ArticlePDF AvailableLiterature Review

Developmental Genes and Regulatory Proteins, Domains of Cognitive Impairment in Schizophrenia Spectrum Psychosis and Implications for Antipsychotic Drug Discovery: The Example of Dysbindin-1 Isoforms and Beyond

Frontiers
Frontiers in Pharmacology
Authors:

Abstract

Alongside positive and negative symptomatology, deficits in working memory, attention, selective learning processes, and executive function have been widely documented in schizophrenia spectrum psychosis. These cognitive abnormalities are strongly associated with impairment across multiple function domains and are generally treatment-resistant. The DTNBP1 (dystrobrevin-binding protein-1) gene, encoding dysbindin, is considered a risk factor for schizophrenia and is associated with variation in cognitive function in both clinical and nonclinical samples. Downregulation of DTNBP1 expression in dorsolateral prefrontal cortex and hippocampal formation of patients with schizophrenia has been suggested to serve as a primary pathophysiological process. Described as a “hub,” dysbindin is an important regulatory protein that is linked with multiple complexes in the brain and is involved in a wide variety of functions implicated in neurodevelopment and neuroplasticity. The expression pattern of the various dysbindin isoforms (-1A, -1B, -1C) changes depending upon stage of brain development, tissue areas and subcellular localizations, and can involve interaction with different protein partners. We review evidence describing how sequence variation in DTNBP1 isoforms has been differentially associated with schizophrenia-associated symptoms. We discuss results linking these isoform proteins, and their interacting molecular partners, with cognitive dysfunction in schizophrenia, including evidence from drosophila through to genetic mouse models of dysbindin function. Finally, we discuss preclinical evidence investigating the antipsychotic potential of molecules that influence dysbindin expression and functionality. These studies, and other recent work that has extended this approach to other developmental regulators, may facilitate identification of novel molecular pathways leading to improved antipsychotic treatments.
Developmental Genes and
Regulatory Proteins, Domains of
Cognitive Impairment in
Schizophrenia Spectrum Psychosis
and Implications for Antipsychotic
Drug Discovery: The Example of
Dysbindin-1 Isoforms and Beyond
John L. Waddington
1,2
*, Xuechu Zhen
2
and Colm M. P. OTuathaigh
1,3
1
School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland,
2
Jiangsu Key
Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College
of Pharmaceutical Sciences, Soochow University, Suzhou, China,
3
Medical Education Unit, School of Medicine, Brookeld
Health Sciences Complex, University College Cork, Cork, Ireland
Alongside positive and negative symptomatology, decits in working memory, attention,
selective learning processes, and executive function have been widely documented in
schizophrenia spectrum psychosis. These cognitive abnormalities are strongly associated
with impairment across multiple function domains and are generally treatment-resistant.
The DTNBP1 (dystrobrevin-binding protein-1) gene, encoding dysbindin, is considered a
risk factor for schizophrenia and is associated with variation in cognitive function in both
clinical and nonclinical samples. Downregulation of DTNBP1 expression in dorsolateral
prefrontal cortex and hippocampal formation of patients with schizophrenia has been
suggested to serve as a primary pathophysiological process. Described as a hub,
dysbindin is an important regulatory protein that is linked with multiple complexes in the
brain and is involved in a wide variety of functions implicated in neurodevelopment and
neuroplasticity. The expression pattern of the various dysbindin isoforms (-1A, -1B, -1C)
changes depending upon stage of brain development, tissue areas and subcellular
localizations, and can involve interaction with different protein partners. We review
evidence describing how sequence variation in DTNBP1 isoforms has been differentially
associated with schizophrenia-associated symptoms. We discuss results linking these
isoform proteins, and their interacting molecular partners, with cognitive dysfunction in
schizophrenia, including evidence from drosophila through to genetic mouse models of
dysbindin function. Finally, we discuss preclinical evidence investigating the antipsychotic
potential of molecules that inuence dysbindin expression and functionality. These
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16381
Edited by:
Adrian Preda,
University of California,
Irvine, United States
Reviewed by:
Kazutaka Ohi,
Kanazawa Medical University,
Japan
Francesco Papaleo,
Italian Institute of Technology (IIT),
Italy
Antonieta Lavin,
Medical University of South Carolina,
United States
*Correspondence:
John L. Waddington
jwadding@rcsi.ie
Specialty section:
This article was submitted to
Neuropharmacology,
a section of the journal
Frontiers in Pharmacology
Received: 28 July 2019
Accepted: 16 December 2019
Published: 29 January 2020
Citation:
Waddington JL, Zhen X and
OTuathaigh CMP (2020)
Developmental Genes and Regulatory
Proteins, Domains of Cognitive
Impairment in Schizophrenia
Spectrum Psychosis and
Implications for Antipsychotic
Drug Discovery: The Example
of Dysbindin-1
Isoforms and Beyond.
Front. Pharmacol. 10:1638.
doi: 10.3389/fphar.2019.01638
REVIEW
published: 29 January 2020
doi: 10.3389/fphar.2019.01638
studies, and other recent work that has extended this approach to other developmental
regulators, may facilitate identication of novel molecular pathways leading to improved
antipsychotic treatments.
Keywords: cognitive decits, developmental gene, schizophrenia, antipsychotic drug development, dysbindin-1
INTRODUCTION
Schizophrenia, Cognitive Impairment, and
Functional Disability
Schizophrenia is a complex psychiatric disorder that is the
exemplar of a broader spectrum of psychotic illness in which
interactive contributions from genetic and environmental factors
play critical roles in etiology and pathobiology (Owen et al.,
2016); hereafter, we use the term schizophrenia as shorthand for
this spectrum of illness. In intimacy with positive, psychotic
symptoms that dene this spectrum, dysfunction in working
memory, attention, processing speed, visual and verbal learning,
reward-related learning, and prominent decits in executive
function have been extensively documented in schizophrenia
(Kahn and Keefe, 2013). Impairment across these domains, in
juxtaposition with negative symptoms, has long been accepted as
core features of schizophrenia that contribute substantively to
disability and are generally treatment refractory (Green
et al., 2019).
Several authors have recommended inclusion of cognitive
impairment in formal diagnostic criteria for this illness, as well as
highlighting the need for research on developing new and more
effective treatments to enhance cognitive abilities therein (Kahn
and Keefe, 2013;Schaefer et al., 2013;Mark and Toulopoulou,
2016). Cognitive decits associated with schizophrenia are
observed in unaffected family members of individuals with the
disorder (Toulopoulou et al., 2019) and the presence of
schizophrenia-associated cognitive impairment in children can
predict increased risk for the illness (Meier et al., 2014;Agnew-
Blais et al., 2015).
Schizophrenia, Cognitive Impairment, and
Genetics
Twin studies continue to afrm a primary role for genetic factors
in the etiology of schizophrenia (Hilker et al., 2018), in
association and interaction with environmental adversities
(Guloksuz et al., 2019). However, despite considerable
endeavour during the past 20 years, genomic studies of
schizophrenia have arguably failed to provide the expected
answers, or have highlighted the difculties in elucidating a
complex, heterogeneous, and polygenic genetic architecture.
The existing literature indicates contributions from multiple
common and occasional rare variants that may interact in
conferring risk for schizophrenia (Bergen et al., 2019;
Weinberger, 2019). A key challenge in this eld involves the
translation of advances in our understanding of the genetics of
schizophrenia and the mechanistic basis of these associations
into tangible improvements in patient-centred care and
antipsychotic drug discovery.
In support of a common disease-common allele hypothesis,
whereby much of risk for schizophrenia is conferred via the
cumulative effect of multiple common alleles, a landmark
genome wide association study of more than 36,000 cases and
over 113,000 controls identied 108 loci for common risk
variants that achieved genome-wide signicance
(Schizophrenia Working Group of the Psychiatric Genomics
Consortium, 2014). These risk variants are involved in several
known processes, including synaptic plasticity and within the
major histocompatibility complex, but also in as-yet unknown
functions. In the most recent analysis completed by the same
consortium, which involved 30,000 additional subjects, the
number of GWAS-signicant loci was expanded to 246
(Weinberger, 2019).
Polygenic risk scores (PRS) represent an aggregate measure of
genetic risk as they consider the additive effects of all signicant
variation across multiple genes and regulatory areas across the
entire genome (Jones et al., 2016;Xavier et al., 2018;
Toulopoulou et al., 2019). The PRS is calculated by summing
all the alleles (weighted by their individual odds ratios) that have
been associated with an illness in the latest GWAS data set for
that illness. In the most recent study of the effect of schizophrenia
risk alleles on cognition (Richards et al., 2019), schizophrenia
PRS were associated more strongly with case-control cognitive
differences as opposed to variation within cases.
Copy number variants (CNVs), both rare de novo and
inherited, make only a minor contribution to population risk
variation despite larger effect sizes (Manolio et al., 2009;
Malhotra and Sebat, 2012;Kotlar et al., 2015;Genovese et al.,
2016). In schizophrenia, these rare variants are found at loci
containing genes implicated in synaptic function as well as
neurodevelopmental processes linked with glutamatergic
function pathways (Kirov et al., 2012;Marshall et al., 2017).
Genovese et al. (2016) reported that genes implicated in synaptic
function potentially explained more than 70% of the exome
enrichment in damaging ultra-rare variants that contribute to
schizophrenia. Some authors have proposed a merging of
common allele and rare variant mechanisms, suggesting that
individuals with schizophrenia having well-characterized
pathogenic CNVs also associate with an excess burden of
common risk alleles (Tansey et al., 2016;Bergen et al., 2019).
More recent hypotheses suggest that the complex genetic
architecture of schizophrenia may be better explained in terms of
an omnigenicframework. This hypothesis (Boyle et al., 2017)
posits that for complex traits such as schizophrenia, GWAS may
identify genes more central to a disease process. However, these
coregenes function in a cellular network that is associated with
the vastly more numerous other peripheralgenes that have less
evident relationship to disease but are able to inuence the
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16382
function of coregenes. Indeed, recent evidence suggests in
schizophrenia a coregene set that appears to contribute to risk
to a greater extent than an omnigenic background effect
(Rammos et al., 2019).
SCHIZOPHRENIA, COGNITIVE
IMPAIRMENT, AND VARIATION IN
DTNBP1
Dysbindin-1 is a coiled-coil-containing protein encoded by
DTNBP1 (Dystrobrevin Binding Protein 1, 6p22.3), a gene that
has been linked with cognitive and anatomical endophenotypes
in both patients with neuropsychiatric disorders as well as
nonclinical samples (Ayalew et al., 2012;Wang et al., 2017;
Savage et al., 2018). An initial report of genetic linkage to
schizophrenia on chromosome 6p24-22 (Straub et al., 1995)
was followed by multiple individual replications and
conrmatory meta-analyses of DTNBP1 (Allen et al., 2008;
Ayalew et al., 2012;Wang et al., 2017); any concern that such
ndings have not been prominent in GWAS studies to date
(Farrell et al., 2015) must be juxtaposed with increasing
recognition that GWAS cannot in itself be considered
denitive on such issues (Tam et al., 2019;seealso
Schizophrenia, Cognitive Impairment, and Genetics above), and
that the GWAS focus on diagnosis and symptom severity scores
is likely to lead to neglect for genes specically linked with
cognitive dysfunction in schizophrenia. Additionally, the
expression of dysbindin-1 and its isoforms is disrupted in
brain tissue from schizophrenia patients (see Schizophrenia
Pathobiology and Dysbindin Isoforms below). Importantly for
issues to be considered further below, variations in DTNBP1
associated with schizophrenia (SNPs and haplotypes) are located
in intron or promoter regions and almost all are located in the N-
terminus of the gene (Guo et al., 2008).
Several studies have investigated the role of this gene in
cognitive decits of schizophrenia. For example, DTNBP1
haplotypes are associated with greater decline in IQ (Burdick
et al., 2007) and impairment in spatial working memory
(Donohoe et al., 2007;Donohoe et al., 2008;Donohoe et al.,
2010) and attentional/vigilance (Baek et al., 2012), verbal and
visual working memory and speed of processing (Varela-Gomez
et al., 2015), as well as executive function (Scheggia et al., 2018).
Such ndings have been less evident on subdividing
schizophrenia patients into cognitive-decit and cognitive-
sparing groups (Peters et al., 2008). Other studies have also
failed to report a relationship between DTNBP1 variants and
general tests of cognitive ability in patients with schizophrenia
and rst-degree relatives compared to controls (Strohmaier et al.,
2010;Chow et al., 2018). Additionally, while some authors have
shown that DTNBP1 variation in healthy controls and patients is
associated with performance across a diverse range of cognitive
tests (e.g. Burdick et al., 2006), a meta-analysis of 11 articles
examining the relationship between dysbindin and general
cognitive ability revealed only a modest relationship (Zhang
et al., 2010). These results support involvement of the
DTNBP1 gene in selective domains of cognition rather than
non-specic cognitive ability (Luciano et al., 2009). Despite
preliminary evidence to suggest dysbindin involvement in
cognitive function in patients with brain tumours (Correa
et al., 2016), there is a paucity of studies examining the role of
dysbindin in other neurological and neuropsychiatric disorders
where symptoms include cognitive dysfunction.
SCHIZOPHRENIA PATHOBIOLOGY AND
DYSBINDIN ISOFORMS
Dysbindin expression levels vary across neuronal populations
throughout the brain and are particularly abundant in the
dentate gyrus of the hippocampal formation (Talbot et al.,
2004;Talbot et al., 2006), with dysbindin localization in the
CNS occurring in both neurons and, at comparable levels, in glia
(Ghiani et al., 2009;Shao et al., 2011). DTNBP1 encodes for three
majorspliceisoformsofdysbindin-1:1A,1B,and1C.
Dysbindin-1A is most highly concentrated in postsynaptic
density fractions and dysbindin-1B is most abundant in
synaptic vesicle fractions; similar to dysbindin-1A, dysbindin-
1C is most highly concentrated in postsynaptic density fractions;
nuclear localization has also been reported for dysbindin-1A and
-1B (Oyama et al., 2009;Talbot et al., 2011).
