Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

SCHRES-05880; No of Pages 11

Schizophrenia Research xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Schizophrenia Research
journal homepage: www.elsevier.com/locate/schres

Review

Genetic underpinnings of white matter ‘connectivity’: Heritability, risk,


and heterogeneity in schizophrenia
Aristotle N. Voineskos ⁎
Kimel Family Translational Imaging-Genetics Laboratory, Research Imaging Centre, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Canada
Institute of Medical Science, University of Toronto, Canada
Department of Psychiatry, University of Toronto, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Schizophrenia is a highly heritable disorder. Thus, the combination of genetics and brain imaging may be a useful
Received 23 January 2014 strategy to investigate the effects of risk genes on anatomical connectivity, and for gene discovery, i.e. discovering
Received in revised form 11 March 2014 the genetic correlates of white matter phenotypes. Following a database search, I review evidence for heritability
Accepted 12 March 2014
of white matter phenotypes. I also review candidate gene investigations, examining association of putative risk
Available online xxxx
variants with white matter phenotypes, as well as the recent flurry of research exploring relationships of
Keywords:
genome-wide significant risk loci with white matter phenotypes. Finally, I review multivariate and polygene
White matter approaches, which constitute a new wave of imaging-genetics research, including large collaborative initiatives
Genetics aiming to discover new genes that may predict aspects of white matter microstructure. The literature supports
Diffusion tensor imaging the heritability of white matter phenotypes. Loci in genes intimately implicated in oligodendrocyte and myelin
Heritability development, growth and maintenance, and neurotrophic systems are associated with white matter microstruc-
Quantitative trait ture. GWAS variants have not yet sufficiently been explored using DTI-based evaluation of white matter to draw
GWAS conclusions, although micro-RNA 137 is promising due to its potential regulation of other GWAS schizophrenia
Schizophrenia
genes. Many imaging-genetic studies only include healthy participants, which, while helping control for certain
confounds, cannot address questions related to disease heterogeneity or symptom expression, and thus more
studies should include participants with schizophrenia. With sufficiently large sample sizes, the future of this
field lies in polygene strategies aimed at risk prediction and heterogeneity dissection of schizophrenia that can
translate to personalized interventions.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction phenotypes using a quantitative trait locus (QTL) based approach. The
field is now in a position to understand the effects of genetic loci that
In the past 5 years, needles in the complex haystack of schizophrenia contribute to variation in white matter neuroimaging phenotypes,
genetics have been uncovered (O'Donovan et al., 2008; Stefansson et al., and in the process map the genetic underpinnings of white matter
2008; Stefansson et al., 2009; 2011; Ripke et al., 2013). Sample sizes in dysconnectivity that is a critical neurobiological feature for many people
the tens of thousands have now convincingly established schizophrenia suffering from schizophrenia (Shenton et al., 2010).
risk loci, just as such large samples were required in other disorders The purpose of this manuscript is to review what is known regarding
(Ripke et al., 2013). Now, as genetic risk loci emerge, the challenge is the underlying genetic architecture of white matter phenotypes (with a
to identify where, when, and how these loci confer risk for schizophre- focus on diffusion tensor imaging phenotypes), based on work done in
nia. Furthermore, some of these risk loci may provide important clues healthy subjects and from schizophrenia patients. While there is little
regarding dissecting the heterogeneity of schizophrenia. The ‘bottom- doubt that there is shared genetic vulnerability that cuts across psychi-
up’ opportunity of imaging-genetics today is to help characterize the atric disorders, a review of the shared and unique genetic factors under-
effects of these risk loci on vulnerable brain circuitry both in healthy lying potential white matter disruption in mental illnesses such as
people and in people with schizophrenia. The ‘top-down’ opportunity bipolar disorder, autism spectrum disorder, or major depressive disor-
of imaging-genetics is to use large sample sizes with both neuroimaging der is beyond the scope of this manuscript.
and genetic information to identify novel loci or confirm existing To find English language references relevant to the present review on
genetic risk loci from schizophrenia GWAS based on neuroimaging imaging-genetics in white matter in schizophrenia, a sensitive search
strategy was conducted in three relevant article databases (MEDLINE
⁎ 7th Floor, 250 College St, Toronto, Ontario M5T 1R8, Canada. Tel.: +1 416 535
(1946–2014), EMBASE (1974–2014) and PsychINFO (1967–2014))
8501x4378; fax: +1 416 979 6936. using the OvidSP platform. Search terms included “heritability”, “white
E-mail address: Aristotle.Voineskos@camh.ca. matter”, “magnetic resonance imaging”, “diffusion tensor imaging”,

http://dx.doi.org/10.1016/j.schres.2014.03.034
0920-9964/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
2 A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx

“genetics”, “genome-wide association”, “schizophrenia”, and insight into the heritability of white matter volumes. A recent meta-
“psychosis”. All titles and abstracts were screened, and full-text versions analysis identified over 50 such studies (many of which also assessed
of articles deemed relevant to the present review were then obtained. gray matter volumes, cortical thickness, and other neuroimaging phe-
Results of the review are described primarily in Sections 2–5, with notypes derived from T1-weighted imaging). The results of this meta-
Table 1 specifically highlighting the results of association studies of analysis (Blokland et al., 2012) supported strong genetic effects on
genome-wide significant variants for schizophrenia with white matter total and regional white matter volumes. Across the lifespan, studies
phenotypes. continue to support heritability of white matter volume in large sam-
ples. A study of 8 year old twins, 57 MZ and 35 DZ (Quebec Newborn
2. Heritability of white matter phenotypes Twins Study, or QNTS) showed additive genetic effects of left and
right brain white matter volume (81%) (Yoon et al., 2010). In neonatal
Strong heritability is a principal tenet in genetic studies relying on twins, heritability of total white matter volume is also very high (85%)
the utility of a phenotype other than the disease phenotype. With re- (Gilmore et al., 2010). While genetic effects on brain structure may
spect to white matter, the neuroimaging phenotype could be either not be as prominent in later-life (Batouli et al., 2013), heritability of
volumetric, based on diffusion-weighted parameters, magnetization white matter volumes toward the end of the human lifespan remain
transfer ratio imaging, related to white matter hyperintensity, or quan- high, and in fact are higher than gray matter or CSF, as shown in the
titative T1 or T2-weighted imaging. Among these neuroimaging mea- older Australian twins study (OATS) which consisted of 154 MZ and
sures, volumetric and diffusion-based parameters have been most 82 DZ individuals, aged 65 or older (Batouli et al., 2014). Therefore, it
studied in schizophrenia. The region (or tract) of interest, the choice appears that white matter volume is a stable and highly heritable phe-
of voxel-wise vs. region-of-interest (ROI) or tract-based analysis, and notype across the lifespan. However, assessment of white matter tissue
the timepoint in the lifespan of the human sample studied can all affect is limited using volumetric approaches, from the perspective of both
the heritability estimate, among other factors. Heritability studies are measurement and visualization. Diffusion-weighted imaging ap-
typically conducted using both monozygotic and dizygotic twins, al- proaches can overcome some of these limitations (Jones, 2008).
though studies using parents and siblings can also be conducted.
2.2. Heritability studies of Diffusion Tensor Imaging (DTI) derived measures
2.1. Heritability studies of white matter volume
Over the past ten years, the field has gravitated toward the use of
One of the first heritability studies to examine white matter volume Diffusion Tensor Imaging, or DTI (Basser et al., 1994) in imaging-
examined 54 monozygotic (MZ) and 58 dizygotic (DZ) twins and 34 of genetics studies of white matter. DTI via measurements of diffusion an-
their full siblings using MRI. The authors estimated that genetic factors isotropy and diffusivity provides an index of tissue microstructure, can
accounted for 88% of the individual differences in white matter volume. localize effects to specific white matter tracts, and can provide informa-
The authors also found that genes influencing gray and white matter tion on tract integrity and potentially on axonal and myelin architec-
overlapped to a large extent and completely determined their pheno- ture. The first DTI heritability study was small: 15 MZ and 18 DZ twin
typic correlation (Baare et al., 2001). Another early study focused on pairs of elderly men were studied from the National Heart, Lung, and
the corpus callosum and showed that high heritability estimates (e.g. Blood Institute Twin study (NHLBI), and FA in the genu and splenium
79% for the midline cross-sectional area) were present through of the corpus callosum were the neuroimaging measures of interest
late adult life (i.e. into the seventh and eighth decades of life) (Pfefferbaum et al., 2001). The authors found that genetic contributions
(Pfefferbaum et al., 2000), supported by another study which found explained 67% and 49% of the total variance in FA respectively in the
even higher heritability (Scamvougeras et al., 2003). One study that ex- genu and splenium. However, a study of 23 MZ and 23 DZ twin pairs
amined MZ twins found very high correlation coefficients (r N 0.90) for from the Queensland Twin Imaging Study (QTIMS) showed that genetic
white matter volumes in MZ twin pairs, a value much higher than that factors explained 75–90% of the variance in FA in almost all white mat-
present in matched controls (White et al., 2002). In children, additive ter regions (Chiang et al., 2009b). Closer to the beginning of the lifespan,
genetic effects accounted for a substantial proportion of the variability the same group examined 705 twins (129 MZ and 170 DZ) and their sib-
in white matter volume (Wallace et al., 2006). When the above initial, lings aged 12 to 29 from QTIMS, and heritability of FA across white mat-
small studies are taken together, one could conclude that white matter ter tracts using a voxel-wise approach was calculated (Chiang et al.,
volumes are highly heritable across the lifespan. 2011b). The authors also considered effects of age, sex, IQ, and socioeco-
Another approach is to study MZ twins discordant for disease to as- nomic status. They found that in those individuals with above average
sess genetic and environmental risk factors. A small study of 11 MZ and IQ, heritability was greater than 80% in thalamus, genu, posterior inter-
11 same-gender DZ twin pairs discordant for schizophrenia found de- nal capsule and corona radiata, while in those with below average IQ, FA
creased white matter volume in discordant twin pairs compared with heritability dipped down to approximately 40%. They also found that in
healthy twin pairs, particularly in the MZ twin pairs. A decrease in adolescents, 70–80% of the variation in FA was due to genetic factors,
gray matter was found in the patients compared with their co-twins while in adults only 30–40% of the variation in FA was similarly attribut-
and compared with the healthy twins. The authors concluded that the able. Finally, male sex was also associated with higher heritability.
decrease in white matter volume reflected the increased genetic risk While this study had a very impressive sample size, the authors used
to develop schizophrenia, whereas the decreases in grey matter volume smoothing in their postprocessing routine and acquired anisotropic
were related to environmental risk factors, highlighting the importance voxels, both of which can confound results (Jones et al., 2002; Jones
of genetic contributions to white matter structures in schizophrenia et al., 2005). Another study from the same sample assessed n = 374
(Hulshoff Pol et al., 2004). The same group, appearing to use the same healthy adults (60 MZ pairs, 45 DZ pairs and 164 mixed sex DZ twins
twin pairs at longitudinal 5-year follow-up reported interesting data ex- and siblings) to understand genetic effects on white mater asymmetry.
ploring percentage volume change over time. The authors found signif- Modest genetic effects (10–37% depending on the structure) were
icant additive genetic influences on the correlations between found. Sex differences in asymmetry were also present (Jahanshad
schizophrenia liability and progressive brain volume changes; however, et al., 2010).
progressive change in white matter did not appear to have genetic influ- An even younger sample of MZ and DZ twins consisting of 9 year old
ence over and above that of gray matter (Brans et al., 2008). children (total n = 185) from the Netherlands Twin Registry (NTR) and
This manuscript does not review all structural MRI studies that have Utrecht Medical Centre Twin Sample (UMCTS), used DTI with fiber
assessed the heritability of white matter volumes. However, recent tractography to assess heritability of FA, radial diffusivity (RD), axial or
studies publishing on large samples can help provide more conclusive longitudinal/axial diffusivity (AD), and of magnetization transfer ratio

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx 3

Table 1
Details of studies examining association of genome-wide significant loci for schizophrenia with white matter phenotypes.

