0.99 (0.97-1.01)
1.00 (0.98-1.03)
1.00 (0.98-1.02)
1.00 (0.98-1.02)
1.01 (0.99-1.03)
1.00 (0.98-1.02)
1.00 (0.98-1.02)
1.00 (0.98-1.02)
1.00 (0.98-1.03)
1.02 (0.99-1.04)
1.01 (0.99-1.03)
0.98 (0.95-1.01)
1.00 (0.97-1.02)
1.00 (0.98-1.03)
1.00 (0.98-1.03)
No data
0.95 (0.89-1.03)
0.99 (0.97-1.01)
No data
0.97 (0.94-0.99)
1.01 (0.99-1.03)
1.03 (1.00-1.06)
1.00 (0.98-1.03)
0.99 (0.96-1.03)
0.97 (0.95-1.00)
0.97 (0.95-1.00)
0.99 (0.97-1.01)
0.99 (0.96-1.03)
1.04 (1.01-1.08)
1.01 (0.97-1.06)
12
PGC P
0.17
0.48
0.55
8.0e-5
0.79
0.17
0.29
0.56
0.035
0.30
0.82
0.88
0.96
0.53
0.81
0.77
0.92
0.82
0.22
0.29
0.13
0.68
0.76
0.88
0.22
0.31
0.019
0.35
0.020
0.74
0.72
0.022
0.021
0.32
0.78
0.012
0.58
PGC Pmin
±25kb
3.0e-4
0.0095
8.0e-5
PGC SNPs
±25kb
330
195
279
0.0096
0.0065
540
419
0.00040
0.015
390
360
0.00095
1652
8.3e-9
369
0.015
0.0026
0.0073
403
287
574
1.0e-10
677
HTR2A
KCNN3
MTHFR
NOTCH4
NRG1
PPP3CC
PRODH
RGS4
SLC6A3
SLC6A4
TNF
ZDHHC8
rs6311
rs6313
1333T/C
rs1801131
rs1801133
rs367398
rs6994992
rs62510682
rs35753505
rs7837713
rs383964
rs10917670
rs951439
rs2661319
rs951436
VNTR
5-HTTVNTR
5-HTTLPR
rs1800629
rs175174
7
26
1
8
11
5
6
6
13
1
4
7
7
8
9
6
9
15
7
4
2717
6384
184
1964
2529
1045
3999
4192
5874
1870
1453
4377
4519
4827
5049
731
1951
2652
1081
1461
1.14 (1.06-1.23)
0.98 (0.93-1.03)
1.12 (0.33-3.76)
0.91 (0.84-0.99)
1.09 (1.01-1.17)
1.00 (0.87-1.15)
1.06 (0.99-1.13)
0.94 (0.88-1.01)
0.95 (0.90-1.01)
0.99 (0.81-1.21)
1.09 (0.88-1.35)
0.94 (0.89-0.99)
0.97 (0.91-1.03)
0.93 (0.88-0.99)
1.01 (0.96-1.07)
0.97 (0.82-1.16)
1.11 (1.01-1.21)
1.01 (0.94-1.09)
1.00 (0.86-1.17)
1.00 (0.90-1.11)
0.0005
0.40
0.86
0.031
0.026
0.99
0.087
0.074
0.076
0.91
0.42
0.033
0.28
0.013
0.73
0.77
0.024
0.75
0.98
0.96
1.01 (0.99-1.04)
1.01 (0.99-1.04)
0.95 (0.93-0.98) †
1.02 (1.00-1.05)
1.01 (0.98-1.03)
No data
1.01 (0.98-1.03)
0.97 (0.95-1.00)
1.00 (0.97-1.02)
1.01 (0.97-1.06)
1.02 (0.97-1.07)
1.00 (0.97-1.02)
1.00 (0.97-1.02)
1.01 (0.99-1.03)
1.01 (0.99-1.03)
0.98 (0.94-1.01) †
0.91 (0.86-0.96) †
1.03 (1.00-1.07) †
0.91 (0.89-0.94)
0.98 (0.96-1.01)
0.18
0.18
3.3e-5
0.030
0.55
0.61
0.024
0.69
0.62
0.41
0.66
0.67
0.47
0.38
0.24
4.2e-4
0.058
5.6e-10
0.17
(rs12704290)
0.011
444
6.8e-6
0.016
713
443
1.1e-18
0.0012
560
4586
0.00017
0.0092
0.0061
387
445
238
0.0103
4.2e-4
497
207
1.7e-18
4.1e-6
294
314
We obtained the SZGene 164 database (http://www.szgene.org) in 11/2009 (active work on SZGene stopped in 12/2011). We selected all study for the 25
genes in Table 1 that met inclusion criteria: (a) case/control design, (b) published before 2009 (we verified that no GWAS were included), (c) subjects of
European ancestry, (d) biallelic markers and complete data. We selected markers that had been in ≥5 studies. If a gene did not have a marker with ≥5
studies, we selected the marker with the largest number of studies so that all 25 genes had at least one genetic marker. We conducted a fixed-effect metaanalysis for each marker. The first six columns show characteristics derived from SZGene. Gene=standard gene name. Marker=name of genetic variant
(converted to rs IDs where possible). Studies=SZGene studies that met inclusion criteria. Ncase=sum of cases. SZG OR=odds ratio (OR) and 95% confidence
interval (CI) from the SZGene meta-analysis. SZG P=meta-analysis P-value. The next four columns show results from the PGC schizophrenia megaanalysis. 165 PGC OR=OR and 95% CI. PGC P= P-value. PGC Pmin=minimum P-value ±25 kb of a gene. PGC SNPs=SNPs ±25 kb of the gene. Alleles were
harmonized between SZGene and PGC so that the ORs are comparable. The PGC and SZGene results are mostly for the same SNP but, for markers that
were not SNPs (†), all PGC SNP results within a range encompassing the variant were obtained, and the one with smallest P-value is shown (BDNF
270C/T, SLC6A3 VNTR, SLC6A4 5-HTTLPR, SLC6A4 5-HTTVNTR, APOE rs429358/rs7412, and KCNN3 1333T/C).
13
Table S2. Independent meta-analyses for historical genes published 2008-2014
Gene
PMID
rSID
Year
Lead Author
OR
P
Note
AKT1
19931325
rs3730358
2010
Lee
1.02 (0.94-1.12)
NS
Pooled across multiple ancestries
AKT1
23747160
rs3730358
2013
Loh
1.004(0.926–1.090)
0.92
Pooled across multiple ancestries
AKT1
18715757
rs3730358
2008
Shi
0.951 (0.854–1.060)
0.36
EUR
BDNF
23438165
270C/T
2013
Watanabe
1.20 (0.93–1.55)
0.16
EUR
COMT
19329282
rs737865
2009
Okochi
1.041 (0.978-1.108)
0.21
Pooled across multiple ancestries
COMT
19329282
rs165599
1.032 (0.960-1.108)
0.40
Pooled across multiple ancestries
COMT
19329282
rs4680
0.989 (0.942-1.039)
0.67
Pooled across multiple ancestries
COMT
20488458
rs4680
2011
Costas
0.947 (0.904-0.993)
0.023
Only over-dominant model reported
DAOA
21443574
rs2391191
2011
Muller
CI includes 1.0
NS
Only forest plot presented
DAOA
24447945
rs2391191
2014
Tan
0.96 (0.89-1.04)
0.30
EUR, random effects model
DAOA
24447945
rs947267
0.97 (0.87-1.09)
0.63
EUR, fixed effects model
DAOA
24447945
rs3918342
1.03 (0.92-1.15)
0.64
EUR, random effects model
DISC1
21483435
rs6675281
0.93
EUR
DISC1
21483435
rs3738401
0.11
EUR
DISC1
21483435
rs821616
0.51
EUR
DRD2
22370928
rs1801028
2012
Liu
1.47 (1.18-1.83)
0.0006
Pooled across multiple ancestries
DRD3
18295456
rs6280
2008
Ma
1.017 (0.943-1.097)
0.67
EUR
HTR2A
23857788
rs6313
2012
Jianliang
1.06 (0.96-1.17)
NS
Chinese
HTR2A
24962835
rs6313
2014
Tan
0.994 (0.903-1.093)
0.89
EUR, random effects model
HTR2A
23404241
rs6313
2013
Gu
1.12 (1.05-1.20)
0.28
EUR
MTHFR
21185933
rs1801131
2011
Peerbooms
1.06 (0.99-1.15)
NS
Pooled across multiple ancestries
MTHFR
24535549
rs1801133
2014
Nishi
1.16 (1.03-1.31)
0.014
Japanese, fixed effects model
MTHFR
24938371
rs1801133
2014
Hu
1.07 (0.98–1.16)
0.14
EUR, random effects model
MTHFR
24938371
rs1801131
1.07 (0.96–1.19)
0.22
EUR, random effects model
NRG1
19362450
rs6994992
1.03(0.97–1.10)
0.33
Pooled across multiple ancestries
NRG1
19362450
rs35753505
1.02(0.92–1.11)
0.76
Pooled across multiple ancestries
NRG1
19362450
rs62510682
1.06(0.98–1.15)
0.12
Pooled across multiple ancestries
2012
2009
Mathieson
Gong
ZDHHC8
20661937
rs175174
2010
Xu
0.97 (0.88-1.07)
0.55
EUR, fixed effects model
We searched PubMed for: ("schizophrenia"[MeSH Terms] OR "schizophrenia"[All Fields]) AND ("meta-analysis"[Publication Type] OR "meta-analysis as topic"[MeSH
Terms] OR "meta-analysis"[All Fields]). All studies 2008-2014 were obtained. NS=not significant at 0.05.
14
Table S3. Candidate gene studies of DAO
Paper
Bass et al, 2009
Ancestry
3 of 4 grandparents were of
English, Irish, Welsh, or
Scottish descent
Canadian, Russian
Phenotype
schizophrenia
Sample size
431 cases
442 controls
Variant
rs3741775, rs3918346,
rs2111902
P-value (OR)
n.s.
schizophrenia
Korean
homicidal behaviors in
schizophrenia
Corvin et al, 2007
Irish origin, i.e., Irish parents
/ grandparents
schizophrenia
373 cases, 812 controls
Fallin et al, 2005
Ashkenazi
274 trios
Gaysina et al, 2010
UK, Canada
schizophrenia,
schizoaffective
disorder
bipolar I disorder
bipolar disorder
rs3741775, rs3918346,
rs2111902, MDAAO-1 (SNP),
MDAAO-2 (SNP), MDAAO-3
(SNP), rs3918347, rs888531
rs2070586, rs2070587,
rs2070588, rs2111902,
rs3918346, rs3741775,
rs3825251
rs3741775, rs888531
rs3918346
rs2111902
rs2070587, rs2070588,
rs3741775, rs3825251,
rs3918346
5 SNPs typed
rs3741775
four SNP markers were associated in Canadian
samples
Chung et al, 2007
213 (Canadian cases)
241 (Canadian controls)
183 (Russian cases)
183 (Russian controls)
188 cases
Jönsson et al, 2009
Danish, Norwegian, Swedish
schizophrenia
rs2070587, rs2070588,
rs3741775, rs3825251
n.s.
Kim et al, 2010
Korean
schizophrenia
rs2070588, rs3741775,
rs3825251, rs3918346,
rs2111902, rs3220845
rs2070586
n.s.
Chumakov et al,
2002
337 trios
515 cases (UK)
1316 controls (UK)
385 cases (Canada)
312 controls (Canada)
420 cases (Denmark)
1004 controls (Denmark)
162 cases (Norway)
177 controls (Norway)
255 cases (Sweden)
292 controls (Sweden)
96 cases
96 controls
rs2070587
Liu et al, 2004
Chinese
schizophrenia
547 cases
536 controls
Liu et al, 2006
Taiwanese
schizophrenia
218 families
Ohnuma et al, 2009
Japanese
schizophrenia
340 cases
340 controls
Ohnuma et al, 2010
Japanese population
schizophrenia
Papagni et al, 2011
native English speakers
regional activation and
functional connectivity
1656 cases
1842 controls
121 subjects
rs3918347
rs2070587, rs2070588,
rs3825251, rs3918346,
rs2111902
rs3741775
rs2111902, rs3918346,
rs3741775
rs4964762, rs11114083,
rs2070586, rs2111902,
rs3918346, rs3741775,
rs11114086, rs11114087,
rs3825251, rs3918347,
rs4964770
rs3825251, rs3918347,
rs4964770
rs3918346
15
n.s.
n.s.
0.0034 (OR 1.43)
0.018 (OR 1.29)
n.s.
highly suggestive (any SNP or haplotype p <0.01)
n.s.
0.005 (allelic, females)
0.014 (genotypic, females)
0.025 (allelic, males)
0.002 (allelic, females)
0.005 (genotypic, females)
0.018 (allelic females)
n.s.
0.000001 (OR 1.59)
n.s.
n.s.
n.s.
Cases with one or two copies of the T allele showed
lower deactivation in the left precuneus and greater
Prata et al, 2008
Roussos et al, 2011
Caucasian with at least 3
grandparents born in
Scotland
Greek
in schizophrenia,
bipolar I, controls
bipolar affective
disorder
activation in the right posterior cingulate gyrus
213 cases, 197 controls
rs2111902, rs3918346,
rs3741775, rs3918347
n.s.
sensorimotor gating,
working memory,
personality in healthy
males
530 individuals
rs2111902, rs3918346,
rs3741775, rs3825251
rs4623951
n.s.
Sacchetti et al, 2013
Italian
schizophrenia
391 patients,488 controls
Schumacher et al,
2004
German
bipolar affective
disorder
schizophrenia
300 cases, 300 controls
schizophrenia
Stefanis et al, 2007
Caucasian, Indian, Native
American, Asian, African,
and mixed ethnicity
Greek
113 nuclear families
168 cases
168 controls
2243 young male military
conscripts
Suliman et al, 2010
German
schizophrenia
Shinkai et al, 2007
schizotypy and
cognitive
endophenotypes
299 cases, 300 controls
531 cases
755 controls
Vilella et al, 2007
Spanish
schizophrenia
589 cases
617 controls
Wirgeres et al, 2008
Norwegian
155 patients
Wood et al, 2007
self-reported whites from
various US regions
PANSS in
schizophrenia
schizophrenia
345 schizophrenia cases
150 schizoaffective cases
352 controls
rs2070586, rs2070587,
rs2111902, rs10861974,
rs3741775, rs3918347
MDAAO4, MDAAO5, MDAAO6
MDAAO4
MDAAO5
MDAAO6
rs3741775, rs3918346,
rs2111902, rs3918347
n.s.
0.026 (allelic)
0.019 (allelic),
0.041 (genotypic)
0.021 (allelic)
n.s.
rs2111902
rs3918346
rs3741775
n.s.