Sequence variation might be a mechanism by which the
function of DTNBP1 differs between schizophrenia cases and
controls. In this scenario, instead of a simple incorrectly encoded
protein, the amount of DTNBP1 expression may be affected.
Indeed, it has been shown that variations in DTNBP1 affect its
expression in the human brain (Bray et al., 2003;Bray et al., 2005;
Bray et al., 2008).
The hippocampal formation plays a fundamental role in
working and episodic memory, highlighting the possible role of
dysbindin in cognitive processes that are believed to be critical to
schizophrenia (Tamminga et al., 2010). Talbot et al. (2004) reported
that schizophrenia patients show reduction of dysbindin-1 protein
in the principal neurons of CA2 and CA3 and especially in the
dentate gyrus of the hippocampal formation. A subsequent study
from the same group reported that dysbindin proteins in the
dentate gyrus are essentially present postsynaptically (Talbot
et al., 2006). Talbot et al. (2011) then reported that in
schizophrenia patients levels of dysbindin-1B and -1C, but not
-1A, are decreased in the hippocampal formation, while levels of
dysbindin-1A, but not -1B or -1C, are decreased in the superior
temporal gyrus. Furthermore, transcripts encoding dysbindin-1B
are upregulated in peripheral bloodleukocytesofpatientswith
schizophrenia relative to controls, with an intronic DTNBP1
variant associated with schizophrenia affecting splicing and
leading to specic over-expression of dysbindin-1B (Xu et al., 2015).
Similarly to the hippocampal formation, the dorsolateral
prefrontal cortex (DLPFC) is crucially involved in working and
episodic memory, highlighting a further possible contribution of
dysbindin to cognitive decits encountered in schizophrenia
(Lewis et al., 2008). Weickert et al. (2004) reported varying
levels of DTNBP1 mRNA within the DLPFC, with higher
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16383
expression found in layers IV and V; in DLPFC of schizophrenia
patients decreases in DTNBP1 mRNA were reported in layers II,
III, V and VI, but not in layers I and IV. Subsequently, decreases
in DTNBP1 mRNA were reported in the hippocampal formation
(Weickert et al., 2008). However, a subsequent study from the
same team could not wholly conrm these ndings (Fung et al.,
2011). Tang et al. (2009) reported reductions in dysbindin-1C,
but not in -1A or -1B, in DLPFC in the absence of changes in
mRNA levels of the three isoforms. It should be noted that
laminar-specic alterations in dysbindin mRNA and proteins
may have escaped detection with the homogenate-based
approach used in both studies (Tang et al., 2009;Fung et al.,
2011). More recently, dysbindin-1B, but not -1A, protein levels
were found to be higher in the DLPFC in schizophrenia
(Konopaske et al., 2018).
SCHIZOPHRENIA, NEURODEVELOPMENT,
NEUROPLASTICITY, AND DYSBINDIN
A wealth of epidemiological, clinical, and biological evidence
now indicates that the origins of schizophrenia are to be found
primarily in the early phases of fetal brain development: during
this period a variety of factors, mainly genetic but also including
environmental adversities, interact to disrupt brain
morphogenesis (Waddington et al., 2012;Birnbaum and
Weinberger, 2017); thereafter, endogenous and exogenous
factors and processes can act to further sculptbrain
development and maturation as a substrate from which the
diagnostic symptoms and disabilities of clinical psychosis
emerge, most typically during early adulthood (Waddington
et al., 2012;Weinberger, 2017).
Regulation of brain development involves a multitude of
genetic processes and associated proteins that act and interact
sequentially to determine normaladult cerebral structure and
function. Among these, dysbindin is one important regulator
that is linked with multiple complexes in the brain and numerous
diverse functions implicated in neurodevelopment and
neuroplasticity. However, while the different dysbindin isoform
proteins might interact with different partners, the majority of
studies do not provide sufcient information to conrm or refute
isoform-specic processes.
Promotion of Cell Growth, Proliferation,
and Antiapoptotic Effects
Dysbindin promotes activation of Akt (Numakawa et al., 2004),
which mediates growth and proliferation of cells (Manning and
Cantley, 2007). Akt has itself been proposed as a risk gene for
schizophrenia and a target for antipsychotic drug development
(Zheng et al., 2012;Enriquez-Barreto and Morales, 2016). Studies
have reported that dysbindin is present in neurites and in axonal
growth cones and that down regulation of dysbindin results in
aberrant organisation of the actin cytoskeleton at the tips of
neurites of differentiating cells, with shortening of such neurites
(Kubota et al., 2009;Taneichi-Kuroda et al., 2009).
BLOC-1 and Endosomal Trafcking
Dysbindin is a stable component of the multi-subunit complex
BLOC-1 (biogenesis of lysosome-related organelles complex-1;
DellAngelica, 2004;Li et al., 2004;Lee et al., 2012). Among the
subunits of BLOC-1, dysbindin interacts directly with pallidin,
MUTED, and snapin (Li et al., 2003;Starcevic and DellAngelica,
2004). Mice containing constitutive deletion of the pallidin gene
show performance decits in two different measures of
recognition memory: the novel object recognition task and a
measure of social novelty recognition (Spiegel et al., 2015).
Studies have also reported an association between markers at
MUTED and schizophrenia (Straub et al., 2005; but see also
Gerrish et al., 2009). Moreover, both dysbindin and MUTED
siRNA increased cell surface dopamine (DA) D2 receptors (D2R)
and blocked DA-induced D2R internalization in human and rat
cells. In contrast, decreased dysbindin altered neither D1
receptors (D1R) levels nor their basal expression or DA-
induced internalisation (Iizuka et al., 2007). Such an increase
in D2R signalling could contribute to the imbalance in DAergic
neurotransmission characteristic of schizophrenia, i.e.,
hyperfunction through D2R and attenuation of such
hyperfunction by current D2R antagonist antipsychotics
(McCutcheon et al., 2019).
A recent review has comprehensively addressed the
relationship and interactions between BLOC-1 genes/proteins
and cognitive phenotypes observed in neurodevelopmental
disorders (Hartwig et al., 2018). The predominant theme is
that BLOC-1 subunits in brain areas linked to cognitive
functions are part of a more complex set of molecular
interactions, including proteins implicated in schizophrenia
such as disrupted-in-schizophrenia-1 and SNARE (Talbot,
2009;Hartwig et al., 2018).
Dysbindin and Dystrophin-Associated
Protein Complex
Dysbindin binds to the dystrobrevins, which are components of the
dystrophin-associated protein complex (DPC) (Benson et al., 2001;
but see also Nazarian et al., 2006). In the brain, several DPC-like
complexes have been implicated in cognitive impairment
commonly found in patients with Duchenne muscular dystrophy
(DMD) (Blake and Kröger, 2000). Indeed, DPC is necessary for
maturation and function of a subset of inhibitory synapses (Grady
et al., 2006). Lack of dystrophin in the mdx mouse model of DMD
produces an altered distribution of dysbindin in the brain,
suggesting a role for dysbindin-1 in the cognitive impairment
observed in DMD patients (Sillitoe et al., 2003).
NONISOFORM- AND ISOFORM-SPECIFIC
GENETIC MUTATION MODELS FOR
DYSBINDIN-1 FUNCTION
Drosophila Mutants
One dysbindin protein has been demonstrated in drosophila,
known as CG6865-PA. While the amino acid sequence of its
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16384
coiled-coil domain is closely related to that of all known
orthologs of dysbindin, neither its N-terminal nor its C-
terminal region is closely related to corresponding regions in
any vertebrate dysbindin ortholog. CG6865-PA shares 28%
amino acid identity with human dysbindin-1A (Guo et al.,
2009). Drosophila dysbindin mutants demonstrate impaired
neurotransmission, disrupted synaptic homeostasis, presynaptic
and postsynaptic morphological alterations, and disruption in
short term memory (Dickman and Davis, 2009;Cheli et al., 2010;
Shao et al., 2011;Gokhale et al., 2015;Mullin et al., 2015;Gokhale
et al., 2016).
Shao et al. (2011) reported that neuronal disruption of
dysbindin function in drosophila was associated with
hypoglutamatergic transmission and decits in working
memory, while disruption of dysbindin function in glial cells
was associated with hyperdopaminergia and locomotor
hyperactivity; this latter effects was mediated via a reduction in
the protein Ebony, a glia-specic beta-alanyl biogenic amine
synthetase involved in metabolic degradation of biogenic
amines in the nervous system. This study reveals distinct
functions of dysbindin in neurons and glial cells and highlights
the potential of new therapeutics for schizophrenia that target
glial cells (Bernstein et al., 2015). As reduction of dysbindin in
drosophila impacts on short-term memory, dysbindin-dependent
pathways may shed further light on the mechanisms of cognitive
dysfunction in schizophrenia (Larimore et al., 2014).
Mutant Mouse Models
Genetic mouse models enable etiologically related investigation
of the pathophysiology of schizophrenia, providing means to
advance target discovery to improve treatment for psychotic
illness (OTuathaigh and Waddington, 2015). The development
of new and effective drug therapies generally requires analysis of
the intact brain in valid preclinical models, most commonly
involving rodents (Dawson et al., 2015). Therefore, integrated
research strategies for the delineation of animal models, based on
characterization of cognitive decits and underlying
mechanisms, have considerable translational potential
(Diamantopoulou and Gogos, 2019). In particular, based on
the role of working memory in supporting a variety of
cognitive abilities and its association with decits in social and
occupational functioning in schizophrenia, characterization of
these processes in any mutant model is fundamental to
understanding the relevance of the experimental model to
cognitive dysfunction in this disorder.
While dysbindin-1A is highly conserved among vertebrates,
there is no ortholog of human dysbindin-1B in mice; dysbindin-
1C is a 270 amino acid protein in humans and a 271 amino acid
protein in mice (Talbot, 2009). A spontaneous deletion mutation
in DTNBP1 occurred in the DBA/2J strain, leading to complete
absence of dysbindin-1A and 1C proteins in homozygous mice
and reduced expression levels in heterozygous mice; the sandy
(sdy) coat colour of homozygous mutants gives the strain its
name (Swank et al., 1991). While the earliest studies were carried
out in mice with the original DBA/2J background (sdy/DBA), the
mutation has been transferred subsequently onto a C57BL/6J
background (sdy/B6). DBA/2J mice are homozygous for at least
six mutations, of which four are associated with neural
impairments (Cox et al., 2009;Talbot, 2009). These appear to
account for a number of auditory and visual decits, when
compared with C57BL/6 mice.
Sandy Mice on a DBA/2J Background
Studies indicated abnormalities related to cognitive impairment
(disturbance of long-delay recognition memory during an object
recognition test) in homozygous sdy/DBA mice without
abnormalities related to basal activity levels or anxiety (Feng
et al., 2008). These authors also conrmed a previous nding of
direct interaction with and decrease in the steady-state level of
snapin (a SNAP-25 binding protein), suggesting an upstream
regulatory role of dysbindin on neurotransmitter release via
snapin. Takao et al. (2008) also reported cognitive decits,
including impairment in long-term memory retention (Barnes
maze test) and in working memory (T-maze, forced alternation
task). Bhardwaj et al. (2009) also demonstrated decits in short-
term memory (object recognition memory test) and stronger
dependent memory for fearful events in sdy/DBA mice. In a
subsequent study such mutants showed impaired recognition
memory in the novel object recognition and social recognition
paradigms (Spiegel et al., 2015).
Kobayashi et al. (2011) demonstrated hypersensitivity to both
serotonergic and DAergic modulation of DG-to-CA3 signal
transmission in 4-month-old to 6-month-old male
homozygous sdy/DBA mice. These authors also reported
decreased expression of D1R mRNA in the hippocampus that
could contribute to changes in synaptic modulation. Jentsch et al.
(2009) reported impaired glutamatergic transmission through
potentially both presynaptic and postsynaptic mechanisms in the
prefrontal cortex (PFC) of heterozygous and homozygous sdy/
DBA mice. While dysbindin deletion appears to decrease
glutamate release at the axon terminal, it may also result in an
increase in excitability that might be due to reduction in
neuronal dendritic branching and/or transmitter release in
GABAergic interneurons. This study also reported
homozygous sdy/DBA mice to show impairment in a spatial
working memory task (delayed nonmatch-to-position test), with
heterozygous mutants showing an intermediate level of
performance. Collectively, these results suggest that dysbindin
dysregulation might contribute to the cognitive symptoms of
schizophrenia by decreasing glutamatergic transmission, at least
in the prefrontal cortex.
Sandy Mice on a C57BL/6J Background
Studies involving the sandy mouse on a C57BL6 background also
show cognitive decits. Cox et al. (2009) rst reported
impairment in spatial memory and/or initial learning and
acquisition (Morris water maze) in sdy/B6 mice. Carlson et al.
(2011) reported reductions in sdy/B6 mice of auditory-evoked
response adaptation, prepulse inhibition, and evoked g-activity,
which is most frequently linked with disrupted inhibition and
reduction in parvalbumin (PV)-positive interneuron activity.
Indeed, subsequent analyses revealed reduction of fast-spiking
GABAergic inhibition and fewer PV cells in the hippocampus of
sdy/B6 mice.
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16385
As previously shown in sdy/DBA mice (Jentsch et al., 2009),
homozygous sdy/B6 mice showed a decrease in working memory
performance (delayed nonmatch-to-position test) compared to
wildtypes; these results correlated with degree of expression of the
NR1 subunit of the NMDA receptor. Other studies have shown an
altered GluN2B-GluN2A switch in the hippocampus and cortex of
dysbindin mutant mice (Sinclair et al., 2016). Genetic loss of
dysbindin-1 expression has also been shown to impact on NMDA
receptor-dependent synaptic plasticity in the hippocampus in both
sdy/DBA and sdy/B6 mice (Glen et al., 2014). Another study
demonstrated that loss of dysbindin-1 leads to reduced mGluR1
signalling in the hippocampus, which is associated with altered
hippocampal synaptic plasticity in sdy/B6 mice (Bhardwaj et al.,
2015). Overall, these results suggest a role for dysbindin in the
regulation of NMDA and cognition that might be associated with
cognitive decits in schizophrenia.