Gene White matter phenotype Sample Finding Reference

ZNF804A T1-weighted MRI N = 39 healthy controls (Caucasian) Association with total white matter Lencz et al. (2010)
Total white matter volume and
lobar white matter volume
ZNF804A DTI tractography–Fronto-temporal N = 62 healthy controls (Caucasian) No association Voineskos et al.
and interhemispheric white matter FA (2011b)
ZNF804A DTI–tract based spatial statistics (TBSS)–FA N = 53 healthy controls No association Sprooten et al.
throughout white matter skeleton; ROI and N = 84 healthy controls (2012)
tractography N = 50 unaffected relatives of bipolar
subjects (Caucasian)
ZNF804A T1-weighted MRI N = 69 healthy controls Genotype by diagnosis interaction Wei et al. (2012)
White matter density N = 80 people with schizophrenia (Risk allele with higher WM density in scz,
(Han Chinese) and lower in healthy in left prefrontal lobe)
ZNF804A DTI–mean FA analysis of lobar N = 75 controls Genotype by diagnosis interaction Kuswanto et al.
white matter N = 125 people with schizophrenia (FA of parietal, temporal, cingulate WM) (2012)
regions (Chinese)
ZNF804A T1-weighted MRI N = 198 controls Risk allele carriers in both group had Wassink et al.
Lobar white matter volumes N = 335 people with a schizophrenia larger WM volumes (2012)
spectrum
disorder (Caucasian)
ZNF804A DTI–TBSS–FA, MD, AD, RD throughout N = 69 controls No association Wei et al. (2013)
white matter skeleton N = 100 people with schizophrenia
(Han Chinese)
CACNA1C DTI–TBSS–predefined hippocampal ROI FA N = 100 controls (Caucasian) Risk allele associated with reduced right Dietsche et al.
hippocampal FA (2014)
Neurogranin T1-weighted MRI N = 263 controls No association Ohi et al. (2012)
VBM N = 99 people with schizophrenia
(Japanese)
Neurogranin T1-weighted MRI N = 140 controls No association Rose et al. (2012)
VBM (Caucasian)
ANK3 DTI–volume of interest in anterior limb of N = 88 controls (Caucasian) Lower FA and AD of anterior limb of internal Linke et al. (2012)
internal capsule followed by tractography capsule in those with risk genotype
MHC T1-weighted MRI N = 173 controls No association Agartz et al. (2011)
Lobar white matter volumes N = 247 schizophrenia spectrum and
bipolar spectrum disorder (Caucasian)
MHC T1-weighted MRI N = 892 controls (Caucasian) No association Walters et al.
ZNF804A Voxel-based morphometry (VBM) (2013)
Cousijn et al. (2012)
CSMD1 T1-weighted MRI N = 150 controls No association Rose et al. (2013)
VBM (Caucasian)
CNNM2 T1-weighted MR N = 159 controls No association Rose et al. (2014)
VBM N = 39 controls
(Caucasian)
MIR137 DTI–TBSS–Whole Brain white N = 121 controls Risk allele homozygotes with schizophrenia had Lett et al. (2013)
matter skeleton mean FA N = 92 people with schizophrenia lower white matter FA compared to protective
(Caucasian) allele carriers
VRK2 T1-weighted MRI N = 286 Association of risk allele with total Li et al. (2012)
VBM Han Chinese white matter volume
RELN DTI–TBSS followed by FA N = 93 No association Tost et al. (2010)
measurement of specific ROIs Caucasian

(MTR) (Brouwer et al., 2010). The authors studied the genu and heritability of 0.92. Other tracts with high heritability included the unci-
splenium of the corpus callosum, and left and right uncinate and supe- nate fasciculus (0.62), genu, body, and splenium of corpus callosum
rior longitudinal fasciculi. Significant genetic influences could not be (.72, .69, .68), and cingulate gyrus (.68). Overall FA heritability, unlike
established for FA. However, genetic factors significantly explained var- RD heritability, did not increase in regions with higher FA. Extending
iation in both longitudinal/axial and radial diffusivity (especially the lat- out toward the end of the lifespan, a family study (San Antonio Family
ter), as well as MTR. Both MTR and radial diffusivity heritability values Heart Study or SAFHS) of 467 healthy individuals, ranging in age from
were highest at the superior longitudinal fasciculus. However, a study 19 to 85, found significant genetic contributions to FA and radial diffu-
of neonatal twin pairs (Geng et al., 2012) found significant heritability sivity (but not longitudinal diffusivity) following TBSS and tract-based
across all three average diffusion parameters studied over whole brain calculations of diffusion parameters with the help of the JHU atlas
white matter. FA had the highest heritability (.60) followed by axial (Kochunov et al., 2010). The authors used 417 markers spaced at ap-
(longitudinal) diffusivity (.57), and radial diffusivity (.53). There was proximately 10-centiMorgan intervals across 22 autosomes. Whole
considerable variation across white matter regions or tracts, which brain FA showed the highest heritability (.52) among all diffusion pa-
might help explain the lack of findings from the previous study of rameters followed by radial diffusivity (.37). The genetic factors that
9 year olds, where only a small number of tracts were examined. In produced increases in radial diffusivity were also responsible for de-
the neonatal study, among white matter tracts, FA heritability was creases in FA. However, there were no significant age by sex or age-
highest at left posterior corona radiata (.82), with highest RD heritabil- squared by sex covariates.
ity (all N 0.80) in left middle and inferior occipital gyrus, right pre- Others have looked at a combination of white matter FA and cogni-
cuneus, right inferior temporal gyrus, deep white matter regions of tive performance in heritability studies. In one of the same studies brief-
right sagittal stratum, right splenium of corpus callosum, and left poste- ly described above (Chiang et al., 2009b), in 92 identical and fraternal
rior corona radiata. For AD, right posterior corona radiata showed twins (23 pairs of each), the anatomical profile of correlations between

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
4 A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx

white matter FA and full-scale, verbal, and performance intelligence (QTL) genomic strategy. This stands in contrast to several studies
quotients was examined. In addition to showing relatively strong corre- using volume of subcortical structures as a quantitative trait, initially
lations of white matter FA with intellectual performance, the authors using the ADNI dataset (e.g. hippocampal volume (Potkin et al., 2009),
implicated the same genes as mediating the correlation between IQ caudate volume (Stein et al., 2011), and now through the ENIGMA
and white matter integrity, using an approach that whereby the IQ of working group (e.g. for hippocampal volume (Bis et al., 2012; Stein
one twin was predicted with high precision from the white matter in- et al., 2012). The study by Stein et al. in n = 21,151 healthy individuals
tegrity of the other twin. In a study using unaffected relatives, examin- revealed significant association of an intergenic variant, rs7294919 with
ing 22 participants at genetic risk for schizophrenia, 23 people with hippocampal volume. The first study using a DTI-based phenotype as a
schizophrenia, and 37 non-psychiatric controls, both reductions and in- QTL analysis in 467 healthy individuals recruited from randomly-
creases in FA were found in different regions in the high-risk partici- ascertained pedigrees of extended families found possible linkage at re-
pants in relation to the comparison participants (Hoptman et al., gions 15q25 and 3q27 (LOD scores of 2.36 and 2.24 respectively). The
2008). In a larger study, including the same participants both white first region contains the major depressive disorder 2 gene as well as
matter FA and cognitive performance were found to be heritable the semaphorinprecursor 4B gene (which is involved in axonal exten-
(Bertisch et al., 2010). sion and growth). The second region includes the adiponectin gene
Meta-analytic studies and multi-site analyses support the heritability and serotonin receptor-like 3D and 3E genes (Kochunov et al., 2010).
of white matter FA. A recently published meta-analysis (Blokland et al., More recently global FA (derived from a principal components analysis
2012), based on two studies and a small number of twin pairs found of 12 white matter tracts) was used as a quantitative phenotype in a
tight confidence intervals for FA based measures with moderate heritabil- GWAS in elderly individuals from the Lothian Birth Cohort (n = 668).
ity values for superior longitudinal fasciculus, and genu and splenium of No single SNP achieved GWAS significance; although there was sugges-
the corpus callosum. A much larger ‘mega-analysis’, a specific data tive evidence (p values of 10− 6) at the ADAMTS18 gene, related to
pooling strategy, of 5 samples (4 twin samples and 1 family sample) tumor suppression and homeostasis and the LOC388630 gene, which
from the ENIGMA working group, using a total sample size of 2,248 indi- is of unknown function (Lopez et al., 2012). The ENIGMA-DTI working
viduals found robust and consistent heritability values for FA of all white group is now positioned, however, to conduct a genome-wide analysis
matter tracts examined except for the cortico-spinal tract, with heritabil- of white matter FA in many thousands of individuals, and other working
ity values mainly ranging from 0.4 to 0.7. The investigators combined groups have also combined DTI and genetic data numbering into the
much of the data from some of the samples described in the present re- thousands (Blokland et al., 2013). Therefore, with this considerable in-
view along with others. One important finding was that datasets with crease in statistical power, new genes associated with white matter FA
smaller numbers of subjects were more likely to produce heritability esti- at genome-wide significant levels will almost certainly be discovered.
mates not representative of the pooled trend. Overall, pooled heritability
estimates across the brain were shown to be regionally uniform with ad- 4. Candidate genes and gene systems: A Focus on Oligodendrocytes,
ditive genetic factors explaining over 50% of inter-subject variance in FA Axons, and Myelin, Neurodevelopmental and Neurotrophic Path-
values (Kochunov et al., in press). The ROI template used in this study ways and Relationship with White matter phenotypes
was shown to be reliable for integrating multiple datasets, and supported
preliminary heritability results in a subset of n = 400 individuals from the Genes involved in oligodendrocyte, myelin, and axonal development
same twin and pedigree samples (Jahanshad et al., 2013). A multi-site col- and maintenance were logical early choices in candidate gene studies of
laborative neuroimaging and genetic effort known as the B-SNIP (Bipolar white matter phenotypes. In particular, genes of the neuregulin1-
Schizophrenia Network on Intermediate Phenotypes) consortium recent- tyrosine kinase receptor ErbB4 (NRG1-ErbB4) gene system and oligo-
ly studied 513 participants with schizophrenia, psychotic bipolar disorder, dendrocyte/myelin (OM) system genes were of high interest, and
their first degree relatives, and healthy controls. The authors found that expression of these systems is coordinated (Georgieva et al., 2006).
both disease groups showed lower FA than comparison subjects in multi- OM genes are predominantly expressed in oligodendrocytes and direct-
ple regions, with more marked differences in schizophrenia. Furthermore, ly involved in their myelination (initiation, deposition, compaction, and
similar, but smaller effects were seen in schizophrenia relatives, with a maintenance) (Davis et al., 2003) and trophic support (Segal et al.,
continuous FA decrease from healthy subjects to relatives to probands. 2007), axonal support (McTigue and Tripathi, 2008), and axo-glial inter-
Overall, the authors found that white matter FA was highly heritable in actions (Pernet et al., 2008). NRG1 plays an important role in cortico-
their sample, supporting its value as a potential endophenotype cortical myelination during neurodevelopment (Chen et al., 2006),
(Skudlarski et al., 2013). When combining samples into larger mega- or and disruption of the NRG1-ErbB4 pathway in oligodendrocytes in ani-
meta-analyses, the use of tractography data generated from different pro- mal models leads to alteration of the myelin sheath of major white mat-
cessing methods (e.g. probabilistic vs. deterministic) might confound ter tracts, reduced conduction velocity, and cognitive changes (Roy
results. One approach to circumvent this issue has been described by et al., 2007). Effects of NRG1 variants on white matter integrity have
the ENIGMA working group through the use of mega-analysis via utiliza- been shown in healthy controls in the anterior limb of the internal cap-
tion of a common ROI-based template. The tradeoff, however, is that more sule and subcortical medial frontal white matter respectively (McIntosh
sophisticated methods of white matter analysis are not presently used in et al., 2008; Winterer et al., 2008), and in schizophrenia patients in the
large mega or meta-analytic imaging-genetic studies. anterior cingulum (Wang et al., 2009). ErbB4 demonstrates coordinated
The unifying result of heritability studies with large sample sizes is expression with OM genes, oligodendrocyte transcription factor-2
that white matter phenotypes are heritable. Both white matter volume (OLIG2) and 2′,3, cyclic nucleotide 3′-phosphodiesterase (CNP), in post-
and white matter FA are heritable. It appears that heritability may vary mortem brain (Georgieva et al., 2006), and a SNP in this gene was asso-
depending on the white matter tract under study; however, moderate ciated with integrity of the uncinate fasciculus (Konrad et al., 2009), as
or high heritability appears to be the norm for nearly all white matter well as with the anterior limb of the internal capsule (Zuliani et al.,
tracts, with the exception possibly, of the corticospinal tract. Such herita- 2011) in healthy individuals. OM genes, i.e. CNP, OLIG2, myelin associat-
bility appears to be relatively consistent throughout the lifespan, although ed glycoprotein (MAG), myelin oligodendrocyte glycoprotein (MOG),
it might be slightly higher during the neonatal period and childhood. and transferrin (TF), an iron gene, came to attention following the first
postmortem transcriptomic studies (Hakak et al., 2001) in schizophre-
3. White matter phenotypes as quantitative trait loci nia. OM genes code for proteins that influence the microstructural com-
ponents of white matter tracts that form the main barriers to water
There are surprisingly few studies addressing the underlying genetic diffusion indexed using DTI. Iron genes also play an important role in
architecture of white matter structure using a quantitative trait locus myelination, and most of the brain's iron is found in oligodendrocytes,

