ArticlePDF Available

Exploring the pathogenetic association between Schizophrenia and type 2 diabetes mellitus diseases based on pathway analysis

Authors:

Abstract and Figures

Background Schizophrenia (SCZ) and type 2 diabetes mellitus (T2D) are both complex diseases. Accumulated studies indicate that schizophrenia patients are prone to present the type 2 diabetes symptoms, but the potential mechanisms behind their association remain unknown. Here we explored the pathogenetic association between SCZ and T2D based on pathway analysis and protein-protein interaction. Results With sets of prioritized susceptibility genes for SCZ and T2D, we identified significant pathways (with adjusted p-value < 0.05) specific for SCZ or T2D and for both diseases based on pathway enrichment analysis. We also constructed a network to explore the crosstalk among those significant pathways. Our results revealed that some pathways are shared by both SCZ and T2D diseases through a number of susceptibility genes. With 382 unique susceptibility proteins for SCZ and T2D, we further built a protein-protein interaction network by extracting their nearest interacting neighbours. Among 2,104 retrieved proteins, 364 of them were found simultaneously interacted with susceptibility proteins of both SCZ and T2D, and proposed as new candidate risk factors for both diseases. Literature mining supported the potential association of partial new candidate proteins with both SCZ and T2D. Moreover, some proteins were hub proteins with high connectivity and interacted with multiple proteins involved in both diseases, implying their pleiotropic effects for the pathogenic association. Some of these hub proteins are the components of our identified enriched pathways, including calcium signaling, g-secretase mediated ErbB4 signaling, adipocytokine signaling, insulin signaling, AKT signaling and type II diabetes mellitus pathways. Through the integration of multiple lines of information, we proposed that those signaling pathways, which contain susceptibility genes for both diseases, could be the key pathways to bridge SCZ and T2D. AKT could be one of the important shared components and may play a pivotal role to link both of the pathogenetic processes. Conclusions Our study is the first network and pathway-based systematic analysis for SCZ and T2D, and provides the general pathway-based view of pathogenetic association between two diseases. Moreover, we identified a set of candidate genes potentially contributing to the linkage between these two diseases. This research offers new insights into the potential mechanisms underlying the co-occurrence of SCZ and T2D, and thus, could facilitate the inference of novel hypotheses for the co-morbidity of the two diseases. Some etiological factors that exert pleiotropic effects shared by the significant pathways of two diseases may have important implications for the diseases and could be therapeutic intervention targets.
Content may be subject to copyright.
RESEARCH Open Access
Exploring the pathogenetic association between
schizophrenia and type 2 diabetes mellitus
diseases based on pathway analysis
Yanli Liu
1
, Zezhi Li
2
, Meixia Zhang
3
, Youping Deng
4
, Zhenghui Yi
5*
, Tieliu Shi
1*
From The 2011 International Conference on Bioinformatics and Computational Biology (BIOCOMP11)
Las Vegas, NV, USA. 18-21 July 2011
Abstract
Background: Schizophrenia (SCZ) and type 2 diabetes mellitus (T2D) are both complex diseases. Accumulated
studies indicate that schizophrenia patients are prone to present the type 2 diabetes symptoms, but the potential
mechanisms behind their association remain unknown. Here we explored the pathogenetic association between
SCZ and T2D based on pathway analysis and protein-protein interaction.
Results: With sets of prioritized susceptibility genes for SCZ and T2D, we identified significant pathways (with adjusted
p-value < 0.05) specific for SCZ or T2D and for both diseases based on pathway enrichment analysis. We also
constructed a network to explore the crosstalk among those significant pathways. Our results revealed that some
pathways are shared by both SCZ and T2D diseases through a number of susceptibility genes. With 382 unique
susceptibility proteins for SCZ and T2D, we further built a protein-protein interaction network by extracting their nearest
interacting neighbours. Among 2,104 retrieved proteins, 364 of them were found simultaneously interacted with
susceptibility proteins of both SCZ and T2D, and proposed as new candidate risk factors for both diseases. Literature
mining supported the potential association of partial new candidate proteins with both SCZ and T2D. Moreover, some
proteins were hub proteins with high connectivity and interacted with multiple proteins involved in both diseases,
implying their pleiotropic effects for the pathogenic association. Some of these hub proteins are the components of
our identified enriched pathways, including calcium signaling, g-secretase mediated ErbB4 signaling, adipocytokine
signaling, insulin signaling, AKT signaling and type II diabetes mellitus pathways. Through the integration of multiple
lines of information, we proposed that those signaling pathways, which contain susceptibility genes for both diseases,
could be the key pathways to bridge SCZ and T2D. AKT could be one of the important shared components and may
play a pivotal role to link both of the pathogenetic processes.
Conclusions: Our study is the first network and pathway-based systematic analysis for SCZ and T2D, and provides
the general pathway-based view of pathogenetic association between two diseases. Moreover, we identified a set
of candidate genes potentially contributing to the linkage between these two diseases. This research offers new
insights into the potential mechanisms underlying the co-occurrence of SCZ and T2D, and thus, could facilitate the
inference of novel hypotheses for the co-morbidity of the two diseases. Some etiological factors that exert
pleiotropic effects shared by the significant pathways of two diseases may have important implications for the
diseases and could be therapeutic intervention targets.
* Correspondence: yizhenghui1971@gmail.com; tieliushi01@gmail.com
1
Center for Bioinformatics and Computational Biology, and The Institute of
Biomedical Sciences, School of Life Sciences, East China Normal University,
500 Dongchuan Road, Shanghai 200241, China
5
Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong
University School of Medicine, 600 Wan Ping Nan Road, Shanghai 200030,
China
Full list of author information is available at the end of the article
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
© 2013 Liu et al.; li censee BioMed Central Ltd. This is an open access article distributed under the terms of the Crea tive Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unr estricted use, distribution, and reproduction in
any medium, provided the original work is properly cited.
Background
Schizophrenia (SCZ) is a chronic, severe, and disabling
brain disorder that has affected people with lifelong dis-
ability. The phenotype is heterogeneous and complex,
with multiple genes and environmental exposures likely
involved. It is characterized by a breakdown of thought
processes and by poor emotional responsiveness. It most
commonly manifests itself as auditory hallucinations,
paranoid or bizarre delusions, or disorganized speech
and thinking, and it is accompanied by significant social
or occupational dysfunction. The onset of symptoms
typically occurs in young adulthood with 1% prevalence
in the general population [1]. Recently, researchers have
identified specific genes/markers and chromosomal
regions for SCZ through numerous genetic studies, such
as linkage scans and their meta-analyses, candidate gene
association analyses, gene expression and genome-wide
association studies (GWAS) [2-5].
Type 2 diabetes mellitus (T2D) is characterized by per-
sistent high blood glucose in the context of insulin resis-
tance and relative insulin deficiency, due to pancreatic
beta-cell dysfunction. Cardiovascular diseases, chronic
renal failure, retinal, and nerve damage are usual compli-
cations of this illness. Many genes and pathways have
also been implicated with the T2D, but the mechanisms
underlying the connections remain further investigation.
Recently studies indicate that the prevalence of T2D
among individuals suffering from schizophrenia or schi-
zoaffective disorders is significant higher than that of the
general population [6,7]. For instance, a recent study
reported that T2D is more common in schizophrenics
than normal controls in Canada, especially in young males
and females [8]. Another recent study also reported an ele-
vated risk of T2D in schizophrenic individuals in Taiwan
[9].