Papaleo et al. (2012) reported differences in reference memory
(T-maze) or a habituation/dishabituation task (olfactory
discrimination test) in sdy/B6 mice, which demonstrated more
rapid acquisition of a working memory task (T-maze) and overall
worse performance on the same test under more demanding
(proactive interference) or more stressful (transfer in a new cage)
conditions relative to controls. Other data from the same group
suggested that interval timing decits may be a crucial component
of abnormal cognition in sdy/B6 mice and that these decits might
be improved with increased training and experience (Carr
et al., 2012).
Subsequent experiments were conducted to evaluate D2R
modulation of these behavioral effects (Papaleo et al., 2012).
While chronic D2R agonist administration did not affect
acquisition in the working memory test, under more
demanding conditions this treatment impaired working
memory performance in a manner comparable to that
observed in sdy/B6 mice. These authors also reported
enhanced excitability and excitatory inputs to pyramidal
neurons in medial-PFC (mPFC) layer II/III (the layers
principally involved in intracortical projections), as well as
decreased excitatory inputs to fast-spiking GABAergic neurons
(Papaleo et al., 2012). DAergic modulation of excitatory synaptic
transmission in layer II/III pyramidal neurons involves CaM-
kinase-II (CaMKII)-dependent mechanisms that have been
implicated in learning and memory processes (Gonzalez-Islas
and Hablitz, 2003). While sdy/B6 mutants displayed lower
CaMKII and CaMKKbprotein levels in mPFC, control mice
chronically treated with a D2R agonist showed the same specic
reductions (with no change in CaMKIV and CaMKKalevels;
Papaleo et al., 2012). Overall, these results suggest that some
effects of dysbindin on cognition, associated with changes in
cortical activity and CaMK components of the mPFC, are
induced via upregulation of D2R. Consistent with a dysbindin-
associated increase in D2 receptors, dysbindin deciency was
associated with an antipsychotic-dependent increase in the ratio
between the D2Short (D2S) and D2Long (D2L) isoforms in the
PFC of mice, which is associated with potentiation of cortical
presynaptic D2R signalling (Scheggia et al., 2018).
It has been proposed that DA signalling and PFC-dependent
cognition follow an inverted U-shaped relationship, by which
both inadequate or excessive DAergic signalling has a disruptive
effect on cognitive performance that reects an imbalance in
dopamine D1/D2R activation (Vijayraghavan et al., 2007).
Consistent with this hypothesis, Papaleo et al. (2014)
investigated the effects on working memory performance of
simultaneous disruption of dysbindin and the gene encoding
the COMT enzyme, which plays a central role in the degradation
of DA in PFC. They showed that while disruption of either
dysbindin or COMT alone produced an improvement in
working memory performance in the discrete paired-trial
variable-delay T-maze task, mice with disruption of both genes
demonstrated impaired working memory performance. These
authors demonstrated a similar epistatic interaction on working
memory performance and accompanying activation of PFC in
humans. Based on the literature, the behavioral effects of this
genetic interaction in both human and murine working memory
paradigms reect the effects of both genes on D1/D2R signalling
in PFC. The same authors recently reported, in both mice and
patients with schizophrenia, an interaction between functional
variation in both dysbindin and D3R genes and working memory
and executive function performance (Leggio et al., 2019).
Specically, simultaneous reduction of D3R and dysbindin
function was associated with improved performance in tasks
accessing these cognitive domains. It was shown that this
epistatic interaction was associated with a shift in the balance
between D2R and D3R receptor expression in the PFC, leading to
an increase in D2R signalling in that brain region.
Dysbindin-1A Mutant Mice
We have recently described the generation of a genetic mouse
model with isoform-specic deletion of dysbindin-1A protein
(Petit et al., 2017). Initial phenotypic characterization showed
sexually dimorphic phenotypes, with female knockouts (KO)
being more reactive to stressful situations and male KO showing
increased exploration during initial exposure to a novel
environment that may be related to some disruption in
habituation and dysregulation of hippocampus-dependent
working memory function. No effect of genotype was observed
during acquisition or during performance in long-term
(olfactory) memory or either a conventional spatial working
memory task (spontaneous alternation) or a low-interference
delay-dependent working memory task; however, in a high-
interference task variant, male KO mutants showed
impairment in vulnerability to interference (Petit et al., 2017).
Dysbindin-1B Mutant Mice
Another recent study, which involved a transgenic mouse model
expressing the human dysbindin-1B isoform, documented the
presence of middle- and late-stage apoptosis in the hippocampus
of dysbindin-1B mutants; in a T-maze alternation task, these
mutants also showed decits in working memory (Yang and Xu,
2017). Dysbindin-1B expression was also shown to impair spatial
learning in the Morris water maze (Yang et al., 2016).
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16386
Dysbindin Gain-of-Function Mutant Mice
A further study described the behavioral phenotype of a gain-of-
function dysbindin mutant (Shintani et al., 2014). Specically,
these authors generated a transgenic model that expressed the
human dysbindin-1A isoform. Such mutants displayed unaltered
sensory or motor behavior and no changes in anxiety,
sensorimotor gating, or exploratory behavior; they evidenced
heightened locomotor responsivity to methamphetamine and
upregulation and downregulation of several genes in PFC,
including decreases in Arc and Egr2 expression.
Spp Dysbindin Mutant Mice
Most recently, behavioral and cognitive phenotypes have been
investigated in mice containing a single point mutation in the salt
and pepper (spp) allele of DTNBP1 (Chang et al., 2018). While
spp mutants did not exhibit any decits in working memory (T-
maze alternation task), some decits in aspects of recognition
memoryperformancewereobserved.Thesemicealso
demonstrated a reduction in the schizophrenia-associated
SNAP-25 protein in PFC. No data are available regarding
relative isoform expression in the brains of spp mutants.
Dysbindin Mutation × Exposure to
Environmental Risk Factors
Emphasis on genetic risk factors for schizophrenia should be
tempered by evidence from investigations in twins (Hilker et al.,
2018) and epidemiological studies indicating a role for
environmental factors that may act both independently and via
gene-environment interactions (Guloksuz et al., 2019).
Therefore, animal models of neuropsychiatric disorders can
and should investigate experimental induction of/exposure to
nongenetic risk factors, including psychosocial stress and
biological insults. Though there is some debate around
common pathological mechanisms underlying the impact of
such manipulations on schizophrenia-related phenotypes
(Bradshaw and Korth, 2019), a combination of dysbindin gene
mutation and postnatal exposure to Poly I:C (a model of immune
activation in relation to schizophrenia) has been investigated. In
sdy/B6 mice, exposure to Poly I:C resulted in decits in prepulse
inhibition and recognition memory, together with ndings
contrary to expectations, such as reduced locomotion; sdy/B6
mice treated with Poly I:C also displayed attenuated contextual
and cue-dependent fear conditioning memory (Al-Shammari
et al., 2018).
DYSBINDIN-1 AND PUTATIVE
TREATMENT STRATEGIES FOR
COGNITIVE DYSFUNCTION IN
SCHIZOPHRENIA
Consistent with the proposed role for the D2R in the PFC in
mediating the putative effects of dysbindin variation on higher
order cognitive processes (Papaleo et al., 2012;Scheggia et al.,
2018;Leggio et al., 2019), a series of elegant translational studies
have shown that patients and mice with genetic variation
associated with decreased dysbindin-1 expression demonstrate
improved responsivity to the effects of antipsychotic medication
on executive function (Scheggia et al., 2018). Mechanistic
interrogation of this interaction revealed that the cognitive
response to antipsychotics was mediated by enhanced
presynaptic D2R in PFC (Scheggia et al., 2018). A subsequent
study revealed a putative role for epistatic D3R-dysbindin-1
interaction and executive function decits in schizophrenia,
highlighting the viability of D3R modulation as a treatment
target for cognitive dysfunction (Leggio et al., 2019).
As outlined above, dysbindin disruption has also been
proposed to underlie cognitive impairment observed in
patients with Duchenne muscular dystrophy. A recent study
investigated the effects of treatment with the cacao avonoid
(-)-epicatechin on brain dysbindin levels in the mdx mutant, a
genetic mouse model of Duchenne muscular dystrophy; such
treatment partially reversed genotype-dependent reductions in
protein levels of the dystrophin-associated protein complex as
well as dysbindin protein in PFC (Estrada-Mena et al., 2017). It
was not reported whether this partial recovery was also observed
at a behavioral level.
Investigation of transcriptional responses of developing
hippocampal neurons in sdy/B6 mutants revealed not only the
characteristic GABAergic interneuron deciency but also
changes in expression of the cation-chloride cotransporters
NKCC1 and KCC2 during hippocampal development
(Larimore et al., 2017). NKCC1 and KCC2 expression changes
have been documented in the brains of patients with
schizophrenia (Hyde et al., 2011;Sullivan et al., 2015)and
NKCC1 agents, including the NKCC1 chloride antagonist
bumetanide, have been studied in schizophrenia. Bumetanide
reduced hallucinations in a case of schizophrenia (Lemonnier
et al., 2016) and in a randomized, double-blind placebo-
controlled clinical trial bumetanide treatment reduced
hallucinations (Rahmanzadeh et al., 2017a) but did not exert
broad antipsychotic activity (Rahmanzadeh et al., 2017b);
cognition was not specically investigated.
Consistent with the observed involvement of dysbindin in
hippocampal and PFC glutamatergic function, a recent study
investigated the impact of pharmacological enhancement of
endogenous levels of brain-derived neurotrophic factor
(BDNF) on dysbindin-related reduction of presynaptic calcium
levels in PFC and social recognition memory in sdy/B6 mice
(Saggu et al., 2013). Systemic treatment with ngolimod, a
sphingosine 1-phosphate receptor modulator that has been
shown to increase endogenous BDNF levels, attenuated decits
in recognition memory and sociability and in presynaptic
calcium and BDNF levels in the PFC of sdy/B6 mice (Becker-
Krail et al., 2017). BDNF levels in schizophrenia patients have
been associated with impairment across multiple cognitive
domains (Man et al., 2018;Yang et al., 2019). A recent review
has highlighted that the BDNF gene should be prioritised for
pharmacogenetic research into antipsychotic drugs and potential
relevance to treatment response and adverse effects (Han and
Deng, 2018).
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16387
Similarly, administration of CDPPB, a positive allosteric
modulator of MGluR5, restored short-term recognition and
spatial memory decits (Morris water maze) in sdy/B6 mice
(Bhardwaj et al., 2015). Given their effects on glutamatergic
signalling, and particularly on NMDA receptor activity,
MGluR5 modulators have been suggested to represent
promising treatment targets for neuropsychiatric disorders that
are characterized by cognitive decits.
Expression of SREBP1, a sterol regulatory element binding
protein (SREBP) that regulates the expression of genes implicated
in biosynthesis of fatty acids, cholesterol, triglycerides, and
phospholipids, is reduced in sdy/B6 mice and in schizophrenia
patients; activation of SREBP1 and Arc, a protein implicated in
memory and cognition, is reduced in sdy/B6 mice and both of
these decits were restored by treatment with clozapine,
suggesting a link with cognitive dysfunction (Chen et al., 2016).
DYSBINDIN-1 AND BEYOND
Given the indicated role for dysbindin-1 in the pathophysiology
of schizophrenia and associated cognitive impairment, it will be
heuristic to search for drug targets and molecules that might
inuence its expression and functionality. Those studies that
indicate relationships between dysbindin-1 function, neuronal
and behavioral processes associated with the pathobiology of
psychotic illness and responsivity to current D2R antagonist
antipsychotic drugs are particularly provocative in this regard;
this is because they offer the prospect of clues to identifying non-
DAergic mechanisms of antipsychotic activity. Importantly,
these concepts generalize beyond dysbindin-1 to other
developmental regulators. For example, in addition to
identifying a role for dysbindin-1A in the regulation of
schizophrenia-related behavioral processes, including
specialised delay/interference-dependent working memory
(Petit et al., 2017), we have recently identied a role for the
developmental gene and regulatory protein neuregulin-1
(NRG1) in a triad with miRNA-143 and D2R that was
revealed through investigation of schizophrenia-related
behavioral abnormalities induced by phencyclidine; miRNA-
143 directly targeted to the 3un-translated region of NRG1
mRNA to reduce protein expression of NRG1 and the D2R
modulated expression of NRG1 in PFC (Wang et al., 2019).
The issues reviewed above constitute a substantive basis for
such a search. Furthermore, they offer tantalising glimpses into
mechanisms and putative target sites relating to DTNBP1/
dysbindin-1A and how these concepts might generalize to a
broader spectrum of developmental genes and regulatory
proteins implicated in the pathobiology of schizophrenia
spectrum psychosis.
AUTHOR CONTRIBUTIONS
JW, XZ, and COT reviewed the relevant literature and wrote this
paper. JW, XZ, and COT revised the manuscript. All the authors
listed agreed to the publication of this paper.
FUNDING
This work was supported by Science Foundation Ireland through
grant 07/IN.1/B960.
REFERENCES
Agnew-Blais, J. C., Buka, S. L., Fitzmaurice, G. M., Smoller, J. W., Goldstein, J. M.,
and Seidman, L. J. (2015). Early childhood IQ trajectories in individuals later
developing schizophrenia and affective psychoses in the New England family
studies. Schizophr. Bull. 41, 817823. doi: 10.1093/schbul/sbv027
Allen, N. C., Bagade, S., McQueen, M. B., Ioannidis, J. P., Kavvoura, F. K., Khoury,
M. J., et al. (2008). Systematic meta-analyses and eld synopsis of genetic
association studies in schizophrenia: the SzGene database. Nat. Genet. 40, 827
834. doi: 10.1038/ng.171
Al-Shammari, A. R., Bhardwaj, S. K., Musaelyan, K., Srivastava, L. K., and
Szele, F. G. (2018). Schizophrenia-related dysbindin-1 gene is required for
innate immune response and homeostasis in the developing subventricular
zone. NPJ Schizophr. 4, 15. doi: 10.1038/s41537-018-0057-5
Ayalew, M., Le-Niculescu, H., Levey, D. F., Jain, N., Changala, B., Patel, S. D., et al.