0.033 (d´-S2B working memory)
0.048 (perceptual aberration scale CAPE, depression)
rs987044, rs3916982,
rs7136681, rs73410945,
rs7980427, rs11114086,
rs3825251, STR-1-dao2, STR2-dao3, SNP-3-exon11,
rs10746135, rs7313861,
rs3918305
rs11114071
rs4964766
rs2070586
rs73193464
rs2111902
rs3918346
rs3741775
rs4623951, rs11114086,
rs2070587, hCV2880411,
rs4964281
rs2070588, rs3741775
rs2070587
rs3741775, rs17041020,
rs4623951
rs2070586
n.s.
rs2111902
16
0.015 (adjusted p-value, PPI 85_30)
0.004 (adjusted p-value, PPI 85_60)
0.004 (adjusted p-value, pooled PPI 60 ms)
n.s.
0.04
0.049
0.015
0.017
0.021
0.039
0.046
n.s.
n.s.
0.036 (PANSS total score)
n.s.
0.02159 (SCZ vs CON, allelic)
0.01219 (SCZ+SCA vs CON, allelic)
0.03973 (SCZ+SCA vs CON, genotypic)
0.00219 (SCZ vs CON, allelic)
0.00667 (SCZ vs CON, genotypic)
0.00056 (SCZ+SCA vs CON, allelic)
0.00192 (SCZ+SCA vs CON, genotypic)
rs3918346
rs3825251
Yamada et al, 2005
Central Japan
schizophrenia
50 cases
52 controls
IVS1a+465G>C
IVS1b+15G>A
570 cases
570 controls
(sample set B)
IVS1a+465G>C,
IVS1b+15G>A,
DAO-01, DAO-02, DAO-03,
DAO-04, DAO-05
DAO-06
124 families
Yang et al, 2013
Taiwan Han Chinese
Table S2 Citations
ratio of D-Serine to
total Serine
schizophrenia
Sustained attention
was assessed using
the unmasked and
masked CPTs (Zd´
and Zmd´)
Four CPT strata
50 cases
52 controls
912 cases, 600 controls
IVS1a+465G>C,
IVS1b+15G>A, DAO-01, DAO02, DAO-03, DAO-04, DAO05, DAO-06
DAO-01, DAO-02, DAO-03,
DAO-04, DAO-05, DAO-06
rs2070585, rs2070587
rs11114083
rs2070586
rs55944529
0.00068 (SCZ vs CON, allelic)
0.00297 (SCZ vs CON, genotypic)
0.00004 (SCZ + SCA vs CON, allelic)
0.00028 (SCZ + SCA vs CON, genotypic)
0.01800 (SCZ vs CON, allelic)
0.04340 (SCZ vs CON, genotypic)
0.00246 (SCZ+SCA vs CON, allelic)
0.00647(SCZ + SCA vs CON, genotypic)
0.038 (genotypic)
0.041 (allelic)
0.026 (genotypic)
n.s.
0.020 (case-control, allelic)
0.023 (case-control, genotypic)
n.s.
n.s.
n.s.
0.0357 (raw p-value, allele-based, Zmd´ >= -2.5)
0.0293 (raw p-value, trend-based, Zmd´>= -2.5)
0.0491 (raw p-value, genotype-based, Zd´>= -2.5)
0.0275 (raw p-value, allele-based, Zmd´ >= -2.5)
0.0252 (raw p-value, trend-based, Zmd´>= -2.5)
0.0459 (raw-p-value, genotype-based, overall)
0.0313 (raw p-value, allele-based, overall)
0.0299 (raw p-value, trend-based, overall)
0.0392 (raw p-value, allele-based, Zd´ < -2.5)
0.0365 (raw p-value, trend-based, Zd´ < -2.5)
Bass NJ, Datta SR, McQuillin A, Puri V, Choudhury K, Thirumalai S, Lawrence J, Quested D, Pimm J, Curtis D, Gurling HM. Evidence for the association of the DAOA (G72) gene with
schizophrenia and bipolar disorder but not for the association of the DAO gene with schizophrenia. Behav Brain Funct. 2009 Jul 8;5:28.
Chumakov I, Blumenfeld M, Guerassimenko O, Cavarec L, Palicio M, Abderrahim H, Bougueleret L, Barry C, Tanaka H, La Rosa P, Puech A, Tahri N, Cohen-Akenine A, Delabrosse S,
Lissarrague S, Picard FP, Maurice K, Essioux L, Millasseau P, Grel P, Debailleul V, Simon AM, Caterina D, Dufaure I, Malekzadeh K, Belova M, Luan JJ, Bouillot M, Sambucy JL, Primas G,
Saumier M, Boubkiri N, Martin-Saumier S, Nasroune M, Peixoto H, Delaye A, Pinchot V, Bastucci M, Guillou S, Chevillon M, Sainz-Fuertes R, Meguenni S, Aurich-Costa J, Cherif D, Gimalac
A, Van Duijn C, Gauvreau D, Ouellette G, Fortier I, Raelson J, Sherbatich T, Riazanskaia N, Rogaev E, Raeymaekers P, Aerssens J, Konings F, Luyten W, Macciardi F, Sham PC, Straub RE,
Weinberger DR, Cohen N, Cohen D. Genetic and physiological data implicating the new human gene G72 and the gene for D-amino acid oxidase in schizophrenia. Proc Natl Acad Sci U S
A. 2002 Oct 15;99(21):13675-80.
Chung S, Jung J, Chung HY, Yoo HK, Kim CY, Joo YH, Choi SE, Hong JP. No association between polymorphisms of DAO and DAOA genes and homicidal behaviors in Korean schizophrenia.
Psychiatr Genet. 2007 Oct;17(5):313.
Corvin A, McGhee KA, Murphy K, Donohoe G, Nangle JM, Schwaiger S, Kenny N, Clarke S, Meagher D, Quinn J, Scully P, Baldwin P, Browne D, Walsh C, Waddington JL, Morris DW, Gill M.
Evidence for association and epistasis at the DAOA/G30 and D-amino acid oxidase loci in an Irish schizophrenia sample. Am J Med Genet B Neuropsychiatr Genet. 2007 Oct
5;144B(7):949-53.
Fallin MD, Lasseter VK, Avramopoulos D, Nicodemus KK, Wolyniec PS, McGrath JA, Steel G, Nestadt G, Liang KY, Huganir RL, Valle D, Pulver AE. Bipolar I disorder and schizophrenia: a 440single-nucleotide polymorphism screen of 64 candidate genes among Ashkenazi Jewish case-parent trios. Am J Hum Genet. 2005 Dec;77(6):918-36.
Gaysina D, Cohen-Woods S, Chow PC, Martucci L, Schosser A, Ball HA, Tozzi F, Perry J, Muglia P, Kennedy JL, King N, Vincent JB, Parikh SV, Strauss J, Craig IW, McGuffin P, Farmer A.
Association analysis of DAOA and DAO in bipolar disorder: results from two independent case-control studies. Bipolar Disord. 2010 Aug;12(5):579-81.
17
Jönsson EG, Saetre P, Vares M, Andreou D, Larsson K, Timm S, Rasmussen HB, Djurovic S, Melle I, Andreassen OA, Agartz I, Werge T, Hall H, Terenius L. DTNBP1, NRG1, DAOA, DAO and
GRM3 polymorphisms and schizophrenia: an association study. Neuropsychobiology. 2009;59(3):142-50.Ohnuma T, Shibata N, Maeshima H, Baba H, Hatano T, Hanzawa R, Arai H.
Association analysis of glycine- and serine-related genes in a Japanese population of patients with schizophrenia. Prog Neuropsychopharm Biol Psychiatry. 2009 Apr 30;33(3):511-8.
Kim B, Kim H, Joo YH, Lim J, Kim CY, Song K. Sex-different association of DAO with schizophrenia in Koreans. Psychiatry Res. 2010 Sep 30;179(2):121-5.
Liu X, He G, Wang X, Chen Q, Qian X, Lin W, Li D, Gu N, Feng G, He L. Association of DAAO with schizophrenia in the Chinese population. Neurosci Lett. 2004 Oct 21;369(3):228-33.
Liu YL, Fann CS, Liu CM, Chang CC, Wu JY, Hung SI, Liu SK, Hsieh MH, Hwang TJ, Chan HY, Chen JJ, Faraone SV, Tsuang MT, Chen WJ, Hwu HG. No association of G72 and D-amino acid
oxidase genes with schizophrenia. Schizophr Res. 2006 Oct;87(1-3):15-20.
Ohnuma T, Shibata N, Baba H, Ohi K, Yasuda Y, Nakamura Y, Okochi T, Naitoh H, Hashimoto R, Iwata N, Ozaki N, Takeda M, Arai H. No association between DAO and schizophrenia in a
Japanese patient population: a multicenter replication study. Schizophr Res. 2010 May;118(1-3):300-2.
Ohnuma T, Shibata N, Maeshima H, Baba H, Hatano T, Hanzawa R, Arai H. Association analysis of glycine- and serine-related genes in a Japanese population of patients with schizophrenia.
Prog Neuropsychopharmacol Biol Psychiatry. 2009 Apr 30;33(3):511-8.
Papagni SA, Mechelli A, Prata DP, Kambeitz J, Fu CH, Picchioni M, Walshe M, Toulopoulou T, Bramon E, Murray RM, Collier DA, Bellomo A, McGuire P. Differential effects of DAAO on regional
activation and functional connectivity in schizophrenia, bipolar disorder and controls. Neuroimage. 2011 Jun 15;56(4):2283-91.
Prata D, Breen G, Osborne S, Munro J, St Clair D, Collier D. Association of DAO and G72(DAOA)/G30 genes with bipolar affective disorder. Am J Med Genet B Neuropsychiatr Genet. 2008
Sep 5;147B(6):914-7.Corvin A, Donohoe G, McGhee K, Murphy K, Kenny N, Schwaiger S, Nangle JM, Morris D, Gill M. D-amino acid oxidase (DAO) genotype and mood symptomatology
in schizophrenia. Neurosci Lett. 2007 Oct 16;426(2):97-100.
Roussos P, Giakoumaki SG, Adamaki E, Georgakopoulos A, Robakis NK, Bitsios P. The association of schizophrenia risk D-amino acid oxidase polymorphisms with sensorimotor gating,
working memory and personality in healthy males. Neuropsychopharmacology. 2011 Jul;36(8):1677-88.
Sacchetti E, Scassellati C, Minelli A, Valsecchi P, Bonvicini C, Pasqualetti P, Galluzzo A, Pioli R, Gennarelli M. Schizophrenia susceptibility and NMDA-receptor mediated signalling: an
association study involving 32 tagSNPs of DAO, DAOA, PPP3CC, and DTNBP1 genes. BMC Med Genet. 2013 Mar 9;14:33.
Schumacher J, Jamra RA, Freudenberg J, Becker T, Ohlraun S, Otte AC, Tullius M, Kovalenko S, Bogaert AV, Maier W, Rietschel M, Propping P, Nöthen MM, Cichon S. Examination of G72 and
D-amino-acid oxidase as genetic risk factors for schizophrenia and bipolar affective disorder. Mol Psychiatry. 2004 Feb;9(2):203-7.
Schumacher J, Abou Jamra R, Becker T, Klopp N, Franke P, Jacob C, Sand P, Fritze J, Ohlraun S, Schulze TG, Rietschel M, Illig T, Propping P, Cichon S, Deckert J, Nöthen MM. Investigation of
the DAOA/G30 locus in panic disorder. Mol Psychiatry. 2005 May;10(5):428-9.
Shinkai T, De Luca V, Hwang R, Müller DJ, Lanktree M, Zai G, Shaikh S, Wong G, Sicard T, Potapova N, Trakalo J, King N, Matsumoto C, Hori H, Wong AH, Ohmori O, Macciardi F, Nakamura J,
Kennedy JL. Association analyses of the DAOA/G30 and D-amino-acid oxidase genes in schizophrenia: further evidence for a role in schizophrenia. Neuromolecular Med.
2007;9(2):169-77.
Stefanis NC, Trikalinos TA, Avramopoulos D, Smyrnis N, Evdokimidis I, Ntzani EE, Ioannidis JP, Stefanis CN. Impact of schizophrenia candidate genes on schizotypy and cognitive
endophenotypes at the population level. Biol Psychiatry. 2007 Oct 1;62(7):784-92.
Suliman H, Schumacher J, Becker T, Cichon S, Schulze TG, Propping P, Rietschel M, Nöthen MM, Jamra RA. Association study of 20 genetic variants at the (D)-amino acid oxidase gene in
schizophrenia. Psychiatr Genet. 2010 Apr;20(2):82-3.
Vilella E, Costas J, Sanjuan J, Guitart M, De Diego Y, Carracedo A, Martorell L, Valero J, Labad A, De Frutos R, Nájera C, Moltó MD, Toirac I, Guillamat R, Brunet A, Vallès V, Pérez L, Leon M, de
Fonseca FR, Phillips C, Torres M. Association of schizophrenia with DTNBP1 but not with DAO, DAOA, NRG1 and RGS4 nor their genetic interaction. J Psychiatr Res. 2008
Mar;42(4):278-88.
Wirgenes KV, Djurovic S, Agartz I, Jonsson EG, Werge T, Melle I, Andreassen OA. Dysbindin and d-amino-acid-oxidase gene polymorphisms associated with positive and negative symptoms
in schizophrenia. Neuropsychobiology.2009;60(1):31-6.
Wood LS, Pickering EH, Dechairo BM. Significant support for DAO as a schizophrenia susceptibility locus: examination of five genes putativelyassociated with schizophrenia. Biol
Psychiatry. 2007 May 15;61(10):1195-9.
Yamada K, Ohnishi T, Hashimoto K, Ohba H, Iwayama-Shigeno Y, Toyoshima M, Okuno A, Takao H, Toyota T, Minabe Y, Nakamura K, Shimizu E, Itokawa M, Mori N, Iyo M, Yoshikawa T.
Identification of multiple serine racemase (SRR) mRNA isoforms and genetic analyses of SRR and DAO in schizophrenia and D-serine levels. Biol Psychiatry. 2005 Jun 15;57(12):1493503.
Yang HC, Liu CM, Liu YL, Chen CW, Chang CC, Fann CS, Chiou JJ, Yang UC, Chen CH, Faraone SV, Tsuang MT, Hwu HG. The DAO gene is associated with schizophrenia and interacts with other
genes in the Taiwan Han Chinese population. PLoS One. 2013;8(3):e60099.