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx 5

which maintain iron homeostasis in the brain. A recent study examined psychiatric genetics consortium (PGC) have identified 22 risk loci for
genotypic variation in NRG1, MOG, and CNP with brain structure using schizophrenia (Ripke et al., 2013). Risk loci identified at GWAS-
volumetric approaches, and showed that an NRG1 SNP and a MOG significant levels (typically a p value of 5 × 10−8) carry a degree of con-
SNP were associated with white matter volume (in schizophrenia pa- fidence in the genetic association result that was not present prior to the
tients and healthy controls respectively) (Cannon et al., 2012). Gene advent of GWAS. The first such variant, rs1344706, situated within the
variants in TF and HFE were examined in association with white matter zinc finger transcription factor 804A, or ZNF804A gene, was identified
FA in 574 individuals (twins and siblings) and revealed a significant as- as a risk locus with a myelin/oligodendrocyte transcription factor bind-
sociation of the HFE rs1799945 SNP with FA in the external capsule, and ing site (Riley et al., 2010). The first imaging-genetics study of this
this allele was associated with decreased levels of serum transferrin GWAS variant showed association in healthy individuals with fronto-
(Jahanshad et al., 2012). FA in cingulum and in the superior longitudinal temporal and prefrontal interhemispheric connectivity phenotypes
fasciculus was also associated with serum transferrin. using functional neuroimaging (Esslinger et al., 2009); thus it might
In animal models, the neurodevelopmental disrupted-in- have been expected that the same variant would be associated with
schizophrenia 1 (DISC1) gene interacts with the OLIG2 gene, as does the corresponding fronto-temporal and interhemispheric white matter
NRG1 to influence oligodendrocyte development (Wood et al., 2009). connections; however, in an investigation using tract-based FA pheno-
In humans, the DSCI1Ser704Cys polymorphism has been associated types, no such association was found, although there was a relationship
with decreased FA in prefrontal white matter of 22 Cys allele carriers with cortical thickness in fronto-temporal regions (Voineskos et al.,
vs. 86 Ser/Ser individuals (Hashimoto et al., 2006), 10 Cys/Cys individ- 2011b). Just prior to that result, others found an association of this var-
uals showed decreased white matter FA throughout much of the brain iant with volumetric measures of gray and white matter (Lencz et al.,
vs. 38 Cys/Ser and 39 Ser/Ser healthy individuals (Sprooten et al., 2010). Further, another group did not find association of the ZNF804A
2011). In a study of n = 278 healthy individuals decreased network ef- SNP and white matter FA, using TBSS (Sprooten et al., 2012) in healthy
ficiency was found in Cys allele carriers compared to Ser homozygotes individuals. Studies of this ZNF804A variant in relation to white matter
(Li et al., 2013), measured using graph theoretical approaches applied phenotypes in patients with schizophrenia have not shown consistent
to FA of fiber tracts throughout the brain. Neurotrophic genes have results. One study found a genotype-by-diagnosis interaction on white
also been investigated in relation to white matter phenotypes, particu- matter density in the left prefrontal lobe (Wei et al., 2012), whereas an-
larly the brain derived neurotrophic factor (BDNF), which interacts other by the same group found no association with ‘white matter integ-
with the p75 neurotrophin receptor (Cosgaya et al., 2002), which itself rity’, on FA, MD, AD, or RD in either schizophrenia patients or healthy
is an intimate binding partner of MAG, MOG, and Nogo to influence controls (Wei et al., 2013) using TBSS. However, a large study found a
myelination (Wang et al., 2002). Although there are negative findings significant main effect association of this SNP, as well as an association
in healthy individuals (Montag et al., 2010), many more studies have in patients with schizophrenia, on total white matter, and particularly
shown association of the val66met with white matter microstructure frontal white matter volume (Wassink et al., 2012), with another
(Kennedy et al., 2009; Chiang et al., 2011a). In the Chiang et al. study, study (Kuswanto et al., 2012) demonstrating a genotype-by-diagnosis
in 455 healthy subjects, composed of twins and families, the val- interaction in FA of white matter in parietal, temporal, and cingulate re-
variant significantly modulated the association between FA and a mea- gions. Yet an even larger study of n = 892 individuals found no associ-
sure of intelligence in the splenium of the corpus callosum. The ation of this SNP with white matter volume (Cousijn et al., 2012).
val66met variant has also been shown using data from 258 healthy The L-type voltage-dependent calcium channel CAv1.2 (alpha-1C
adult twins and their non twin siblings to explain nearly all the total var- subunit) (CACNA1C) gene was initially discovered in bipolar GWAS
iance in FA in the posterior cingulate gyrus (Chiang et al., 2009a). In the and then subsequently was also found to be associated with schizophre-
Kennedy et al. study, no genotype-related differences were observed in nia at a genome-wide significant level (Green et al., 2010). Although
the genu of the corpus callosum, while the splenium demonstrated most of the imaging-genetics studies of this gene have used fMRI para-
genotype-dependent age-effects. Another study showed genotype-by- digms (Bigos et al., 2010), a recent DTI study showed that the CACNA1C
age interactions for FA of white matter tracts connecting to the medial SNP (rs1006737) was associated with FA in the right hippocampal for-
temporal lobe (Voineskos et al., 2011a). The NTRK1 gene (also known mation using a hypothesis-driven approach (Dietsche et al., 2014).
as TRKA) encodes a high-affinity receptor for NGF, a neurotrophin in- The neurogranin gene is another schizophrenia risk gene discovered
volved in nervous system development and myelination which has by GWAS (Stefansson et al., 2009). The neurogranin protein is a post-
been associated with risk for schizophrenia. A SNP at NTRK1, rs6336 synaptic protein that is expressed in the human brain and involved in
was recently associated with FA in a study of 391 young healthy adults the regulation of calmodulin availability in neurons (Huang et al.,
(Braskie et al., 2012). The same group also found evidence for associa- 2004). A small number of MRI studies have been completed in relation
tion of NTRK3 variants (the NTRK3 gene is also known as TRKC) to this gene, although none have used DTI. Two studies examined the ef-
which is implicated in oligodendrocyte and myelin development, with fect of this neurogranin SNP (rs12807809) on white matter volume, but
white matter FA in a widespread manner (corpus callosum, inferior lon- no association was found (Ohi et al., 2012; Rose et al., 2012). The anky-
gitudinal fasciculus) (Braskie et al., 2013). When taken together these rin 3 (ANK3) gene has genome-wide supported evidence for association
results suggest that variation in OM genes, the NRG1-ErbB4 system, with both schizophrenia and bipolar disorder (2011). ANK3 is a mem-
and neurotrophin genes is associated with white matter microstruc- ber of the ankyrin family of proteins that link integral membrane pro-
ture. However, given the main effects shown by these gene variants, teins to the underlying spectrin-actin cytoskeleton and is expressed in
it is not entirely clear whether these associations are relevant to CNS. ANK3 also directs the localization of potassium channel proteins
schizophrenia, and may not be disease specific. Furthermore, only to the nodes of Ranvier (Judy et al., 2013). Healthy carriers of the
a subset of studies included schizophrenia patients, although those ANK3 rs10994336 risk genotype were shown to have lower FA and lon-
that did suggest a main effect of these variants on microstructural gitudinal diffusivity in the anterior limb of the internal capsule (Linke
integrity of white matter, rather than one more prominent in either et al., 2012).
patients or controls. Perhaps surprisingly, while the major histocompatability complex
(MHC) region provides the strongest statistical association with schizo-
5. GWAS variants and relationship with white matter phenotypes phrenia in GWAS studies (Stefansson et al., 2009; Ripke et al., 2013),
few neuroimaging studies have been completed examining association
Since the first schizophrenia GWAS published in 2008 (O'Donovan with GWAS SNPs (e.g. rs6904071) within MHC and brain structure.
et al., 2008), collaborative initiatives have provided increasing sample None have been published with a focus on white matter imaging pheno-
sizes and accompanying power, such that the latest results of the types, although two large volumetric studies (Agartz et al., 2011;

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
6 A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx

Walters et al., 2013) did explore brain volumes and no association with 6. Rare variants and highly penetrant mutations
white matter volume was reported. Another GWAS SNP, part of the
transcription factor 4 (TCF4) gene, was investigated in relation The 22q11 deletion syndrome is reviewed in another manuscript in
to neuroimaging phenotypes, but not white matter phenotypes this special issue. Genome-wide approaches also can identify genomic
(Wirgenes et al., 2012). TCF4 may play an important role in regulation deletions and duplications, i.e. copy number variation. Ideally a number
of oligodendrocyte differentiation and myelination (Emery, 2010) and of subjects with a deletion or duplication in the same genomic region
thus variation in this gene may influence white matter phenotypes in can be collected for study, i.e. for ‘bottom-up’ phenotyping. However,
schizophrenia either through TCF4 or through interaction with other since it is so challenging to identify a sufficient number of individuals
genes. The rs10503253 SNP within the CUB and Sushi multiple with the same genomic change, a SNP with functional relevance in that
domains-1 (CSMD1) gene, a tumor suppressor gene has also been iden- region could instead be used to understand effects of variation on white
tified as genome-wide significant for schizophrenia (2011). A voxel matter phenotypes. For instance, deletions and duplications of genes in
based morphometry investigation of white matter volume showed no the neurexin superfamily increase risk for schizophrenia (O'Dushlaine
association (Rose et al., 2013). This gene is thought to be involved in et al., 2011). The contactin-associated protein-2 (CNTNAP2) gene, a
complement control (Havik et al., 2011). The same group recently pub- member of the neurexin superfamily is on chromosome 7 and has
lished on the cyclin M2 (CNNM2) gene (Rose et al., 2014). The authors shown associations with autism and schizophrenia. The rs271026 SNP
did not find any association between the rs7914558 SNP and white mat- from CTNNAP2 was associated with FA in the uncinate fasciculus
ter volume. CNNM2 is a divalent metal cation transporter and may be (Clemm von Hohenberg et al., 2013). Another SNP in the same gene,
part of a proteomic network centered on CAv2. rs779475, was associated with FA in anterior thalamic radiation in fe-
Among schizophrenia GWAS genes, the microRNA-137 (MIR137) males, and with FA of rostral fronto-occipital fasciculus in males. Similar-
gene may be of particular importance. A SNP just upstream of this gene, ly, a SNP in neurexin1, rs1045881, located in the 3′ untranslated region of
rs1625579, demonstrated genome-wide significance and was the ‘top the gene, and serving as a microRNA binding site, was associated with
hit’ after the MHC region in a recent GWAS that also included meta- frontal lobe white matter volume in healthy individuals (Voineskos
analysis (Ripke et al., 2013). MIR137 may be of particular importance, be- et al., 2011c). It remains unclear whether ‘tagging’ a genomic deletion
cause it appears to relate intimately to other schizophrenia GWAS genes, with a common genetic variant is a tractable approach; however due to
possibly suggesting a central or prominent role for this gene in disease ex- the high odds ratios conferred by a structural genetic change within
pression. Of note, this microRNA has as its targets schizophrenia GWAS these susceptible regions, further exploration of susceptible genes in rela-
genes CSMD1, C10orf26, CACNA1C, and TCF4 (Kwon et al., 2013), as tion to white matter phenotypes is definitely warranted.
well as ZNF804A (Kim et al., 2012). Thus, MIR137 may act through
other schizophrenia GWAS risk genes. Since microRNAs regulate transla- 7. Multi/polygene approaches
tion of other mRNAs they may affect phenotypic expression of disease.
Accordingly, this variant has been found to influence age-at-onset, hippo- Since several risk loci have only been discovered very recently, some
campal and lateral ventricle volume, and whole brain white matter FA in have not yet been examined in relation to neuroimaging phenotypes.
people with schizophrenia, thus serving as an explanation for the hetero- However, collaborative efforts, and a transition to polygenic, rather
geneity of phenotypic expression of disease (Lett et al., 2013). Notably, than single variant-at-a-time investigations will likely change our
this variant was not significantly associated with structural neuroimaging understanding of the effects of proven risk loci on white matter pheno-
phenotypes in healthy individuals in the same study. Consistent associa- types. Multivariate approaches such as independent components analy-
tion of this variant with structural brain phenotypes in patients was sis (ICA) and partial least squares (PLS) can be used to simultaneously
shown across the adult lifespan, where risk allele homozygotes had signif- assess the relationship between a group of SNPs and a number of neuro-
icant impairments in brain structure from the first episode of illness. This imaging phenotypes. One recent paper (Voineskos et al., 2013a)
study demonstrates that the consequences of risk variation may differ in assessed the relationship of SNPs in oligodendrocyte and myelin genes
patient and healthy control groups, supported by a recent functional neu- with FA of several white matter tracts both in healthy people and in
roimaging study (van Erp et al., 2014), with another study investigating people with schizophrenia. Gene variants in MAG and CNP were
controls only (Mothersill et al., 2013). Furthermore, from a clinical stand- associated with fronto-temporal and interhemispheric white matter
point, the finding by Lett et al. raises the potential for disease prognostica- tract FA. Furthermore, relationships of some of these SNPs with
tion and staging among patients with schizophrenia using a combination neurocognitive performance were mediated through white matter FA.
of imaging and genetics. Overall, however, neuroimaging studies of A small study using parallel ICA explored the relationship of 367 SNPs
GWAS variants leave much to be desired. Many studies, despite their re- with optimized VBM data from MRI scans, but focused only on gray
cent publication dates, are using basic white matter volumetric pheno- matter. No white matter results were reported (Jagannathan et al.,
types rather than DTI-based phenotypes, and few studies are including 2010).
patients with schizophrenia. Perhaps the most straightforward approach for examining effects of
In many ways the GWAS field in schizophrenia is still in its earliest several gene variants was demonstrated in a recent study. The authors
days. Most studies have been completed in Caucasians only. Sex effects pre-selected one SNP from each of 6 genes (Kohannim et al., 2012),
are rarely studied. Epistatic interactions between or among genes have which had previously been associated with white matter FA (BDNF,
not yet been reported. Therefore, clarification and translatability to peo- NTRK1, ErbB4, Clusterin (CLU) (Braskie et al., 2011), HFE, and COMT
ple of other ethnic backgrounds and sex effects will be important future (Thomason et al., 2010), and in this case used FA of the corpus callosum
directions, and subsequently can inform studies combining brain imag- as their main phenotype. The authors found that 5 of the 6 SNPs exam-
ing and genetics. For instance, in a Han Chinese sample, a genome-wide ined explained nearly 6% of the variability in mean callosal FA, using a
significant SNP, from the vaccinia-related kinase (VRK2) gene, linear mixed effects model, and also displayed extensive, significant ef-
rs2312146 was associated with white matter volume, and with up- fects on 82% of the volume of the corpus callosum. In a somewhat differ-
regulation in schizophrenia patients (Li et al., 2012). With respect to ent approach, from a sample of 472 twins and their non-twin siblings
sex, a GWAS study identified that the rs7341475 SNP in the reelin (Chiang et al., 2012), the authors used their sample to identify patterns
gene (RELN), was associated with schizophrenia only in women of brain regions where white matter FA was under strong and relatively
(Shifman et al., 2008). However, a comprehensive association study of homogeneous genetic control. Then, they performed GWA from a num-
this SNP with brain structure (including DTI), function, and postmortem ber of ROIs to identify SNPs associated with FA in these regions. SNP
tissue expression revealed no association with any of these phenotypes pairs were then discovered that tended to jointly affect brain regions.
either via main effect, or gene by sex interaction (Tost et al., 2010). Network topology analysis was then applied to this SNP association