(2012). Convergent functional genomics of schizophrenia: from
comprehensive understanding to genetic risk prediction. Mol. Psychiatry 17,
887905. doi: 10.1038/mp.2012.37
Baek, J. H., Kim, J. S., Ryu, S., Oh, S., Noh, J., Lee, W. K., et al. (2012). Association
of genetic variations in DTNBP1 with cognitive function in schizophrenia
patients and healthy subjects. Am. J. Med. Genet. Part B: Neuropsychiatr. Genet.
7, 841849. doi: 10.1002/ajmg.b.32091
Becker-Krail, D., Farrand, A. Q., Boger, H. A., and Lavin, A. (2017). Effects of
ngolimod administration in a genetic model of cognitive decits. J. Neurosci.
Res. 95, 11741181. doi: 10.1002/jnr.23799
Benson, M. A., Newey, S. E., Martin-Rendon, E., Hawkes, R., and Blake, D. J.
(2001). Dysbindin, a novel coiled-coil-containing protein that interacts with
the dystrobrevins in muscle and brain. J. Biol. Chem. 276, 2423224241. doi:
10.1074/jbc.M010418200
Bergen, S. E., Ploner, A., Howrigan, D., ODonovan, M. C., Smoller, J. W., et al.
(2019). CNV Analysis Group and the Schizophrenia Working Group of the
Psychiatric Genomics Consortium, Joint contributions of rare copy number
variants and common SNPs to risk for schizophrenia. Am. J. Psychiatry 176,
2935. doi: 10.1176/appi.ajp.2018.17040467
Bernstein, H. G., Steiner, J., Guest, P. C., Dobrowolny, H., and Bogerts, B. (2015).
Glial cells as key players in schizophrenia pathology: recent insights and
concepts of therapy. Schizophr. Res. 161, 418. doi: 10.1016/j.schres.2014.03.035
Bhardwaj, S. K., Baharnoori, M., Sharif-Askari, B., Kamath, A., Williams, S., and
Srivastava, L. K. (2009). Behavioral characterization of dysbindin-1 decient
sandy mice. Behav. Brain Res. 197, 435441. doi: 10.1016/j.bbr.2008.10.011
Bhardwaj, S. K., Ryan, R. T., Wong, T. P., and Srivastava, L. K. (2015). Loss of
dysbindin-1, a risk gene for schizophrenia, leads to impaired group 1
metabotropic glutamate receptor function in mice. Front. Behav. Neurosci. 9,
72. doi: 10.3389/fnbeh.2015.00072
Birnbaum, R., and Weinberger, D. R. (2017). Genetic insights into the
neurodevelopmental origins of schizophrenia. Nat. Rev. Neurosci. 18, 727
740. doi: 10.1038/nrn.2017.125
Blake, D. J., and Kröger, S. (2000). The neurobiology of Duchenne muscular
dystrophy: learning lessons from muscle? Trends Neurosci. 23, 9299. doi:
10.1016/s0166-2236(99)01510-6
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16388
Boyle, E. A., Li, Y. I., and Pritchard, J. K. (2017). An expanded view of complex
traits: from polygenic to omnigenic. Cell 169, 11771186. doi: 10.1016/
j.cell.2017.05.038
Bradshaw, N. J., and Korth, C. (2019). Protein misassembly and aggregation as
potential convergence points for nongenetic causes of chronic mental illness.
Mol. Psychiatry 24, 936951. doi: 10.1038/s41380-018-0133-2
Bray, N. J., Buckland, P. R., Owen, M. J., and ODonovan, M. C. (2003). Cis-acting
variation in the expression of a high proportion of genes in human brain. Hum.
Genet. 113, 149153. doi: 10.1007/s00439-003-0956-y
Bray, N. J., Preece, A., Williams, N. M., Moskvina, V., Buckland, P. R., Owen, M. J.,
et al. (2005). Haplotypes at the dystrobrevin binding protein 1 (DTNBP1) gene
locus mediate risk for schizophrenia through reduced DTNBP1 expression.
Hum. Mol. Genet. 14, 19471954. doi: 10.1093/hmg/ddi199
Bray, N. J., Holmans, P. A., van den Bree, M. B., Jones, L., Elliston, L. A., Hughes,
G., et al. (2008). Cis-and trans-loci inuence expression of the schizophrenia
susceptibility gene DTNBP1. Hum. Mol. Genet. 17, 11691174. doi: 10.1093/
hmg/ddn006
Burdick, K. E., Lencz, T., Funke, B., Finn, C. T., Szeszko, P. R., Kane, J. M., et al.
(2006). Genetic variation in DTNBP1 inuences general cognitive ability.
Hum. Mol. Genet. 15, 15631568. doi: 10.1093/hmg/ddi48
Burdick, K. E., Goldberg, T. E., Funke, B., Bates, J. A., Lencz, T., Kucherlapati, R.,
et al. (2007). DTNBP1 genotype inuences cognitive decline in schizophrenia.
Schizophr. Res. 89, 169172. doi: 10.1016/j.schres.2006.09.008
Carlson, G. C., Talbot, K., Halene, T. B., Gandal, M. J., Kazi, H. A., Schlosser, L.,
et al. (2011). Dysbindin-1 mutant mice implicate reduced fast-phasic
inhibition as a nal common disease mechanism in schizophrenia. Proc.
Natl. Acad. Sci. U.S.A. 108, E962E970. doi: 10.1073/pnas.1109625108
Carr, G. V., Jenkins, K. A., Weinberger, D. R., and Papaleo, F. (2012). Loss of
dysbindin-1 in mice impairs reward-based operant learning by increasing
impulsive and compulsive behavior. Behav. Brain Res. 241, 173184. doi:
10.1016/j.bbr.2012.12.021
Chang,E. H., Fernando, K., Yeung,L. W. E., Barbari, K., Chandon,T. S., and Malhotra,
A. K. (2018). Single point mutation on the gene encoding dysbindin results in
recognition decits. Genes Brain Behav. 17, e12449. doi: 10.1111/gbb.12449
Cheli, V. T., Daniels, R. W., Godoy, R., Hoyle, D. J., Kandachar, V., Starcevic, M.,
et al. (2010). Genetic modiers of abnormal organelle biogenesis in a
Drosophila model of BLOC-1 deciency. Hum. Mol. Genet. 19, 861878.
doi: 10.1093/hmg/ddp555
Chen, Y., Bang, S., McMullen, M. F., Kazi, H., Talbot, K., Ho, M. X., et al. (2016).
Neuronal activity-induced sterol regulatory element binding protein-1
(SREBP1) is disrupted in dysbindin-null mice-potential link to cognitive
impairment in schizophrenia. Mol. Neurobiol. 54, 16991709. doi: 10.1007/
s12035-016-9773-x
Chow, T. J., Tee, S. F., Loh, S. Y., Yong, H. S., Abu Bakar, A. K., and Tang, P. Y.
(2018). Variants in ZNF804A and DTNBP1 assessed for cognitive impairment
in schizophrenia using a multiplex family-based approach. Psychiatry Res. 270,
11661167. doi: 10.1016/j.psychres.2018.04.051
Correa, D. D., Satagopan, J., Cheung, K., Arora, A. K., Kryza-Lacombe, M., Xu, Y.,
et al. (2016). COMT, BDNF, and DTNBP1 polymorphisms and cognitive
functions in patients with brain tumors. Neuro-Oncol. 18, 14251433. doi:
10.1093/neuonc/now057
Cox, M., Tucker, A., Tang, J., Talbot, K., Richer, D., Yeh, L., et al. (2009).
Neurobehavioral abnormalities in the dysbindin-1 mutant, sandy, on a
C57BL/6J genetic background. Genes Brain Behav. 8, 390397. doi: 10.1111/
j.1601-183X.2009.00477.x
Dawson, N., Morris, B. J., and Pratt, J. A. (2015). Functional brain connectivity
phenotypes for schizophrenia drug discovery. J. Psychopharmacol. 29, 169177.
doi: 10.1177/0269881114563635
DellAngelica, E. C. (2004). The building BLOC(k)s of lysosomes and related
organelles. Curr. Op. Cell Biol. 16, 458464. doi: 10.1016/j.ceb.2004.05.001
Diamantopoulou, A., and Gogos, J. A. (2019). Neurocognitive and perceptual
processing in genetic mouse models of schizophrenia: emerging lessons.
Neuroscientist Jan 17. doi: 1073858418819435
Dickman, D. K., and Davis, G. W. (2009). The schizophrenia susceptibility gene
dysbindin controls synaptic homeostasis. Science 326, 11271130. doi: 10.1126/
science.1179685
Donohoe, G., Morris, D. W., Clarke, S., McGhee, K. A., Schwaiger, S., Nangle, J.
M., et al. (2007). Variance in neurocognitive performance is associated with
dysbindin-1 in schizophrenia: a preliminary study. Neuropsychologia 45, 454
458. doi: 10.1016/j.neuropsychologia.2006.06.016
Donohoe, G., Morris, D. W., De Sanctis, P., Magno, E., Montesi, J. L.,
Garavan, H. P., et al. (2008). Early visual processing decits in dysbindin-
associated schizophrenia. Biol. Psychiatry 63, 484489. doi: 10.1016/
j.biopsych.2007.07.022
Donohoe, G., Frodl, T., Morris, D., Spoletini, I., Cannon, D. M., Cherubini, A.,
et al. (2010). Reduced occipital and prefrontal brain volumes in dysbindin-
associated schizophrenia. Neuropsychopharmacology 35, 368373. doi:
10.1038/npp.2009.140
Enriquez-Barreto, L., and Morales, M. (2016). The PI3K signaling pathway as a
pharmacological target in Autism related disorders and Schizophrenia. Mol.
Cell Ther. 4, 2. doi: 10.1186/s40591-016-0047-9
Estrada-Mena, F. J., Rodriguez, A., Mendoza-Lorenzo, P., Neri-Gomez, T.,
Manjarrez-Gutierrez, G., Perez-Ortiz, A. C., et al. (2017). Effects of
(-)-epicatechin on frontal cortex DAPC and dysbindin of the mdx mice.
Neurosci. Lett. 658, 142149. doi: 10.1016/j.neulet.2017.08.056
Farrell, M. S., Werge, T., Sklar, P., Owen, M. J., Ophoff, R. A., ODonovan, M. C.,
et al. (2015). Evaluating historical candidate genes for schizophrenia. Mol.
Psychiatry 20, 555562. doi: 10.1038/mp.2015.16
Feng, Y.-Q., Zhou, Z.-Y., He, X., Wang, H., Guo, X.-L., Hao, C.-J., et al. (2008).
Dysbindin deciency in sandy mice causes reduction of snapin and displays
behaviors related to schizophrenia. Schizophr. Res. 106, 218228. doi: 10.1016/
j.schres.2008.07.018
Fung, S. J., Sivagnanasundaram, S., and Weickert, C. S. (2011). Lack of change in
markers of presynatic terminal abundance alongside subtle reductions in
markers of presynaptic terminal plasticity in prefrontal cortex of
schizophrenia patients. Biol. Psychiatry 69, 7179. doi: 10.1016/
j.biopsych.2010.09.036
Genovese, G., Fromer, M., Stahl, E. A., Ruderfer, D. M., Chambert, K., Landén, M.,
et al. (2016). Increased burden of ultra-rare protein-altering variants among
4,877 individuals with schizophrenia. Nat. Neurosci. 19, 14331441. doi:
10.1038/nn.4402
Gerrish, A., Williams, H., Moskvina, V., Owen, M. J., ODonovan, M. C., and
Williams, N. M. (2009). An examination of MUTED as a schizophrenia
susceptibility gene. Schizophr. Res. 107, 110111. doi: 10.1016/
j.schres.2008.08.011
Ghiani,C.,Starcevic,M.,Rodriguez-Fernandez,I.,Nazarian,R.,Cheli,V.,
Chan, L., et al. (2009). The dysbindin-containing complex (BLOC-1) in
brain: developmental regulation, interaction with SNARE proteins and role
in neurite outgrowth. Mol. Psychiatry 15, 204215. doi: 10.1038/mp.2009.58
Glen,W.B.Jr,Horowitz,B.,Carlson, G. C., Cannon, T. D., Talbot, K.,
Jentsch, J. D., et al. (2014). Dysbindin-1 loss compromises NMDAR-
dependent synaptic plasticity and contextual fear conditioning. Hippocampus
24, 204213. doi: 10.1002/hipo.22215
Gokhale, A., Mullin, A. P., Zlatic, S. A., Easley, C. A.4th, Merritt, M. E., Raj, N.,
et al. (2015). The N-ethylmaleimide-sensitive factor and dysbindin interact to
modulate synaptic plasticity. J. Neurosci. 35, 76437653. doi: 10.1523/
JNEUROSCI.4724-14.2015
Gokhale, A., Hartwig, C., Freeman, A. H., Das, R., Zlatic, S. A., Vistein, R., et al.
(2016). The proteome of BLOC-1 genetic defects identies the Arp2/3 Actin
polymerization complex to function downstream of the schizophrenia
susceptibility factor dysbindin at the synapse. J. Neurosci. 36, 1239312411.
doi: 10.1523/JNEUROSCI.1321-16.2016
Gonzalez-Islas, C., and Hablitz, J. J. (2003). Dopamine enhances EPSCs in layer II-
III pyramidal neurons in rat prefrontal cortex. J. Neurosci. 23, 867875. doi:
10.1523/JNEUROSCI.23-03-00867.2003
Grady, R. M., Wozniak, D. F., Ohlemiller, K. K., and Sanes, J. R. (2006). Cerebellar
synaptic defects and abnormal motor behavior in mice lacking a-and b-
dystrobrevin. J. Neurosci. 26, 28412851. doi: 10.1523/JNEUROSCI.4823-
05.2006
Green, M. F., Horan, W. P., and Lee, J. (2019). Nonsocial and social cognition in
schizophrenia: current evidence and future directions. World Psychiatry 18,
146161. doi: 10.1002/wps.20624
Guloksuz, S., Pries, L. K., Delespaul, P., Kenis, G., Luykx, J. J., Lin, B. D., et al.