18
Table S4. Candidate gene studies of DAOA
Paper
Addington et al, 2004
Andreou et al, 2012
Bakker et al, 2007
Bass et al, 2009
Ancestry
mixed
Caucasian
patients had at least three
Dutch-born Caucasian
grandparents
three of the grandparents
were of English, Irish,
Welsh, or Scottish descent
Phenotype
childhood-onset schizophrenia
and psychosis NOS
Sample size
98 children and
adolescents
5-HIAA, HVA, and MHPG
concentration in cerebrospinal
fluid in healthy individuals
132 individuals
deficit and non-deficit
schizophrenia
308 cases
580 controls
schizophrenia
431 cases
442 controls
bipolar disorder
303 cases
442 controls
734 cases
442 controls
225 individuals
301 cases
604 controls
139 cases (116
´enriched cases´
(linkage to 13q33),
113 controls
Bousman et al, 2013
Australian
schizophrenia and bipolar
disorder combined
transition to psychosis in
individuals at ultra-high risk for
psychosis
Carrera et al, 2010
Spanish Caucasian origin
schizophrenia
Chen et al, 2004
US birth, self-reported as
´white´
bipolar affective disorder
Chen et al, 2013
Chinese
schizophrenia
454 cases
480 controls
Chen et al, 2012
Chinese
resting-state brain activity in
major depression
488 cases
480 controls
Chiesa et al, 2011
Korean
Chiesa et al, 2012
Korean
Chumakov et al, 2002
Canadian, Russian
schizophrenia, treatment
outcomes
major depressive disorder
(MDD), bipolar disorder (BD)
221 cases
145 cases (MDD)
132 cases (BD)
170 controls
213 (Canadian cases)
241 (Canadian
controls)
183 (Russian cases)
183 (Russian controls)
19
Variant
rs746187, rs3916964, rs778294, rs778293,
rs1935062, rs1935058
rs2391191
rs3916967
rs2391191, rs778294
P-value (OR)
n.s.
rs3918342
0.03 (5-HIAA)
0.001 (HVA)
0.004 (HVA)
n.s.
rs1421292
rs3918342, rs2391191, rs3916967, rs1421292
rs3916965, rs3916967, rs2391191, rs947267,
rs778294, rs778293, rs1421292
rs3918342
rs2391191, rs947267, rs778294, rs1421292,
rs1341402, rs1935062, rs954581
rs3918342
rs3918342
rs1421292
rs7320588, rs6416389, rs1935057, rs1341402,
rs2391191, rs1539070, rs1935062, rs6491961,
rs7986339, rs2153674, rs7989006, rs7984976,
rs947267, rs778294, rs778293, rs3918342
rs9586843, rs7329966, rs7324448, rs1575633,
rs1570709
rs1935058, rs2391191, rs954581
rs1935062
rs947267
rs3916965, rs3916966, rs3916967, rs2391191,
rs3918341, rs9558562, rs947267, rs778294,
rs3918342, rs1421292
rs3916965, rs3916966, rs3916967, rs9558562,
rs947267, rs3918342, rs1421292
rs2391191
rs3918341
rs778294
rs3916966, rs3916967, rs2391191, rs3916968,
rs778293
rs7139958
rs9558571
rs3916966, rs3916967, rs2391191, rs3916968,
rs7139958, rs9558571, rs778293
DAOA_M06 (SNP), rs746187 (M07),
DAOA_M08 (SNP), DAOA_M09 (SNP),
DAOA_M10 (SNP), rs3916964 (M11),
rs3916966 (M13), rs3918341 (M16),
DAOA_M17 (SNP), rs947267 (M18), rs778294
(M19), rs3916971 (M21), rs3916970 (M20)
rs3918342 (M23)
0.031
0.015
n.s.
n.s.
0.016
n.s.
0.038
0.008
0.044
n.s.
n.s.
n.s.
0.0072 (OR 1.67)
0.0048 (enriched, OR 1.77)
0.0403 (all cases, OR 0.69)
0.0295 (enriched cases, OR 0.66)
n.s.
n.s.
0.01 (allelic OR 1.27)
0.04 (OR 1.09)
0.02 (1.37)
n.s.
0.005 PANSS positive at baseline)
0.008 (allelic, PANSS at baseline)
n.s.
n.s.
0.019 (allelic, Canadian)
rs2391191 (M15)
rs778293 (M22)
rs3916967 (M14)
rs3916965 (M12)
rs1421292 (M24)
Chung et al, 2007
Korean
Corvin et al, 2007
Irish
Corvin et al, 2007
Irish origin, i.e., Irish
parents / grandparents
Dalvie et al, 2010
mixed ancestry,
Caucasian
Detera-Wadleigh et al,
2006
various ethnicities
homicidal behaviors in
schizophrenia
depression/anxiety factor in
schizophrenia
schizophrenia
188 cases
bipolar 1: age at onset, #
depressive episodes, #
hospitalizations for depression
schizophrenia and bipolar
disorder
191 cases
schizophrenia
bipolar disorder
196 schizophrenia, 59
schizoaffective
373 cases, 812
controls
meta-analysis
meta-analysis
meta-analysis
Di Maria et al, 2009
Italian descent
psychosis
185 Alzheimer´s
disease
Donohoe et al, 2007
Irish
episodic memory
93 cases
Fallin et al, 2005
Ashkenazi
schizophrenia / schizoaffective
disorder
274 trios
rs3918341, rs9301030, rs3916968, rs947267,
rs701567, rs778294
rs3918346
0.01
rs778293, rs3918342, rs1421292
rs2391191
rs3916965
rs701567
n.s.
0.01 (corrected p-value, OR 1.31)
0.005 (corrected p-value, OR 1.34)
n.s.
rs746187, rs3916964, rs1341402, rs3916967,
rs3918341, rs947267, rs778294, rs954581,
rs3916970, rs3916971
rs3916965
rs3916966
rs1935058
rs2391191
rs1935062
rs778293
rs3918342
rs1421292
rs746187, rs3916964, rs3916966, rs1935058,
rs1341402, rs3916967, rs3918341, rs1935062,
rs947267, rs778294, rs954581, rs3916970,
rs3916971
rs3916965
rs2391191
rs778293
rs3918342
rs1421292
rs746187, rs3916964, rs3916965, rs3916966,
rs1935058, rs1341402, rs3916967, rs2391191,
rs3918341, rs947267, rs778294, rs954581,
rs3916970, rs3916971, rs778293, rs1421292
rs1935062
rs3918342
rs1341402, rs1341403, rs6491961, rs9558571,
rs778294, rs12864685
rs2153674
rs2391191
n.s.
rs2391191, rs947267, rs778294, rs3916970,
rs3916968, rs1935062, rs954580, rs2893229,
rs1341406, rs1341402, rs701567,
DAOA_GCintronic, DAOA_3'UTR873,
20
0.017 (allelic, Russian)
0.032 (allelic, Canadian)
0.003 (allelic, Canadian)
0.007 (genotypic, Canadian)
0.038 (allelic, Canadian)
0.007 (allelic, Canadian)
0.020 (genotypic, Canadian)
0.019 (allelic, Canadian)
0.044 (genotypic, Canadian)
0.006 (genotypic, Russian)
n.s.
0.0001
0.0413
0.0015
0.0000
0.0143
0.0003
0.0000
0.0010
n.s.
0.0002
0.0006
0.0033
0.0006
0.0092
n.s.
0.0019
0.0309
n.s.
0.033
0.028 (immediate recall)
0.015 (delayed recall)
gene is suggestive (any SNP or
haplotype minimal p value
0.01<p<0.05)
DAOA_3'UTR1746
Gawlik et al, 2010
Gaysina et al, 2010
Goldberg et al, 2006
Gomez et al, 2008
Grigoroiu-Serbanescu
et al, 2010
Hall et al, 2004
Hall et al, 2008
Hattori et al, 2003
bipolar I disorder
337 trios
14 SNPs
German Caucasian
descent
UK, Canada
affective psychosis
Americans of European
extraction (Caucasian
according to self report)
schizophrenia, cognitive
variables
248 cases
188 controls
515 cases (UK)
1316 controls (UK)
385 cases (Canada)
312 controls (Canada)
up to 106 trios
>600 subjects
(cognitive function)
14 normal individuals
(fMRI)
30 normal individuals
(fMRI)
rs3916966, rs1935058, rs2391191, rs1935062,
rs947267, rs3918342, rs9558575
rs3916965, rs12584489, rs2391191,
rs1935062, rs947267, rs778292, rs3918342,
rs1421292
rs9558562
M01, M02, M03, rs2391191 (M04), rs1935058
(M05), rs1935062 (M06), rs778294 (M07), M08,
M09, M10, M11
M10
646 nuclear families
rs1341402, rs12874006, rs1539070,
rs9284226, rs2153674, rs9558567, rs947267,
rs3918342
rs2391191
rs3918341
rs1935062
rs3916965, rs1935057, rs1341402, rs3916967,
rs2391191, rs1935062, rs67705083, rs778294,
rs954581, rs3916971, rs778293, G72_z6:1117,
rs3918342, rs1421292
rs778293
rs3916965
rs1935057
rs3916967
rs2391191
M7, M12, M14, M23, M25
M15
rs3918342, rs1421292
Hungarian
Romanian
South African Afrikaner,
USA
Scottish
bipolar disorder
juvenile onset mood disorders
bipolar I global diagnosis,
psychotic bipolar I subtype
198 cases
180 controls
bipolar I with delusions
bipolar I in non-psychotic
123 cases
38 cases
schizophrenia
169 South African trios
210 USA trios
61 individuals
hippocampal function in
subjects at high risk of
schizophrenia
bipolar disorder
22 multiplex families
21
rs1998654, rs2181953, rs978714, rs1359387,
M-13, rs2391191, rs947267, rs954581,
rs778334, rs2012887, M-23
rs1815686
rs1935058
rs1341402
rs1935062
rs778294
suggestive (any SNP or haplotype
minimal p value 0.01<p<0.05)
n.s.
n.s.
0.05 (UK sample)
n.s.
0.03 (genotypic, continuous
performance test)
0.04 (genotypic, n-back [one back]
working memory test)
0.05 (genotypic, verbal paired
associate learning)
fMRI: strong effect on BOLD
activation in hippocampus during
episodic memory paradigm
n.s.
0.025
0.029
0.026
n.s.
0.042 (dominant model)
0.044 (OR 1.82)
0.037 (OR 1.88)
0.043 (OR 1.82)
0.043 (OR 1.82)
n.s.
0.045 (S African trios; n.s. US trios
differences in the activation of the
left hippocampus and parahippocampus in the contrast of
sentence completion versus rest,
and in recruitment of right inferior
prefrontal cortex in relation to
increasing task difficulty
n.s.
0.041
0.00077
0.0075
0.0078
0.018
Hong et al, 2006
Hukic et al, 2013
Chinese
Swedish
schizophrenia
cognitive manic symptoms
(CMS) in bipolar I disorder
152 families (NIMH
pedigrees)
216 families
488 cases
1044 controls
rs1935058, rs1341402, rs2391191, rs1935062,
rs947267, rs778294, rs954581
rs947267
rs3916967
rs2391191
Jansen et al, 2009
European descent
cognitive functions
423 healthy individuals
rs1935062
rs3918342
rs1421292
Jansen et al, 2010
Jönsson et al, 2009
European descent
Danish, Norwegian,
Swedish
memory encoding, retrieval
schizophrenia
Korostishevsky et al,
2004
Ashkenazi
schizophrenia
Korostishevsky et al,
2006
Kotaka et al, 2009
Palestinian Arab
Japanese
Krug et al, 2011
western- or middle
European descent
Li et al, 2007
various ethnicities
schizophrenia
methamphetamine induced
psychosis
brain activation in right middle
temporal gyrus during verbal
fluency task
schizophrenia (meta-analysis)
83 healthy subjects
420 cases (DK), 1004
controls (DK), 162
cases (Norway), 177
controls (Norway), 255
cases (Sweden), 292
controls (Sweden)
60 cases
130 controls
56 families from
northern Israel
136 families from
central Israel
31 families from
southern Israel
209 cases
291 controls
rs3918342, rs1421292
rs2391191, rs778294, rs3918342,
M9924634108M24
rs3916965, rs3916967, rs2391191, rs3918341,
rs947267, rs778294, rs3916970
rs3918342
rs778293
rs3916971
rs3916966
rs3916965, rs3916966, rs3916967, rs2391191,
rs3918341, rs947267, rs778294 , rs3916971,
rs778293, rs3918342
rs3916970
rs3916965, rs2391191, rs947267, rs3918342,
rs1421292
rs778293
96 healthy individuals
rs3918342, rs1421292
2138 cases, 2228
controls, 463 parentoffspring trios
rs3916966, rs3916967, rs3918341, rs947267,
rs778294, rs3916970, rs3916971, rs778293,
rs1935062
rs3916965
rs2391191
rs3918342
rs1421292
rs3916965, rs3916966, rs3916967, rs2391191,
rs947267, rs778294, rs3916970, rs3916971,
rs778293, rs3918342, rs3916968
rs3916965, rs3916967, rs2391191
rs778294
rs3918342
rs778293
Liu et al, 2006
Taiwanese
schizophrenia
218 families
Ma et al, 2006
Han Chinese, Scottish
schizophrenia
588 (Han Chinese
cases) 588 (Han
Chinese controls)
183 (Scottish cases)
182 (Scottish controls)
22
n.s.
0.016
0.018 (CMS vs. Non-CMS)
0.026 (CMS vs controls)
0.039 (CMS vs. Non-CMS)
0.019 (CMS vs controls)
n.s.
0.002 (letter-number-span test)
0.001 (d2-test for attention)
0.001 (letter-number-span test)
0.001 (d2-test for attention)
n.s.
n.s.
n.s.
0.001 (genotypic)
0.001 (allelic)
0.005 (genotypic)
0.04 (genotypic)
0.04 (genotypic)
n.s.
0.018 (haplotype relative risk,
northern Israel)
0.014 (transmission disequilibrium
test, northern Israel)
n.s.
0.0015 (allelic)
0.00016 (genotypic)
rs1421292 positively correlated with
brain activation in right middle
temporal gyrus and right procuneus
n.s.
0.0225 (OR 1.12)
0.0086 (OR 1.15)
0.0136 (OR 0.88)
0.0010 (OR 0.8)
n.s.
n.s.