Molecular inference and GWAS studies also point out
that SCZ shares substantial polygenetic component with
T2D. Increased attention is now being given to a possible
genetic basis for co-morbidity of SCZ and T2D [10]. The
pathogenetic association between SCZ and T2D has been
recognized but the potential mechanism behind the asso-
ciation has not been fully explored [10]. Recently, more
and more researchers have paid their attentions to iden-
tify the candidate genes for human diseases, including
T2D and SCZ, mainly through genome-wide association,
transcriptomic and proteomic expression studies. These
have greatly facilitated the research of genetic basis for
pathogenetic association between SCZ and T2D. It is
well accepted that genes or proteins usually interact with
each other to form complexes or pathways within a cell,
rather than function alone to carry out biological func-
tions [11]. Considering that SCZ and T2D are both com-
plex diseases, their pathogenesis is believed coupled with
lots of factors. Lin has proposed three models for hypoth-
eses concerning the co-morbidity between SCZ and T2D
[10]. One of the models suggested that T2D and SCZ are
caused by shared etiological factors, which is consistent
with other research result that T2D and SCZ are caused
by multiple genetic variants [12]. From this perspective,
we can link these two diseases by their shared susceptibil-
ity genes. Those genes may exert pleiotropic effects; it
means they play roles in two different pathological path-
ways, one related to SCZ and the other associated with
T2D. For example, TCF7L2, one of the best confirmed
susceptibility genes for T2D, has been also inferred to
strongly relate to SCZ. On one hand, TCF7L2 acts a role
in pancreatic beta cell function; on the other hand, it is a
transcription factor involved in the Wnt/beta-catenin sig-
naling [13]. Since Wnt signaling pathway plays a role in
the development of central nervous system (CNS) [14],
and has been also associated with SCZ [15,16], TCF7L2
may contribute to the co-morbidity of SCZ and T2D
through Wnt signaling pathway [17]. In addition to
genetic factors, environmental factors may also influence
susceptibility to both SCZ and T2D, and anti-psychotic
medications can also trigger the pathogenetic association
between SCZ and T2D. Although significant attentions
have been paid to explore the association between SCZ
and T2D, not much progress has been made and the
potential mechanisms remain unclear.
It is hypothesized that many genes may contribute
major risk to SCZ through their interaction and com-
bined effects, with each gene might contribute a small or
moderate risk. Similarly, T2D has also been regarded as a
complex disease and associated with the dysfunctions of
multiple genes. Therefore, we assumed that proteins that
interact with both SCZ proteins and T2D proteins should
also be the potential ones to contribute to both diseases.
Accordingly, in this study, we used those susceptibility
genes that have been implicated for SCZ or T2D in gen-
ome wide association study (GWAS) as the basis and
retrieved their nearest interactive partners from human
protein interaction data to construct a protein-protein
interaction network. Next, we selected those novel candi-
date genes from the network that interact with both SCZ
related proteins and T2D related proteins. In this way,
we prioritized a set of new candidate genes related to
both diseases. Moreover, considering that different biolo-
gical processes for these two diseases may share the same
susceptibility genes, we conducted pathway enrichment
analysis with those susceptibility genes related to two dis-
eases, and identified the pathways common to these two
diseases and those genes participating into those path-
ways. Through the pathway analysis, we tried to link the
pathogenetic association between the two diseases at the
molecular level.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 2 of 14
Materials and methods
Susceptibility gene sets of SCZ and T2D
SCZ susceptibility genes were extracted from two publicly
available databases: Genetic Association Database [18,19]
and A Catalog of Published Genome-Wide Association
Studies [20,21]. The former is an archive of human genetic
association studies of complex diseases and disorders,
which includes summary data extracted from published
papers in peer reviewed journals on candidate gene and
GWAS studies (updated to June 9, 2012); the latter is an
online catalogue of SNP-trait associations from published
genome-wide association studies for use in investigating
genomic characteristics of trait/disease-associated SNPs
(TASs) (updated by June 22, 2012). T2D susceptibility
genes were collected from three main sources: the first
was Type 2 Diabetes Genetic Association Database
[22,23], and this database provides specialized information
on the genetic risk factors involved in the development of
T2D. Among the data in this database, we only selected
genes reported in more than two independent studies.
The other two data sources were the same as SCZ genes.
The follow-up analyses are based on these two susceptibil-
ity genes sets. A detailed flow chart of my methodology is
illustrated in Figure 1.
Significant pathway enrichment analysis
To carry out the pathway enrichment analysis, we
uploaded SCZ and T2D susceptibility genes into Cytos-
cape as cluster 1 and cluster 2, respectively, and ClueGO
was used for pathway enrichment analysis for all those
genes [24]. Two pathway databases, Kyoto Encyclopedia of
Genes and Genomes (KEGG) pathway [25,26] and Bio-
Carta pathway [27,28], were selected for pathway enrich-
ment analysis. Those susceptibility genes were mapped to
their enriched pathways based on the hypergeometric test,
and p-value was corrected by Benjamini-Hochberg
method [29]. It is possible that genes from both clusters
are associated with one pathway, but in different propor-
tions. Here we defined an enriched pathway specific to
one of the clusters if over 66% genes in the pathway are
from this cluster. Pathways with adjusted p-value < 0.05
were regarded as significant enriched pathways and were
selected for further analysis.
Pathway-pathway interaction network construction
To visually represent relationships between the selected
significant pathways, a pathway-pathway interaction net-
work was created, in which the node represented the sig-
nificant pathway, the edge between the significant
pathways was defined according to kappa scores which
were calculated based on any pathway pair shared genes
in a similar way as described by DAVID software [30].
The different proportion of the genes from the analyzed
clusters was represented with a colour gradient from blue
for the first cluster genes, to red for the second cluster.
Approximately equal proportions of the two clusters
were represented in light-yellow. The genes shared by
any pathway pair and those mapped to corresponding
significant pathways were also displayed in this network
as small nodes with different colours to distinguish them
from pathway nodes. The network was automatically laid
out using the Organic layout algorithm supported by
Cytoscape.
Protein-protein interaction data
Protein-Protein interaction data was downloaded from
Human Protein Reference Database (HPRD, version: 13
April, 2010) [31,32]. After removing self interactions and
disperse nodes, we ended up with 36,727 interactions
which cover 9,205 human genes. All proteins encoded by
unique susceptibility genes of two diseases were mapped
into HPRD, and then we extracted those proteins that
directly interact with our susceptibility proteins, and con-
structed a protein-protein interaction network in which a
node is a protein and an edge represents interaction
between two proteins.
New candidate genes prediction
Among all the nearest interacting proteins, those simul-
taneously interacting with both SCZ and T2D suscept-
ibility gene products were selected, then we constructed
a sub-network with them and their interacted susceptibil-
ity proteins. Next, we performed extensively literature
mining in PubMed to determine whether the relationship
between a candidate protein and SCZ or T2D has been
supported by previous studies. Based on these two
aspects evidence we predicted those genes with pleiotro-
pic effects as the risk factors that may contribute to the
pathogenetic association between SCZ and T2D.
Results
SCZ and T2D susceptibility gene sets
All the susceptibility genes were selected based on the
Genome-Wide Association Studies (GWAS). For SCZ
susceptibility genes, we retrieved 169 genes from
Genetic Association Database and 57 genes from data-
base of A Catalog of Published Genome-Wide Associa-
tion Studies. For T2D related genes, we extracted 26
genes and 79 genes from each of above databases,
respectively. In addition, we collected 143 genes from
Type 2 Diabetes Genetic Association Database. After
removing redundancy, we obtained 196 susceptibility
genes for SCZ and 200 for T2D, among them, 14 genes
are in common for both diseases (Additional file 1).