(2019). Examining the independent and joint effects of molecular genetic
liability and environmental exposures in schizophrenia: results from the
EUGEI study. World Psychiatry 18, 173182. doi: 10.1002/wps.20629
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 16389
Guo, A., Sun, J., Riley, B., Thiselton, D., Kendler, K., and Zhao, Z. (2008). The
dystrobrevin-binding protein 1 gene: features and networks. Mol. Psychiatry
14, 1829. doi: 10.1038/mp.2008.88
Guo, A. Y., Sun, J., Riley, B. P., Thiselton, D. L., Kendler, K. S., Zhao, Z., et al.
(2009). The dystrobrevin-binding protein 1 gene: features and networks. Mol.
Psychiatry. 14, 1829. doi: 10.1038/mp.2008.88
Han, M., and Deng, C. (2018). BDNF as a pharmacogenetic target for
antipsychotic treatment of schizophrenia. Neurosci. Lett. 9, 133870. doi:
10.1016/j.neulet.2018.10.015
Hartwig, C., Monis, W. J., Chen, X., Dickman, D. K., Pazour, G. J., and Faundez, V.
(2018). Neurodevelopmental disease mechanisms, primary cilia, and
endosomes converge on the BLOC-1 and BORC complexes. Dev. Neurobiol.
78, 311330. doi: 10.1002/dneu.22542
Hilker, R., Helenius, D., Fagerlund, B., Skytthe, A., Christensen, K., Werge, T. M.,
et al. (2018). Heritability of schizophrenia and schizophrenia spectrum based
on the nationwide Danish twin register. Biol. Psychiatry 83, 492498. doi:
10.1016/j.biopsych.2017.08.017
Hyde, T. M., Lipska, B. K., Ali, T., Mathew, S. V., Law, A. J., Metitiri, O. E., et al.
(2011). Expression of GABA signaling molecules KCC2, NKCC1, and GAD1 in
cortical development and schizophrenia. J. Neurosci. 31, 1108811095. doi:
10.1523/JNEUROSCI.1234-11.2011
Iizuka, Y., Sei, Y., Weinberger, D. R., and Straub, R. E. (2007). Evidence that the
BLOC-1 protein dysbindin modulates dopamine D2 receptor internalization
and signaling but not D1 internalization. J. Neurosci. 27, 1239012395. doi:
10.1523/JNEUROSCI.1689-07.2007
Jentsch, J. D., Trantham-Davidson, H., Jairl, C., Tinsley, M., Cannon, T. D., and
Lavin, A. (2009). Dysbindin modulates prefrontal cortical glutamatergic
circuits and working memory function in mice. Neuropsychopharmacology
34, 26012608. doi: 10.1038/npp.2009.90
Jones, H. J., Stergiakouli, E., Tansey, K. E., Hubbard, L., Heron, J., Cannon, M.,
et al. (2016). Phenotypic manifestation of genetic risk for schizophrenia during
adolescence in the general population. JAMA Psychiatry 73, 221228. doi:
10.1001/jamapsychiatry.2015.3058
Kahn,R.S.,andKeefe,R.S.(2013).Schizophreniaisacognitiveillness:timeforachange
in focus. JAMA Psychiatry 70, 11071112. doi: 10.1001/jamapsychiatry.2013.155
Kirov, G., Pocklington, A. J., Holmans, P., Ivanov, D., Ikeda, M., Ruderfer, D., et al.
(2012). De novo CNV analysis implicates specic abnormalities of
postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol.
Psychiatry 17, 142153. doi: 10.1038/mp.2011.154
Kobayashi, K., Umeda-Yano, S., Yamamori, H., Takeda, M., Suzuki, H., and
Hashimoto, R. (2011). Correlated alterations in serotonergic and dopaminergic
modulations at the hippocampal mossy ber synapse in mice lacking
dysbindin. PloS One 6, e18113. doi: 10.1371/journal.pone.0018113
Konopaske, G. T., Balu, D. T., Presti, K. T., Chan, G., Benes, F. M., and Coyle, J. T.
(2018). Dysbindin-1 contributes to prefrontal cortical dendritic arbor
pathology in schizophrenia. Schizophr. Res. 201, 270277. doi: 10.1016/
j.schres.2018.04.042
Kotlar, A. V., Mercer, K. B., Zwick, M. E., and Mulle, J. G. (2015). New discoveries
in schizophrenia genetics reveal neurobiological pathways: a review of recent
ndings. Eur. J. Med. Genet. 58, 704714. doi: 10.1016/j.ejmg.2015.10.008
Kubota, K., Kumamoto, N., Matsuzaki, S., Hashimoto, R., Hattori, T., Okuda, H.,
et al. (2009). Dysbindin engages in c-Jun N-terminal kinase activity and
cytoskeletal organization. Biochem. Biophys. Res. Commun. 379, 191195.
doi: 10.1016/j.bbrc.2008.12.017
Larimore, J., Zlatic, S. A., Gokhale, A., Tornieri, K., Singleton, K. S., Mullin, A. P.,
et al. (2014). Mutations in the BLOC-1 subunits dysbindin and muted generate
divergent and dosage-dependent phenotypes. J. Biol. Chem. 289, 1429114300.
doi: 10.1074/jbc.M114.553750
Larimore, J., Zlatic, S. A., Arnold, M., Singleton, K. S., Cross, R., Rudolph, H., et al.
(2017). Dysbindin deciency modies the expression of GABA neuron and ion
permeation transcripts in the developing hippocampus. Front. Genet. 8, 28. doi:
10.3389/fgene.2017.00028
Lee, H. H., Nemecek, D., Schindler, C., Smith, W. J., Ghirlando, R., Steven, A. C.,
et al. (2012). Assembly and architecture of biogenesis of lysosome-related
organelles complex-1 (BLOC-1). J. Biol. Chem. 287, 58825890. doi: 10.1074/
jbc.M111.325746
Leggio, G. M., Torrisi, S. A., Mastrogiacomo, R., Mauro, D., Chisari, M., Devroye,
C., et al. (2019). The epistatic interaction between the dopamine D3 receptor
and dysbindin-1 modulates higher-order cognitive functions in mice and
humans. Mol. Psychiatry. doi: 10.1038/s41380-019-0511-4
Lemonnier, E., Lazartigues, A., and Ben-Ari, Y. (2016). Treating schizophrenia
with the diuretic bumetanide: a case report. Clin. Neuropharmacol. 39, 115
117. doi: 10.1097/WNF.0000000000000136
Lewis, D. A., Hashimoto, T., and Morris, H. M. (2008). Cell and receptor type-
specic alterations in markers of GABA neurotransmission in the prefrontal
cortex of subjects with schizophrenia. Neurotox. Res. 14, 237248. doi: 10.1007/
BF03033813
Li, W., Zhang, Q., Oiso, N., Novak, E. K., Gautam, R., OBrien, E. P., et al. (2003).
Hermansky-Pudlak syndrome type 7 (HPS-7) results from mutant dysbindin, a
member of the biogenesis of lysosome-related organelles complex 1 (BLOC-1).
Nat. Genet. 35, 8489. doi: 10.1038/ng1229
Li, W., Rusiniak, M. E., Chintala, S., Gautam, R., Novak, E. K., and Swank, R. T.
(2004). Murine HermanskyPudlak syndrome genes: regulators of lysosome-
related organelles. Bioessays 26, 616628. doi: 10.1002/bies.20042
Luciano, M., Miyajima, F., Lind, P. A., Bates, T. C., Horan, M., Harris, S. E., et al.
(2009). Variation in the dysbindin gene and normal cognitive function in three
independent population samples. Genes Brain Behav. 8, 218227. doi: 10.1111/
j.1601-183X.2008.00462.x
Malhotra, D., and Sebat, J. (2012). CNVs: harbingers of a rare variant revolution in
psychiatric genetics. Cell 148, 12231241. doi: 10.1016/j.cell.2012.02.039
Man, L., Lv, X., Du, X. D., Yin, G., Zhu, X., Zhang, Y., et al. (2018). Cognitive
impairments and low BDNF serum levels in rst-episode drug-naive patients
with schizophrenia. Psychiatry Res. 263, 16. doi: 10.1016/
j.psychres.2018.02.034
Manning, B. D., and Cantley, L. C. (2007). AKT/PKB signaling: navigating
downstream. Cell 129, 12611274. doi: 10.1016/j.cell.2007.06.009
Manolio, T. A., Collins, F. S., Cox, N. J., Goldstein, D. B., Hindorff, L. A., Hunter,
D. J., et al. (2009). Finding the missing heritability of complex diseases. Nature
461, 747753. doi: 10.1038/nature08494
Mark, W., and Toulopoulou, T. (2016). Cognitive intermediate phenotype and
genetic risk for psychosis. Curr. Opin. Neurobiol. 36, 2330. doi: 10.1016/
j.conb.2015.08.008
Marshall, C. R., Howrigan, D. P., Merico, D., Thiruvahindrapuram, B., Wu, W.,
Greer, D. S., et al. (2017). Contribution of copy number variants to
schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 49,
2735. doi: 10.1038/ng.3725
McCutcheon, R. A., Abi-Dargham, A., and Howes, O. D. (2019). Schizophrenia,
dopamine and the striatum: from biology to symptoms. Trends Neurosci. 42,
205220. doi: 10.1002/wps.20693
Meier, M. H., Caspi, A., Reichenberg, A., Keefe, R. S., Fisher, H. L., Harrington, H.,
et al. (2014). Neuropsychological decline in schizophrenia from the premorbid
to the postonset period: evidence from a population-representative
longitudinal study. Am.J.Psychiatry171, 91101. doi: 10.1176/
appi.ajp.2013.12111438
Mullin, A. P., Sadanandappa, M. K., Ma, W., Dickman, D. K., VijayRaghavan, K.,
Ramaswami, M., et al. (2015). Gene dosage in the dysbindin schizophrenia
susceptibility network differentially affects synaptic function and plasticity. J.
Neurosci. 35, 325338. doi: 10.1523/JNEUROSCI.3542-14.2015
Nazarian, R., Starcevic, M., Spencer, M. J., and DellAngelica, E. C. (2006).
Reinvestigation of the dysbindin subunit of BLOC-1 (biogenesis of
lysosome-related organelles complex-1) as a dystrobrevin-binding protein.
Biochem. J. 395, 587598. doi: 10.1042/BJ20051965
Numakawa, T., Yagasaki, Y., Ishimoto, T., Okada, T., Suzuki, T., Iwata, N., et al.
(2004). Evidence of novel neuronal functions of dysbindin, a susceptibility gene
for schizophrenia. Hum. Mol. Genet. 13, 26992708. doi: 10.1093/hmg/ddh280
OTuathaigh, C. M., and Waddington, J. L. (2015). Closing the translational gap
between mutant mouse models and the clinical reality of psychotic illness.
Neurosci. Biobehav. Rev. 58, 1935. doi: 10.1016/j.neubiorev.2015.01.016
Owen, M. J., Sawa, A., and Mortensen, P. B. (2016). Schizophrenia. Lancet 388,
8697. doi: 10.1016/S0140-6736(15)01121-6
Oyama, S., Yamakawa, H., Sasagawa, N., Hosoi, Y., Futai, E., and Ishiura, S. (2009).
Dysbindin-1, a schizophrenia-related protein, functionally interacts with the
DNA-dependent protein kinase complex in an isoform-dependent manner.
PloS One 4, e4199. doi: 10.1371/journal.pone.0004199
Papaleo, F., Yang, F., Garcia, S., Chen, J., Lu, B., Crawley, J., et al. (2012).
Dysbindin-1 modulates prefrontal cortical activity and schizophrenia-like
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 163810
behaviors via dopamine/D2 pathways. Mol. Psychiatry 7, 8598. doi: 10.1038/
mp.2010.106
Papaleo, F., Burdick, M. C., Callicott, J. H., and Weinberger, D. R. (2014). Epistatic
interaction between COMT and DTNBP1 modulates prefrontal function in
mice and in humans. Mol. Psychiatry 19, 311316. doi: 10.1038/mp.2013.133
Peters, K., Wiltshire, S., Henders, A. K., Dragović, M., Badcock, J. C., Chandler, D.,
et al. (2008). Comprehensive analysis of tagging sequence variants in DTNBP1
shows no association with schizophrenia or with its composite neurocognitive
endophenotypes. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 1159
1166. doi: 10.1002/ajmg.b.30741
Petit, E. I., Michalak, Z., Cox, R., OTuathaigh, C. M., Clarke, N., Tighe, O., et al.
(2017). Dysregulation of specialized delay/interference-dependent working
memory following loss of dysbindin-1A in schizophrenia-related
phenotypes. Neuropsychopharmacology 42, 13491360. doi: 10.1038/
npp.2016.282
Rahmanzadeh, R., Eftekhari, S., Shahbazi, A., Khodaei Ardakani, M. R.,
Rahmanzade, R., Mehrabi, S., et al. (2017a). Effect of bumetanide, a selective
NKCC1 inhibitor, on hallucinations of schizophrenic patients; a double-blind
randomized clinical trial. Schizophr. Res. 184, 145146. doi: 10.1016/
j.schres.2016.12.002
Rahmanzadeh, R., Shahbazi, A., Ardakani, M. K., Mehrabi, S., Rahmanzade, R.,
and Joghataei, M. T. (2017b). Lack of the effect of bumetanide, a selective
NKCC1 inhibitor, in patients with schizophrenia: A double-blind randomized
trial. Psychiatry Clin. Neurosci. 71, 7273. doi: 10.1111/pcn.12475
Rammos, A., Gonzalez, L. A. N.Schizophrenia Working Group of the Psychiatric
Genomics Consortium, , Weinberger, D. R., Mitchell, K. J., Nicodemus, K. K.