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx 7

matrix and a SNP network that influences white matter integrity was Little work has been done in this area to date, but the emergence of the
isolated. Hub SNPs in the network interacted with each other in their ef- ENIGMA working group, and other working groups (e.g. B-SNIP,
fects on white matter integrity, and possibly influenced intellectual per- IMAGEN), means that new genome-wide hits for association with
formance. SNPs from genes within a number of networks were white matter FA are imminent. Similarly, polygenic analyses are now
identified including cell adhesion, synapse, molecular functions, nerve possible, and results using such an approach are starting to emerge. Lit-
fiber integrity, and others. tle to no gene–gene interaction work has been done, unlike with fMRI
Finally, another approach that takes the effects of multiple genetic for instance (e.g. (Callicott et al., 2013), and this is an important area
variants into consideration is the polygenic risk score, which is gaining of future investigation using white matter phenotypes. Acceleration in
traction across different types of genetic studies of psychiatric disorders. bioinformatic approaches to functionally annotate the genome
In a study examining shared susceptibility for schizophrenia and brain (Bernstein et al., 2012; Birney, 2012) (e.g. ENCODE) facilitates opportu-
volumes (Terwisscha van Scheltinga et al., 2013), odds ratios for nities to understand the role of a risk locus at a molecular level. Publical-
genome-wide SNP data were calculated from a sample of 8690 patients ly available databases (e.g. BRAINCLOUD) (Colantuoni et al., 2011) offer
and 11831 control subjects. This information was used to calculate indi- the opportunity to explore putative mechanistic effects of risk loci via
vidual polygenic schizophrenia (risk) scores in an independent sample association with mRNA expression. The developing Allen Brain Atlas
of 152 patients and 142 controls with MRI data. The polygenic risk documents gene expression in specific brain regions (Shen et al.,
score was significantly associated with white matter volume equally in 2012)). Bridging the gap from gene to brain is now at our finger-tips.
patients and control subjects. The polygenic schizophrenia score was spe- Clinical translation is the ultimate goal. Using polygenic models of
cifically associated with reduced white matter volume but did not explain risk prediction combined with neuroimaging, the field can begin mov-
variance in gray matter volume. While 14,751 SNPs modulated disease ing toward disease risk prediction for high-risk adolescents or family
risk (i.e. explained variance in disease status), a smaller subset of 2,020 members in a clinical setting. However, prior to such tangible progress,
SNPs explained most of the variance in white matter. From the set of further clarification and work is needed to definitively establish the ‘in-
2020 SNPS, 186 showed most evidence for association with white matter termediate findings’ in ultra high risk subjects, such that quantitatively-
volume. Genes of these 186 SNPs were groupable into seven clusters, based disease staging (i.e. preclinical vs. clinical) can occur accurately.
with an immunoglobulin-like cell-adhesion molecule cluster as enriched At the beginning of evident clinical disease expression (e.g. the first ep-
compared with the reference database. isode of psychosis), new findings suggest that imaging-genetic data
from carefully characterized patient samples could be used to aid in
8. Conclusion prognosis (Lett et al., 2013). However, it will be useful to further clarify
genetic effects at different timepoints in the lifespan, such that investi-
A major challenge but also source of potential progress, is the iden- gators can appropriately ‘hone in’ to use imaging-genetics to address
tification of the genetic risk factors of disease as they pertain to the questions related to identifying disease risk (in childhood and adoles-
brain structures or circuitry vulnerable in schizophrenia (Akil et al., cence) and disease heterogeneity (in adulthood). Another important
2010). With heritability established, white matter FA is a reasonable area of clinical application is treatment innovation. With research prior-
phenotype to study in schizophrenia, since many studies have found ities at the National Institute of Mental Health moving toward a Re-
FA reductions in patients, and intermediate FA reductions are present search Domain Criteria (RDoC) style approach (Insel et al., 2010),
in first-degree relatives. Most genetic association studies of white mat- brain circuits are becoming foci for therapeutic innovation. One could
ter FA have used healthy participants only. Early studies used small envision a scenario in which, either prior to psychotic illness onset
sample sizes, and only recently are large, and sometimes very large (e.g. a high-risk state), or during illness, certain genetic risk carriers
(through the combination of data from different sites) samples being are administered a specific treatment. If the gene (or genes) in those in-
used. Candidate gene studies were initially conducted, and the focus dividuals predisposes to vulnerability or impairment in a specific circuit
has now moved to genetic variants with genome-wide significant evi- that is relevant to a specific behavioral impairment, e.g. prefrontal inter-
dence for a role in schizophrenia. No specific neuroanatomic pattern hemispheric impairment and working memory impairment, then treat-
of association has emerged with any one of these genes, possibly due ment response could be assessed via early changes in circuit structure or
to some studies hypothesizing (and examining) association with only circuit function. The addition of genetically mediated differences in cir-
one or a small number of white matter tracts, while others have used cuit structure and function will be an important component of such in-
a ‘brain-wide’ approach with existing analytic tools such as TBSS. novation. Overall, this field is moving rapidly, and replicable discoveries
Other white matter imaging techniques such as Magnetization Transfer are starting to be made. Given that schizophrenia is a brain disease, and
Ratio, which can better index myelin integrity compared to DTI, have that genomic discoveries are beginning to translate into therapeutic
been underutilized in imaging-genetics studies. Advanced diffusion im- progress in other areas of medicine, it should only be a short matter of
aging techniques such as diffusion spectrum imaging, which can pro- time before imaging-genomics does the same for schizophrenia.
vide superior neuroanatomic characterization of brain white matter,
will be an important area of future investigation that the field of Role of the funding source
imaging-genetics can benefit from. None of the funders played any role in the design, conceptualization, or writing of the
Only a small number of imaging-genetic GWAS variant–white mat- manuscript.
ter studies have examined association in patients. This is an important
issue, because gene systems may predict neural variation and behaviour Contributors
(cognition) in a different manner in a disease population compared to a Dr. Voineskos designed, conceptualized, and wrote the manuscript.
healthy population. That is to say that utilization of the intermediate
phenotype approach in a healthy population does not conclusively Conflict of interest
prove how a gene variant might predict brain structure or function in Dr. Voineskos has no conflicts of interest to disclose. He would like to acknowledge
an individual with disease. Using imaging-genetic strategies in patient funding from the Canadian Institutes of Health Research, Ontario Mental Health Founda-
tion, Brain and Behavior Research Foundation, National Institute of Mental Health
populations should help provide biological clues regarding the consid-
(R01MH099167), and the Koerner, Labatt, and Kimel Families via the CAMH Foundation.
erable heterogeneity of disease expression among people with schizo-
phrenia. (Gottesman and Gould, 2003; Meyer-Lindenberg and
Weinberger, 2006; Voineskos et al., 2013b). The collection of large References
datasets now means that QTL approaches are now possible, where Agartz, I., Brown, A.A., Rimol, L.M., Hartberg, C.B., Dale, A.M., Melle, I., Djurovic, S.,
genome-wide association with DTI-based phenotypes can be examined. Andreassen, O.A., 2011. Common sequence variants in the major histocompatibility

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
8 A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx

complex region associate with cerebral ventricular size in schizophrenia. Biol. Psychi- Chiang, M.C., Barysheva, M., Toga, A.W., Medland, S.E., Hansell, N.K., James, M.R.,
atry 70 (7), 696–698. McMahon, K.L., de Zubicaray, G.I., Martin, N.G., Wright, M.J., Thompson, P.M., 2011a.
Akil, H., Brenner, S., Kandel, E., Kendler, K.S., King, M.C., Scolnick, E., Watson, J.D., Zoghbi, BDNF gene effects on brain circuitry replicated in 455 twins. Neuroimage 55 (2),
H.Y., 2010. Medicine. The future of psychiatric research: genomes and neural circuits. 448–454.
Science 327 (5973), 1580–1581. Chiang, M.C., McMahon, K.L., de Zubicaray, G.I., Martin, N.G., Hickie, I., Toga, A.W., Wright,
Baare, W.F., Hulshoff Pol, H.E., Boomsma, D.I., Posthuma, D., de Geus, E.J., Schnack, H.G., M.J., Thompson, P.M., 2011b. Genetics of white matter development: a DTI study of
van Haren, N.E., van Oel, C.J., Kahn, R.S., 2001. Quantitative genetic modeling of vari- 705 twins and their siblings aged 12 to 29. Neuroimage 54 (3), 2308–2317.
ation in human brain morphology. Cereb. Cortex 11 (9), 816–824. Chiang, M.C., Barysheva, M., McMahon, K.L., de Zubicaray, G.I., Johnson, K., Montgomery,
Basser, P.J., Mattiello, J., LeBihan, D., 1994. MR diffusion tensor spectroscopy and imaging. G.W., Martin, N.G., Toga, A.W., Wright, M.J., Shapshak, P., Thompson, P.M., 2012.
Biophys. J. 66 (1), 259–267. Gene network effects on brain microstructure and intellectual performance identified
Batouli, S.A., Trollor, J.N., Wen, W., Sachdev, P.S., 2013. The heritability of volumes of brain in 472 twins. J. Neurosci. 32 (25), 8732–8745.
structures and its relationship to age: a review of twin and family studies. Ageing Res. Clemm von Hohenberg, C., Wigand, M.C., Kubicki, M., Leicht, G., Giegling, I., Karch, S.,
Rev. 13C, 1–9. Hartmann, A.M., Konte, B., Friedl, M., Ballinger, T., Eckbo, R., Bouix, S., Jager, L.,
Batouli, S.A., Sachdev, P.S., Wen, W., Wright, M.J., Ames, D., Trollor, J.N., 2014. Heritability Shenton, M.E., Rujescu, D., Mulert, C., 2013. CNTNAP2 polymorphisms and structural
of brain volumes in older adults: the Older Australian Twins Study. Neurobiol. Aging brain connectivity: a diffusion-tensor imaging study. J. Psychiatr. Res. 47 (10),
35 (4), 937 (e935-937 e918). 1349–1356.
Bernstein, B.E., Birney, E., Dunham, I., Green, E.D., Gunter, C., Snyder, M., 2012. An inte- Colantuoni, C., Lipska, B.K., Ye, T., Hyde, T.M., Tao, R., Leek, J.T., Colantuoni, E.A., Elkahloun,
grated encyclopedia of DNA elements in the human genome. Nature 489 (7414), A.G., Herman, M.M., Weinberger, D.R., Kleinman, J.E., 2011. Temporal dynamics and
57–74. genetic control of transcription in the human prefrontal cortex. Nature 478 (7370),
Bertisch, H., Li, D., Hoptman, M.J., Delisi, L.E., 2010. Heritability estimates for cognitive fac- 519–523.
tors and brain white matter integrity as markers of schizophrenia. Am. J. Med. Genet. Cosgaya, J.M., Chan, J.R., Shooter, E.M., 2002. The neurotrophin receptor p75NTR as a pos-
B Neuropsychiatr. Genet. 153B (4), 885–894. itive modulator of myelination. Science 298 (5596), 1245–1248.
Bigos, K.L., Mattay, V.S., Callicott, J.H., Straub, R.E., Vakkalanka, R., Kolachana, B., Hyde, T.M., Cousijn, H., Rijpkema, M., Harteveld, A., Harrison, P.J., Fernandez, G., Franke, B., Arias-
Lipska, B.K., Kleinman, J.E., Weinberger, D.R., 2010. Genetic variation in CACNA1C af- Vasquez, A., 2012. Schizophrenia risk gene ZNF804A does not influence macro-
fects brain circuitries related to mental illness. Arch. Gen. Psychiatry 67 (9), 939–945. scopic brain structure: an MRI study in 892 volunteers. Mol. Psychiatry 17
Birney, E., 2012. The making of ENCODE: lessons for big-data projects. Nature 489 (7414), (12), 1155–1157.
49–51. Davis, K.L., Stewart, D.G., Friedman, J.I., Buchsbaum, M., Harvey, P.D., Hof, P.R., Buxbaum, J.,
Bis, J.C., DeCarli, C., Smith, A.V., van der Lijn, F., Crivello, F., Fornage, M., Debette, S., Shulman, Haroutunian, V., 2003. White matter changes in schizophrenia: evidence for myelin-
J.M., Schmidt, H., Srikanth, V., Schuur, M., Yu, L., Choi, S.H., Sigurdsson, S., Verhaaren, B. related dysfunction. Arch. Gen. Psychiatry 60 (5), 443–456.
F., DeStefano, A.L., Lambert, J.C., Jack Jr., C.R., Struchalin, M., Stankovich, J., Ibrahim- Dietsche, B., Backes, H., Laneri, D., Weikert, T., Witt, S.H., Rietschel, M., Sommer, J., Kircher,
Verbaas, C.A., Fleischman, D., Zijdenbos, A., den Heijer, T., Mazoyer, B., Coker, L.H., T., Krug, A., 2014. The impact of a CACNA1C gene polymorphism on learning and hip-
Enzinger, C., Danoy, P., Amin, N., Arfanakis, K., van Buchem, M.A., de Bruijn, R.F., pocampal formation in healthy individuals: a diffusion tensor imaging study.
Beiser, A., Dufouil, C., Huang, J., Cavalieri, M., Thomson, R., Niessen, W.J., Chibnik, L.B., Neuroimage 89, 256–261.
Gislason, G.K., Hofman, A., Pikula, A., Amouyel, P., Freeman, K.B., Phan, T.G., Oostra, Emery, B., 2010. Regulation of oligodendrocyte differentiation and myelination. Science
B.A., Stein, J.L., Medland, S.E., Vasquez, A.A., Hibar, D.P., Wright, M.J., Franke, B., 330 (6005), 779–782.
Martin, N.G., Thompson, P.M., Nalls, M.A., Uitterlinden, A.G., Au, R., Elbaz, A., Beare, R. Esslinger, C., Walter, H., Kirsch, P., Erk, S., Schnell, K., Arnold, C., Haddad, L., Mier, D., Opitz
J., van Swieten, J.C., Lopez, O.L., Harris, T.B., Chouraki, V., Breteler, M.M., De Jager, P.L., von Boberfeld, C., Raab, K., Witt, S.H., Rietschel, M., Cichon, S., Meyer-Lindenberg, A.,
Becker, J.T., Vernooij, M.W., Knopman, D., Fazekas, F., Wolf, P.A., van der Lugt, A., 2009. Neural mechanisms of a genome-wide supported psychosis variant. Science
Gudnason, V., Longstreth Jr., W.T., Brown, M.A., Bennett, D.A., van Duijn, C.M., 324 (5927), 605.
Mosley, T.H., Schmidt, R., Tzourio, C., Launer, L.J., Ikram, M.A., Seshadri, S., 2012. Com- Geng, X., Prom-Wormley, E.C., Perez, J., Kubarych, T., Styner, M., Lin, W., Neale, M.C.,
mon variants at 12q14 and 12q24 are associated with hippocampal volume. Nat. Gilmore, J.H., 2012. White matter heritability using diffusion tensor imaging in neo-
Genet. 44 (5), 545–551. natal brains. Twin Res. Hum. Genet. 15 (3), 336–350.
Blokland, G.A., de Zubicaray, G.I., McMahon, K.L., Wright, M.J., 2012. Genetic and environ- Georgieva, L., Moskvina, V., Peirce, T., Norton, N., Bray, N.J., Jones, L., Holmans, P.,
mental influences on neuroimaging phenotypes: a meta-analytical perspective on Macgregor, S., Zammit, S., Wilkinson, J., Williams, H., Nikolov, I., Williams, N.,
twin imaging studies. Twin Res. Hum. Genet. 15 (3), 351–371. Ivanov, D., Davis, K.L., Haroutunian, V., Buxbaum, J.D., Craddock, N., Kirov, G., Owen,
Blokland, G.A., CONSORTIUM, G., Petryshen, T.L., 2013. The GENUS consortium: genetics M.J., O'Donovan, M.C., 2006. Convergent evidence that oligodendrocyte lineage tran-
of endophenotypes of neurofunction to understand schizophrenia. World Congress scription factor 2 (OLIG2) and interacting genes influence susceptibility to schizo-
of Psychiatric Genetics, Boston, MA. phrenia. Proc. Natl. Acad. Sci. U. S. A. 103 (33), 12469–12474.
Brans, R.G., van Haren, N.E., van Baal, G.C., Schnack, H.G., Kahn, R.S., Hulshoff Pol, H.E., Gilmore, J.H., Schmitt, J.E., Knickmeyer, R.C., Smith, J.K., Lin, W., Styner, M., Gerig, G., Neale,
2008. Heritability of changes in brain volume over time in twin pairs discordant for M.C., 2010. Genetic and environmental contributions to neonatal brain structure: a
schizophrenia. Arch. Gen. Psychiatry 65 (11), 1259–1268. twin study. Hum. Brain Mapp. 31 (8), 1174–1182.
Braskie, M.N., Jahanshad, N., Stein, J.L., Barysheva, M., McMahon, K.L., de Zubicaray, G.I., Gottesman, I.I., Gould, T.D., 2003. The endophenotype concept in psychiatry: etymology
Martin, N.G., Wright, M.J., Ringman, J.M., Toga, A.W., Thompson, P.M., 2011. Common and strategic intentions. Am. J. Psychiatry 160 (4), 636–645.
Alzheimer's disease risk variant within the CLU gene affects white matter microstruc- Green, E.K., Grozeva, D., Jones, I., Jones, L., Kirov, G., Caesar, S., Gordon-Smith, K., Fraser, C.,
ture in young adults. J. Neurosci. 31 (18), 6764–6770. Forty, L., Russell, E., Hamshere, M.L., Moskvina, V., Nikolov, I., Farmer, A., McGuffin, P.,
Braskie, M.N., Jahanshad, N., Stein, J.L., Barysheva, M., Johnson, K., McMahon, K.L., de Holmans, P.A., Owen, M.J., O'Donovan, M.C., Craddock, N., 2010. The bipolar disorder
Zubicaray, G.I., Martin, N.G., Wright, M.J., Ringman, J.M., Toga, A.W., Thompson, P.M., risk allele at CACNA1C also confers risk of recurrent major depression and of schizo-
2012. Relationship of a variant in the NTRK1 gene to white matter microstructure in phrenia. Mol. Psychiatry 15 (10), 1016–1022.
young adults. J. Neurosci. 32 (17), 5964–5972. Hakak, Y., Walker, J.R., Li, C., Wong, W.H., Davis, K.L., Buxbaum, J.D., Haroutunian, V.,
Braskie, M.N., Kohannim, O., Jahanshad, N., Chiang, M.C., Barysheva, M., Toga, A.W., Fienberg, A.A., 2001. Genome-wide expression analysis reveals dysregulation of
Ringman, J.M., Montgomery, G.W., McMahon, K.L., de Zubicaray, G.I., Martin, N.G., myelination-related genes in chronic schizophrenia. Proc. Natl. Acad. Sci. U. S. A. 98
Wright, M.J., Thompson, P.M., 2013. Relation between variants in the neurotrophin (8), 4746–4751.
receptor gene, NTRK3, and white matter integrity in healthy young adults. Hashimoto, R., Numakawa, T., Ohnishi, T., Kumamaru, E., Yagasaki, Y., Ishimoto, T., Mori, T.,
Neuroimage 82, 146–153. Nemoto, K., Adachi, N., Izumi, A., Chiba, S., Noguchi, H., Suzuki, T., Iwata, N., Ozaki, N.,
Brouwer, R.M., Mandl, R.C., Peper, J.S., van Baal, G.C., Kahn, R.S., Boomsma, D.I., Hulshoff Taguchi, T., Kamiya, A., Kosuga, A., Tatsumi, M., Kamijima, K., Weinberger, D.R., Sawa,
Pol, H.E., 2010. Heritability of DTI and MTR in nine-year-old children. Neuroimage A., Kunugi, H., 2006. Impact of the DISC1 Ser704Cys polymorphism on risk for major
53 (3), 1085–1092. depression, brain morphology and ERK signaling. Hum. Mol. Genet. 15 (20),
Callicott, J.H., Feighery, E.L., Mattay, V.S., White, M.G., Chen, Q., Baranger, D.A., Berman, K.F., 3024–3033.
Lu, B., Song, H., Ming, G.L., Weinberger, D.R., 2013. DISC1 and SLC12A2 interaction af- Havik, B., Le Hellard, S., Rietschel, M., Lybaek, H., Djurovic, S., Mattheisen, M., Muhleisen, T.
fects human hippocampal function and connectivity. J. Clin. Invest. 123 (7), W., Degenhardt, F., Priebe, L., Maier, W., Breuer, R., Schulze, T.G., Agartz, I., Melle, I.,
2961–2964. Hansen, T., Bramham, C.R., Nothen, M.M., Stevens, B., Werge, T., Andreassen, O.A.,
Cannon, D.M., Walshe, M., Dempster, E., Collier, D.A., Marshall, N., Bramon, E., Murray, R. Cichon, S., Steen, V.M., 2011. The complement control-related genes CSMD1 and
M., McDonald, C., 2012. The association of white matter volume in psychotic disor- CSMD2 associate to schizophrenia. Biol. Psychiatry 70 (1), 35–42.
ders with genotypic variation in NRG1, MOG and CNP: a voxel-based analysis in af- Hoptman, M.J., Nierenberg, J., Bertisch, H.C., Catalano, D., Ardekani, B.A., Branch, C.A.,
fected individuals and their unaffected relatives. Transl. Psychiatry 2, e167. Delisi, L.E., 2008. A DTI study of white matter microstructure in individuals at high ge-
Chen, S., Velardez, M.O., Warot, X., Yu, Z.X., Miller, S.J., Cros, D., Corfas, G., 2006. netic risk for schizophrenia. Schizophr. Res. 106 (2–3), 115–124.
Neuregulin 1-erbB signaling is necessary for normal myelination and sensory func- Huang, K.P., Huang, F.L., Jager, T., Li, J., Reymann, K.G., Balschun, D., 2004. Neurogranin/
tion. J. Neurosci. 26 (12), 3079–3086. RC3 enhances long-term potentiation and learning by promoting calcium-mediated
Chiang, M.C., Avedissian, C., Barysheva, M., Toga, A.W., McMahon, K.L., de Zubicaray, G.I., signaling. J. Neurosci. 24 (47), 10660–10669.
Wright, M.J., Thompson, P.M., 2009a. Extending genetic linkage analysis to diffusion Hulshoff Pol, H.E., Brans, R.G., van Haren, N.E., Schnack, H.G., Langen, M., Baare, W.F., van
tensor images to map single gene effects on brain fiber architecture. Med. Image Oel, C.J., Kahn, R.S., 2004. Gray and white matter volume abnormalities in monozy-
Comput. Comput. Assist. Interv. 12 (Pt 2), 506–513. gotic and same-gender dizygotic twins discordant for schizophrenia. Biol. Psychiatry
Chiang, M.C., Barysheva, M., Shattuck, D.W., Lee, A.D., Madsen, S.K., Avedissian, C., 55 (2), 126–130.
Klunder, A.D., Toga, A.W., McMahon, K.L., de Zubicaray, G.I., Wright, M.J., Srivastava, Insel, T., Cuthbert, B., Garvey, M., Heinssen, R., Pine, D.S., Quinn, K., Sanislow, C., Wang, P.,
A., Balov, N., Thompson, P.M., 2009b. Genetics of brain fiber architecture and intellec- 2010. Research domain criteria (RDoC): toward a new classification framework for
tual performance. J. Neurosci. 29 (7), 2212–2224. research on mental disorders. Am. J. Psychiatry 167 (7), 748–751.