0.025 (OR 0.69, Scottish sample)
0.0005 (OR 0.60, Scottish)
0.022 (OR 0.71, Scottish)
0.0013 (OR 0.76, Han Chinese)
Ma et al, 2009
Chinese
Maheshwari et al,
2009
Caucasian
126 family trios
bipolar disorder
19 families with 146
samples from
Statistical Explanation
for Positional Cloning
(STEPC) analysis
376 bipolar parentproband trios
555 cases, 564
controls
Mössner et al, 2010
Mulle et al, 2005
Müller et al, 2011
German
samples from NIMH
Genetics Initiative and the
Western Psychiatric
Institute and Clinic at the
University of Pittsburgh
Canadian
progression of prodromal
syndromes to first episode
psychosis
82 probands
schizophrenia
159 trios
rs778294
rs3918342
rs778293
rs1935058
rs1341402
rs9301030
rs1815686
rs12862108
rs1935062
rs778294
rs778326
rs978714, rs2025522, rs3916964, rs9558551,
rs7981258, rs9301029, rs1253464, rs3916965,
rs9519671, rs3916966, rs1935057, rs1935058,
ss107796322, rs1341402, rs2391191,
ss107796323, ss107796324, rs12862108,
rs9301030, rs1935062, rs9519683,
ss104807115, rs7986339, rs947267, rs701567,
rs778294, rs7987770, ss107796325,
rs9558581, rs778326, rs954581, rs9301034,
rs778336, rs778334, rs778293, rs2012887,
rs3918342, rs1421292
variants identified by sequencing DAOA exons:
ss104807091, ss104807092, ss104807093,
ss104807094, ss104807095, ss104807096,
ss104807097, ss104807098, ss104807099,
ss104807100, ss104807101, ss104807102,
ss104807103, rs2391191, ss104807104,
ss104807105, ss104807106, ss104807107,
rs9558562, ss104807108, ss104807109,
ss104807110, ss104807111, ss104807112,
ss104807113, rs9519683, ss104807114,
ss104807115, ss104807116, ss104807117,
ss104807118, rs9582999, ss104807119,
ss104807120, rs9670704, ss104807121,
ss104807122, ss104807123, rs778294,
ss104807124, ss104807125, ss104807126,
ss104807127, ss104807128, ss104807129,
ss104807130, ss104807131, rs9558573,
ss104807132, ss104807133, ss104807134,
ss104807135, ss104807136
rs1935058, rs2391191, rs1935062, rs947267,
rs778293, rs3918342, rs1421292
rs1341402
rs778294
M-1, M-2, M-3, M-5, M-11, M-12, M-13, M-14,
M-15, M-16, M-21, M-22, M-23, M-24
M-4
n.s.
0.02 (all)
(all), 0.041 (male)
0.0009
0.0083
0.031
0.039
0.020
0.0074
0.017
0.026
n.s.
No significant excess of rare
variants in cases compared to
controls
n.s.
0.002
0.030
n.s.
0.04
bipolar disorder
303 core families
rs1341402, rs2391191, rs947267, rs778294
rs1935062
n.s.
0.02 (total sample)
bipolar disorder with psychotic
symptoms
157 families
rs1935062
0.01 (European sample)
n.s.
23
Ohi et al, 2009
Pae et al, 2010
Prata et al, 2008
Prata et al, 2012
Rethelyi et al, 2009
Rietschel et al, 2008
Japanese
Korean
bipolar disorder
meta-analysis
schizophrenia
meta-analysis
schizophrenia
antipsychotic response after
switch to aripiprazole
1774 (cases)
2092 (controls)
86 patients
rs1341402, rs2391191, rs1935062, rs947267,
rs778294
rs1341402, rs2391191, rs1935062, rs947267,
rs778294
rs2391191
rs3916965, rs3916967, rs2391191, rs778294,
rs3916970
rs778293
rs947267
rs3918342
rs746187, rs3916966, rs2391191, rs3916972
brain function during verbal
fluency
50 healthy volunteers
rs746187
Hungarian Caucasian
descent
German
schizophrenia
280 cases,
230 controls
500 major depression
patients, 1030 controls
rs2391191, rs3918342
schizophrenia
391 patients,
488 controls
Sanders et al, 2008
European ancestry
schizophrenia
1870 cases
2002 controls
Schulz et al, 2011
Caucasian
Schulze et al, 2005
German, Polish
reduced cortical thickness in
schizophrenia
bipolar disorder
52 patients,
42 controls
300 patients
(German), 300
controls (German),
294 patients (Polish),
24
0.019 (allelic)
0.005 (Schedule for the
Assessment of Negative Symptoms
[SANS] score)
0.02 (Brief Psychiatric Rating Scale
[BPRS] score)
n.s.
n.s.
213 bipolar I cases,
197 controls
Italian
0.03 (fixed effects model, East
Asians)
n.s.
0.034 (genotypic)
0.030 (allelic)
0.037 (allelic)
0.02 (Brief Psychiatric Rating Scale
[BPRS] score)
bipolar disorder
Saccetti et al, 2013
n.s.
rs3918342
rs1421292
rs2391191
Caucasian with at least 3
of 4 grandparents born in
Scotland
English native speakers
major depression, neuroticism
n.s.
Significant effect on activation in the
left postcentral and supramarginal
gyri (FWE p<0.05)
n.s.
rs3916965, rs1935058, rs1341402, rs3916967,
rs2391191, rs1935062, DAO_3UTR_SNP12,
rs3916971, rs3918342, rs1421292
rs778293
G72_z6:1117
n.s.
rs1341402, rs1341403, rs6491961, rs9558571,
rs778294
rs12864685
rs778294, rs3918342, rs2391191, rs778293,
rs3916968, rs3916967, rs3916966, rs3916965,
rs1421292, rs701567, rs778334, rs17713906,
rs10492528, rs12864685, rs12874006,
rs1935060, rs2039934, rs7139958, rs8002132,
rs9284226, rs9301030, rs9555175, rs9558573,
rs9670704, rs1361562, G72GD486 (SNP),
G72RS425 (SNP)(SNP rs identifiers from SZ
gene database)
rs1539070
rs1557072
rs2391191
n.s.
rs1341402, rs2391191
rs3918342
rs3918342
rs3918342
0.02 (major depression)
0.04 (major depression)
0.05 (recessive model)
n.s.
0.05 (OR 1.13)
0.02 (OR 1.84)
associated with distinct cortical
thinning
n.s.
<0.02 (bipolar disorder)
0.005 (bipolar with psychosis)
<0.0004 (bipolar with delusions)
Schumacher et al,
2004
Caucasian
schizophrenia
bipolar disorder
Schumacher et al,
2005
German descent
panic disorder
311 controls (Polish)
299 cases
300 controls
300 cases
300 controls
152 cases, 208
controls
Seifuddin et al, 2012
various ethnicities
bipolar disorder
meta-analysis
Shi et al, 2009
Han Chinese
schizophrenia
561 families
Shi et al, 2008
various ethnicities
schizophrenia
4304 cases, 5423
controls, 1384 families
(Meta-analysis)
rs1421292
rs1341402, rs1935062, rs778294
M23
rs2391191
M12
M24
M12, rs1341402, rs2391191, rs1935062,
rs778294, M24
M23
rs2391191, rs3916972
rs421292
rs778293
rs3918342
rs1421292, rs2391191, rs778294
rs3918342
rs947267
rs778293
rs3916965, rs3916966, rs1935058, rs1341402,
rs3916967, rs2391191, rs3918341, rs1935062,
rs778294, rs3916970, rs3916971, rs778293,
rs3918342, rs1421292
rs947267
rs778293
rs1421292
bipolar disorder
Shin et al, 2007
Korean
schizophrenia
Shinkai et al, 2007
Canadian population,
various ethnicities
schizophrenia
Soronen et al, 2008
Finnish
visuospatial ability in bipolar
disorder
1145 cases, 1829
controls, 174 families
388 patients, 367
controls
113 nuclear families,
168 cases, 168
controls
180 families
rs2391191, rs778294, rs1341402, rs1935058,
rs1935062
rs947267
rs778294
rs3916965, rs3916966, rs3916967, rs2391191,
rs3918341, rs3916970
rs3916971
rs947267, rs2391191, rs778293, rs3916967 ,
rs3916965, rs1421292, rs3916972
rs746187
rs3918342
rs3916966
rs2391191
25
<0.007 (bipolar delusions)
n.s.
0.033 (allelic), (OR 1.28)
0.037 (allelic), (OR 1.28)
0.048 (allelic), (OR 1.25)
0.036 (allelic), (OR 1.27)
n.s.
0.013 (allelic, OR 1.33)
0.025 (genotypic, OR 1.68)
n.s.
0.024 (OR 2.12, dominant)
0.031 (OR 1.39, allelic model)
0.049 (OR 1.35, allelic model)
0.047 (OR 1.35, allelic model)
n.s.
0.05 (OR 1.14)
n.s.
0.0012
n.s.
0.03 (OR 0.87, all), 5.62E-6 (OR
0.71, Asians)
0.0002 (OR 0.81, Asians)
0.027 (OR 1.11, all ethnicities)
0.031 (OR 1.12, Europeans)
n.s.
0.008
0.009
n.s.
0.03
n.s.
0.013 (OR 0.67, case-control)
0.029 (transmission disequilibrium
test)
0.0015 (General ability)
0.0010 (Abstraction)
0.0013 (Psychomotor speed)
4.00E-06 (Visuospatial ability)
0.0091 (Auditory attention)
0.0298 (Verbal working memory)
0.0047 (Immediate visual memory)
0.0005 (Delayed visual memory)
0.0032 (Free short delay recall)
0.0443 (Free long delay recall)
0.0173 (Recognition memory)
0.0162 (Stroop Interference score)
0.0195 (Semantic fluency)
0.0002 (General ability)
0.0003 (Abstraction)
Soronen et al, 2011
Finnish
Mood disorder (MOOD),
bipolar disorder, major
depressive disorder (MDD),
anxiety, alcoholism, psychotic
symptoms, suicide attempts,
more time ill
178 bipolar I and II
patients, 272 major
depressive disorder
patients, 1322 controls
rs2391191
rs778336
Stefanis et al, 2007
Greek
2243 male conscripts
rs2391191, rs778293, rs3918342
Tan et al, 2014
Caucasian, Asian
n.s.
Spanish
white persons from the
United Kingdom
rs746187, rs3916967, rs2391191, rs947267,
rs778293, rs1421292, rs2181953
rs3916965, rs2391191, rs778294, rs778293,
rs1421292, rs1935058, rs1341402, rs954581,
DAOA_3'UTR_SNP12
rs2391191, rs778294, rs778293, rs1421292,
rs1935058, rs954581,
rs391695
rs1341402
DAOA_3'UTR_SNP12
rs3916967, rs778293, rs3918342, rs1935062
rs2391191
n.s.
Williams et al, 2006
17515 cases, 25189
controls
589 cases
617 controls
709 cases
1416 controls
rs2391191, rs947267, rs3918342
Vilella et al, 2007
schizotypy and cognitive
endophenotypes
schizophrenia, bipolar
disorder, depressive disorder
schizophrenia
0.0034 (Psychomotor speed)
5.00E-06 (Visuospatial ability)
0.0088 (Auditory attention)
0.0213 (Verbal working memory)
0.0047 (Immediate visual memory)
0.0010 (Delayed visual memory)
0.0024 (Free short delay recall)
0.0206 (Free long delay recall)
0.0079 (Recognition memory)
0.0060 (Stroop Interference score)
0.0239 (Semantic fluency)
0.0086 (General ability)
0.0063 (Abstraction)
0.0369 (Phonemic fluency)
0.0083 (General ability)
0.0173 (Abstraction)
0.0125 (Psychomotor speed)
0.0108 (Visuospatial ability)
0.0047 (Stroop Interference score)
0.0168 (Semantic fluency)
0.0051 (General ability)
0.0392 (Visuospatial ability)
0.0465 (MOOD, all)
0.0310 (MOOD, all)
0.0209 (MOOD, positive FHx)
0.0406 (MDD, all)
0.0407 (MDD, positive FHx)
0.0447 (anxiety)
0.0114 (time ill)
n.s.
rs2153674
rs701567
rs954580
schizophrenia
bipolar I disorder
Wang et al, 2004
Han Chinese
schizophrenia
706 cases
1416 controls
537 cases
538 controls
rs3916965
Wood et al, 2007
self-reported whites from
various US regions
schizophrenia
345 schizophrenia
cases, 150
schizoaffective cases,
352 controls
Yang et al, 2013
Taiwan´s Han Chinese
Population
schizophrenia
912 cases, 600
controls
26
rs2391191, rs947267, rs3918342, rs1935062,
rs1935058, rs954580, rs1341402, rs9301036,
rs9301035, rs778336, rs778334, rs778332,
rs778331, rs778330, rs17713906, rs1642681,
rs1341401, rs11618600, rs10492527
rs3916966, rs45476401, rs113692101,
rs2391191, rs778294, rs9558573
n.s.
n.s.
0.047
0.03
0.01
n.s.
0.0010 (OR 1.33, all)
0.020 (OR 1.33, males)
0.021, OR 1.34, females)
0.019 (OR 1.23, all)
0.045 (OR 1.29, females)
n.s.
n.s.
Yue et al, 2006
Chinese Han descendants
schizophrenia (SZ), early
onset schizophrenia (EOS),
male schizophrenia
Yue et al, 2007
Han Chinese
schizophrenia (SZ), clinical
symptoms
Zhang et al, 2009
Chinese Han
bipolar disorder
Zuliani et al, 2009
UK population
bipolar disorder, temporal lobe
and amygdala structure
Zou et al, 2005
Han Chinese
216 cases, 321
controls (SZ), 88
cases, 131 controls
(EOS), 120 cases, 184
controls (men)
359 cases
359 controls
237 trios
475 cases,
588 controls
38 cases, 81 controls
233 trios
rs2391191, rs778294
n.s.
rs947267
0.006 (OR 1.43, SZ)
0.012 (OR 1.67, EOS)
0.034 (OR 1.45, males)
n.s.
0.004 (OR 1.38)
0.019 (, trios)
n.s.
0.047 (OR 0.77)
0.003 (OR 1.33)
rs1421292: decreased gray matter
density left temporal pole in bipolar
disorder. rs3918342: reductions in
right amygdala gray matter density
n.s.
0.0108
0.0018
rs2391191, rs778294, rs3918342
rs947267
rs3916965, rs2391191, rs3918342
rs778294
rs778293
rs1421292, rs3918342
rs3916967
rs2391191
rs3916965
Addington AM, Gornick M, Sporn AL, Gogtay N, Greenstein D, Lenane M, Gochman P, Baker N, Balkissoon R, Vakkalanka RK, Weinberger DR, Straub RE, Rapoport JL. Polymorphisms in the
13q33.2 gene G72/G30 are associated with childhood-onset schizophrenia and psychosis not otherwise specified. Biol Psychiatry. 2004 May 15;55(10):976-80.
Andreou D, Saetre P, Werge T, Andreassen OA, Agartz I, Sedvall GC, Hall H, Terenius L, Jönsson EG. D-amino acid oxidase activator gene (DAOA) variation affects cerebrospinal fluid
homovanillic acid concentrations in healthy Caucasians. Eur Arch Psychiatry Clin Neurosci. 2012 Oct;262(7):549-56.
Bakker SC, Hoogendoorn ML, Hendriks J, Verzijlbergen K, Caron S, Verduijn W, Selten JP, Pearson PL, Kahn RS, Sinke RJ. The PIP5K2A and RGS4 genes are differentially associated with
deficit and non-deficit schizophrenia. Genes Brain Behav. 2007 Mar;6(2):113-9.
Bass NJ, Datta SR, McQuillin A, Puri V, Choudhury K, Thirumalai S, Lawrence J, Quested D, Pimm J, Curtis D, Gurling HM. Evidence for the association of the DAOA (G72) gene with
schizophrenia and bipolar disorder but not for the association of the DAO gene with schizophrenia. Behav Brain Funct. 2009 Jul 8;5:28.