(2019). The role of polygenic risk score gene-set analysis in the context of the
omnigenic model of schizophrenia. Neuropsychopharmacology. doi: 10.1038/
s41386-019-0410-z
Richards, A. L., Pardiñas, A. F., Frizzati, A., Tansey, K. E., Lynham, A. J., Holmans,
P., et al. (2019). The relationship between polygenic risk scores and cognition
in schizophrenia. Schizophr. Bull., pii: sbz061. doi: 10.1093/schbul/sbz061
Saggu, S., Cannon, T. D., Jentsch, J. D., and Lavin, A. (2013). Potential molecular
mechanisms for decreased synaptic glutamate release in dysbindin-1 mutant
mice. Schizophr. Res. 146, 254263. doi: 10.1016/j.schres.2013.01.037
Savage, J. E., Jansen, P. R., Stringer, S., Watanabe, K., Bryois, J., de Leeuw, C. A.,
et al. (2018). Genome-wide association meta-analysis in 269,867 individuals
identies new genetic and functional links to intelligence. Nat. Genet. 50, 912
919. doi: 10.1038/s41588-018-0152-6
Schaefer, J., Giangrande, E., Weinberger, D. R., and Dickinson, D. (2013). The
global cognitive impairment in schizophrenia: consistent over decades and
around the world. Schizophr. Res. 150, 4250. doi: 10.1016/j.schres.2013.07.009
Scheggia, D., Mastrogiacomo, R., Mereu, M., Sannino, S., Straub, R. E., Armando,
M., et al. (2018). Variations in Dysbindin-1 are associated with cognitive
response to antipsychotic drug treatment. Nat. Commun. 9, 2265. doi: 10.1038/
s41467-018-04711-w
Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014).
Biological insights from 108 schizophrenia-associated genetic loci. Nature 511,
421427. doi: 10.1038/nature13595
Shao, L., Shuai, Y., Wang, J., Feng, S., Lu, B., Li, Z., et al. (2011). Schizophrenia
susceptibility gene dysbindin regulates glutamatergic and dopaminergic
functions via distinctive mechanisms in Drosophila. Proc. Natl. Acad. Sci.
U.S.A. 108, 1883118836. doi: 10.1073/pnas.1114569108
Shintani, N., Onaka, Y., Hashimoto, R., Takamura, H., Nagata, T., Umeda-Yano,
S., et al. (2014). Behavioral characterization of mice overexpressing human
dysbindin-1. Mol. Brain 7, 74. doi: 10.1186/s13041-014-0074-x
Sillitoe, R. V., Benson, M. A., Blake, D. J., and Hawkes, R. (2003). Abnormal
dysbindin expression in cerebellar mossy ber synapses in the mdx mouse
model of Duchenne muscular dystrophy. J. Neurosci. 23, 65766585. doi:
10.1523/JNEUROSCI.23-16-06576.2003
Sinclair, D., Cesare, J., McMullen, M., Carlson, G. C., Hahn, C. G., and Borgmann-
Winter, K. E. (2016). Effects of sex and DTNBP1 (dysbindin) null gene
mutation on the developmental GluN2B-GluN2A switch in the mouse
cortex and hippocampus. J. Neurodev. Disord. 8, 14. doi: 10.1186/s11689-
016-9148-7
Spiegel, S., Chiu, A., James, A. S., Jentsch, J. D., and Karlsgodt, K. H. (2015).
Recognition decits in mice carrying mutations of genes encoding BLOC-1
subunits pallidin or dysbindin. Genes Brain Behav. 14, 618624. doi: 10.1111/
gbb.12240
Starcevic, M., and DellAngelica, E. C. (2004). Identication of snapin and three
novel proteins (BLOS1, BLOS2, and BLOS3/reduced pigmentation) as subunits
of biogenesis of lysosome-related organelles complex-1 (BLOC-1). J. Biol.
Chem. 279, 28393401.
Straub, R. E., MacLean, C. J., ONeill, F. A., Burke, J., Murphy, B., Duke, F., et al.
(1995). A potential vulnerability locus for schizophrenia on chromosome
6p2422: evidence for genetic heterogeneity. Nat. Genet. 11, 287293. doi:
10.1038/ng1195-287
Strohmaier, J., Frank, J., Wendland, J. R., Schumacher, J., Jamra, R. A., Treutlein, J.,
et al. (2010). A reappraisal of the association between Dysbindin (DTNBP1)
and schizophrenia in a large combined case-control and family-based sample
of German ancestry. Schizophr. Res.118, 98105. doi: 10.1016/
j.schres.2009.12.025
Sullivan, C. R., Funk, A. J., Shan, D., Haroutunian, V., and McCullumsmith, R. E.
(2015). Decreased chloride channel expression in the dorsolateral prefrontal
cortex in schizophrenia. PloS One 10, e0123158. doi: 10.1371/
journal.pone.0123158
Straub, R. E., Mayhew, M. B., Vakkalanka, R. K., Kolachana, B., Goldberg, T. E.,
Egan, M. F., et al. (2005). MUTED, a protein that binds to dysbindin
(DTNBP1), is associated with schizophrenia. Am.J.Med.Genet.B
Neuropsychiatr. Genet. 138B, 136.
Swank, R. T., Sweet, H. O., Davisson, M. T., Reddington, M., and Novak, E. K.
(1991). Sandy: a new mouse model for platelet storage pool deciency. Genet.
Res. 58, 5162. doi: 10.1017/s0016672300029608
Takao, K., Toyama, K., Nakanishi, K., Hattori, S., Takamura, H., Takeda, M., et al.
(2008). Impaired long-term memory retention and working memory in sdy
mutant mice with a deletion in Dtnbp1, a susceptibility gene for schizophrenia.
Mol. Brain 1, 1123. doi: 10.1186/1756-6606-1-11
Talbot, K., Eidem, W. L., Tinsley, C. L., Benson, M. A., Thompson, E. W.,
Smith, R. J., et al. (2004). Dysbindin-1 is reduced in intrinsic, glutamatergic
terminals of the hippocampal formation in schizophrenia. J. Clin. Invest. 113,
13531363. doi: 10.1172/JCI20425
Talbot, K., Cho, D.-S., Ong, W.-Y., Benson, M. A., Han, L.-Y., Kazi, H. A., et al.
(2006). Dysbindin-1 is a synaptic and microtubular protein that binds brain
snapin. Hum. Mol. Genet. 15, 30413054. doi: 10.1093/hmg/ddl246
Talbot, K., Louneva, N., Cohen, J. W., Kazi, H., Blake, D. J., and Arnold, S. E.
(2011). Synaptic dysbindin-1 reductions in schizophrenia occur in an isoform-
specic manner indicating their subsynaptic location. PloS One 6, e16886. doi:
10.1371/journal.pone.0016886
Talbot, K. (2009). The sandy (sdy) mouse: a dysbindin-1 mutant relevant to
schizophrenia research. Prog.Brain Res. 179, 8794. doi: 10.1016/S0079-6123
(09)17910-4
Tam, V., Patel, N., Turcotte, M., Bossé, Y., Paré, G., and Meyre, D. (2019). Benets
and limitations of genome-wide association studies. Nat. Rev. Genet. 20, 467
484. doi: 10.1038/s41576-019-0127-1
Tamminga, C. A., Stan, A. D., and Wagner, A. D. (2010). The hippocampal
formation in schizophrenia. Am. J. Psychiatry 167, 11781193. doi: 10.1176/
appi.ajp.2010.09081187
Taneichi-Kuroda, S., Taya, S., Hikita, T., Fujino, Y., and Kaibuchi, K. (2009).
Direct interaction of Dysbindin with the AP-3 complex via its mu subunit.
Neurochem. Int. 54, 431438. doi: 10.1016/j.neuint.2009.01.014
Tang, J., LeGros, R. P., Louneva, N., Yeh, L., Cohen, J. W., Hahn, C.-G., et al.
(2009). Dysbindin-1 in dorsolateral prefrontal cortex of schizophrenia cases is
reduced in an isoform-specic manner unrelated to dysbindin-1 mRNA
expression. Hum. Mol. Genet. 18, 38513863. doi: 10.1093/hmg/ddp329
Tansey, K. E., Rees, E., Linden, D. E., Ripke, S., Chambert, K. D., Moran, J. L., et al.
(2016). Common alleles contribute to schizophrenia in CNV carriers. Mol.
Psychiatry 21, 1153. doi: 10.1038/mp.2015.170
Toulopoulou, T., Zhang, X., Cherny, S., Dickinson, D., Berman, K. F., Straub, R. E.,
et al. (2019). Polygenic risk score increases schizophrenia liability through
cognition-relevant pathways. Brain 142, 471485. doi: 10.1093/brain/awy279
Varela-Gomez, N., Mata, I., Perez-Iglesias, R., Rodriguez-Sanchez, J. M., Ayesa, R.,
Fatjo-Vilas, M., et al. (2015). Dysbindin gene variability is associated with
cognitive abnormalities in rst-episode non-affective psychosis. Cogn.
Neuropsychiatry 20, 14456. doi: 10.1080/13546805.2014.991780
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 163811
Vijayraghavan, S., Wang, M., Birnbaum, S. G., Williams, G. V., and Arnsten, A. F.
(2007). Inverted-U dopamine D1 receptor actions on prefrontal neurons
engaged in working memory. Nat. Neurosci. 10, 376384. doi: 10.1038/nn1846
Waddington, J. L., Hennessy, R. J., OTuathaigh, C. M. P., Owoeye, O., and Russell,
V. (2012). Schizophrenia and the lifetime trajectory of psychotic illness:
developmental neuroscience and pathobiology, redux, in The origins of
schizophrenia. Eds. A. S. Brown and P. H. Patterson (New York: Columbia
University Press), pp. 321.
Wang, H., Xu, J., Lazarovici, P., and Zheng, W. (2017). Dysbindin-1 involvement
in the etiology of schizophrenia. Int. J. Mol. Sci. 18, pii: E2044. doi: 10.3390/
ijms18102044
Wang, P., Cao, T., Chen, J., Jiang, Y., Wang, C., Waddington, J. L., et al. (2019). D2
receptor-mediated miRNA-143 expression is associated with the effects of
antipsychotic drugs on phencyclidine-induced schizophrenia-related
locomotor hyperactivity and with neuregulin-1 expression in mice.
Neuropharmacology. 157, 107675. doi: 10.1016/j.neuropharm.2019.107675
Weickert, C. S., Straub, R. E., McClintock, B. W., Matsumoto, M., Hashimoto, R.,
Hyde, T. M., et al. (2004). Human dysbindin (DTNBP1) gene expression in
normal brain and in schizophrenic prefrontal cortex and midbrain. Arch. Gen.
Psychiatry 61, 544545. doi: 10.1001/archpsyc.61.6.544
Weickert, C. S., Rothmond, D. A., Hyde, T. M., Kleinman, J. E., and Straub, R. E. (2008).
Reduced DTNBP1 (dysbindin-1) mRNA in the hippocampal formation of
schizophrenia patients. Schizophr. Res. 98, 105110. doi: 10.1016/j.schres.2007.05.041
Weinberger, D. R. (2017). Future of days past: neurodevelopment and
schizophrenia. Schizophr. Bull. 43, 11641168. doi: 10.1093/schbul/sbx118
Weinberger, D. R. (2019). Thinking about schizophrenia in an era of genomic
medicine. Am. J. Psychiatry 176, 1220. doi: 10.1176/appi.ajp.2018.18111275
Xavier,R.M.,Dungan,J.R.,Keefe,R.S.E.,andVorderstrasse,A.(2018).Polygenicsignal
for symptom dimensions and cognitive performance in patients with chronic
schizophrenia. Schizophr. Res. Cogn. 12, 1119. doi: 10.1016/j.scog.2018.01.001
Xu, Y., Sun, Y., Ye, H., Zhu, L., Liu, J., Wu, X., et al. (2015). Increased dysbindin-1B
isoform expression in schizophrenia and its propensity in aggresome
formation. Cell. Discovery 1, 15032. doi: 10.1038/celldisc.2015.32
Yang, W., and Xu, Q. (2017). Hippocampal apoptosis and cognitive function in
Dysbindin-1B mice. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 39, 307311. doi:
10.3881/j.issn.1000-503X.2017.03.002
Yang, W., Zhu, C., Shen, Y., and Xu, Q. (2016). The pathogenic mechanism of
dysbindin-1B toxic aggregation: BLOC-1 and intercellular vesicle trafcking.
Neuroscience 333, 7891. doi: 10.1016/j.neuroscience.2016.07.008
Yang, Y., Liu, Y., Wang, G., Hei, G., Wang, X., Li, R., et al. (2019). Brain-derived
neurotrophic factor is associated with cognitive impairments in rst-episode
and chronic schizophrenia. Psychiatry Res. 273, 528536. doi: 10.1016/
j.psychres.2019.01.051
Zhang, J. P., Burdick, K. E., Lencz, T., and Malhotra, A. K. (2010). Meta-analysis of
genetic variation in DTNBP1 and general cognitive ability. Biol. Psychiatry 68,
11261133. doi: 10.1016/j.biopsych.2010.09.016
Zheng, W., Wang, H., Zeng, Z., Lin, J., Little, P. J., Srivastava, L. K., et al. (2012).
The possible role of the Akt signaling pathway in schizophrenia. Brain Res.
1470, 145148. doi: 10.1016/j.brainres.2012.06.032
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Waddington, Zhen and OTuathaigh. This is an open-access article
distributed under the terms of the Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other forums is permitted, provided the
original author(s) and the copyright owner(s) are credited and that the original
publicationin this journal is cited, in accordance with accepted academic practice. No
use, distribution or reproduction is permitted which does not comply with these terms.
Waddington et al. Developmental Genes and Antipsychotic Drug Discovery
Frontiers in Pharmacology | www.frontiersin.org January 2020 | Volume 10 | Article 163812
... Dysbindin-1 (Dys1) protein, coded by the Dystrobrevin Binding Protein 1 (DTNBP1) gene, has been implicated in cognitive processes [1,2], responses to antipsychotic drugs [1,3,4], and genetic risk for schizophrenia [3][4][5]. In agreement, dystrophin and its binding partners (Dys1 being a component of the dystrophin complex [6]) are implicated in schizophrenia by genome-wide studies [7], in line with previous linkage studies [8][9][10][11], and with evidence of reduced Dys1 levels in post-mortem brain samples from patients with schizophrenia [12,13]. ...