Enrichment pathway analysis
To perform functional enrichment tests of the suscept-
ibility genes, we uploaded SCZ and T2D related genes,
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 3 of 14
named as cluster 1 and cluster 2 respectively, into
ClueGO, a Cytoscape plug-in to decipher biological net-
works, and mapped them to their enrichment pathways.
Here, considering the incomplete of each pathway anno-
tation system, we selected two main pathway databases,
KEGG and BioCarta to conduct our analysis. As a result,
we ended up with 10 significant pathways (with adjusted
p-value < 0.05) specific to SCZ, 11 significant pathways
specific to T2D, and 7 pathways for both diseases (Table
1). Here we defined an enriched pathway specific to one
of the clusters if over 66% genes (the default value in
the system) in the pathway are from this cluster. Inter-
estingly, some of the enriched pathways, even though
they were classified as one of the clusters based on the
statistics, they included genes for both SCZ and T2D,
such as Adipocytokine signaling pathway and PPAR sig-
naling pathway, both of them were clustered as T2D
pathways. In fact, for 18 susceptibility genes in the Adi-
pocytokine signaling pathway, 4 of them are related to
SCZ, while 12 of them are identified to T2D related
genes, and the rest 2 genes have been linked to both
SCZ and T2D. PPAR signaling pathway includes 13
T2D related genes and 2 SCZ related genes. Neuroactive
ligand-receptor interaction pathway and Calcium
signaling pathway [33] were enriched as SCZ pathways.
There are 35 genes in Neuroactive ligand-receptor inter-
action pathway, and 26 of them are related to SCZ,
while the rest 9 genes come from T2D gene list. Cal-
cium signaling pathway contains 18 genes implicated to
SCZ, and 5 genes linked to T2D.
Next, to explore the association and crosstalk between
those different enriched pathways, we constructed a
pathway-based network with all those 28 significant path-
ways in which a large node is a pathway and an edge
represents crosstalk between two pathways through their
shared genes (Figure 2). The genes shared by any path-
way pair and those mapped to corresponding significant
pathways were displayed in this network as small nodes
with different colours to distinguish them from pathway
nodes. From the pathway-pathway interaction network, it
can be observed that many genes are shared by multiple
pathways, such as TNF shared by over 12 different signal-
ing pathways, AKT1 participating into 4 different signal-
ling pathways (Additional file 2).
New candidate risk gene inference
To infer new genes associated with both SCZ and T2D,
we conducted network analysis based on protein-protein
Genetic Association Database ˖169
A Catalog of Published Genome-Wide Association Studies˖57
196 SCZ susceptibility genes sources
Genetic Association Database ˖26
A Catalog of Published Genome-Wide Association Studies˖79
Type 2 Diabetes Genetic Association Database:143
200 T2D susceptibility genes sources
Unique SCZ susceptibility genes ˖182
Unique T2D susceptibility genes˖186
Common susceptibility genes:14
Pathway Enrichment Analysis
Cytoscape:ClueGO
The Hypergeometric Test
Significant Pathways Specific to SCZ:10
Common Significant
Pathways:7
Significant Pathways Specific to T2D:11
P-value< 0.05
Adjusted by Benjamini-Hochberg metho
d
Pathway-Pathway Interaction Network
Protein-Protein Interaction Network
Human Protein Reference Database
SCZ and T2D Molecular Network
Systematic Literature Mining
PubMed
New Candidate Genes Prediction
PubMed
Figure 1 A detailed flow chart for the analysis process. The green, pink, blue and yellow denote gene sources, pathway enrichment analysis,
network construction and new candidate gene prediction, respectively.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 4 of 14
interaction (PPI). First, we downloaded human PPI data
from HPRD. Next, we mapped all 382 unique SCZ and
T2D susceptibility gene related proteins (susceptibility
proteins) to the human PPI data, only proteins that have
their interacting partners in the HPRD data were selected
in our further analysis. Then we retrieved those suscept-
ibility proteins with their nearest interacting neighbours
from the PPI data. After removing self-interaction and
duplicates, the final network included a total of 2,104
nodes and 3,155 interactions (Additional file 3). Those
2,104 proteins included 143 SCZ susceptibility proteins,
138 T2D susceptibility proteins, 12 common susceptibil-
ity proteins and 1,811 their direct interaction partners.
Among the 1,811 protein partners, there were 1,108 pro-
teins that interact with more than one SCZ susceptibility
proteins, 1,067 proteins with more than one T2D
susceptibility proteins, and 364 proteins with both dis-
easessusceptibility proteins. We proposed those 364
proteins as new candidate risk factors for both SCZ and
T2D according to function association (guilt by associa-
tion) rule. Function association refers to that if two pro-
teins interact with one another, they usually participate
in the same, or related, cellular functions. Based on this
assumption, new functions of proteins can be inferred
with their interaction partners.
The 364 candidate proteins and their interacted suscept-
ibility proteins may provide new relationship for elucidat-
ing the common molecular pathways that may underlie
both SCZ and T2D. So we extracted those 364 candidate
proteins and their interacted susceptibility proteins from
the entire network to construct a sub-network (Additional
file 4). In this sub-network, among all 364 candidate
Table 1 The 28 significant pathways analysed using ClueGO.
Significant Pathways Specific To
cluster
Adjust P-
Value
Total
number
Gene source
unique SCZ
gene count
unique T2D
gene count
Common
gene count
Adipocytokine signaling pathway T2D 3.91E-09 18 4 12 2
Neuroactive ligand-receptor interaction SCZ 4.01E-09 35 26 9 0
Maturity onset diabetes of the young T2D 1.94E-08 11 0 11 0
Type II diabetes mellitus T2D 1.98E-08 14 1 12 1
PPAR signaling pathway T2D 9.33E-07 15 2 13 0
Calcium signaling pathway SCZ 5.03E-06 23 18 5 0
Visceral Fat Deposits and the Metabolic Syndrome T2D 6.57E-05 5 0 4 1
Type I diabetes mellitus SCZ 7.12E-05 10 8 1 1
Corticosteroids and cardioprotection BOTH 8.96E-05 6 3 2 1
Low-density lipoprotein (LDL) pathway during
atherogenesis
T2D 3.28E-04 4 0 4 0
Insulin signaling pathway T2D 9.20E-04 16 1 15 0
Graft-versus-host disease SCZ 0.001587097 8 6 1 1
IL-10 Anti-inflammatory Signaling Pathway BOTH 0.001671861 4 1 1 2
Basic mechanism of action of PPARa, PPARb(d) and
PPARg and effects on gene expression
T2D 0.001780493 3 0 3 0
Actions of Nitric Oxide in the Heart BOTH 0.002369651 5 2 3 0
Erythropoietin mediated neuroprotection through NF-kB BOTH 0.003426765 4 2 1 1
Allograft rejection SCZ 0.003509 7 5 0 2
g-Secretase mediated ErbB4 Signaling Pathway SCZ 0.005740212 3 3 0 0
Msp/Ron Receptor Signaling Pathway BOTH 0.005740212 3 1 1 1
Autoimmune thyroid disease SCZ 0.020029548 7 6 0 1
Alzheimers disease BOTH 0.020096651 5 1 2 2
Free Radical Induced Apoptosis SCZ 0.020961818 3 2 0 1
Role of PPAR-gamma Coactivators in Obesity and
Thermogenesis
T2D 0.020961818 3 0 3 0
Asthma SCZ 0.023911463 5 3 0 2
AKT Signaling Pathway SCZ 0.024540904 4 3 1 0
Hematopoietic cell lineage BOTH 0.027660527 9 4 4 1
Regulation of PGC-1a T2D 0.029865431 3 1 2 0
Role of ERBB2 in Signal Transduction and Oncology T2D 0.030339933 4 1 3 0
SCZ: schizophrenia; T2D: type 2 diabetes mellitus. The p-value is adjusted by Benjamini-Hochberg method.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 5 of 14
proteins, 9 proteins closely interacted with both multiple
SCZ and T2D susceptibility proteins (with both interacting
partners 5) and were regarded as hub proteins, these
hub proteins include SRC, PRKACA, PRKCA, GRB2,
PTPN11, SMAD3, YWHAZ, PIK3R1 and PLCG1 (Figure
3). Some of these hub proteins are the components of our
identified enriched pathways (Table 2).