Bousman CA, Yung AR, Pantelis C, Ellis JA, Chavez RA, Nelson B, Lin A, Wood SJ, Amminger GP, Velakoulis D, McGorry PD, Everall IP, Foley DL. Effects of NRG1 and DAOA genetic variation
on transition to psychosis in individuals at ultra-high risk for psychosis. Transl Psychiatry. 2013 Apr 30;3:e251.
Carrera N, Arrojo M, Paz E, Ramos-Ríos R, Agra S, Páramo M, Brenlla J, Costas J. Testing the antagonistic pleiotropy model of schizophrenia susceptibility by analysis of DAOA, PPP1R1B, and
APOL1 genes. Psychiatry Res. 2010 Sep 30;179(2):126-9.
Chen J, Xu Y, Zhang J, Liu Z, Xu C, Zhang K, Shen Y, Xu Q. Genotypic association of the DAOA gene with resting-state brain activity in major depression. Mol Neurobiol. 2012 Oct;46(2):36173.
Chen J, Xu Y, Zhang J, Liu Z, Xu C, Zhang K, Shen Y, Xu Q. A combined study of genetic association and brain imaging on the DAOA gene in schizophrenia. Am J Med Genet B Neuropsychiatr
Genet. 2013 Mar;162B(2):191-200.
Chen YS, Akula N, Detera-Wadleigh SD, Schulze TG, Thomas J, Potash JB, DePaulo JR, McInnis MG, Cox NJ, McMahon FJ. Findings in an independent sample support an association between
bipolar affective disorder and the G72/G30 locus on chromosome 13q33. Mol Psychiatry. 2004 Jan;9(1):87-92.
Chiesa A, Pae CU, Porcelli S, Han C, Lee SJ, Patkar AA, Park MH, Serretti A. DAOA variants and schizophrenia: influence on diagnosis and treatment outcomes. Int J Psychiatry Clin Pract.
2011 Nov;15(4):303-10.
Chiesa A, Pae CU, Porcelli S, Han C, Lee SJ, Patkar AA, Park MH, Jun TY, Serretti A. DAOA variants on diagnosis and response to treatment in patients with major depressive disorder and
bipolar disorder. J Int Med Res. 2012;40(1):258-65.
Chumakov I, Blumenfeld M, Guerassimenko O, Cavarec L, Palicio M, Abderrahim H, Bougueleret L, Barry C, Tanaka H, La Rosa P, Puech A, Tahri N, Cohen-Akenine A, Delabrosse S,
Lissarrague S, Picard FP, Maurice K, Essioux L, Millasseau P, Grel P, Debailleul V, Simon AM, Caterina D, Dufaure I, Malekzadeh K, Belova M, Luan JJ, Bouillot M, Sambucy JL, Primas G,
Saumier M, Boubkiri N, Martin-Saumier S, Nasroune M, Peixoto H, Delaye A, Pinchot V, Bastucci M, Guillou S, Chevillon M, Sainz-Fuertes R, Meguenni S, Aurich-Costa J, Cherif D, Gimalac
A, Van Duijn C, Gauvreau D, Ouellette G, Fortier I, Raelson J, Sherbatich T, Riazanskaia N, Rogaev E, Raeymaekers P, Aerssens J, Konings F, Luyten W, Macciardi F, Sham PC, Straub RE,
Weinberger DR, Cohen N, Cohen D. Genetic and physiological data implicating the new human gene G72 and the gene for D-amino acid oxidase in schizophrenia. Proc Natl Acad Sci U S
A. 2002 Oct 15;99(21):13675-80.
Chung S, Jung J, Chung HY, Yoo HK, Kim CY, Joo YH, Choi SE, Hong JP. No association between polymorphisms of DAO and DAOA genes and homicidal behaviors in Korean schizophrenia.
Psychiatr Genet. 2007 Oct;17(5):313.
27
Corvin A, McGhee KA, Murphy K, Donohoe G, Nangle JM, Schwaiger S, Kenny N, Clarke S, Meagher D, Quinn J, Scully P, Baldwin P, Browne D, Walsh C, Waddington JL, Morris DW, Gill M.
Evidence for association and epistasis at the DAOA/G30 and D-amino acid oxidase loci in an Irish schizophrenia sample. Am J Med Genet B Neuropsychiatr Genet. 2007 Oct
5;144B(7):949-53.
Corvin A, Donohoe G, McGhee K, Murphy K, Kenny N, Schwaiger S, Nangle JM, Morris D, Gill M. D-amino acid oxidase (DAO) genotype and mood symptomatology in schizophrenia. Neurosci
Lett. 2007 Oct 16;426(2):97-100.
Dalvie S, Horn N, Nossek C, van der Merwe L, Stein DJ, Ramesar R. Psychosis and relapse in bipolar disorder are related to GRM3, DAOA, and GRIN2B genotype. Afr J Psychiatry
(Johannesbg). 2010 Sep;13(4):297-301.
Detera-Wadleigh SD, McMahon FJ. G72/G30 in schizophrenia and bipolar disorder: review and meta-analysis. Biol Psychiatry. 2006 Jul 15;60(2):106-14.
Di Maria E, Bonvicini C, Bonomini C, Alberici A, Zanetti O, Gennarelli M. Genetic variation in the G720/G30 gene locus (DAOA) influences the occurrence of psychotic symptoms in patients
with Alzheimer's disease. J Alzheimers Dis. 2009;18(4):953-60.
Donohoe G, Morris DW, Robertson IH, McGhee KA, Murphy K, Kenny N, Clarke S, Gill M, Corvin AP. DAOA ARG30LYS and verbal memory function in schizophrenia. Mol Psychiatry. 2007
Sep;12(9):795-6.
Fallin MD, Lasseter VK, Avramopoulos D, Nicodemus KK, Wolyniec PS, McGrath JA, Steel G, Nestadt G, Liang KY, Huganir RL, Valle D, Pulver AE. Bipolar I disorder and schizophrenia: a 440single-nucleotide polymorphism screen of 64 candidate genes among Ashkenazi Jewish case-parent trios. Am J Hum Genet. 2005 Dec;77(6):918-36.
Gawlik M, Wehner I, Mende M, Jung S, Pfuhlmann B, Knapp M, Stöber G. The DAOA/G30 locus and affective disorders: haplotype based association study in a polydiagnostic approach. BMC
Psychiatry. 2010 Jul 29;10:59.
Gaysina D, Cohen-Woods S, Chow PC, Martucci L, Schosser A, Ball HA, Tozzi F, Perry J, Muglia P, Kennedy JL, King N, Vincent JB, Parikh SV, Strauss J, Craig IW, McGuffin P, Farmer A.
Association analysis of DAOA and DAO in bipolar disorder: results from two independent case-control studies. Bipolar Disord. 2010 Aug;12(5):579-81.
Goldberg TE, Straub RE, Callicott JH, Hariri A, Mattay VS, Bigelow L, Coppola R, Egan MF, Weinberger DR. The G72/G30 gene complex and cognitive abnormalities in schizophrenia.
Neuropsychopharmacology. 2006 Sep;31(9):2022-32.
Gomez L, Wigg K, Feng Y, Kiss E, Kapornai K, Tamás Z, Mayer L, Baji I, Daróczi G, Benák I, Kothencné VO, Dombovári E, Kaczvinszk E, Besnyo M, Gádoros J, King N, Székely J, Kovacs M, Vetró
A, Kennedy JL, Barr CL. G72/G30 (DAOA) and juvenile-onset mood disorders. Am J Med Genet B Neuropsychiatr Genet. 2009 Oct 5;150B(7):1007-12.
Grigoroiu-Serbanescu M, Herms S, Diaconu CC, Jamra RA, Meier S, Bleotu C, Neagu AI, Prelipceanu D, Sima D, Gherghel M, Mihailescu R, Rietschel M, Nöthen MM, Cichon S, Mühleisen TW.
Possible association of different G72/G30 SNPs with mood episodes and persecutory delusions in bipolar I Romanian patients. Prog Neuropsychopharmacol Biol Psychiatry. 2010 May
30;34(4):657-63.
Hall D, Gogos JA, Karayiorgou M. The contribution of three strong candidate schizophrenia susceptibility genes in demographically distinct populations. Genes Brain Behav. 2004
Aug;3(4):240-8.
Hall J, Whalley HC, Moorhead TW, Baig BJ, McIntosh AM, Job DE, Owens DG, Lawrie SM, Johnstone EC. Genetic variation in the DAOA (G72) gene modulates hippocampal function in subjects
at high risk of schizophrenia. Biol Psychiatry. 2008 Sep 1;64(5):428-33.
Hattori E, Liu C, Badner JA, Bonner TI, Christian SL, Maheshwari M, Detera-Wadleigh SD, Gibbs RA, Gershon ES. Polymorphisms at the G72/G30 gene locus, on 13q33, are associated with
bipolar disorder in two independent pedigree series. Am J Hum Genet. 2003 May;72(5):1131-40.
Hong CJ, Hou SJ, Yen FC, Liou YJ, Tsai SJ. Family-based association study between G72/G30 genetic polymorphism and schizophrenia. Neuroreport. 2006 Jul 17;17(10):1067-9.
Hukic DS, Frisén L, Backlund L, Lavebratt C, Landén M, Träskman-Bendz L, Edman G, Schalling M, Ösby U. Cognitive manic symptoms in bipolar disorder associated with polymorphisms in
the DAOA and COMT genes. PLoS One. 2013 Jul 5;8(7):e67450.
Jansen A, Krach S, Krug A, Markov V, Eggermann T, Zerres K, Thimm M, Nöthen MM, Treutlein J, Rietschel M, Kircher T. Effect of the G72 (DAOA) putative risk haplotype on cognitive
functions in healthy subjects. BMC Psychiatry. 2009 Sep 24;9:60.
Jansen A, Krach S, Krug A, Markov V, Thimm M, Paulus FM, Zerres K, Stöcker T, Shah NJ, Nöthen MM, Treutlein J, Rietschel M, Kircher T. The effect of G72 genotype on neural correlates of
memory encoding and retrieval. Neuroimage. 2010 Nov 15;53(3):1001-6.
Jönsson EG, Saetre P, Vares M, Andreou D, Larsson K, Timm S, Rasmussen HB, Djurovic S, Melle I, Andreassen OA, Agartz I, Werge T, Hall H, Terenius L. DTNBP1, NRG1, DAOA, DAO and
GRM3 polymorphisms and schizophrenia: an association study. Neuropsychobiology. 2009;59(3):142-50.
Korostishevsky M, Kaganovich M, Cholostoy A, Ashkenazi M, Ratner Y, Dahary D, Bernstein J, Bening-Abu-Shach U, Ben-Asher E, Lancet D, Ritsner M, Navon R. Is the G72/G30 locus
associated with schizophrenia? single nucleotide polymorphisms,haplotypes, and gene expression analysis. Biol Psychiatry. 2004 Aug 1;56(3):169-76.
Korostishevsky M, Kremer I, Kaganovich M, Cholostoy A, Murad I, Muhaheed M, Bannoura I, Rietschel M, Dobrusin M, Bening-Abu-Shach U, Belmaker RH, Maier W, Ebstein RP, Navon R.
Transmission disequilibrium and haplotype analyses of the G72/G30 locus: suggestive linkage to schizophrenia in Palestinian Arabs living in the North of Israel. Am J Med Genet B
Neuropsychiatr Genet. 2006 Jan 5;141B(1):91-5.
Kotaka T, Ujike H, Okahisa Y, Takaki M, Nakata K, Kodama M, Inada T, Yamada M, Uchimura N, Iwata N, Sora I, Iyo M, Ozaki N, Kuroda S. G72 gene is associated with susceptibility to
methamphetamine psychosis. Prog Neuropsychopharmacol Biol Psychiatry. 2009 Aug 31;33(6):1046-9.
Krug A, Markov V, Krach S, Jansen A, Zerres K, Eggermann T, Stöcker T, Shah NJ, Nöthen MM, Georgi A, Strohmaier J, Rietschel M, Kircher T. Genetic variation in G72 correlates with brain
activation in the right middle temporal gyrus in a verbal fluency task in healthy individuals. Hum Brain Mapp. 2011 Jan;32(1):118-26.
28
Li D, He L. G72/G30 genes and schizophrenia: a systematic meta-analysis of association studies. Genetics. 2007 Feb;175(2):917-22.
Maheshwari M, Shi J, Badner JA, Skol A, Willour VL, Muzny DM, Wheeler DA, Gerald FR, Detera-Wadleigh S, McMahon FJ, Potash JB, Gershon ES, Liu C, Gibbs RA. Common and rare variants
of DAOA in bipolar disorder. Am J Med Genet B Neuropsychiatr Genet. 2009 Oct 5;150B(7):960-6.
Liu YL, Fann CS, Liu CM, Chang CC, Wu JY, Hung SI, Liu SK, Hsieh MH, Hwang TJ, Chan HY, Chen JJ, Faraone SV, Tsuang MT, Chen WJ, Hwu HG. No association of G72 and D-amino acid
oxidase genes with schizophrenia. Schizophr Res. 2006 Oct;87(1-3):15-20.
Ma J, Qin W, Wang XY, Guo TW, Bian L, Duan SW, Li XW, Zou FG, Fang YR, Fang JX, Feng GY, Gu NF, St Clair D, He L. Further evidence for the association between G72/G30 genes and
schizophrenia in two ethnically distinct populations. Mol Psychiatry. 2006 May;11(5):479-87.
Ma J, Sun J, Zhang H, Zhang R, Kang WH, Gao CG, Liu HS, Ma XH, Min ZX, Zhao WX, Ning QL, Wang SH, Zhang YC, Guo TW, Lu SM. Evidence for transmission disequilibrium at the DAOA gene
locus in a schizophrenia family sample. Neurosci Lett. 2009 Sep 22;462(2):105-8.
Maheshwari M, Shi J, Badner JA, Skol A, Willour VL, Muzny DM, Wheeler DA, Gerald FR, Detera-Wadleigh S, McMahon FJ, Potash JB, Gershon ES, Liu C, Gibbs RA. Common and rare variants
of DAOA in bipolar disorder. Am J Med Genet B Neuropsychiatr Genet. 2009 Oct 5;150B(7):960-6.
Mössner R, Schuhmacher A, Wagner M, Quednow BB, Frommann I, Kühn KU, Schwab SG, Rietschel M, Falkai P, Wölwer W, Ruhrmann S, Bechdolf A, Gaebel W, Klosterkötter J, Maier W.
DAOA/G72 predicts the progression of prodromal syndromes to first episode psychosis. Eur Arch Psychiatry Clin Neurosci. 2010 Apr;260(3):209-15.
Mulle JG, Chowdari KV, Nimgaonkar V, Chakravarti A. No evidence for association to the G72/G30 locus in an independent sample of schizophrenia families. Mol Psychiatry. 2005
May;10(5):431-3.