... Dysbindin-1 (Dys1) protein, coded by the Dystrobrevin Binding Protein 1 (DTNBP1) gene, has been implicated in cognitive processes [1,2], responses to antipsychotic drugs [1,3,4], and genetic risk for schizophrenia [3][4][5]. In agreement, dystrophin and its binding partners (Dys1 being a component of the dystrophin complex [6]) are implicated in schizophrenia by genome-wide studies [7], in line with previous linkage studies [8][9][10][11], and with evidence of reduced Dys1 levels in post-mortem brain samples from patients with schizophrenia [12,13]. ...
... In agreement, dystrophin and its binding partners (Dys1 being a component of the dystrophin complex [6]) are implicated in schizophrenia by genome-wide studies [7], in line with previous linkage studies [8][9][10][11], and with evidence of reduced Dys1 levels in post-mortem brain samples from patients with schizophrenia [12,13]. These clinical implications have been linked to Dys1-related modulation of neuronal dopaminergic and glutamatergic signaling [3,4,[13][14][15][16], through receptor-mediated intracellular trafficking mechanisms, including D2-like receptors [1,3,6,17]. ...
Article
Full-text available
The mechanisms underlying the dichotomic cortical/basal ganglia dopaminergic abnormalities in schizophrenia are unclear. Astrocytes are important non-neuronal modulators of brain circuits, but their role in dopaminergic system remains poorly explored. Microarray analyses, immunohistochemistry, and two-photon laser scanning microscopy revealed that Dys1 hypofunction increases the reactivity of astrocytes, which express only the Dys1A isoform. Notably, behavioral and electrochemical assessments in mice selectively lacking the Dys1A isoform unraveled a more prominent impact of Dys1A in behavioral and dopaminergic/D2 alterations related to basal ganglia, but not cortical functioning. Ex vivo electron microscopy and protein expression analyses indicated that selective Dys1A disruption might alter intracellular trafficking in astrocytes, but not in neurons. In agreement, Dys1A disruption only in astrocytes resulted in decreased motivation and sensorimotor gating deficits, increased astrocytic dopamine D2 receptors and decreased dopaminergic tone within basal ganglia. These processes might have clinical relevance because the caudate, but not the cortex, of patients with schizophrenia shows a reduction of the Dys1A isoform. Therefore, we started to show a hitherto unknown role for the Dys1A isoform in astrocytic-related modulation of basal ganglia behavioral and dopaminergic phenotypes, with relevance to schizophrenia.
... Dysbindin-1 is a coiled protein having cognitive endophenotype which DTNBP1 (dystrobrevinbinding protein-1) gene encodes and is present on chromosome 6p22.3. Dysbindin exists in three isoforms as 1A, 1B, and 1C, which has been recognized as a risk factor for schizophrenia [85]. Dysbindin 1A is mostly located at post synapse, which is believed to be associated with dendritic spine density and causing disturbances in WM in the HC. ...
... The association of dysbindin-1 with schizophrenia causing primary pathology has been related with downregulation of DTNBP1 expression in dorsolateral PFC and HC formation. DTNBP1 gene has been associated with dysregulated cognitive function in both clinical and non-clinical samples [85]. Allelic variation of DTNBP1 has been linked with schizophrenia causing white matter integrity impairments in healthy adult subjects, decrease in grey matter volume in preteenagers, anomalies in neurite outgrowth and morphology, resulting in cognitive deficits [87]. ...
Article
Full-text available
Schizophrenia affects millions of people worldwide and is a major challenge for the scientific community. Like most psychotic diseases, it is also considered a complicated mental disorder caused by an imbalance in neurotransmitters. Due to the complexity of neuropathology, it is always a complicated disorder. The lack of proper understanding of the pathophysiology makes the disorder unmanageable in clinical settings. However, due to recent advances in animal models, we hope we can have better therapeutic approaches with more success in clinical settings. Dopamine, glutamate, GABA, and serotonin are the neurotransmitters involved in the pathophysiology of schizophrenia. Various animal models have been put forward based on these neurotransmitters, including pharmacological, neurodevelopmental, and genetic models. Polymorphism of genes such as dysbindin, DICS1, and NRG1 has also been reported in schizophrenia. Hypothesis based on dopamine, glutamate, and serotonin are considered successful models of schizophrenia on which drug therapies have been designed to date. New targets like the orexin system, muscarinic and nicotinic receptors, and cannabinoid receptors have been approached to alleviate the negative and cognitive symptoms. The non-pharmacological models like the post-weaning social isolation model (maternal deprivation), the isolation rearing model etc. have been also developed to mimic the symptoms of schizophrenia and to create and test new approaches of drug therapy which is a breakthrough at present in psychiatric disorders. Different behavioral tests have been evaluated in these specific models. This review will highlight the currently available animal models and behavioral tests in psychic disorders concerning schizophrenia.
... This gene encodes the dysbindin-1 protein, which plays a relevant role in neurotransmission and neurodevelopment. 111 Dysbindin-1 has been found in presynaptic and postsynaptic locations in several brain areas of interest in schizophrenia, including the hippocampus, the pre-frontal cortex and the midbrain. 112,113 Because dysbindin-1 interacts with different proteins involved in the release of neurotransmitters, its alteration could affect synaptic homeostasis. ...
... 55 Furthermore, a risk haplotype that included 2 of the above SNPs (rs2619522 and rs760761) was associated with a greater decline in IQ in patients with SSD. 51 The mechanism responsible for this association is unknown, but these polymorphisms might influence intelligence by reducing DTNBP1 expression in the prefrontal cortex, hippocampus and midbrain, 111,112 thus affecting the glutamatergic system. 113 However, other studies did not find this association, 52,54 possibly because of the great variety of SNPs analyzed. ...
Article
Full-text available
Background: To study whether there is genetic overlap underlying the risk for schizophrenia spectrum disorders (SSDs) and low intelligence quotient (IQ), we reviewed and summarized the evidence on genetic variants associated with both traits. Methods: We performed this review in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) and preregistered it in PROSPERO. We searched the Medline databases via PubMed, PsycInfo, Web of Science and Scopus. We included studies in adults with a diagnosis of SSD that explored genetic variants (single nucleotide polymorphisms [SNPs], copy number variants [CNVs], genomic insertions or genomic deletions), estimated IQ and studied the relationship between genetic variability and both traits (SSD and IQ). We synthesized the results and assessed risk of bias using the Quality of Genetic Association Studies (Q-Genie) tool. Results: Fifty-five studies met the inclusion criteria (45 case-control, 9 cross-sectional, 1 cohort), of which 55% reported significant associations for genetic variants involved in IQ and SSD. The SNPs more frequently explored through candidate gene studies were in COMT, DTNBP1, BDNF and TCF4. Through genome-wide association studies, 2 SNPs in CHD7 and GATAD2A were associated with IQ in patients with SSD. The studies on CNVs suggested significant associations between structural variants and low IQ in patients with SSD. Limitations: Overall, primary studies used heterogeneous IQ measurement tools and had small samples. Grey literature was not screened. Conclusion: Genetic overlap between SSD and IQ supports the neurodevelopmental hypothesis of schizophrenia. Most of the risk polymorphisms identified were in genes relevant to brain development, neural proliferation and differentiation, and synaptic plasticity.
... Lower COMT activity combined with lower DTNBP1 expression is associated with poorer performance in mice and lower PFC efficiency measured with functional magnetic resonance imaging (fMRI) during the performance of a working memory task in humans, compared with carriers of a single genetic variation either in COMT or DTNBP1 genes. These mechanisms have been associated with the genetic risk for schizophrenia, particularly, with impaired working memory [15,23], and altered cognitive responses to antipsychotic drugs [9,15,24]. Estrogens have been proposed as mediating factors of the relationship between sex and cognition and antipsychotic treatment response via dopamine signaling modulation [25,26]. Estrogens can control COMT activity [27,28] through estrogen response elements (ERE) sequences that are present in the promoter of the COMT gene [29] and inhibit its transcription [30,31]. ...
Article
Full-text available
Cognitive dysfunctions are core-enduring symptoms of schizophrenia, with important sex-related differences. Genetic variants of the DTBPN1 gene associated with reduced dysbindin-1 protein (Dys) expression negatively impact cognitive functions in schizophrenia through a functional epistatic interaction with Catechol-O-methyltransferase (COMT). Dys is involved in the trafficking of dopaminergic receptors, crucial for prefrontal cortex (PFC) signaling regulation. Moreover, dopamine signaling is modulated by estrogens via inhibition of COMT expression. We hypothesized a sex dimorphism in Dys-related cognitive functions dependent on COMT and estrogen levels. Our multidisciplinary approach combined behavioral-molecular findings on genetically modified mice, human postmortem Dys expression data, and in vivo fMRI during a working memory task performance. We found cognitive impairments in male mice related to genetic variants characterized by reduced Dys protein expression (pBonferroni = 0.0001), as well as in male humans through a COMT/Dys functional epistatic interaction involving PFC brain activity during working memory (t(23) = −3.21; pFDR = 0.004). Dorsolateral PFC activity was associated with lower working memory performance in males only (p = 0.04). Also, male humans showed decreased Dys expression in dorsolateral PFC during adulthood (pFDR = 0.05). Female Dys mice showed preserved cognitive performances with deficits only with a lack of estrogen tested in an ovariectomy model (pBonferroni = 0.0001), suggesting that genetic variants reducing Dys protein expression could probably become functional in females when the protective effect of estrogens is attenuated, i.e., during menopause. Overall, our results show the differential impact of functional variants of the DTBPN1 gene interacting with COMT on cognitive functions across sexes in mice and humans, underlying the importance of considering sex as a target for patient stratification and precision medicine in schizophrenia.
... Moreover, sometimes depression precedes Parkinson's disease [9]. People with schizophrenia are prone to develop Parkinson's disease, while parkinsonism can lead to psychoses [10]. Some signs of schizophrenia can also be observed in AD, and some researchers point to the same risk factors for AD and schizophrenia [11]. ...
Article
Full-text available
Mental disorders represent common brain diseases characterized by substantial impairments of social and cognitive functions. The neurobiological causes and mechanisms of psychopathologies still have not been definitively determined. Various forms of brain proteinopathies, which include a disruption of protein conformations and the formation of protein aggregates in brain tissues, may be a possible cause behind the development of psychiatric disorders. Proteinopathies are known to be the main cause of neurodegeneration, but much less attention is given to the role of protein impairments in psychiatric disorders’ pathogenesis, such as depression and schizophrenia. For this reason, the aim of this review was to discuss the potential contribution of protein illnesses in the development of psychopathologies. The first part of the review describes the possible mechanisms of disruption to protein folding and aggregation in the cell: endoplasmic reticulum stress, dysfunction of chaperone proteins, altered mitochondrial function, and impaired autophagy processes. The second part of the review addresses the known proteins whose aggregation in brain tissue has been observed in psychiatric disorders (amyloid, tau protein, α-synuclein, DISC-1, disbindin-1, CRMP1, SNAP25, TRIOBP, NPAS3, GluA1, FABP, and ankyrin-G).
... Dysbindin-1 is a critically important protein encoded by DTNBP1 that has implications in neuronal neurodevelopment and neuroplasticity. Its abnormal functioning has been linked to schizophrenia [47,48]. Dysbindin-1 is abundant in the prefrontal area and hippocampus in clinically healthy individuals. ...
Article
Full-text available
The article is a review of the latest meta-analyses regarding the genetic spectrum in schizophrenia, discussing the risks given by the disrupted-in-schizophrenia 1 (DISC1), catechol-O-methyltransferase (COMT), monoamine oxidases-A∕B (MAO-A∕B), glutamic acid decarboxylase 67 (GAD67) and neuregulin 1 (NRG1) genes, and dysbindin-1 protein. The DISC1 polymorphism significantly increases the risk of schizophrenia, as well injuries from the prefrontal cortex that affect connectivity. NRG1 is one of the most important proteins involved. Its polymorphism is associated with the reduction of areas in the corpus callosum, right uncinate, inferior lateral fronto-occipital fascicle, right external capsule, fornix, right optic tract, gyrus. NRG1 and the ErbB4 receptor (tyrosine kinase receptor) are closely related to the N-methyl-D-aspartate receptor (NMDAR) (glutamate receptor). COMT is located on chromosome 22 and together with interleukin-10 (IL-10) have an anti-inflammatory and immunosuppressive function that influences the dopaminergic system. MAO gene methylation has been associated with mental disorders. MAO-A is a risk gene in the onset of schizophrenia, more precisely a certain type of single-nucleotide polymorphism (SNP), at the gene level, is associated with schizophrenia. In schizophrenia, we find deficits of the γ-aminobutyric acid (GABA)ergic neurotransmitter, the dysfunctions being found predominantly at the level of the substantia nigra. In schizophrenia, missing an allele at GAD67, caused by a SNP, has been correlated with decreases in parvalbumin (PV), somatostatin receptor (SSR), and GAD ribonucleic acid (RNA). Resulting in the inability to mature PV and SSR neurons, which has been associated with hyperactivity.
... Findings of a GWAS on a smaller sample including 726 patients with schizophrenia and 667 healthy controls suggested that variants in a few genes may modulate cognitive phenotypes: NRG3 (neuregulin 3) for abstraction and mental flexibility, SHANK3 (SH3 And Multiple Ankyrin Repeat Domains 3) and HCN1 (hyperpolarization activated cyclic nucleotide gated potassium channel 1) for spatial memory (Greenwood et al., 2019). Other studies focused on the role of single genes in cognitive function in patients with schizophrenia (e.g., dysbindin-1 and dopamine D3 receptor genes; Leggio et al., 2021;Waddington et al., 2020). ...