To verify whether the function association approach is
reasonable to infer the function relationships of those
proteins to the two diseases, we performed systematic lit-
erature mining to survey whether those candidate genes
are reported in PubMed articles for SCZ and T2D. As a
result, we found that 59 candidate genes have been con-
nected to SCZ [34-38], 77 candidate genes have been
linked to T2D [39-43], while 25 candidate genes [44-49]
have been implicated to both SCZ and T2D with various
studies (Additional file 5). Totally, 161 candidate genes
(~45% of all candidate genes) have been related to either
SCZ or T2D or both diseases with various experimental
approaches, further proving the rationale of function
associationin the application of disease related gene
inference. We proposed that genes encoding those 33
proteins (9 hub proteins and 25 proteins, with one com-
mon protein) could be high-priority candidate genes con-
tributing to pathogenetic association between SCZ and
T2D.
SCZ and T2D molecular network construction
Last, to explore the potential relationships of those iden-
tified genes and two diseases, based on our constructed
GAD2
GAD1
HNF4A
HNF1A
NEUROD1
HHEX
VEGFA
PDE3B
HLA-E
KEGG:05330
TNF
CTLA4
INS
KEGG:05320
HLA-DQA1
IL10RA
BioCarta:163
PRKAA2
IRS2
KEGG:04910
ANXA1
AKT1
BioCarta:41
CHRNA7
BioCarta:141
NOS3
CACNA1C
KEGG:04080
GNAS
CACNA1E
SORBS1
POMC
BioCarta:135
ADIPOR1
KEGG:04930
KEGG:04920
ADIPOR2
NPY
GCK
PDX1
SLC2A2
PRKCZ
INSR
KCNJ11
KEGG:04950
ABCC8
IAPP
NEUROG3
HNF1B
HLA-DQB1
KEGG:04940
BioCarta:213
IRS1
SREBF1
PTPN1
PPP1R3A
FOXO1
HLA-A
IL10
IL1B
HLA-DRB1
KEGG:05310
CSF2RA
KEGG:05332
KEGG:05010
IDE APOE
IL3RA
KEGG:04640
DBI
BioCarta:312
BioCarta:203
PLIN1
KEGG:03320
APOA5
RETN
APOC3
IL6
RXRB
ACSL6
PCK1
PRNP
ADIPOQ
PPARGC1A
UCP1
PPARA
FABP2
LEP
GRIN1
ADRB2
BioCarta:119
CHRM1
BioCarta:232
HIF1A
SOD2
PPP3CC
NFKB1
NOS1
YWHAH
KEGG:04020
LEPR
DRD5
GRIN2A
ADRA1A
ADRB3
HTR4
HTR2C
HTR2A
HTR5A
CCKAR
GRM8
GRIK3
GABBR1
GRIN2B
MC3R
GRM3
GABRB2
GCGR
GRIA4
BioCarta:121
DRD3
CNR1
GABRA5
NRG2
NRG3
DRD4
DRD2
BioCarta:241
ERBB4
MC4R GRM5
AGTR1
DRD1
PARD3
TAAR6
MTNR1B
LDLR
ESR1
PPARD
IL6R
PPARG
BioCarta:16
BioCarta:153
CCL2
CD14
CR2
LPL
BioCarta:243
KEGG:04920: Adipocytokine signaling pathway
KEGG:04080: Neuroactive ligand-receptor interaction
KEGG:04950Maturity onset diabetes of the young
KEGG:04930: Type II diabetes mellitus
KEGG:03320: PPAR signaling pathway
KEGG:04020: Calcium signaling pathway
BioCarta:312: Visceral Fat Deposits and the Metabolic Syndrome
KEGG:04940: Type I diabetes mellitus
BioCarta:141: Corticosteroids and cardioprotection
BioCarta:16: Low-density lipoprotein (LDL) pathway during atherogenesis
KEGG:04910: Insulin signaling pathway
KEGG:05332: Graft-versus-host disease
BioCarta:163: IL-10 Anti-inflammatory Signaling Pathway
BioCarta:241: Basic mechanism of action of PPARa, PPARb(d) and PPARg and
effects on gene expression
BioCarta:213: Actions of Nitric Oxide in the Heart
BioCarta:119: Erythropoietin mediated neuroprotection through NF-kB
KEGG:05330: Allograft rejection
BioCarta:121g-Secretase mediated ErbB4 Signaling Pathway 
BioCarta:203: Msp/Ron Receptor Signaling Pathway
KEGG:05320Autoimmune thyroid disease
KEGG:05010: Alzheimer's disease
BioCarta:135: Free Radical Induced Apoptosis
BioCarta:243: Role of PPAR-gamma Coactivators in Obesity and Thermogenesi
s
KEGG:05310: Asthma
BioCarta:41: AKT Signaling Pathway
KEGG:04640: Hematopoietic cell lineage
BioCarta:232: Regulation of PGC-1a
BioCarta:153: Role of ERBB2 in Signal Transduction and Oncology
Figure 2 Pathway-pathway interaction network. The large node represents pathway, blue for SCZ, red for T2D, light-yellow for both. The
small node represents gene mapped to corresponding pathways. Small nodes in green and purple are genes from SCZ and T2D susceptibility
gene lists, respectively. The edge represents crosstalk between any two pathways. The network is automatically laid out using the Organic layout
algorithm supported by Cytoscape.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 6 of 14
pathway network, protein-protein interaction and litera-
ture survey, we developed a SCZ & T2D molecular net-
work (STMN), in which the relationships between those
susceptibility genes/proteins and the two diseases have
been inferred (Figure 4).
Discussion
As complex diseases, both SCZ and T2D have attracted
more and more attentions in the research communities
for their significant increasing prevalence during past
decades. Clinical studied have reported that the risk of
T2D is increased in schizophrenic patients and T2D is
one of the leading causes of morbidity and mortality in
individuals affected with SCZ-related disorders (i.e.,
SCZ, schizoaffective disorder, and schizophreniform dis-
order) [50,51]. There have been numerous reports of
susceptibility genes or loci to SCZ or T2D, however, few
genes have been confirmed to link to the two diseases
MPZL1
GRIN2B
CXCR4
PTPN11
SLAMF
1
LEPR
DDR1
GRIN1
CTLA4
INSR
PLCG1
PKN2
ALK
AGTR1
CSF2RA
CHRNA7
NOTCH4
HNF4A
TGM2
SPRY2
YWHAH
ADRB3
HNF1A
IL6R
PPARD
PDE3B
TH
LMNA
IRS1
IRS2
SYK
GRIN2A
TPH1 ESR1
SRC
UCP2 YWHAZ
GABBR1
SMAD3
PARD3
UCP3
FOXO1
ANXA1
WWOX
PTPN1
ADRB2
PRKCA
GRB2
EGF
ERBB4
DRD3
PRNP
GRM5
OGG1
GFPT1
NRGN
PPARA
GRIA4
PRKACA
SLC2A2
ABCA1
NFKB1
MC4R
PLIN1
SYN2
CACNA1C
NOS1 PRKCZ
HLA-A DRD4
HMGA2
AKT1
PIK3R1
ANK3
VDR
Figure 3 The sub-network of 9 hub proteins and their interacted susceptibility proteins. Nodes in blue represent SCZ susceptibility
proteins; nodes in light-green represent T2D susceptibility proteins; nodes in yellow represent SCZ and T2D common susceptibility proteins.