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx 9

Jagannathan, K., Calhoun, V.D., Gelernter, J., Stevens, M.C., Liu, J., Bolognani, F., McIntosh, A.M., Moorhead, T.W., Job, D., Lymer, G.K., Munoz Maniega, S., McKirdy, J.,
Windemuth, A., Ruano, G., Assaf, M., Pearlson, G.D., 2010. Genetic associations of Sussmann, J.E., Baig, B.J., Bastin, M.E., Porteous, D., Evans, K.L., Johnstone, E.C.,
brain structural networks in schizophrenia: a preliminary study. Biol. Psychiatry 68 Lawrie, S.M., Hall, J., 2008. The effects of a neuregulin 1 variant on white matter den-
(7), 657–666. sity and integrity. Mol. Psychiatry 13 (11), 1054–1059.
Jahanshad, N., Lee, A.D., Barysheva, M., McMahon, K.L., de Zubicaray, G.I., Martin, N.G., McTigue, D.M., Tripathi, R.B., 2008. The life, death, and replacement of oligodendrocytes in
Wright, M.J., Toga, A.W., Thompson, P.M., 2010. Genetic influences on brain asymme- the adult CNS. J. Neurochem. 107 (1), 1–19.
try: a DTI study of 374 twins and siblings. Neuroimage 52 (2), 455–469. Meyer-Lindenberg, A., Weinberger, D.R., 2006. Intermediate phenotypes and genetic
Jahanshad, N., Kohannim, O., Hibar, D.P., Stein, J.L., McMahon, K.L., de Zubicaray, G.I., mechanisms of psychiatric disorders. Nat. Rev. Neurosci. 7 (10), 818–827.
Medland, S.E., Montgomery, G.W., Whitfield, J.B., Martin, N.G., Wright, M.J., Toga, A. Montag, C., Schoene-Bake, J.C., Faber, J., Reuter, M., Weber, B., 2010. Genetic variation on
W., Thompson, P.M., 2012. Brain structure in healthy adults is related to serum trans- the BDNF gene is not associated with differences in white matter tracts in healthy
ferrin and the H63D polymorphism in the HFE gene. Proc. Natl. Acad. Sci. U. S. A. 109 humans measured by tract-based spatial statistics. Genes Brain Behav. 9 (8),
(14), E851–E859. 886–891.
Jahanshad, N., Kochunov, P.V., Sprooten, E., Mandl, R.C., Nichols, T.E., Almasy, L., Blangero, J., Mothersill, O., Morris, D.W., Kelly, S., Rose, E.J., Fahey, C., O'Brien, C., Lyne, R., Reilly, R., Gill,
Brouwer, R.M., Curran, J.E., de Zubicaray, G.I., Duggirala, R., Fox, P.T., Hong, L.E., M., Corvin, A.P., Donohoe, G., 2013. Effects of MIR137 on fronto-amygdala functional
Landman, B.A., Martin, N.G., McMahon, K.L., Medland, S.E., Mitchell, B.D., Olvera, R.L., connectivity. Neuroimage 90C, 189–195.
Peterson, C.P., Starr, J.M., Sussmann, J.E., Toga, A.W., Wardlaw, J.M., Wright, M.J., O'Donovan, M.C., Craddock, N., Norton, N., Williams, H., Peirce, T., Moskvina, V., Nikolov, I.,
Hulshoff Pol, H.E., Bastin, M.E., McIntosh, A.M., Deary, I.J., Thompson, P.M., Glahn, D. Hamshere, M., Carroll, L., Georgieva, L., Dwyer, S., Holmans, P., Marchini, J.L., Spencer,
C., 2013. Multi-site genetic analysis of diffusion images and voxelwise heritability C.C., Howie, B., Leung, H.T., Hartmann, A.M., Moller, H.J., Morris, D.W., Shi, Y., Feng, G.,
analysis: a pilot project of the ENIGMA-DTI working group. Neuroimage 81, 455–469. Hoffmann, P., Propping, P., Vasilescu, C., Maier, W., Rietschel, M., Zammit, S.,
Jones, D.K., 2008. Studying connections in the living human brain with diffusion MRI. Cor- Schumacher, J., Quinn, E.M., Schulze, T.G., Williams, N.M., Giegling, I., Iwata, N., Ikeda,
tex 44 (8), 936–952. M., Darvasi, A., Shifman, S., He, L., Duan, J., Sanders, A.R., Levinson, D.F., Gejman, P.V.,
Jones, D.K., Williams, S.C., Gasston, D., Horsfield, M.A., Simmons, A., Howard, R., 2002. Iso- Cichon, S., Nothen, M.M., Gill, M., Corvin, A., Rujescu, D., Kirov, G., Owen, M.J., Buccola,
tropic resolution diffusion tensor imaging with whole brain acquisition in a clinically N.G., Mowry, B.J., Freedman, R., Amin, F., Black, D.W., Silverman, J.M., Byerley, W.F.,
acceptable time. Hum. Brain Mapp. 15 (4), 216–230. Cloninger, C.R., 2008. Identification of loci associated with schizophrenia by genome-
Jones, D.K., Symms, M.R., Cercignani, M., Howard, R.J., 2005. The effect of filter size on wide association and follow-up. Nat. Genet. 40 (9), 1053–1055.
VBM analyses of DT-MRI data. Neuroimage 26 (2), 546–554. O'Dushlaine, C., Kenny, E., Heron, E., Donohoe, G., Gill, M., Morris, D., Corvin, A., 2011. Mo-
Judy, J.T., Seifuddin, F., Pirooznia, M., Mahon, P.B., Jancic, D., Goes, F.S., Schulze, T., Cichon, lecular pathways involved in neuronal cell adhesion and membrane scaffolding con-
S., Noethen, M., Rietschel, M., Depaulo Jr., J.R., Potash, J.B., Zandi, P.P., 2013. Converg- tribute to schizophrenia and bipolar disorder susceptibility. Mol. Psychiatry 16 (3),
ing evidence for epistasis between ANK3 and potassium channel gene KCNQ2 in bi- 286–292.
polar disorder. Front. Genet. 4, 87. Ohi, K., Hashimoto, R., Yasuda, Y., Nemoto, K., Ohnishi, T., Fukumoto, M., Yamamori, H.,
Kennedy, K.M., Rodrigue, K.M., Land, S.J., Raz, N., 2009. BDNF Val66Met polymorphism in- Umeda-Yano, S., Okada, T., Iwase, M., Kazui, H., Takeda, M., 2012. Impact of the
fluences age differences in microstructure of the corpus callosum. Front. Hum. genome wide supported NRGN gene on anterior cingulate morphology in schizo-
Neurosci. 3, 19. phrenia. PLoS ONE 7 (1), e29780.
Kim, A.H., Parker, E.K., Williamson, V., McMichael, G.O., Fanous, A.H., Vladimirov, V.I., Pernet, V., Joly, S., Christ, F., Dimou, L., Schwab, M.E., 2008. Nogo-A and myelin-associated
2012. Experimental validation of candidate schizophrenia gene ZNF804A as target glycoprotein differently regulate oligodendrocyte maturation and myelin formation.
for hsa-miR-137. Schizophr. Res. 141 (1), 60–64. J. Neurosci. 28 (29), 7435–7444.
Kochunov, P., Glahn, D.C., Lancaster, J.L., Winkler, A.M., Smith, S., Thompson, P.M., Almasy, Pfefferbaum, A., Sullivan, E.V., Swan, G.E., Carmelli, D., 2000. Brain structure in men re-
L., Duggirala, R., Fox, P.T., Blangero, J., 2010. Genetics of microstructure of cerebral mains highly heritable in the seventh and eighth decades of life. Neurobiol. Aging
white matter using diffusion tensor imaging. Neuroimage 53 (3), 1109–1116. 21 (1), 63–74.
Kochunov, P., Jahanshad, N., Sprooten, E., Nichols, T., Mandl, R., Almasy, L., Booth, T., Pfefferbaum, A., Sullivan, E.V., Carmelli, D., 2001. Genetic regulation of regional micro-
Brouwer, R.M., Curran, J.E., de Zubicaray, G., Dimitrova, R., Duggirala, R., Fox, P., structure of the corpus callosum in late life. Neuroreport 12 (8), 1677–1681.
Hong, E.J., Landman, B.A., Lemaitre, H., Lopez, l, Martin, N.G., McMahon, K.L., Potkin, S.G., Guffanti, G., Lakatos, A., Turner, J.A., Kruggel, F., Fallon, J.H., Saykin, A.J., Orro,
Mitchell, B., Olvera, R.L., Peterson, C.P., Starr, J.M., Sussmann, J., Toga, A., Wardlaw, J. A., Lupoli, S., Salvi, E., Weiner, M., Macciardi, F., 2009. Hippocampal atrophy as a quan-
M., Wright, M.J., Wright, S., Bastin, M.E., McIntosh, A., Boomsma, D., Kahn, R.S., den titative trait in a genome-wide association study identifying novel susceptibility
Braber, A., de Geus, E.J., Deary, I.J., Hulshoff Pol, H., Williamson, D., Blangero, J., genes for Alzheimer's disease. PLoS ONE 4 (8), e6501.
van 't Ent, D., Thompson, P., Glahn, D.C., 2014. Multi-site study of additive genetic ef- Riley, B., Thiselton, D., Maher, B.S., Bigdeli, T., Wormley, B., McMichael, G.O., Fanous, A.H.,
fects on fractional anisotropy of cerebral white matter: comparing meta and mega Vladimirov, V., O'Neill, F.A., Walsh, D., Kendler, K.S., 2010. Replication of association
analytical approaches for data pooling. Neuroimage (in press). between schizophrenia and ZNF804A in the Irish Case-Control Study of Schizophre-
Kohannim, O., Jahanshad, N., Braskie, M.N., Stein, J.L., Chiang, M.C., Reese, A.H., Hibar, D.P., nia sample. Mol. Psychiatry 15 (1), 29–37.
Toga, A.W., McMahon, K.L., de Zubicaray, G.I., Medland, S.E., Montgomery, G.W., Ripke, S., O'Dushlaine, C., Chambert, K., Moran, J.L., Kahler, A.K., Akterin, S., Bergen, S.E.,
Martin, N.G., Wright, M.J., Thompson, P.M., 2012. Predicting white matter integrity Collins, A.L., Crowley, J.J., Fromer, M., Kim, Y., Lee, S.H., Magnusson, P.K., Sanchez, N.,
from multiple common genetic variants. Neuropsychopharmacology 37 (9), Stahl, E.A., Williams, S., Wray, N.R., Xia, K., Bettella, F., Borglum, A.D., Bulik-Sullivan,
2012–2019. B.K., Cormican, P., Craddock, N., de Leeuw, C., Durmishi, N., Gill, M., Golimbet, V.,
Konrad, A., Vucurevic, G., Musso, F., Stoeter, P., Dahmen, N., Winterer, G., 2009. ErbB4 ge- Hamshere, M.L., Holmans, P., Hougaard, D.M., Kendler, K.S., Lin, K., Morris, D.W.,
notype predicts left frontotemporal structural connectivity in human brain. Mors, O., Mortensen, P.B., Neale, B.M., O'Neill, F.A., Owen, M.J., Milovancevic, M.P.,
Neuropsychopharmacology 34 (3), 641–650. Posthuma, D., Powell, J., Richards, A.L., Riley, B.P., Ruderfer, D., Rujescu, D.,
Kuswanto, C.N., Woon, P.S., Zheng, X.B., Qiu, A., Sitoh, Y.Y., Chan, Y.H., Liu, J., Williams, H., Sigurdsson, E., Silagadze, T., Smit, A.B., Stefansson, H., Steinberg, S., Suvisaari, J.,
Ong, W.Y., Sim, K., 2012. Genome-wide supported psychosis risk variant in ZNF804A Tosato, S., Verhage, M., Walters, J.T., Levinson, D.F., Gejman, P.V., Laurent, C.,
gene and impact on cortico-limbic WM integrity in schizophrenia. Am. J. Med. Genet. Mowry, B.J., O'Donovan, M.C., Pulver, A.E., Schwab, S.G., Wildenauer, D.B.,
B Neuropsychiatr. Genet. 159B (3), 255–262. Dudbridge, F., Shi, J., Albus, M., Alexander, M., Campion, D., Cohen, D., Dikeos, D.,
Kwon, E., Wang, W., Tsai, L.H., 2013. Validation of schizophrenia-associated genes CSMD1, Duan, J., Eichhammer, P., Godard, S., Hansen, M., Lerer, F.B., Liang, K.Y., Maier, W.,
C10orf26, CACNA1C and TCF4 as miR-137 targets. Mol. Psychiatry 18 (1), 11–12. Mallet, J., Nertney, D.A., Nestadt, G., Norton, N., Papadimitriou, G.N., Ribble, R.,
Lencz, T., Szeszko, P.R., DeRosse, P., Burdick, K.E., Bromet, E.J., Bilder, R.M., Malhotra, A.K., Sanders, A.R., Silverman, J.M., Walsh, D., Williams, N.M., Wormley, B., Arranz, M.J.,
2010. A schizophrenia risk gene, ZNF804A, influences neuroanatomical and Bakker, S., Bender, S., Bramon, E., Collier, D., Crespo-Facorro, B., Hall, J., Iyegbe, C.,
neurocognitive phenotypes. Neuropsychopharmacology 35 (11), 2284–2291. Jablensky, A., Kahn, R.S., Kalaydjieva, L., Lawrie, S., Lewis, C.M., Linszen, D.H., Mata, I.,
Lett, T.A., Chakavarty, M.M., Felsky, D., Brandl, E.J., Tiwari, A.K., Goncalves, V.F., Rajji, T.K., McIntosh, A., Murray, R.M., Ophoff, R.A., Van Os, J., Walshe, M., Weisbrod, M.,
Daskalakis, Z.J., Meltzer, H.Y., Lieberman, J.A., Lerch, J.P., Mulsant, B.H., Kennedy, J.L., Wiersma, D., Donnelly, P., Barroso, I., Blackwell, J.M., Brown, M.A., Casas, J.P., Corvin,
Voineskos, A.N., 2013. The genome-wide supported microRNA-137 variant predicts A.P., Deloukas, P., Duncanson, A., Jankowski, J., Markus, H.S., Mathew, C.G., Palmer, C.
phenotypic heterogeneity within schizophrenia. Mol. Psychiatry 18 (4), 443–450. N., Plomin, R., Rautanen, A., Sawcer, S.J., Trembath, R.C., Viswanathan, A.C., Wood, N.
Li, M., Wang, Y., Zheng, X.B., Ikeda, M., Iwata, N., Luo, X.J., Chong, S.A., Lee, J., Rietschel, M., W., Spencer, C.C., Band, G., Bellenguez, C., Freeman, C., Hellenthal, G., Giannoulatou,
Zhang, F., Muller-Myhsok, B., Cichon, S., Weinberger, D.R., Mattheisen, M., Schulze, T. E., Pirinen, M., Pearson, R.D., Strange, A., Su, Z., Vukcevic, D., Langford, C., Hunt, S.E.,
G., Martin, N.G., Mitchell, P.B., Schofield, P.R., Liu, J.J., Su, B., 2012. Meta-analysis and Edkins, S., Gwilliam, R., Blackburn, H., Bumpstead, S.J., Dronov, S., Gillman, M., Gray,
brain imaging data support the involvement of VRK2 (rs2312147) in schizophrenia E., Hammond, N., Jayakumar, A., McCann, O.T., Liddle, J., Potter, S.C., Ravindrarajah,
susceptibility. Schizophr. Res. 142 (1–3), 200–205. R., Ricketts, M., Tashakkori-Ghanbaria, A., Waller, M.J., Weston, P., Widaa, S.,
Li, Y., Liu, B., Hou, B., Qin, W., Wang, D., Yu, C., Jiang, T., 2013. Less efficient information Whittaker, P., McCarthy, M.I., Stefansson, K., Scolnick, E., Purcell, S., McCarroll, S.A.,
transfer in Cys-allele carriers of DISC1: a brain network study based on diffusion Sklar, P., Hultman, C.M., Sullivan, P.F., 2013. Genome-wide association analysis iden-
MRI. Cereb. Cortex 23 (7), 1715–1723. tifies 13 new risk loci for schizophrenia. Nat. Genet. 45 (10), 1150–1159.
Linke, J., Witt, S.H., King, A.V., Nieratschker, V., Poupon, C., Gass, A., Hennerici, M.G., Rose, E.J., Morris, D.W., Fahey, C., Robertson, I.H., Greene, C., O'Doherty, J., Newell, F.N.,
Rietschel, M., Wessa, M., 2012. Genome-wide supported risk variant for bipolar disor- Garavan, H., McGrath, J., Bokde, A., Tropea, D., Gill, M., Corvin, A.P., Donohoe, G.,
der alters anatomical connectivity in the human brain. Neuroimage 59 (4), 2012. The effect of the neurogranin schizophrenia risk variant rs12807809 on brain
3288–3296. structure and function. Twin Res. Hum. Genet. 15 (3), 296–303.
Lopez, L.M., Bastin, M.E., Maniega, S.M., Penke, L., Davies, G., Christoforou, A., Valdes Rose, E.J., Morris, D.W., Hargreaves, A., Fahey, C., Greene, C., Garavan, H., Gill, M., Corvin, A.,
Hernandez, M.C., Royle, N.A., Tenesa, A., Starr, J.M., Porteous, D.J., Wardlaw, J.M., Donohoe, G., 2013. Neural effects of the CSMD1 genome-wide associated schizophrenia
Deary, I.J., 2012. A genome-wide search for genetic influences and biological path- risk variant rs10503253. Am. J. Med. Genet. B Neuropsychiatr. Genet. 162B (6), 530–537.
ways related to the brain's white matter integrity. Neurobiol. Aging 33 (8), 1847 Rose, E.J., Hargreaves, A., Morris, D., Fahey, C., Tropea, D., Cummings, E., Caltagirone, C.,
(e1841-1814). Bossu, P., Chiapponi, C., Piras, F., Spalletta, G., Gill, M., Corvin, A., Donohoe, G., 2014.