Müller DJ, Zai CC, Shinkai T, Strauss J, Kennedy JL. Association between the DAOA/G72 gene and bipolar disorder and meta-analyses in bipolar disorder and schizophrenia. Bipolar Disord.
2011 Mar;13(2):198-207.
Ohi K, Hashimoto R, Yasuda Y, Yoshida T, Takahashi H, Iike N, Fukumoto M, Takamura H, Iwase M, Kamino K, Ishii R, Kazui H, Sekiyama R, Kitamura Y, Azechi M, Ikezawa K, Kurimoto R,
Kamagata E, Tanimukai H, Tagami S, Morihara T, Ogasawara M, Okochi M, Tokunaga H, Numata S, Ikeda M, Ohnuma T, Ueno S, Fukunaga T, Tanaka T, Kudo T, Arai H, Ohmori T, Iwata
N, Ozaki N, Takeda M. Association study of the G72 gene with schizophrenia in a Japanese population: a multicenter study. Schizophr Res. 2009 Apr;109(1-3):80-5.
Pae CU, Chiesa A, Serretti A. Influence of DAOA gene variants on antipsychotic response after switch to aripiprazole. Psychiatry Res. 2010 Jul 30;178(2):430-2.
Prata D, Breen G, Osborne S, Munro J, St Clair D, Collier D. Association of DAO and G72(DAOA)/G30 genes with bipolar affective disorder. Am J Med Genet B Neuropsychiatr Genet. 2008 Sep
5;147B(6):914-7.
Prata DP, Papagni SA, Mechelli A, Fu CH, Kambeitz J, Picchioni M, Kane F, Kalidindi S, McDonald C, Kravariti E, Toulopoulou T, Bramon E, Walshe M, Murray R, Collier DA, McGuire PK. Effect
of D-amino acid oxidase activator (DAOA; G72) on brain function during verbal fluency. Hum Brain Mapp. 2012 Jan;33(1):143-53.
Réthelyi JM, Bakker SC, Polgár P, Czobor P, Strengman E, Pásztor PI, Kahn RS, Bitter I. Association study of NRG1, DTNBP1, RGS4, G72/G30, and PIP5K2A with schizophrenia and symptom
severity in a Hungarian sample. Am J Med Genet B Neuropsychiatr Genet. 2010 Apr 5;153B(3):792-801.
Rietschel M, Beckmann L, Strohmaier J, Georgi A, Karpushova A, Schirmbeck F, Boesshenz KV, Schmäl C, Bürger C, Jamra RA, Schumacher J, Höfels S, Kumsta R, Entringer S, Krug A, Markov
V, Maier W, Propping P, Wüst S, Kircher T, Nöthen MM, Cichon S, Schulze TG. G72 and its association with major depression and neuroticism in large population-based groups from
Germany. Am J Psychiatry. 2008 Jun;165(6):753-62.
Sacchetti E, Scassellati C, Minelli A, Valsecchi P, Bonvicini C, Pasqualetti P, Galluzzo A, Pioli R, Gennarelli M. Schizophrenia susceptibility and NMDA-receptor mediated signalling: an
association study involving 32 tagSNPs of DAO, DAOA, PPP3CC, and DTNBP1 genes. BMC Med Genet. 2013 Mar 9;14:33.
Sanders AR, Duan J, Levinson DF, Shi J, He D, Hou C, Burrell GJ, Rice JP, Nertney DA, Olincy A, Rozic P, Vinogradov S, Buccola NG, Mowry BJ, Freedman R, Amin F, Black DW, Silverman JM,
Byerley WF, Crowe RR, Cloninger CR, Martinez M, Gejman PV. No significant association of 14 candidate genes with schizophrenia in a large European ancestry sample: implications
for psychiatric genetics. Am J Psychiatry. 2008 Apr;165(4):497-506.
Schultz CC, Nenadic I, Koch K, Wagner G, Roebel M, Schachtzabel C, Mühleisen TW, Nöthen MM, Cichon S, Deufel T, Kiehntopf M, Rietschel M, Reichenbach JR, Sauer H, Schlösser RG.
Reduced cortical thickness is associated with the glutamatergic regulatory gene risk variant DAOA Arg30Lys in schizophrenia. Neuropsychopharmacology. 2011 Jul;36(8):1747-53.
Schulze TG, Ohlraun S, Czerski PM, Schumacher J, Kassem L, Deschner M, Gross M, Tullius M, Heidmann V, Kovalenko S, Jamra RA, Becker T, Leszczynska-Rodziewicz A, Hauser J, Illig T,
Klopp N, Wellek S, Cichon S, Henn FA, McMahon FJ, Maier W, Propping P, Nöthen MM, Rietschel M. Genotype-phenotype studies in bipolar disorder showing association between the
DAOA/G30 locus and persecutory delusions: a first step toward a molecular genetic classification of psychiatric phenotypes. Am J Psychiatry. 2005 Nov;162(11):2101-8.
Schumacher J, Jamra RA, Freudenberg J, Becker T, Ohlraun S, Otte AC, Tullius M, Kovalenko S, Bogaert AV, Maier W, Rietschel M, Propping P, Nöthen MM, Cichon S. Examination of G72 and
D-amino-acid oxidase as genetic risk factors for schizophrenia and bipolar affective disorder. Mol Psychiatry. 2004 Feb;9(2):203-7.
Schumacher J, Abou Jamra R, Becker T, Klopp N, Franke P, Jacob C, Sand P, Fritze J, Ohlraun S, Schulze TG, Rietschel M, Illig T, Propping P, Cichon S, Deckert J, Nöthen MM. Investigation of
the DAOA/G30 locus in panic disorder. Mol Psychiatry. 2005 May;10(5):428-9.
Seifuddin F, Mahon PB, Judy J, Pirooznia M, Jancic D, Taylor J, Goes FS, Potash JB, Zandi PP. Meta-analysis of genetic association studies on bipolar disorder. Am J Med Genet B
Neuropsychiatr Genet. 2012 Jul;159B(5):508-18.
Shi J, Badner JA, Gershon ES, Liu C. Allelic association of G72/G30 with schizophrenia and bipolar disorder: a comprehensive meta-analysis. Schizophr Res. 2008 Jan;98(1-3):89-97.
Shi J, Badner JA, Gershon ES, Chunyu L, Willour VL, Potash JB. Further evidence for an association of G72/G30 with schizophrenia in Chinese. Schizophr Res. 2009 Feb;107(2-3):324-6.
Shin HD, Park BL, Kim EM, Lee SO, Cheong HS, Lee CH, Kim SG, Sohn JW, Park CS, Kim JW, Kim BH, Kim IY, Choi IG, Woo SI. Association analysis of G72/G30 polymorphisms with
schizophrenia in the Korean population. Schizophr Res. 2007 Nov;96(1-3):119-24.
29
Shinkai T, De Luca V, Hwang R, Müller DJ, Lanktree M, Zai G, Shaikh S, Wong G, Sicard T, Potapova N, Trakalo J, King N, Matsumoto C, Hori H, Wong AH, Ohmori O, Macciardi F, Nakamura J,
Kennedy JL. Association analyses of the DAOA/G30 and D-amino-acid oxidase genes in schizophrenia: further evidence for a role in schizophrenia. Neuromolecular Med.
2007;9(2):169-77.
Soronen P, Silander K, Antila M, Palo OM, Tuulio-Henriksson A, Kieseppä T, Ellonen P, Wedenoja J, Turunen JA, Pietiläinen OP, Hennah W, Lönnqvist J, Peltonen L, Partonen T, Paunio T.
Association of a nonsynonymous variant of DAOA with visuospatial ability in a bipolar family sample. Biol Psychiatry. 2008 Sep 1;64(5):438-42.
Soronen P, Mantere O, Melartin T, Suominen K, Vuorilehto M, Rytsälä H, Arvilommi P, Holma I, Holma M, Jylhä P, Valtonen HM, Haukka J, Isometsä E, Paunio T. P2RX7 gene is associated
consistently with mood disorders and predicts clinical outcome in three clinical cohorts. Am J Med Genet B Neuropsychiatr Genet. 2011 Jun;156B(4):435-47.
Stefanis NC, Trikalinos TA, Avramopoulos D, Smyrnis N, Evdokimidis I, Ntzani EE, Ioannidis JP, Stefanis CN. Impact of schizophrenia candidate genes on schizotypy and cognitive
endophenotypes at the population level. Biol Psychiatry. 2007 Oct 1;62(7):784-92.
Suliman H, Schumacher J, Becker T, Cichon S, Schulze TG, Propping P, Rietschel M, Nöthen MM, Jamra RA. Association study of 20 genetic variants at the (D)-amino acid oxidase gene in
schizophrenia. Psychiatr Genet. 2010 Apr;20(2):82-3.
Tan J, Lin Y, Su L, Yan Y, Chen Q, Jiang H, Wei Q, Gu L. Association between DAOA gene polymorphisms and the risk of schizophrenia, bipolar disorder and depressive disorder. Prog
Neuropsychopharmacol Biol Psychiatry. 2014 Jun 3;51:89-98.
Vilella E, Costas J, Sanjuan J, Guitart M, De Diego Y, Carracedo A, Martorell L, Valero J, Labad A, De Frutos R, Nájera C, Moltó MD, Toirac I, Guillamat R, Brunet A, Vallès V, Pérez L, Leon M, de
Fonseca FR, Phillips C, Torres M. Association of schizophrenia with DTNBP1 but not with DAO, DAOA, NRG1 and RGS4 nor their genetic interaction. J Psychiatr Res. 2008
Mar;42(4):278-88.
Williams NM, Green EK, Macgregor S, Dwyer S, Norton N, Williams H, Raybould R, Grozeva D, Hamshere M, Zammit S, Jones L, Cardno A, Kirov G, Jones I, O'Donovan MC, Owen MJ, Craddock
N. Variation at the DAOA/G30 locus influences susceptibility to major mood episodes but not psychosis in schizophrenia and bipolar disorder. Arch Gen Psychiatry. 2006
Apr;63(4):366-73.
Wang X, He G, Gu N, Yang J, Tang J, Chen Q, Liu X, Shen Y, Qian X, Lin W, Duan Y, Feng G, He L. Association of G72/G30 with schizophrenia in the Chinese population. Biochem Biophys Res
Commun. 2004 Jul 9;319(4):1281-6.
Wood LS, Pickering EH, Dechairo BM. Significant support for DAO as a schizophrenia susceptibility locus: examination of five genes putatively associated with schizophrenia. Biol
Psychiatry. 2007 May 15;61(10):1195-9.
Yang HC, Liu CM, Liu YL, Chen CW, Chang CC, Fann CS, Chiou JJ, Yang UC, Chen CH, Faraone SV, Tsuang MT, Hwu HG. The DAO gene is associated with schizophrenia and interacts with other
genes in the Taiwan Han Chinese population. PLoS One. 2013;8(3):e60099.
Yue W, Liu Z, Kang G, Yan J, Tang F, Ruan Y, Zhang J, Zhang D. Association of G72/G30 polymorphisms with early-onset and male schizophrenia. Neuroreport. 2006 Dec 18;17(18):1899902.
Yue W, Kang G, Zhang Y, Qu M, Tang F, Han Y, Ruan Y, Lu T, Zhang J, Zhang D. Association of DAOA polymorphisms with schizophrenia and clinical symptoms or therapeutic effects.
Neurosci Lett. 2007 Apr 6;416(1):96-100.
Zhang Z, Li Y, Zhao Q, Huang K, Wang P, Yang P, Li S, Feng G, Lindpaintner K, He L, Shi Y. First evidence of association between G72 and bipolar disorder in the Chinese Han population.
Psychiatr Genet. 2009 Jun;19(3):151-3.
Zuliani R, Moorhead TW, Job D, McKirdy J, Sussmann JE, Johnstone EC, Lawrie SM, Brambilla P, Hall J, McIntosh AM. Genetic variation in the G72 (DAOA) gene affects temporal lobe and
amygdala structure in subjects affected by bipolar disorder. Bipolar Disord. 2009 Sep;11(6):621-7.
Zou F, Li C, Duan S, Zheng Y, Gu N, Feng G, Xing Y, Shi J, He L. A family-based study of the association between the G72/G30 genes and schizophrenia in the Chinese population. Schizophr
Res. 2005 Mar 1;73(2-3):257-61.
30
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
Emamian, E.S., Hall, D., Birnbaum, M.J., Karayiorgou, M. & Gogos, J.A. Convergent
evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat Genet 36,
131-7 (2004).
Gilman, S.R. et al. Diverse types of genetic variation converge on functional gene
networks involved in schizophrenia. Nature neuroscience 15, 1723-8 (2012).
Freedman, R. et al. Linkage of a neurophysiological deficit in schizophrenia to a
chromosome 15 locus. Proceedings of the National Academy of Sciences of the United
States of America 94, 587-592 (1997).
Leonard, S. et al. Association of promoter variants in the alpha 7 nicotinic
acetylcholine receptor subunit gene with an inhibitory deficit found in
schizophrenia. Arch. Gen. Psych. 59, 1085-1096 (2002).
Freedman, R., Hall, M., Adler, L.E. & Leonard, S. Evidence in postmortem brain tissue
for decreased numbers of hippocampal nicotinic receptors in schizophrenia. Biol.
Psych. 38, 22-33 (1995).
Stephens, S.H. et al. Association of the 5'-upstream regulatory region of the α7
nicotinic acetylcholine receptor subunit gene (CHRNA7) with schizophrenia.
Schizo.Res. 109, 102-112 (2009).
Stone, J.L. et al. Rare chromosomal deletions and duplications increase risk of
schizophrenia. Nature 455, 237-241 (2008).
Stefansson, H. et al. Large recurrent microdeletions associated with schizophrenia.
Nature 455, 232-237 (2008).
Gault, J. et al. Genomic organization and partial duplication of the human α7
neuronal nicotinic acetylcholine receptor gene. Genomics 52, 173-185 (1998).
Araud, T. et al. The duplicated alpha 7 nicotinic receptor gene CHRFAM7A is a
dominant negative regulator of CHRNA7 expression. Biochem. Pharmacol. 82, 904914 (2011).
Li, T. et al. Preferential transmission of the high activity allele of COMT in
schizophrenia. Psychiatr Genet 6, 131-133 (1996).
Allen, N.C. et al. Systematic meta-analyses and field synopsis of genetic association
studies in schizophrenia: the SzGene database. Nat Genet 40, 827-834 (2008).
Sullivan, P.F. et al. Genomewide association for schizophrenia in the CATIE study:
results of stage 1. Mol Psychiatry 13, 570-584 (2008).
Gothelf, D. et al. Biological effects of COMT haplotypes and psychosis risk in 22q11.2
deletion syndrome. Biol Psychiatry 75, 406-413 (2014).
Gothelf, D. et al. COMT genotype predicts longitudinal cognitive decline and
psychosis in 22q11.2 deletion syndrome. Nat Neurosci 8, 1500-2 (2005).