Large nodes in red are 9 hub proteins. The size of hub proteins reflects the degree in the network.
Table 2 Six hub proteins that involved in previous enriched pathways.
Previous enriched pathways Adjusted P-Value Hub proteins
Calcium signaling pathway 7.42E-06 PLCG1 PRKACA PRKCA
g-Secretase mediated ErbB4 Signaling Pathway 4.33E-03 PRKCA
Adipocytokine signaling pathway 6.41E-08 PTPN11
Insulin signaling pathway 5.46E-06 GRB2 PIK3R1 PRKACA
AKT Signaling Pathway 4.26E-04 PIK3R1
Type II diabetes mellitus 6.67E-03 PRKACA PIK3R1
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 7 of 14
and the mechanisms for the association remain unclear.
The limited success in detection of genetic factors for
both diseases has indicated that the diseases are not
caused by the dysfunction of a specific molecule or
pathway, most likely both diseases are caused by the
altered function or expression of many genes, which
mayindividuallycontributetoonlyasmallrisk,but
their collective dysfunctional effects interfere with the
function of several biological pathways that eventually
produce the clinical outcome [52]. Therefore, studies
based on network and pathway interaction naturally are
the choice for both of the diseases and their association.
To our knowledge, our study is the first network and
pathway-based systematic analyses for the pathogenetic
association between SCZ and T2D by using susceptibil-
ity genes generated from various researches. For many
Figure 4 The SCZ & T2D molecular network. The potential underlying common molecular mechanism between SCZ and T2D. Nodes in blue,
pink and green background are related to SCZ, T2D and common progression, respectively. Solid and dashed lines represent direct and indirect
regulation; lines with arrow and spot represent activation and inhibition, respectively.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 8 of 14
complex diseases, including SCZ and T2D, there are no
applicable gene signatures in clinical to detect them in
early stages. The new discovered common susceptible
genes related to the pathogenetic association between
SCZ and T2D could be potentially used as candidates to
signify the co-occurrence of SCZ with T2D.
From our enrichment pathway analysis results and the
pathway-pathway interaction network, we observed that
many genes are shared by several pathways, such as
TNF shared by 12 enriched pathways and AKT1 shared
by 4 enriched pathways. Those genes that participate in
several pathways could be the key components for the
pathway crosstalks and the potential risk factors for the
SCZ and T2D association.
As a serine/threonine kinase, AKT is a key regulator of
many signal transduction processes mediated by protein
phosphorylation and a central molecule in regulating mul-
tiple cellular processes such as glucose metabolism, tran-
scription, apoptosis, cell proliferation, angiogenesis, and
cell motility [53]. AKT is activated by phosphoinositide
3-kinase (PI3K), which itself is activated by several
upstream signaling pathways, Neuroactive ligand-receptor
interaction pathway is the major one for the activation of
PI3K. Through PI3K, AKT is regulated by many proteins,
such as insulin receptors, receptor tyrosine kinases, G pro-
tein coupled receptors, cytokine receptors, etc., then con-
trols diverse biological responses such as programmed cell
death, cell proliferation, migration, and metabolic pro-
cesses. Recently, accumulating evidences suggest that
impaired AKT signaling plays a role in the pathogenesis of
SCZ [54]. The potential molecular mechanisms underlying
the role of AKT signaling in SCZ has contributed to the
AKT dysfunction. Activated AKT can phosphorylate a
number of other molecules, one of them is the strong
clinically relevant target, glycogen synthase kinase-3
(GSK3) [53]. GSK3 has been confirmed to play several
roles in glucose metabolism, differentiation and develop-
ment, intracellular trafficking, apoptosis, and regulation of
gene transcription [55]. In the brain, both GSK3 and AKT
have been proposed to modulate synaptic plasticity [56].
AKT1 activation has been reported to be reduced in the
hippocampus and frontal cortex of SCZ patients compared
with healthy controls [54]. Other studies have further pro-
vided the evidence of a reduction of AKT1 mRNA and
protein levels in peripheral blood, prefrontal cortex, and
hippocampus in SCZ patients [57,58]. Moreover, the single
SNP that is associated with reduced expression of AKT1
in peripheral lymphocytes is associated with brain volume
reductions in caudate and right prefrontal cortex [59].
The AKT signaling pathway also plays a pivotal role in
the metabolic functions of insulin in the liver. AKT regu-
lates glycogenesis through the phosphorylation of GSK3,
GSK3 phosphorylates glycogen synthase and converts it
to the less-active (glucose-6-phosphate dependent) form,
thus inhibits glycogen synthesis. In contrast to the phos-
phorylation of AKT for its activation [53], constitutively
activated GSK3 in resting cells requires phosphorylation
by kinases such as AKT to inactivate it [60]. Interestingly,
68% less expression of AKT1 has been detected in the
lymphocytes of SCZ patients compared with healthy con-
trols [54]. Significant reduction of AKT1 expression and
deregulation of AKT1-associated pathways have recently
also been reported in peripheral blood cells of schizo-
phrenia patients [61]. The impaired activation of AKT in
SCZ patients could result in the higher activity of GSK3
in blood, which eventually causes the reduction of glyco-
gen and inhibition of glucose with the increase of blood
glucose levels. In addition, AKT1 has also been associated
with other signaling pathways, such Dopamine pathways,
Wnt signalling pathway and Adipocytokine signaling
pathway. The dysfunction of these signaling pathways
with impaired AKT1 all has significant impact on the
SCZ or T2D, which is consistent with our analysis result.
Taken together, AKT signaling pathway could be one of
the pivotal pathways to bridge the association between
SCZ and T2D, AKT1 gene, together with GSK3 gene in
this pathway, may be responsible for the co-occurrence
of SCZ and T2D.
Leptin (LEP) gene (Figure 2) is involved in the pathways
of Neuroactive ligand-receptor interaction and Adipocyto-
kine signaling in our pathway-pathway interaction net-
work. Leptin is secreted by adipose tissue and signifies the
endocrine function of adipose tissue. An increase in leptin
signals can affect the neuronal targets in the hypothala-
mus. Leptin activates Janus-activating kinase2 (Jak2) and
STAT3, leading to activate alpha-MSH and CART in
POMC/CART neuron, and inhibit NPY and AGRP in
NPY/AGRP neuron. The Neuroactive ligand-receptor
interaction pathway contains G protein-coupled receptors
(GPCRs) of dopamine and serotonin which have been pro-
posed to play an important role in the pathophysiology of
SCZ. Previous studies have suggested that LEP may associ-
ate with SCZ [62,63]. Adipocytokine signaling pathway has
been specifically linked to T2D. As a component for Adi-
pocytokine signalling pathway, LEP is considered to be an
important regulator in the pathophysiology of T2D dis-
eases. In our constructed STMN, we also observed a
crosstalk between leptin and insulin in the hypothalamus.