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
10 A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx

Effects of a novel schizophrenia risk variant rs7914558 at CNNM2 on brain structure H.G., Schnell, K., Seiferth, N., Smith, C., Steen, V.M., Valdes Hernandez, M.C., Van den
and attributional style. Br. J. Psychiatry 204, 115–121. Heuvel, M., van der Wee, N.J., Van Haren, N.E., Veltman, J.A., Volzke, H., Walker, R.,
Roy, K., Murtie, J.C., El-Khodor, B.F., Edgar, N., Sardi, S.P., Hooks, B.M., Benoit-Marand, Westlye, L.T., Whelan, C.D., Agartz, I., Boomsma, D.I., Cavalleri, G.L., Dale, A.M.,
M., Chen, C., Moore, H., O'Donnell, P., Brunner, D., Corfas, G., 2007. Loss of erbB Djurovic, S., Drevets, W.C., Hagoort, P., Hall, J., Heinz, A., Jack Jr., C.R., Foroud, T.M., Le
signaling in oligodendrocytes alters myelin and dopaminergic function, a poten- Hellard, S., Macciardi, F., Montgomery, G.W., Poline, J.B., Porteous, D.J., Sisodiya, S.M.,
tial mechanism for neuropsychiatric disorders. Proc. Natl. Acad. Sci. U. S. A. 104 Starr, J.M., Sussmann, J., Toga, A.W., Veltman, D.J., Walter, H., Weiner, M.W., Bis, J.C.,
(19), 8131–8136. Ikram, M.A., Smith, A.V., Gudnason, V., Tzourio, C., Vernooij, M.W., Launer, L.J., DeCarli,
Scamvougeras, A., Kigar, D.L., Jones, D., Weinberger, D.R., Witelson, S.F., 2003. Size of the C., Seshadri, S., Andreassen, O.A., Apostolova, L.G., Bastin, M.E., Blangero, J., Brunner, H.
human corpus callosum is genetically determined: an MRI study in mono and dizy- G., Buckner, R.L., Cichon, S., Coppola, G., de Zubicaray, G.I., Deary, I.J., Donohoe, G., de
gotic twins. Neurosci. Lett. 338 (2), 91–94. Geus, E.J., Espeseth, T., Fernandez, G., Glahn, D.C., Grabe, H.J., Hardy, J., Hulshoff Pol,
Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium, 2011. H.E., Jenkinson, M., Kahn, R.S., McDonald, C., McIntosh, A.M., McMahon, F.J., McMahon,
Genome-wide association study identifies five new schizophrenia loci. Nat. Genet. K.L., Meyer-Lindenberg, A., Morris, D.W., Muller-Myhsok, B., Nichols, T.E., Ophoff, R.A.,
43 (10), 969–976. Paus, T., Pausova, Z., Penninx, B.W., Potkin, S.G., Samann, P.G., Saykin, A.J., Schumann,
Segal, D., Koschnick, J.R., Slegers, L.H., Hof, P.R., 2007. Oligodendrocyte pathophysiology: a G., Smoller, J.W., Wardlaw, J.M., Weale, M.E., Martin, N.G., Franke, B., Wright, M.J.,
new view of schizophrenia. Int. J. Neuropsychopharmacol. 10 (4), 503–511. Thompson, P.M., 2012. Identification of common variants associated with human hip-
Shen, E.H., Overly, C.C., Jones, A.R., 2012. The Allen Human Brain Atlas: comprehensive pocampal and intracranial volumes. Nat. Genet. 44 (5), 552–561.
gene expression mapping of the human brain. Trends Neurosci. 35 (12), 711–714. Terwisscha van Scheltinga, A.F., Bakker, S.C., van Haren, N.E., Derks, E.M., Buizer-Voskamp,
Shenton, M.E., Whitford, T.J., Kubicki, M., 2010. Structural neuroimaging in schizophrenia: J.E., Boos, H.B., Cahn, W., Hulshoff Pol, H.E., Ripke, S., Ophoff, R.A., Kahn, R.S., 2013. Ge-
from methods to insights to treatments. Dialogues Clin. Neurosci. 12 (3), 317–332. netic schizophrenia risk variants jointly modulate total brain and white matter vol-
Shifman, S., Johannesson, M., Bronstein, M., Chen, S.X., Collier, D.A., Craddock, N.J., Kendler, K. ume. Biol. Psychiatry 73 (6), 525–531.
S., Li, T., O'Donovan, M., O'Neill, F.A., Owen, M.J., Walsh, D., Weinberger, D.R., Sun, C., Flint, Thomason, M.E., Dougherty, R.F., Colich, N.L., Perry, L.M., Rykhlevskaia, E.I., Louro, H.M.,
J., Darvasi, A., 2008. Genome-wide association identifies a common variant in the reelin Hallmayer, J.F., Waugh, C.E., Bammer, R., Glover, G.H., Gotlib, I.H., 2010. COMT geno-
gene that increases the risk of schizophrenia only in women. PLoS Genet. 4 (2), e28. type affects prefrontal white matter pathways in children and adolescents.
Skudlarski, P., Schretlen, D.J., Thaker, G.K., Stevens, M.C., Keshavan, M.S., Sweeney, J.A., Neuroimage 53 (3), 926–934.
Tamminga, C.A., Clementz, B.A., O'Neil, K., Pearlson, G.D., 2013. Diffusion tensor imag- Tost, H., Lipska, B.K., Vakkalanka, R., Lemaitre, H., Callicott, J.H., Mattay, V.S., Kleinman, J.E.,
ing white matter endophenotypes in patients with schizophrenia or psychotic bipolar Marenco, S., Weinberger, D.R., 2010. No effect of a common allelic variant in the
disorder and their relatives. Am. J. Psychiatry 170 (8), 886–898. reelin gene on intermediate phenotype measures of brain structure, brain function,
Sprooten, E., Sussmann, J.E., Moorhead, T.W., Whalley, H.C., Ffrench-Constant, C., and gene expression. Biol. Psychiatry 68 (1), 105–107.
Blumberg, H.P., Bastin, M.E., Hall, J., Lawrie, S.M., McIntosh, A.M., 2011. Association van Erp, T.G., Guella, I., Vawter, M.P., Turner, J., Brown, G.G., McCarthy, G., Greve, D.N.,
of white matter integrity with genetic variation in an exonic DISC1 SNP. Mol. Psychi- Glover, G.H., Calhoun, V.D., Lim, K.O., Bustillo, J.R., Belger, A., Ford, J.M., Mathalon, D.
atry 16 (7), 685 (688-689). H., Diaz, M., Preda, A., Nguyen, D., Macciardi, F., Potkin, S.G., 2014. Schizophrenia
Sprooten, E., McIntosh, A.M., Lawrie, S.M., Hall, J., Sussmann, J.E., Dahmen, N., Konrad, A., miR-137 locus risk genotype is associated with dorsolateral prefrontal cortex hyper-
Bastin, M.E., Winterer, G., 2012. An investigation of a genomewide supported psycho- activation. Biol. Psychiatry 75 (5), 398–405.
sis variant in ZNF804A and white matter integrity in the human brain. Magn. Reson. Voineskos, A.N., Lerch, J.P., Felsky, D., Shaikh, S., Rajji, T.K., Miranda, D., Lobaugh, N.J.,
Imaging 30 (10), 1373–1380. Mulsant, B.H., Pollock, B.G., Kennedy, J.L., 2011a. The brain-derived neurotrophic fac-
Stefansson, H., Rujescu, D., Cichon, S., Pietilainen, O.P., Ingason, A., Steinberg, S., Fossdal, R., tor Val66Met polymorphism and prediction of neural risk for Alzheimer disease.
Sigurdsson, E., Sigmundsson, T., Buizer-Voskamp, J.E., Hansen, T., Jakobsen, K.D., Arch. Gen. Psychiatry 68 (2), 198–206.
Muglia, P., Francks, C., Matthews, P.M., Gylfason, A., Halldorsson, B.V., Gudbjartsson, Voineskos, A.N., Lerch, J.P., Felsky, D., Tiwari, A., Rajji, T.K., Miranda, D., Lobaugh, N.J., Pollock, B.
D., Thorgeirsson, T.E., Sigurdsson, A., Jonasdottir, A., Jonasdottir, A., Bjornsson, A., G., Mulsant, B.H., Kennedy, J.L., 2011b. The ZNF804A gene: characterization of a novel
Mattiasdottir, S., Blondal, T., Haraldsson, M., Magnusdottir, B.B., Giegling, I., Moller, neural risk mechanism for the major psychoses. Neuropsychopharmacology 36 (9),
H.J., Hartmann, A., Shianna, K.V., Ge, D., Need, A.C., Crombie, C., Fraser, G., Walker, N., 1871–1878.
Lonnqvist, J., Suvisaari, J., Tuulio-Henriksson, A., Paunio, T., Toulopoulou, T., Bramon, Voineskos, A.N., Lett, T.A., Lerch, J.P., Tiwari, A.K., Ameis, S.H., Rajji, T.K., Muller, D.J.,
E., Di Forti, M., Murray, R., Ruggeri, M., Vassos, E., Tosato, S., Walshe, M., Li, T., Mulsant, B.H., Kennedy, J.L., 2011c. Neurexin-1 and frontal lobe white matter: an
Vasilescu, C., Muhleisen, T.W., Wang, A.G., Ullum, H., Djurovic, S., Melle, I., Olesen, J., overlapping intermediate phenotype for schizophrenia and autism spectrum disor-
Kiemeney, L.A., Franke, B., Sabatti, C., Freimer, N.B., Gulcher, J.R., Thorsteinsdottir, U., ders. PLoS ONE 6 (6), e20982.
Kong, A., Andreassen, O.A., Ophoff, R.A., Georgi, A., Rietschel, M., Werge, T., Voineskos, A.N., Felsky, D., Kovacevic, N., Tiwari, A.K., Zai, C., Chakravarty, M.M., Lobaugh,
Petursson, H., Goldstein, D.B., Nothen, M.M., Peltonen, L., Collier, D.A., St Clair, D., N.J., Shenton, M.E., Rajji, T.K., Miranda, D., Pollock, B.G., Mulsant, B.H., McIntosh, A.R.,