Caspi, A. et al. Moderation of the effect of adolescent-onset cannabis use on adult
psychosis by a functional polymorphism in the catechol-O-methyltransferase gene:
longitudinal evidence of a gene X environment interaction. Biol Psychiatry 57, 111727 (2005).
31
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
Henquet, C. et al. An experimental study of catechol-o-methyltransferase Val158Met
moderation of delta-9-tetrahydrocannabinol-induced effects on psychosis and
cognition. Neuropsychopharmacology 31, 2748-2757 (2006).
Egan, M.F. et al. Effect of COMT Val108/158 Met genotype on frontal lobe function
and risk for schizophrenia. Proc Natl Acad Sci U S A 98, 6917-6922 (2001).
Kaenmaki, M. et al. Quantitative role of COMT in dopamine clearance in the
prefrontal cortex of freely moving mice. J Neurochem 114, 1745-1755 (2010).
Papaleo, F., Erickson, L., Liu, G., Chen, J. & Weinberger, D.R. Effects of sex and COMT
genotype on environmentally modulated cognitive control in mice. Proc Natl Acad
Sci U S A 109, 20160-20165 (2012).
Papaleo, F. et al. Genetic dissection of the role of catechol-O-methyltransferase in
cognition and stress reactivity in mice. J Neurosci 28, 8709-8723 (2008).
Risbrough, V., Ji, B., Hauger, R. & Zhou, X. Generation and Characterization of
Humanized Mice Carrying COMT158 Met/Val Alleles. Neuropsychopharmacology
(2014).
Bray, N.J. et al. A haplotype implicated in schizophrenia susceptibility is associated
with reduced COMT expression in human brain. Am J Hum Genet 73, 152-161
(2003).
Chen, J. et al. Functional analysis of genetic variation in catechol-Omethyltransferase (COMT): effects on mRNA, protein, and enzyme activity in
postmortem human brain. Am J Hum Genet 75, 807-821 (2004).
Nackley, A.G. et al. Human catechol-O-methyltransferase haplotypes modulate
protein expression by altering mRNA secondary structure. Science 314, 1930-1933
(2006).
Cools, R. & D'Esposito, M. Inverted-U-shaped dopamine actions on human working
memory and cognitive control. Biol Psychiatry 69, 113-125 (2011).
Papaleo, F., Burdick, M.C., Callicott, J.H. & Weinberger, D.R. COMT-Dysbindin
epistatic interaction. Mol Psychiatry 19, 273-273 (2014).
Costas, J. et al. Interaction between COMT haplotypes and cannabis in schizophrenia:
a case-only study in two samples from Spain. Schizophr Res 127, 22-7 (2011).
Chumakov, I. et al. Genetic and physiological data implicating the new human gene
G72 and the gene for D-amino acid oxidase in schizophrenia. Proc Natl Acad Sci U S A
99, 13675-80 (2002).
Schumacher, J. et al. Examination of G72 and D-amino-acid oxidase as genetic risk
factors for schizophrenia and bipolar affective disorder. Mol Psychiatry 9, 203-7
(2004).
Detera-Wadleigh, S.D. & McMahon, F.J. G72/G30 in schizophrenia and bipolar
disorder: review and meta-analysis. Biol Psychiatry 60, 106-14 (2006).
Shi, J., Badner, J.A., Gershon, E.S. & Liu, C. Allelic association of G72/G30 with
schizophrenia and bipolar disorder: a comprehensive meta-analysis. Schizophr Res
98, 89-97 (2008).
Muller, D.J., Zai, C.C., Shinkai, T., Strauss, J. & Kennedy, J.L. Association between the
DAOA/G72 gene and bipolar disorder and meta-analyses in bipolar disorder and
schizophrenia. Bipolar Disord 13, 198-207 (2011).
32
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
Tan, J. et al. Association between DAOA gene polymorphisms and the risk of
schizophrenia, bipolar disorder and depressive disorder. Prog
Neuropsychopharmacol Biol Psychiatry 51, 89-98 (2014).
Millar, J.K. et al. Disruption of two novel genes by a translocation co-segregating
with schizophrenia. Human Molecular Genetics 9, 1415-23 (2000).
Blackwood, D.H. et al. Schizophrenia and affective disorders--cosegregation with a
translocation at chromosome 1q42 that directly disrupts brain-expressed genes:
clinical and P300 findings in a family. American Journal of Human Genetics 69, 42833 (2001).
Brandon, N.J. & Sawa, A. Linking neurodevelopmental and synaptic theories of
mental illness through DISC1. Nat Rev Neurosci 12, 707-22 (2011).
Bradshaw, N.J. & Porteous, D.J. DISC1-binding proteins in neural development,
signalling and schizophrenia. Neuropharmacology 62, 1230-41 (2012).
Song, W. et al. Identification of high risk DISC1 structural variants with a 2%
attributable risk for schizophrenia. Biochemical and Biophysical Research
Communications 367, 700-706 (2008).
Thomson, P.A. et al. 708 Common and 2010 rare DISC1 locus variants identified in
1542 subjects: analysis for association with psychiatric disorder and cognitive traits.
Mol Psychiatry (2013).
Sanders, S.J. et al. De novo mutations revealed by whole-exome sequencing are
strongly associated with autism. Nature 485, 237-41 (2012).
Mathieson, I., Munafo, M.R. & Flint, J. Meta-analysis indicates that common variants
at the DISC1 locus are not associated with schizophrenia. Molecular Psychiatry 17,
634-41 (2012).
Ripke, S. et al. Genome-wide association analysis identifies 13 new risk loci for
schizophrenia. Nat Genet 45, 1150-9 (2013).
Soares, D.C., Carlyle, B.C., Bradshaw, N.J. & Porteous, D.J. DISC1: Structure, Function,
and Therapeutic Potential for Major Mental Illness. ACS Chem Neurosci 2, 609-632
(2011).
Hennah, W. & Porteous, D. The DISC1 pathway modulates expression of
neurodevelopmental, synaptogenic and sensory perception genes. PLoS One 4,
e4906 (2009).
Carless, M.A. et al. Impact of DISC1 variation on neuroanatomical and
neurocognitive phenotypes. Molecular Psychiatry 16, 1096-104, 1063 (2011).
Zhou, M. et al. mTOR Inhibition ameliorates cognitive and affective deficits caused
by Disc1 knockdown in adult-born dentate granule neurons. Neuron 77, 647-54
(2013).
Leliveld, S.R. et al. Oligomer Assembly of the C-Terminal DISC1 Domain (640−854)
Is Controlled by Self-Association Motifs and Disease-Associated Polymorphism
S704C. Biochemistry 48, 7746-7755 (2009).
Singh, K.K. et al. Common DISC1 polymorphisms disrupt Wnt/GSK3beta signaling
and brain development. Neuron 72, 545-58 (2011).
Malavasi, E.L., Ogawa, F., Porteous, D.J. & Millar, J.K. DISC1 variants 37W and 607F
disrupt its nuclear targeting and regulatory role in ATF4-mediated transcription.
Human Molecular Genetics 21, 2779-92 (2012).
33
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
Ogawa, F. et al. DISC1 complexes with TRAK1 and Miro1 to modulate anterograde
axonal mitochondrial trafficking. Hum Mol Genet (2013).
Bertram, L., Lill, C.M. & Tanzi, R.E. The genetics of Alzheimer disease: back to the
future. Neuron 68, 270-81 (2010).
Corripio, I. et al. Density of striatal D2 receptors in untreated first-episode
psychosis: an I123-IBZM SPECT study. Eur Neuropsychopharmacol 21, 861-6 (2011).
Seeman, P. Schizophrenia and dopamine receptors. Eur Neuropsychopharmacol 23,
999-1009 (2013).
Seeman, P. et al. Dopamine supersensitivity correlates with D2High states, implying
many paths to psychosis. Proc Natl Acad Sci U S A 102, 3513-8 (2005).
Ward, L.D. & Kellis, M. HaploReg: a resource for exploring chromatin states,
conservation, and regulatory motif alterations within sets of genetically linked
variants. Nucleic Acids Res 40, D930-4 (2012).
Boyle, A.P. et al. Annotation of functional variation in personal genomes using
RegulomeDB. Genome Res 22, 1790-7 (2012).
Xu, Z. & Taylor, J.A. SNPinfo: integrating GWAS and candidate gene information into
functional SNP selection for genetic association studies. Nucleic Acids Res 37, W6005 (2009).
Yilmaz, Z. et al. Antipsychotics, dopamine D(2) receptor occupancy and clinical
improvement in schizophrenia: a meta-analysis. Schizophr Res 140, 214-20 (2012).
Zhang, J.P., Lencz, T. & Malhotra, A.K. D2 receptor genetic variation and clinical
response to antipsychotic drug treatment: a meta-analysis. Am J Psychiatry 167,
763-72 (2010).
Zai, C.C. et al. Meta-analysis of two dopamine D2 receptor gene polymorphisms with
tardive dyskinesia in schizophrenia patients. Mol Psychiatry 12, 794-5 (2007).
Bakker, P.R., van Harten, P.N. & van Os, J. Antipsychotic-induced tardive dyskinesia
and polymorphic variations in COMT, DRD2, CYP1A2 and MnSOD genes: a metaanalysis of pharmacogenetic interactions. Mol Psychiatry 13, 544-56 (2008).
Muller, D.J. et al. Systematic analysis of dopamine receptor genes (DRD1-DRD5) in
antipsychotic-induced weight gain. Pharmacogenomics J 12, 156-64 (2012).
Collins, A.L. et al. Hypothesis-driven candidate genes for schizophrenia compared to
genome-wide association results. Psychol Med 42, 607-16 (2012).
Girgenti, M.J., LoTurco, J.J. & Maher, B.J. ZNF804a regulates expression of the
schizophrenia-associated genes PRSS16, COMT, PDE4B, and DRD2. PLoS One 7,
e32404 (2012).
Ferguson, S.S. Evolving concepts in G protein-coupled receptor endocytosis: the role
in receptor desensitization and signaling. Pharmacol Rev 53, 1-24 (2001).
Williams, J. et al. Association between schizophrenia and T102C polymorphism of
the 5-hydroxytryptamine type 2a-receptor gene. European Multicentre Association
Study of Schizophrenia (EMASS) Group. Lancet 347, 1294-6 (1996).
Hawi, Z. et al. No association or linkage between the 5-HT2a/T102C polymorphism
and schizophrenia in Irish families. Am J Med Genet 74, 370-3 (1997).
Erdmann, J. et al. Systematic screening for mutations in the human serotonin-2A (5HT2A) receptor gene: identification of two naturally occurring receptor variants and
association analysis in schizophrenia. Hum Genet 97, 614-9 (1996).
34
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
Arranz, M.J., Lin, M.W., Powell, J., Kerwin, R. & Collier, D. 5HT 2a receptor T102C
polymorphism and schizophrenia. Lancet 347, 1831-2 (1996).
Ishigaki, T. et al. Intact 5-HT2A receptor exons and the adjoining intron regions in
schizophrenia. Neuropsychopharmacology 14, 339-47 (1996).
Malhotra, A.K., Goldman, D., Buchanan, R., Breier, A. & Pickar, D. 5HT 2a receptor
T102C polymorphism and schizophrenia. Lancet 347, 1830-1 (1996).
Sasaki, T., Hattori, M., Fukuda, R., Kunugi, H. & Nanko, S. 5HT 2a receptor T102C
polymorphism and schizophrenia. Lancet 347, 1832 (1996).
Saiz, P.A. et al. Association study of serotonin 2A receptor (5-HT2A) and serotonin
transporter (5-HTT) gene polymorphisms with schizophrenia. Prog
Neuropsychopharmacol Biol Psychiatry 31, 741-5 (2007).
Penas-Lledo, E.M., Dorado, P., Caceres, M.C., de la Rubia, A. & Llerena, A. Association
between T102C and A-1438G polymorphisms in the serotonin receptor 2A (5HT2A) gene and schizophrenia: relevance for treatment with antipsychotic drugs.
Clin Chem Lab Med 45, 835-8 (2007).
Joober, R. et al. T102C polymorphism in the 5HT2A gene and schizophrenia: relation
to phenotype and drug response variability. J Psychiatry Neurosci 24, 141-6 (1999).
Ramanathan, S. & Glatt, S.J. Serotonergic system genes in psychosis of Alzheimer
dementia: meta-analysis. Am J Geriatr Psychiatry 17, 839-46 (2009).
Arranz, M.J. et al. Evidence for association between polymorphisms in the promoter
and coding regions of the 5-HT2A receptor gene and response to clozapine. Mol
Psychiatry 3, 61-6 (1998).
Arranz, M. et al. Association between clozapine response and allelic variation in 5HT2A receptor gene. Lancet 346, 281-2 (1995).
Masellis, M. et al. Serotonin subtype 2 receptor genes and clinical response to
clozapine in schizophrenia patients. Neuropsychopharmacology 19, 123-32 (1998).
Lin, C.H. et al. No evidence for association of serotonin-2A receptor variant (102T/C)
with schizophrenia or clozapine response in a Chinese population. Neuroreport 10,
57-60 (1999).
Malhotra, A.K. et al. Lack of association between polymorphisms in the 5-HT2A
receptor gene and the antipsychotic response to clozapine. Am J Psychiatry 153,
1092-4 (1996).
Nothen, M.M. et al. Genetic variation of the 5-HT2A receptor and response to
clozapine. Lancet 346, 908-9 (1995).
Schumacher, J., Schulze, T.G., Wienker, T.F., Rietschel, M. & Nothen, M.M.
Pharmacogenetics of the clozapine response. Lancet 356, 506-7 (2000).
Masellis, M. et al. Genetic variation of 5-HT2A receptor and response to clozapine.
Lancet 346, 1108 (1995).
Kirchheiner, J. et al. Pharmacogenetics of antidepressants and antipsychotics: the
contribution of allelic variations to the phenotype of drug response. Mol Psychiatry
9, 442-73 (2004).
Filippini, N. et al. Influence of serotonin receptor 2A His452Tyr polymorphism on
brain temporal structures: a volumetric MR study. Eur J Hum Genet 14, 443-9
(2006).
Schott, B.H. et al. Genetic variation of the serotonin 2a receptor affects hippocampal
novelty processing in humans. PLoS One 6, e15984 (2011).
35
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106.
Olajossy-Hilkesberger, L. et al. Polymorphisms of the 5-HT2A receptor gene and
clinical response to olanzapine in paranoid schizophrenia. Neuropsychobiology 64,
202-10 (2011).
Blasi, G. et al. Converging evidence for the association of functional genetic variation
in the serotonin receptor 2a gene with prefrontal function and olanzapine
treatment. JAMA Psychiatry 70, 921-30 (2013).
Ozaki, N. et al. A naturally occurring amino acid substitution of the human serotonin
5-HT2A receptor influences amplitude and timing of intracellular calcium
mobilization. J Neurochem 68, 2186-93 (1997).