In addition, leptin can activate AKT1 through the activa-
tion of PI3K, and possibly through JAK2, thus providing a
mechanism for regulation of target genes, the same as in
Insulin signaling pathway. Therefore, the crosstalk
between above two pathways also implies the underlying
pathogenetic association between SCZ and T2D.
Corticosteroids and cardioprotection pathway, a path-
way both for SCZ and T2D, was reported to be asso-
ciated with SCZ [64,65] and T2D [66]. It interlinks to
Calcium signaling pathway and Insulin signaling
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 9 of 14
pathway. Interestingly, the crosstalk between Corticos-
teroids and cardioprotection pathway and Insulin signal-
ing pathway is mediated by AKT according to our
pathway-based network. Previous study also has shown
that Calcium signaling pathway is associated with dopa-
mine-induced cortical neuron apoptosis which is consid-
ered as an important mechanism in SCZ pathogenesis
[67]. Meanwhile, Actions of Nitric Oxide in the Heart,
another pathway for both SCZ and T2D, is a crosstalk
between Calcium signaling pathway and Insulin signal-
ing pathway either. Previous study indicated that Nitric
oxide was involved in pathophysiology of SCZ [68].
IL-10 Anti-inflammatory signaling pathway is an
immune-related pathway. Accumulated evidence from
epidemiological, clinical and animal studies suggests that
immune-related pathway may play a key role in the
development of mental diseases including SCZ and
mood disorders [69,70]. IL-10 Anti-inflammatory Signal-
ing Pathway has been reported previously to be involved
in pathophysiology of SCZ [71] and T2D [72], respec-
tively. Therefore, the above evidence suggests that IL-10
Anti-inflammatory signaling pathway may be involved in
the pathogenetic association between SCZ and T2D. In
another perspective, due to inflammation contributes to
injury or enhances CNS vulnerability, and acute inflam-
mation can also be shifted to a chronic inflammatory
state and adversely affect brain development, therefore,
through efficient anti-inflammatory and reparative pro-
cesses, inflammation may resolve without any harmful
effects on the brain. Alternatively, intervention of TNF-
a, before the progressive loss of beta cell function, may
yield promising results in the treatment of T2D. Since
IL-10 is a cytokine with potent anti-inflammatory prop-
erties, it represses the expression of inflammatory cyto-
kines such as TNF-a, IL-6 and IL-1 by activated
macrophages. The anti-inflammatory actions of IL-10
may be therapeutically useful by intervention of TNF-a,
IL-1 or IL-6 to avoid inflammatory response, then to
decrease the CNS vulnerability, further to reduce the
chance to trigger T2D.
In our inferred new candidate risk factors, 9 proteins
interact with multiple proteins involved in both diseases
with high connectivity, 6 of them are found to be the
components of our enriched pathways (Table 2). Among
them, PRKACA is shared by Type II diabetes mellitus,
Insulin signaling pathway and Calcium signaling path-
ways; PIK3R1 is a common molecule of AKT signaling,
Insulin signaling and Type II diabetes mellitus pathways;
PRKCA is a component for both of Calcium signaling
and g-Secretase mediated ErbB4 signaling pathways
while PLCG1 for Calcium signaling pathway, PTPN11 for
Adipocytokine signaling pathway and GRB2 for Insulin
signaling pathway. All of those proteins may be asso-
ciated with both SCZ and T2D through participating into
related signaling pathways and interacting with other dis-
ease related susceptibility genes, then further enhancing
the linkage between SCZ and T2D.
For the rest of three hub proteins, SRC, SMAD3 and
YWHAZ, they may also play some role in contributing to
pathogenic association between SCZ and T2D. Src is a
tyrosine kinase. In the sub-network, it interacts with 7
and 13 SCZ and T2D related proteins, respectively. Src
has been associated with SCZ, the potential molecular
mechanism is that the NRG1-ErbB4 pathway, which is a
candidate pathway participated in cognitive dysfunction
in SCZ, affects NMDAR hypofunction through modula-
tion of Src activity. In mouse model, NRG1-ErbB4 signal-
ing blocks Src enhancement of NMDAR-mediated
synaptic currents [73]. Although there has no report
about Src implicated with T2D, from the sub-network,
we observed that Src links to multiple T2D related pro-
teins, such as INSR, an insulin receptor, and AKT1.
Given that the Src protein is a tyrosine kinase, which
plays critical roles in the activiation of multiple signaling
pathways [74], we speculate that SRC is a potential candi-
date gene with pleiotropic effects that affects both SCZ
and T2D.
SMAD3 is a member of SMAD protein family that are
signal transducers and transcriptional modulators that
mediate multiple signaling pathways. One of those sig-
naling pathway is the transforming growth factor beta
(TGF-b) pathway [75], TGF-bplays an important role in
regulation of insulin gene transcription and b-cell func-
tion [39], it is also a key mediator in the development of
diabetic complications. TGF-bexerts its biological
effects by activating downstream mediators, called
Smad2 and Smad3. Recent studies have demonstrated
that under disease conditions Smad3 act as signal inte-
grators and interact with other signaling pathways, such
as the MAPK and NF-B pathways [76]. In adult Smad3
null mice, TGF-bsignaling through Smad3 is needed to
maintain the rate of cell division of neuronal precursors
in the adult brain and hence the amount of neurogen-
esis [77]. Another Smad family member - Smad4 has
been proven to be related to SCZ, since forebrain-speci-
fic Smad4 knock-out mice shows typical endophenotype
of schizophrenia [78]. Taken together, these data add
new evidence to support our hypothesis that the Smad3
may link to both SCZ and T2D by interacting with mul-
tiple signaling pathways as a signal integrator.
YWHAZ gene product belongs to the 14-3-3 family of
proteins which mediate signal transduction by binding to
phosphoserine-containing proteins. The encoded protein
interacts with IRS1 protein, and is a negative regulator
for insulin signal transduction, suggesting its role in regu-
lating insulin sensitivity [79]. Previous study has also
indicated that the YWHAZ gene is a potential risk factor
for paranoid SCZ, although the potential mechanism of
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 10 of 14
how this gene affected biological functions in the brain is
unknown [80]. Therefore, our hypothesis tentatively
assumes that the YWHAZ may also be a pleiotropic
gene, which participates in the pathogenetic linkage
between SCZ and T2D diseases.
For the rest of new candidate genes, although the
number of interaction partners for them is various and
less than those hub proteins in the PPI network, 25 of
them, including well known genes, TP53, GSK3 and
RXRA, are still supportedly associated with SCZ and
T2D by text mining. Various data have indicated that
they all have been implicated in both of the diseases
(Additional file 5). For those genes without literature
support, they may also be involved in differential but
intertwined SCZ and T2D pathogenetic processes.
Further experiments need to carry out to verify those
associations.
The new candidate genes are inferred from the PPI,
however, it is worth pointing out that the PPI we used
in the study represents a static relationship between
each protein pair. In real biological processes, such as
pathogenetic conditions or different development stages,
gene expression has spatiotemporal pattern, the same as
protein-protein interaction. Therefore, different impli-
cated genes may participate into SCZ and T2D diseases
in different stages and play different roles in the associa-
tion with the SCZ and T2D. By integrating multiple
dimensional data, it can be expected that network-based
approach, combined with other multiple resources, will
provide great help to decipher the coordination and
functional roles of those implicated genes in complex
diseases. Furthermore, it is well known that many pro-
teins in signaling pathways are drug targets. Our path-
way-based network has revealed that many susceptible
genes linking SCZ and T2D participate into different
signaling pathways and have pleiotropic effects, their
encoded proteins could be good candidates as drug tar-
gets to treat this complex disease, and selectively target-
ing those dysfunctional proteins in different signaling
pathways with synergetic effect could potentially have
better treatment outcome.