Stefansson, K., 2008. Large recurrent microdeletions associated with schizophrenia. Kennedy, J.L., 2013a. Oligodendrocyte genes, white matter tract integrity, and cogni-
Nature 455 (7210), 232–236. tion in schizophrenia. Cereb. Cortex 23 (9), 2044–2057.
Stefansson, H., Ophoff, R.A., Steinberg, S., Andreassen, O.A., Cichon, S., Rujescu, D., Werge, T. Voineskos, A.N., Foussias, G., Lerch, J., Felsky, D., Remington, G., Rajji, T.K., Lobaugh, N.,
, Pietilainen, O.P., Mors, O., Mortensen, P.B., Sigurdsson, E., Gustafsson, O., Nyegaard, M. Pollock, B.G., Mulsant, B.H., 2013b. Neuroimaging evidence for the deficit subtype of
, Tuulio-Henriksson, A., Ingason, A., Hansen, T., Suvisaari, J., Lonnqvist, J., Paunio, T., schizophrenia. JAMA Psychiatry 70 (5), 472–480.
Borglum, A.D., Hartmann, A., Fink-Jensen, A., Nordentoft, M., Hougaard, D., Wallace, G.L., Eric Schmitt, J., Lenroot, R., Viding, E., Ordaz, S., Rosenthal, M.A., Molloy, E.A.,
Norgaard-Pedersen, B., Bottcher, Y., Olesen, J., Breuer, R., Moller, H.J., Giegling, I., Clasen, L.S., Kendler, K.S., Neale, M.C., Giedd, J.N., 2006. A pediatric twin study of brain
Rasmussen, H.B., Timm, S., Mattheisen, M., Bitter, I., Rethelyi, J.M., Magnusdottir, B.B., morphometry. J. Child Psychol. Psychiatry 47 (10), 987–993.
Sigmundsson, T., Olason, P., Masson, G., Gulcher, J.R., Haraldsson, M., Fossdal, R., Walters, J.T., Rujescu, D., Franke, B., Giegling, I., Vasquez, A.A., Hargreaves, A., Russo, G.,
Thorgeirsson, T.E., Thorsteinsdottir, U., Ruggeri, M., Tosato, S., Franke, B., Strengman, Morris, D.W., Hoogman, M., Da Costa, A., Moskvina, V., Fernandez, G., Gill, M.,
E., Kiemeney, L.A., Melle, I., Djurovic, S., Abramova, L., Kaleda, V., Sanjuan, J., de Corvin, A., O'Donovan, M.C., Donohoe, G., Owen, M.J., 2013. The role of the major his-
Frutos, R., Bramon, E., Vassos, E., Fraser, G., Ettinger, U., Picchioni, M., Walker, N., tocompatibility complex region in cognition and brain structure: a schizophrenia
Toulopoulou, T., Need, A.C., Ge, D., Yoon, J.L., Shianna, K.V., Freimer, N.B., Cantor, R. GWAS follow-up. Am. J. Psychiatry 170 (8), 877–885.
M., Murray, R., Kong, A., Golimbet, V., Carracedo, A., Arango, C., Costas, J., Jonsson, E. Wang, K.C., Kim, J.A., Sivasankaran, R., Segal, R., He, Z., 2002. P75 interacts with the
G., Terenius, L., Agartz, I., Petursson, H., Nothen, M.M., Rietschel, M., Matthews, P.M., Nogo receptor as a co-receptor for Nogo, MAG and OMgp. Nature 420 (6911),
Muglia, P., Peltonen, L., St Clair, D., Goldstein, D.B., Stefansson, K., Collier, D.A., 2009. 74–78.
Common variants conferring risk of schizophrenia. Nature 460 (7256), 744–747. Wang, F., Jiang, T., Sun, Z., Teng, S.L., Luo, X., Zhu, Z., Zang, Y., Zhang, H., Yue, W., Qu, M., Lu,
Stein, J.L., Hibar, D.P., Madsen, S.K., Khamis, M., McMahon, K.L., de Zubicaray, G.I., Hansell, T., Hong, N., Huang, H., Blumberg, H.P., Zhang, D., 2009. Neuregulin 1 genetic variation
N.K., Montgomery, G.W., Martin, N.G., Wright, M.J., Saykin, A.J., Jack Jr., C.R., Weiner, and anterior cingulum integrity in patients with schizophrenia and healthy controls.
M.W., Toga, A.W., Thompson, P.M., 2011. Discovery and replication of dopamine- J. Psychiatry Neurosci. 34 (3), 181–186.
related gene effects on caudate volume in young and elderly populations (N = Wassink, T.H., Epping, E.A., Rudd, D., Axelsen, M., Ziebell, S., Fleming, F.W., Monson, E., Ho,
1198) using genome-wide search. Mol. Psychiatry 16 (9), 927–937 (881). B.C., Andreasen, N.C., 2012. Influence of ZNF804a on brain structure volumes and
Stein, J.L., Medland, S.E., Vasquez, A.A., Hibar, D.P., Senstad, R.E., Winkler, A.M., Toro, R., symptom severity in individuals with schizophrenia. Arch. Gen. Psychiatry 69 (9),
Appel, K., Bartecek, R., Bergmann, O., Bernard, M., Brown, A.A., Cannon, D.M., 885–892.
Chakravarty, M.M., Christoforou, A., Domin, M., Grimm, O., Hollinshead, M., Holmes, Wei, Q., Kang, Z., Diao, F., Shan, B., Li, L., Zheng, L., Guo, X., Liu, C., Zhang, J., Zhao, J., 2012.
A.J., Homuth, G., Hottenga, J.J., Langan, C., Lopez, L.M., Hansell, N.K., Hwang, K.S., Association of the ZNF804A gene polymorphism rs1344706 with white matter den-
Kim, S., Laje, G., Lee, P.H., Liu, X., Loth, E., Lourdusamy, A., Mattingsdal, M., Mohnke, sity changes in Chinese schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry
S., Maniega, S.M., Nho, K., Nugent, A.C., O'Brien, C., Papmeyer, M., Putz, B., Ramasamy, 36 (1), 122–127.
A., Rasmussen, J., Rijpkema, M., Risacher, S.L., Roddey, J.C., Rose, E.J., Ryten, M., Shen, L., Wei, Q., Kang, Z., Diao, F., Guidon, A., Wu, X., Zheng, L., Li, L., Guo, X., Hu, M., Zhang, J.,
Sprooten, E., Strengman, E., Teumer, A., Trabzuni, D., Turner, J., van Eijk, K., van Erp, T. Liu, C., Zhao, J., 2013. No association of ZNF804A rs1344706 with white matter in-
G., van Tol, M.J., Wittfeld, K., Wolf, C., Woudstra, S., Aleman, A., Alhusaini, S., Almasy, L., tegrity in schizophrenia: a tract-based spatial statistics study. Neurosci. Lett. 532,
Binder, E.B., Brohawn, D.G., Cantor, R.M., Carless, M.A., Corvin, A., Czisch, M., Curran, J. 64–69.
E., Davies, G., de Almeida, M.A., Delanty, N., Depondt, C., Duggirala, R., Dyer, T.D., Erk, S., White, T., Andreasen, N.C., Nopoulos, P., 2002. Brain volumes and surface morphology in
Fagerness, J., Fox, P.T., Freimer, N.B., Gill, M., Goring, H.H., Hagler, D.J., Hoehn, D., monozygotic twins. Cereb. Cortex 12 (5), 486–493.
Holsboer, F., Hoogman, M., Hosten, N., Jahanshad, N., Johnson, M.P., Kasperaviciute, Winterer, G., Konrad, A., Vucurevic, G., Musso, F., Stoeter, P., Dahmen, N., 2008. Associa-
D., Kent Jr., J.W., Kochunov, P., Lancaster, J.L., Lawrie, S.M., Liewald, D.C., Mandl, R., tion of 5′ end neuregulin-1 (NRG1) gene variation with subcortical medial frontal mi-
Matarin, M., Mattheisen, M., Meisenzahl, E., Melle, I., Moses, E.K., Muhleisen, T.W., crostructure in humans. Neuroimage 40 (2), 712–718.
Nauck, M., Nothen, M.M., Olvera, R.L., Pandolfo, M., Pike, G.B., Puls, R., Reinvang, I., Wirgenes, K.V., Sonderby, I.E., Haukvik, U.K., Mattingsdal, M., Tesli, M., Athanasiu, L.,
Renteria, M.E., Rietschel, M., Roffman, J.L., Royle, N.A., Rujescu, D., Savitz, J., Schnack, Sundet, K., Rossberg, J.I., Dale, A.M., Brown, A.A., Agartz, I., Melle, I., Djurovic, S.,

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034
A.N. Voineskos / Schizophrenia Research xxx (2014) xxx–xxx 11

Andreassen, O.A., 2012. TCF4 sequence variants and mRNA levels are associated Yoon, U., Fahim, C., Perusse, D., Evans, A.C., 2010. Lateralized genetic and environmental
with neurodevelopmental characteristics in psychotic disorders. Transl. Psychia- influences on human brain morphology of 8-year-old twins. Neuroimage 53 (3),
try 2, e112. 1117–1125.
Wood, J.D., Bonath, F., Kumar, S., Ross, C.A., Cunliffe, V.T., 2009. Disrupted-in-schizophre- Zuliani, R., Moorhead, T.W., Bastin, M.E., Johnstone, E.C., Lawrie, S.M., Brambilla, P.,
nia 1 and neuregulin 1 are required for the specification of oligodendrocytes and O'Donovan, M.C., Owen, M.J., Hall, J., McIntosh, A.M., 2011. Genetic variants in the
neurones in the zebrafish brain. Hum. Mol. Genet. 18 (3), 391–404. ErbB4 gene are associated with white matter integrity. Psychiatry Res. 191 (2), 133–137.

Please cite this article as: Voineskos, A.N., Genetic underpinnings of white matter ‘connectivity’: Heritability, risk, and heterogeneity in
schizophrenia, Schizophr. Res. (2014), http://dx.doi.org/10.1016/j.schres.2014.03.034

You might also like