Article
Full-text available
Background: Cognitive dysfunction is a core manifestation of schizophrenia and one of the best predictors of long-term disability. Genes increasing risk for schizophrenia may partly act through the modulation of cognition. Methods: We imputed the expression of 130 genes recently prioritized for association with schizophrenia, using PsychENCODE variant weights and genotypes of patients with schizophrenia in CATIE. Processing speed, reasoning, verbal memory, working memory, vigilance, and a composite cognitive score were used as phenotypes. We performed linear regression models for each cognitive measure and gene expression score, adjusting for age, years of education, antipsychotic treatment, years since the first antipsychotic treatment and population principal components. Results: We included 425 patients and expression scores of 91 genes (others had no heritable expression; Bonferroni corrected alpha = 5.49e-4). No gene expression score was associated with cognitive measures, though ENOX1 expression was very close to the threshold for verbal memory (p = 6e-4) and processing speed (p = 7e-4). Other genes were nominally associated with multiple phenotypes (MAN2A1 and PCGF3). Conclusion: A better understanding of the mechanisms mediating cognitive dysfunction in schizophrenia may help in the definition of disease prognosis and in the identification of new treatments, as the treatment of cognitive impairment remains an unmet therapeutic need.
Article
The authors present the data indicating that the formation of psychopathological symptoms of schizophrenia is due to complex and diverse genetic factors associated with various functional and metabolic pathways at different stages of ontogenesis. Despite the fact that at present the genetic basis of positive and negative symptoms as the main pathophysiological manifestations of schizophrenia remains largely unknown, the current level of research allows the identification of some common and unique associations for positive and negative disorders. Based on the analysis of the literature, the specificity of the association of genetic variants with negative symptoms of schizophrenia is shown. It has been also suggested that genes of the immune system may be specifically associated with negative symptoms of schizophrenia. The relevance of studying the relationship of immune system genes, in particular, pro- and anti-inflammatory cytokines, with dimensional characteristics of negative symptoms (abulia-apathy and expressive deficit) is substantiated. Studies of this type have not yet been conducted, despite accumulating data indicating that the heterogeneity of negative symptoms is based on different neurobiological mechanisms. It is concluded that the immunological and molecular genetic study of the subdomains of psychopathological symptoms can be promising as part of the transition to deep phenotyping, which seems to be especially relevant for the study of such an extremely heterogeneous disease from a clinical point of view as schizophrenia. The development of this area is important for solving the problems of precision medicine, which aims to provide the most effective therapy for a particular patient by stratifying the disease into subclasses, taking into account their biological basis.
Article
Full-text available
Improving the quality of life in developed countries has contributed to an increase in its duration, which has led to an increase in the number of reported cases of Alzheimer's disease (AD) and Parkinson's disease (PD) in the world. Today, there are 26.6 million patients with AD in the world and it is suspected that by 2050 the number of such patients may increase four times. Additionally, PD in different countries is recorded among people above 60-65 years old at a level of 167 to 5703 per 100.000 population. The latest studies have made it possible to formulate the main mechanisms of the «microbiota-gut-brain» axis associated with the pathogenesis of some neurodegenerative diseases. In this review, we summarize the currently available information on the possible role of the gut microbiota in the AD and PD development. It was shown that oxidative stress is one of the main pathogenetic mechanisms of the development of neurodegenerative diseases. In addition, the deposition of lipopolysaccharides of gram-negative bacteria and amyloid of microbial origin in the brain tissue of patients with impaired permeability of the intestinal barrier plays an important role in AD. In PD, the synthesis of α-synuclein produced by bacteria and neuroinflammation are of the greatest importance. Knowledge of these mechanisms will allow the development of psychobiotics, which will reduce the risk of neurodegeneration in AD and PD.
Article
Full-text available
The dopamine D2 and D3 receptors are implicated in schizophrenia and its pharmacological treatments. These receptors undergo intracellular trafficking processes that are modulated by dysbindin-1 (Dys). Indeed, Dys variants alter cognitive responses to antipsychotic drugs through D2-mediated mechanisms. However, the mechanism by which Dys might selectively interfere with the D3 receptor subtype is unknown. Here, we revealed an interaction between functional genetic variants altering Dys and D3. Specifically, both in patients with schizophrenia and in genetically modified mice, concomitant reduction in D3 and Dys functionality was associated with improved executive and working memory abilities. This D3/Dys interaction produced a D2/D3 imbalance favoring increased D2 signaling in the prefrontal cortex (PFC) but not in the striatum. No epistatic effects on the clinical positive and negative syndrome scale (PANSS) scores were evident, while only marginal effects on sensorimotor gating, locomotor functions, and social behavior were observed in mice. This genetic interaction between D3 and Dys suggests the D2/D3 imbalance in the PFC as a target for patient stratification and procognitive treatments in schizophrenia.
Article
Full-text available
Background: Cognitive impairment is a clinically important feature of schizophrenia. Polygenic risk score (PRS) methods have demonstrated genetic overlap between schizophrenia, bipolar disorder (BD), major depressive disorder (MDD), educational attainment (EA), and IQ, but very few studies have examined associations between these PRS and cognitive phenotypes within schizophrenia cases. Methods: We combined genetic and cognitive data in 3034 schizophrenia cases from 11 samples using the general intelligence factor g as the primary measure of cognition. We used linear regression to examine the association between cognition and PRS for EA, IQ, schizophrenia, BD, and MDD. The results were then meta-analyzed across all samples. A genome-wide association studies (GWAS) of cognition was conducted in schizophrenia cases. Results: PRS for both population IQ (P = 4.39 × 10-28) and EA (P = 1.27 × 10-26) were positively correlated with cognition in those with schizophrenia. In contrast, there was no association between cognition in schizophrenia cases and PRS for schizophrenia (P = .39), BD (P = .51), or MDD (P = .49). No individual variant approached genome-wide significance in the GWAS. Conclusions: Cognition in schizophrenia cases is more strongly associated with PRS that index cognitive traits in the general population than PRS for neuropsychiatric disorders. This suggests the mechanisms of cognitive variation within schizophrenia are at least partly independent from those that predispose to schizophrenia diagnosis itself. Our findings indicate that this cognitive variation arises at least in part due to genetic factors shared with cognitive performance in populations and is not solely due to illness or treatment-related factors, although our findings are consistent with important contributions from these factors.
Article
Full-text available
A recent development in the genetic architecture of schizophrenia suggested that an omnigenic model may underlie the risk for this disorder. The aim of our study was to use polygenic profile scoring to quantitatively assess whether a number of experimentally derived sets would contribute to the disorder above and beyond the omnigenic effect. Using the PGC2 secondary analysis schizophrenia case-control cohort (N = 29,125 cases and 34,836 controls), a robust polygenic signal was observed from gene sets based on TCF4, FMR1, upregulation from MIR137 and downregulation from CHD8. Additional analyses revealed a constant floor effect in the amount of variance explained, consistent with the omnigenic model. Thus, we report that putative core gene sets showed a significant effect above and beyond the floor effect that might be linked with the underlying omnigenic background. In addition, we demonstrate a method to quantify the contribution of specific gene sets within the omnigenic context.
Article
Full-text available
Schizophrenia is a heritable complex phenotype associated with a background risk involving multiple common genetic variants of small effect and a multitude of environmental exposures. Early twin and family studies using proxy‐genetic liability measures suggest gene‐environment interaction in the etiology of schizophrenia spectrum disorders, but the molecular evidence is scarce. Here, by analyzing the main and joint associations of polygenic risk score for schizophrenia (PRS‐SCZ) and environmental exposures in 1,699 patients with a diagnosis of schizophrenia spectrum disorders and 1,542 unrelated controls with no lifetime history of a diagnosis of those disorders, we provide further evidence for gene‐environment interaction in schizophrenia. Evidence was found for additive interaction of molecular genetic risk state for schizophrenia (binary mode of PRS‐SCZ above 75% of the control distribution) with the presence of lifetime regular cannabis use and exposure to early‐life adversities (sexual abuse, emotional abuse, emotional neglect, and bullying), but not with the presence of hearing impairment, season of birth (winter birth), and exposure to physical abuse or physical neglect in childhood. The sensitivity analyses replacing the a priori PRS‐SCZ at 75% with alternative cut‐points (50% and 25%) confirmed the additive interaction. Our results suggest that the etiopathogenesis of schizophrenia involves genetic underpinnings that act by making individuals more sensitive to the effects of some environmental exposures.
Article
Neuroticism is an important risk factor for psychiatric traits, including depression 1, anxiety 2,3, and schizophrenia 4-6 . At the time of analysis, previous genome-wide association studies 7-12 (GWAS) reported 16 genomic loci associated to neuroticism 10-12 . Here we conducted a large GWAS meta-analysis (n = 449,484) of neuroticism and identified 136 independent genome-wide significant loci (124 new at the time of analysis), which implicate 599 genes. Functional follow-up analyses showed enrichment in several brain regions and involvement of specific cell types, including dopaminergic neuroblasts (P = 3.49 × 10-8), medium spiny neurons (P = 4.23 × 10-8), and serotonergic neurons (P = 1.37 × 10-7). Gene set analyses implicated three specific pathways: neurogenesis (P = 4.43 × 10-9), behavioral response to cocaine processes (P = 1.84 × 10-7), and axon part (P = 5.26 × 10-8). We show that neuroticism's genetic signal partly originates in two genetically distinguishable subclusters 13 ('depressed affect' and 'worry'), suggesting distinct causal mechanisms for subtypes of individuals. Mendelian randomization analysis showed unidirectional and bidirectional effects between neuroticism and multiple psychiatric traits. These results enhance neurobiological understanding of neuroticism and provide specific leads for functional follow-up experiments.
Article
Genome-wide association studies (GWAS) involve testing genetic variants across the genomes of many individuals to identify genotype–phenotype associations. GWAS have revolutionized the field of complex disease genetics over the past decade, providing numerous compelling associations for human complex traits and diseases. Despite clear successes in identifying novel disease susceptibility genes and biological pathways and in translating these findings into clinical care, GWAS have not been without controversy. Prominent criticisms include concerns that GWAS will eventually implicate the entire genome in disease predisposition and that most association signals reflect variants and genes with no direct biological relevance to disease. In this Review, we comprehensively assess the benefits and limitations of GWAS in human populations and discuss the relevance of performing more GWAS.
Article
Cognitive impairment in schizophrenia involves a broad array of nonsocial and social cognitive domains. It is a core feature of the illness, and one with substantial implications for treatment and prognosis. Our understanding of the causes, consequences and interventions for cognitive impairment in schizophrenia has grown substantially in recent years. Here we review a range of topics, including: a) the types of nonsocial cognitive, social cognitive, and perceptual deficits in schizophrenia; b) how deficits in schizophrenia are similar or different from those in other disorders; c) cognitive impairments in the prodromal period and over the lifespan in schizophrenia; d) neuroimaging of the neural substrates of nonsocial and social cognition, and e) relationships of nonsocial and social cognition to functional outcome. The paper also reviews the considerable efforts that have been directed to improve cognitive impairments in schizophrenia through novel psychopharmacology, cognitive remediation, social cognitive training, and alternative approaches. In the final section, we consider areas that are emerging and have the potential to provide future insights, including the interface of motivation and cognition, the influence of childhood adversity, metacognition, the role of neuroinflammation, computational modelling, the application of remote digital technology, and novel methods to evaluate brain network organization. The study of cognitive impairment has provided a way to approach, examine and comprehend a wide range of features of schizophrenia, and it may ultimately affect how we define and diagnose this complex disorder.
Article
Cognitive deficit is thought to represent, at least in part, genetic mechanisms of risk for schizophrenia, with recent evidence from statistical modelling of twin data suggesting direct causality from the former to the latter. However, earlier evidence was based on inferences from twin not molecular genetic data and it is unclear how much genetic influence 'passes through' cognition on the way to diagnosis. Thus, we included direct measurements of genetic risk (e.g. schizophrenia polygenic risk scores) in causation models to assess the extent to which cognitive deficit mediates some of the effect of polygenic risk scores on the disorder. Causal models of family data tested relationships among key variables and allowed parsing of genetic variance components. Polygenic risk scores were calculated from summary statistics from the current largest genome-wide association study of schizophrenia and were represented as a latent trait. Cognition was also modelled as a latent trait. Participants were 1313 members of 1078 families: 416 patients with schizophrenia, 290 unaffected siblings, and 607 controls. Modelling supported earlier findings that cognitive deficit has a putatively causal role in schizophrenia. In total, polygenic risk score explained 8.07% [confidence interval (CI) 5.45-10.74%] of schizophrenia risk in our sample. Of this, more than a third (2.71%, CI 2.41-3.85%) of the polygenic risk score influence was mediated through cognition paths, exceeding the direct influence of polygenic risk score on schizophrenia risk (1.43%, CI 0.46-3.08%). The remainder of the polygenic risk score influence (3.93%, CI 2.37-4.48%) reflected reciprocal causation between schizophrenia liability and cognition (e.g. mutual influences in a cyclical manner). Analysis of genetic variance components of schizophrenia liability indicated that 26.87% (CI 21.45-32.57%) was associated with cognition-related pathways not captured by polygenic risk score. The remaining variance in schizophrenia was through pathways other than cognition-related and polygenic risk score. Although our results are based on inference through statistical modelling and do not provide an absolute proof of causality, we find that cognition pathways mediate a significant part of the influence of cumulative genetic risk on schizophrenia. We estimate from our model that 33.51% (CI 27.34-43.82%) of overall genetic risk is mediated through influences on cognition, but this requires further studies and analyses as the genetics of schizophrenia becomes better characterized.
Article
Brain-derived neurotrophic factor (BDNF) may be related to the pathophysiology of schizophrenia. This study aims to examine the relation between plasma BDNF levels and the cognition of patients with schizophrenia. We recruited 31 patients with chronic schizophrenia, 34 first-episode patients, and 35 healthy control subjects. We examined the MATRICS Consensus Cognitive Battery (MCCB) and the plasma BDNF levels in all groups. The schizophrenic symptoms were assessed using the positive and negative syndrome scale. The BDNF levels of schizophrenic patients were remarkably lower than those of the controls. The cognitive MCCB global composite scores and part index scores of schizophrenic patients were remarkably lower than those of the controls. Moreover, remarkable correlations were observed between BDNF levels and partial cognitive dimensions, such as visual learning, memory, and processing speed. Therefore, BDNF may be involved in the pathophysiology and cognitive impairment of schizophrenia.