There are certain limitations in our study. First, those
prioritized SCZ genes and T2D related genes we used
are all from GWAS. Considering the inherent drawbacks
of GWAS approach with its noise and high false positive
rate, some of the genes may not be truly associated with
both of the diseases, which will certainly affect the path-
way enrichment analysis result and our inference of new
candidate risk genes for the association of SCZ and
T2D. Second, the incomplete pathway annotation sys-
tems for each pathway database could also negatively
contribute to the pathway network construction and the
pathway crosstalk interpretation. Nevertheless, our
results still present novel and promising explanation for
the association between SCZ and T2D, these novel rela-
tionships could offer new insights into these two dis-
easesetiology.
Conclusions
We have successfully built the pathogenetic association
between SCZ and T2D based on their enriched pathway
crosstalk. Through the integration of multiple level ana-
lysis results, including pathway crosstalk, PPI and litera-
ture survey, we revealed some potential molecular
mechanisms and multiple susceptibility genes that could
exert pleiotropic effects shared by two diseases. Totally
364 candidate proteins that directly interacted with both
our SCZ and T2D susceptibility proteins have been
identified, 33 of them have been prioritized as high sig-
nificant genes linking to both of SCZ and T2D.
Although there are certain limitations for our analysis
processes, our preliminary findings can provide an alter-
native direction for other researchers to explore the
linkage between these two diseases.
Currently, many chromosomal intergenic regions and
SNPs on human genome have been associated with dis-
eases. However, no gene has been identified in those
regions or to host those SNPs. It can be anticipated that
with the emergence and significant progress of new
technologies, such as next generation sequence technol-
ogy [81,82], more and more genes and transcribed
regions will be discovered in human genome [83,84] and
those unrealized expression genes in the current inter-
genic regions will be indentified and linked to the dis-
eases. Those will definitely facilitate the investigation of
those complex diseases, and help us to reshape the
potential underlying genetic mechanisms for those com-
plex diseases.
Additional material
Additional file 1: 196 SCZ and 200 T2D susceptibility gene sets.
Additional file 2: Pathway shared genes and their involved
pathways.
Additional file 3: Protein-Protein Interaction Network. This network
consists of 2,104 nodes and 3,155 edges, nodes represent proteins, node
size stands for its degree, edges represent interaction between two
proteins. Nodes in blue are 143 SCZ susceptibility proteins; nodes in
green are 138 T2D susceptibility proteins; nodes in yellow are common
susceptibility proteins; remainder nodes in purple are 1,811 candidate
proteins.
Additional file 4: Sub-network extracted from Additional file 3. This
network consists of 580 nodes and 1,266 edges, node attributes refer to
Additional file 3.
Additional file 5: Literature mining results for the 364 new
candidate genes. 364 candidate genes are in the first column; the
second column hosts the number of interactions from SCZ susceptibility
proteins; the third column lists the PubMed ID for the reported protein
associated with SCZ; the fourth column is the interactions of SCZ. The
corresponding results of T2D are displayed in the following columns.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 11 of 14
Genes in pink background are 25 candidate genes which have been
implicated in both SCZ and T2D with various studies.
Authorscontributions
TS and ZY conceived and designed the study. YL, ZL, MZ performed
analyses. TS, ZY, YL, ZL and YD wrote the manuscript, TS finalized the
manuscript.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
This work was supported by the National 973 Key Basic Research Program
(Grant Nos. 2010CB945401 and 2012CB910400), the National Natural Science
Foundation of China (Grant No. 31171264, 31071162, 31000590 and
81171272) and the Science and Technology Commission of Shanghai
Municipality (11DZ2260300).
This article has been published as part of BMC Medical Genomics Volume 6
Supplement 1, 2013: Proceedings of the 2011 International Conference on
Bioinformatics and Computational Biology (BIOCOMP11). The full contents
of the supplement are available online at http://www.biomedcentral.com/
bmcmedgenomics/supplements/6/S1. Publication of this supplement has
been supported by the International Society of Intelligent Biological
Medicine.
Author details
1
Center for Bioinformatics and Computational Biology, and The Institute of
Biomedical Sciences, School of Life Sciences, East China Normal University,
500 Dongchuan Road, Shanghai 200241, China.
2
Department of Neurology,
Shanghai Changhai Hospital, Secondary Military Medical University, 168
Changhai Road, Shanghai, China.
3
Department of Ophthalmology, West
China Hospital, Sichuan University 37 Guoxuexiang, Chengdu, Sichuan,
610041, China.
4
Rush University Cancer Center, Department of Internal
Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
5
Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong
University School of Medicine, 600 Wan Ping Nan Road, Shanghai 200030,
China.
Published: 23 January 2013
References
1. Peraala J, Suvisaari J, Saarni SI, Kuoppasalmi K, Isometsa E, Pirkola S,
Partonen T, Tuulio-Henriksson A, Hintikka J, Kieseppa T, et al:Lifetime
prevalence of psychotic and bipolar I disorders in a general population.
Arch Gen Psychiat 2007, 64(1):19-28.
2. Ng MY, Levinson DF, Faraone SV, Suarez BK, DeLisi LE, Arinami T, Riley B,
Paunio T, Pulver AE, Irmansyah , et al:Meta-analysis of 32 genome-wide
linkage studies of schizophrenia. Mol Psychiatry 2009, 14(8):774-785.
3. Purcell SM, Wray NR, Stone JL, Visscher PM, ODonovan MC, Sullivan PF,
Sklar P: Common polygenic variation contributes to risk of schizophrenia
and bipolar disorder. Nature 2009, 460(7256):748-752.
4. Shi J, Levinson DF, Duan J, Sanders AR, Zheng Y, Peer I, Dudbridge F,
Holmans PA, Whittemore AS, Mowry BJ, et al:Common variants on
chromosome 6p22.1 are associated with schizophrenia. Nature 2009,
460(7256):753-757.
5. Stefansson H, Ophoff RA, Steinberg S, Andreassen OA, Cichon S, Rujescu D,
Werge T, Pietilainen OP, Mors O, Mortensen PB, et al:Common variants
conferring risk of schizophrenia. Nature 2009, 460(7256):744-747.
6. Coclami T, Cross M: Psychiatric co-morbidity with type 1 and type 2
diabetes mellitus. East Mediterr Health J 2011, 17(10):777-783.
7. Schoepf D, Potluri R, Uppal H, Natalwala A, Narendran P, Heun R: Type-2
diabetes mellitus in schizophrenia: increased prevalence and major risk
factor of excess mortality in a naturalistic 7-year follow-up. Eur Psychiatry
2012, 27(1):33-42.
8. Bresee LC, Majumdar SR, Patten SB, Johnson JA: Prevalence of
cardiovascular risk factors and disease in people with schizophrenia: a
population-based study. Schizophr Res 2010, 117(1):75-82.
9. Chien IC, Chang KC, Lin CH, Chou YJ, Chou P: Prevalence of diabetes in
patients with bipolar disorder in Taiwan: a population-based national
health insurance study. Gen Hosp Psychiatry 2010, 32(6):577-582.