Hazelwood, L.A. & Sanders-Bush, E. His452Tyr polymorphism in the human 5-HT2A
receptor destabilizes the signaling conformation. Mol Pharmacol 66, 1293-300
(2004).
Bray, N.J., Buckland, P.R., Hall, H., Owen, M.J. & O'Donovan, M.C. The serotonin-2A
receptor gene locus does not contain common polymorphism affecting mRNA levels
in adult brain. Mol Psychiatry 9, 109-14 (2004).
Smith, R.M. et al. Multiple regulatory variants modulate expression of 5hydroxytryptamine 2A receptors in human cortex. Biol Psychiatry 73, 546-54
(2013).
Colantuoni, C. et al. Temporal dynamics and genetic control of transcription in the
human prefrontal cortex. Nature 478, 519-23 (2011).
Blouin, J.L. et al. Schizophrenia susceptibility loci on chromosomes 13q32 and 8p21.
Nat Genet 20, 70-73 (1998).
Pulver, A.E. et al. Schizophrenia: a genome scan targets chromosomes 3p and 8p as
potential sites of susceptibility genes. Am J Med Genet 60, 252-260 (1995).
Badner, J.A. & Gershon, E.S. Meta-analysis of whole-genome linkage scans of bipolar
disorder and schizophrenia. Mol Psychiatry 7, 405-411 (2002).
Stefansson, H. et al. Neuregulin 1 and susceptibility to schizophrenia. Am J Hum
Genet 71, 877-92 (2002).
Stefansson, H. et al. Association of neuregulin 1 with schizophrenia confirmed in a
Scottish population. Am J Hum Genet 72, 83-87 (2003).
Corvin, A.P. et al. Confirmation and refinement of an 'at-risk' haplotype for
schizophrenia suggests the EST cluster, Hs.97362, as a potential susceptibility gene
at the Neuregulin-1 locus. Mol Psychiatry 9, 208-213 (2004).
Li, D., Collier, D.A. & He, L. Meta-analysis shows strong positive association of the
neuregulin 1 (NRG1) gene with schizophrenia. Hum Mol Genet 15, 1995-2002
(2006).
Munafo, M.R., Thiselton, D.L., Clark, T.G. & Flint, J. Association of the NRG1 gene and
schizophrenia: a meta-analysis. Mol Psychiatry 11, 539-546 (2006).
Gong, Y.G. et al. A two-method meta-analysis of Neuregulin 1(NRG1) association and
heterogeneity in schizophrenia. Schizophr Res 111, 109-114 (2009).
Agim, Z.S. et al. Discovery, validation and characterization of Erbb4 and Nrg1
haplotypes using data from three genome-wide association studies of schizophrenia.
PLoS One 8(2013).
Harrison, P.J. & Law, A.J. Neuregulin 1 and schizophrenia: genetics, gene expression,
and neurobiology. Biol Psychiatry 60, 132-140 (2006).
36
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
Hall, J. et al. A neuregulin 1 variant associated with abnormal cortical function and
psychotic symptoms. Nat Neurosci 9, 1477-1478 (2006).
Nicodemus, K.K. et al. Biological validation of increased schizophrenia risk with
NRG1, ERBB4, and AKT1 epistasis via functional neuroimaging in healthy controls.
Arch Gen Psychiatry 67, 991-991001 (2010).
Knickmeyer, R.C. et al. Common variants in psychiatric risk genes predict brain
structure at birth. Cereb Cortex 24, 1230-1246 (2014).
Hashimoto, R. et al. Expression analysis of neuregulin-1 in the dorsolateral
prefrontal cortex in schizophrenia. Mol Psychiatry 9, 299-307 (2004).
Law, A.J. et al. Neuregulin 1 transcripts are differentially expressed in schizophrenia
and regulated by 5' SNPs associated with the disease. Proc Natl Acad Sci U S A 103,
6747-6752 (2006).
Moon, E. et al. Lack of association to a NRG1 missense polymorphism in
schizophrenia or bipolar disorder in a Costa Rican population. Schizophr Res 131,
52-57 (2011).
Law, A.J. unpublished observations.
Deakin, I.H. et al. Transgenic overexpression of the type I isoform of neuregulin 1
affects working memory and hippocampal oscillations but not long-term
potentiation. Cereb Cortex 22, 1520-1529 (2012).
Yin, D.-M. et al. Reversal of behavioral deficits and synaptic dysfunction in mice
overexpressing neuregulin 1. Neuron 78, 644-657 (2013).
Nicodemus, K.K. et al. Serious obstetric complications interact with hypoxiaregulated/vascular-expression genes to influence schizophrenia risk. Mol Psychiatry
13, 873-877 (2008).
Karl, T. Neuregulin 1: a prime candidate for research into gene-environment
interactions in schizophrenia? Insights from genetic rodent models. Front Behav
Neurosci 7, 106 (2013).
Norton, N. et al. Evidence that interaction between neuregulin 1 and its receptor
erbB4 increases susceptibility to schizophrenia. Am J Med Genet B Neuropsychiatr
Genet 141B, 96-9101 (2006).
Bani-Fatemi, A. et al. Analysis of CpG SNPs in 34 genes: association test with suicide
attempt in schizophrenia. Schizophr Res 147, 262-8 (2013).
So, H.C. et al. An association study of RGS4 polymorphisms with clinical phenotypes
of schizophrenia in a Chinese population. Am J Med Genet B Neuropsychiatr Genet
147B, 77-85 (2008).
Winantea, J. et al. A summary statistic approach to sequence variation in noncoding
regions of six schizophrenia-associated gene loci. Eur J Hum Genet 14, 1037-43
(2006).
Talkowski, M.E. et al. Evaluation of a susceptibility gene for schizophrenia: genotype
based meta-analysis of RGS4 polymorphisms from thirteen independent samples.
Biol Psychiatry 60, 152-62 (2006).
Fallin, M.D. et al. Bipolar I disorder and schizophrenia: a 440-single-nucleotide
polymorphism screen of 64 candidate genes among Ashkenazi Jewish case-parent
trios. Am J Hum Genet 77, 918-36 (2005).
Zhang, F. et al. Association analysis of the RGS4 gene in Han Chinese and Scottish
populations with schizophrenia. Genes Brain Behav 4, 444-8 (2005).
37
125.
126.
127.
128.
129.
130.
131.
132.
133.
134.
135.
136.
137.
138.
139.
140.
141.
Prasad, K.M. et al. Genetic polymorphisms of the RGS4 and dorsolateral prefrontal
cortex morphometry among first episode schizophrenia patients. Mol Psychiatry 10,
213-9 (2005).
Chen, X. et al. Regulator of G-protein signaling 4 (RGS4) gene is associated with
schizophrenia in Irish high density families. Am J Med Genet B Neuropsychiatr Genet
129B, 23-6 (2004).
Morris, D.W. et al. Confirming RGS4 as a susceptibility gene for schizophrenia. Am J
Med Genet B Neuropsychiatr Genet 125B, 50-3 (2004).
Williams, N.M. et al. Support for RGS4 as a susceptibility gene for schizophrenia. Biol
Psychiatry 55, 192-5 (2004).
Chowdari, K.V. et al. Association and linkage analyses of RGS4 polymorphisms in
schizophrenia. Hum Mol Genet 11, 1373-80 (2002).
Jonsson, E.G. et al. Lack of association between the regulator of G-protein signaling 4
(RGS4) rs951436 polymorphism and schizophrenia. Psychiatr Genet 22, 263-4
(2012).
Rethelyi, J.M. et al. Association study of NRG1, DTNBP1, RGS4, G72/G30, and
PIP5K2A with schizophrenia and symptom severity in a Hungarian sample. Am J
Med Genet B Neuropsychiatr Genet 153B, 792-801 (2010).
Sanders, A.R. et al. No significant association of 14 candidate genes with
schizophrenia in a large European ancestry sample: implications for psychiatric
genetics. Am J Psychiatry 165, 497-506 (2008).
Ishiguro, H. et al. RGS4 is not a susceptibility gene for schizophrenia in Japanese:
association study in a large case-control population. Schizophr Res 89, 161-4 (2007).
Guo, S. et al. RGS4 polymorphisms and risk of schizophrenia: an association study in
Han Chinese plus meta-analysis. Neurosci Lett 406, 122-7 (2006).
Li, D. & He, L. Association study of the G-protein signaling 4 (RGS4) and proline
dehydrogenase (PRODH) genes with schizophrenia: a meta-analysis. Eur J Hum
Genet 14, 1130-5 (2006).
Liu, Y.L. et al. Evaluation of RGS4 as a candidate gene for schizophrenia. Am J Med
Genet B Neuropsychiatr Genet 141B, 418-20 (2006).
Rizig, M.A. et al. Failure to confirm genetic association between schizophrenia and
markers on chromosome 1q23.3 in the region of the gene encoding the regulator of
G-protein signaling 4 protein (RGS4). Am J Med Genet B Neuropsychiatr Genet 141B,
296-300 (2006).
Sobell, J.L., Richard, C., Wirshing, D.A. & Heston, L.L. Failure to confirm association
between RGS4 haplotypes and schizophrenia in Caucasians. Am J Med Genet B
Neuropsychiatr Genet 139B, 23-7 (2005).
Cordeiro, Q. et al. Association and linkage analysis of RGS4 polymorphisms with
schizophrenia and bipolar disorder in Brazil. Genes Brain Behav 4, 45-50 (2005).
Volk, D.W., Eggan, S.M. & Lewis, D.A. Alterations in metabotropic glutamate receptor
1alpha and regulator of G protein signaling 4 in the prefrontal cortex in
schizophrenia. Am J Psychiatry 167, 1489-98 (2010).
Mirnics, K., Middleton, F.A., Stanwood, G.D., Lewis, D.A. & Levitt, P. Disease-specific
changes in regulator of G-protein signaling 4 (RGS4) expression in schizophrenia.
Mol Psychiatry 6, 293-301 (2001).
38
142.
143.
144.
145.
146.
147.
148.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
Ding, L. & Hegde, A.N. Expression of RGS4 splice variants in dorsolateral prefrontal
cortex of schizophrenic and bipolar disorder patients. Biol Psychiatry 65, 541-5
(2009).
Bowden, N.A., Scott, R.J. & Tooney, P.A. Altered expression of regulator of G-protein
signalling 4 (RGS4) mRNA in the superior temporal gyrus in schizophrenia.
Schizophr Res 89, 165-8 (2007).
Erdely, H.A., Tamminga, C.A., Roberts, R.C. & Vogel, M.W. Regional alterations in
RGS4 protein in schizophrenia. Synapse 59, 472-9 (2006).
Schwendt, M., Sigmon, S.A. & McGinty, J.F. RGS4 overexpression in the rat dorsal
striatum modulates mGluR5- and amphetamine-mediated behavior and signaling.
Psychopharmacology (Berl) 221, 621-35 (2012).
Vrajova, M., Pekova, S., Horacek, J. & Hoschl, C. The effects of siRNA-mediated RGS4
gene silencing on the whole genome transcription profile: implications for
schizophrenia. Neuro Endocrinol Lett 32, 246-52 (2011).
Chowdari, K.V. et al. Linkage disequilibrium patterns and functional analysis of RGS4
polymorphisms in relation to schizophrenia. Schizophr Bull 34, 118-26 (2008).
Gu, Z., Jiang, Q. & Yan, Z. RGS4 modulates serotonin signaling in prefrontal cortex
and links to serotonin dysfunction in a rat model of schizophrenia. Mol Pharmacol
71, 1030-9 (2007).
Ebert, P.J., Campbell, D.B. & Levitt, P. Bacterial artificial chromosome transgenic
analysis of dynamic expression patterns of regulator of G-protein signaling 4 during
development. II. Subcortical regions. Neuroscience 142, 1163-81 (2006).
Jaen, C. & Doupnik, C.A. RGS3 and RGS4 differentially associate with G proteincoupled receptor-Kir3 channel signaling complexes revealing two modes of RGS
modulation. Precoupling and collision coupling. J Biol Chem 281, 34549-60 (2006).
Gong, Y. et al. Polymorphisms in microRNA target sites influence susceptibility to
schizophrenia by altering the binding of miRNAs to their targets. Eur
Neuropsychopharmacol 23, 1182-9 (2013).
Cheng, Y.C. et al. Zebrafish rgs4 is essential for motility and axonogenesis mediated
by Akt signaling. Cell Mol Life Sci 70, 935-50 (2013).
Paspalas, C.D., Selemon, L.D. & Arnsten, A.F. Mapping the regulator of G protein
signaling 4 (RGS4): presynaptic and postsynaptic substrates for neuroregulation in
prefrontal cortex. Cereb Cortex 19, 2145-55 (2009).
Kang, G. et al. Two-stage designs to identify the effects of SNP combinations on
complex diseases. J Hum Genet 53, 739-46 (2008).
Nicodemus, K.K. et al. Evidence for statistical epistasis between catechol-Omethyltransferase (COMT) and polymorphisms in RGS4, G72 (DAOA), GRM3, and
DISC1: influence on risk of schizophrenia. Hum Genet 120, 889-906 (2007).
Lipska, B.K. et al. RGS4 mRNA expression in postmortem human cortex is associated
with COMT Val158Met genotype and COMT enzyme activity. Hum Mol Genet 15,
2804-12 (2006).
Buckholtz, J.W. et al. Allelic variation in RGS4 impacts functional and structural
connectivity in the human brain. J Neurosci 27, 1584-93 (2007).
Kattoulas, E. et al. Schizophrenia-related RGS4 gene variations specifically disrupt
prefrontal control of saccadic eye movements. Psychol Med 42, 757-67 (2012).
39
159.
160.
161.
162.
163.
164.
165.
Prasad, K.M. et al. RGS4 polymorphisms associated with variability of cognitive
performance in a family-based schizophrenia sample. Schizophr Bull 36, 983-90
(2010).
Stefanis, N.C. et al. Association of RGS4 variants with schizotypy and cognitive
endophenotypes at the population level. Behav Brain Funct 4, 46 (2008).
Lane, H.Y. et al. RGS4 polymorphisms predict clinical manifestations and responses
to risperidone treatment in patients with schizophrenia. J Clin Psychopharmacol 28,
64-8 (2008).
Campbell, D.B. et al. Ethnic stratification of the association of RGS4 variants with
antipsychotic treatment response in schizophrenia. Biol Psychiatry 63, 32-41
(2008).
Rivero, G. et al. Brain RGS4 and RGS10 protein expression in schizophrenia and
depression. Effect of drug treatment. Psychopharmacology (Berl) 226, 177-88
(2013).
Allen, N. et al. Systematic meta-analyses and field synopsis of genetic association
studies in schizophrenia: The SzGene Database. Nature Genetics 40, 827-34 (2008).
Schizophrenia Working Group of the Psychiatric Genomics Consortium. Genomewide association analysis identifies more than 100 loci for schizophrenia.
(Submitted).
40
Download