10. Lin PI, Shuldiner AR: Rethinking the genetic basis for comorbidity of
schizophrenia and type 2 diabetes. Schizophr Res 2010, 123:(2-3):234-243.
11. Barabasi AL, Oltvai ZN: Network biology: understanding the cells
functional organization. Nat Rev Genet 2004, 5(2):101-U115.
12. Gough SC, ODonovan MC: Clustering of metabolic comorbidity in
schizophrenia: a genetic contribution? J Psychopharmacol 2005, 19(6
Suppl):47-55.
13. Struewing I, Boyechko T, Barnett C, Beildeck M, Byers SW, Mao CD: The
balance of TCF7L2 variants with differential activities in Wnt-signaling is
regulated by lithium in a GSK3beta-independent manner. Biochem
Biophys Res Commun 2010, 399(2):245-250.
14. Freyberg Z, Ferrando SJ, Javitch JA: Roles of the Akt/GSK-3 and Wnt
signaling pathways in schizophrenia and antipsychotic drug action. Am J
Psychiatry 2010, 167(4):388-396.
15. Backman M, Machon O, Mygland L, van den Bout CJ, Zhong W, Taketo MM,
Krauss S: Effects of canonical Wnt signaling on dorso-ventral
specification of the mouse telencephalon. Dev Biol 2005, 279(1):155-168.
16. Brinkmeier ML, Potok MA, Davis SW, Camper SA: TCF4 deficiency expands
ventral diencephalon signaling and increases induction of pituitary
progenitors. Dev Biol 2007, 311(2):396-407.
17. Alkelai A, Greenbaum L, Lupoli S, Kohn Y, Sarner-Kanyas K, Ben-Asher E,
Lancet D, Macciardi F, Lerer B: Association of the type 2 diabetes mellitus
susceptibility gene, TCF7L2, with schizophrenia in an Arab-Israeli family
sample. PLoS One 2012, 7(1):e29228.
18. Genetic association database. [http://geneticassociationdb.nih.gov/].
19. Becker KG, Barnes KC, Bright TJ, Wang SA: The genetic association
database. Nat Genet 2004, 36(5):431-432.
20. A catalog of published genome-wide association studies. [http://www.
genome.gov/gwastudies/].
21. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS,
Manolio TA: Potential etiologic and functional implications of genome-
wide association loci for human diseases and traits. Proc Natl Acad Sci
USA 2009, 106(23):9362-9367.
22. Type 2 diabetes genetic association database. [http://t2db.khu.ac.kr:8080/].
23. Lim JE, Hong KW, Jin HS, Kim YS, Park HK, Oh B: Type 2 diabetes genetic
association database manually curated for the study design and odds
ratio. BMC Med Inform Decis Mak 2010, 10:76.
24. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A,
Fridman WH, Pages F, Trajanoski Z, Galon J: ClueGO: a Cytoscape plug-in
to decipher functionally grouped gene ontology and pathway
annotation networks. Bioinformatics 2009, 25(8):1091-1093.
25. Kyoto encyclopedia of genes and genomes. [http://www.genome.jp/
kegg/pathway.html].
26. Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M: KEGG: Kyoto
Encyclopedia of Genes and Genomes. Nucleic Acids Res 1999, 27(1):29-34.
27. BioCarta. [http://www.biocarta.com/].
28. Nishimura D: BioCarta. Biotech Software & Internet Report 2001, 2(3):117-120.
29. Benjamini Y, Hochberg Y: Controlling the false discovery rate - a practical
and powerful approach to multiple testing. J Roy Stat Soc B Met 1995,
57(1):289-300.
30. Huang DW, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J,
Stephens R, Baseler MW, Lane HC, Lempicki RA: The DAVID Gene
Functional Classification Tool: a novel biological module-centric
algorithm to functionally analyze large gene lists. Genome Biol 2007, 8(9).
31. Human Protein Reference Database. [http://www.hprd.org/index_html].
32. Prasad TS, Kandasamy K, Pandey A: Human protein reference database
and human Proteinpedia as discovery tools for systems biology. Methods
Mol Biol 2009, 577:67-79.
33. Lee HC: Cyclic ADP-ribose and NAADP: fraternal twin messengers for
calcium signaling. Sci China Life Sci 2011, 54(8):699-711.
34. Pitcher GM, Kalia LV, Ng D, Goodfellow NM, Yee KT, Lambe EK, Salter MW:
Schizophrenia susceptibility pathway neuregulin 1-ErbB4 suppresses Src
upregulation of NMDA receptors. Nat Med 2011, 17(4):470-478.
35. Carroll LS, Williams NM, Moskvina V, Russell E, Norton N, Williams HJ,
Peirce T, Georgieva L, Dwyer S, Grozeva D, et al:Evidence for rare and
common genetic risk variants for schizophrenia at protein kinase C,
alpha. Mol Psychiatr 2010, 15(11):1101-1111.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 12 of 14
36. Sun JC, Wan CL, Jia PL, Fanous AH, Kendler KS, Riley BP, Zhao ZM:
Application of systems biology approach identifies and validates GRB2
as a risk gene for schizophrenia in the Irish Case Control Study of
Schizophrenia (ICCSS) sample. Schizophrenia Research 2011, 125(2-
3):201-208.
37. Carter CJ: Schizophrenia susceptibility genes directly implicated in the
life cycles of pathogens: cytomegalovirus, influenza, herpes simplex,
rubella, and Toxoplasma gondii. Schizophr Bull 2009, 35(6):1163-1182.
38. Hashimoto R, Ohi K, Yasuda Y, Fukumoto M, Yamamori H, Takahashi H,
Iwase M, Okochi T, Kazui H, Saitoh O, et al:Variants of the RELA gene are
associated with schizophrenia and their startle responses.
Neuropsychopharmacol 2011, 36(9):1921-1931.
39. Lin HM, Lee JH, Yadav H, Kamaraju AK, Liu E, Zhigang D, Vieira A, Kim SJ,
Collins H, Matschinsky F, et al:Transforming growth factor-beta/Smad3
signaling regulates insulin gene transcription and pancreatic islet beta-
cell function. J Biol Chem 2009, 284(18):12246-12257.
40. Malodobra M, Pilecka A, Gworys B, Adamiec R: Single nucleotide
polymorphisms within functional regions of genes implicated in insulin
action and association with the insulin resistant phenotype. Mol Cell
Biochem 2011, 349(1-2):187-193.
41. Kinoshita T, Doi K, Sugiyama H, Kinoshita S, Wada M, Naruto S,
Tomonaga A: Knowledge-based identification of the ERK2/STAT3 signal
pathway as a therapeutic target for type 2 diabetes and drug discovery.
Chem Biol Drug Des 2011, 78(3):471-476.
42. Costes S, Vandewalle B, Tourrel-Cuzin C, Broca C, Linck N, Bertrand G, Kerr-
Conte J, Portha B, Pattou F, Bockaert J, et al:Degradation of cAMP-
responsive element-binding protein by the ubiquitin-proteasome
pathway contributes to glucotoxicity in beta-cells and human pancreatic
islets. Diabetes 2009, 58(5):1105-1115.
43. Shen N, Yu X, Pan FY, Gao X, Xue B, Li CJ: An early response transcription
factor, Egr-1, enhances insulin resistance in type 2 diabetes with chronic
hyperinsulinism. J Biol Chem 2011, 286(16):14508-14515.
44. Jia Y, Yu X, Zhang B, Yuan Y, Xu Q, Shen Y: An association study between
polymorphisms in three genes of 14-3-3 (tyrosine 3-monooxygenase/
tryptophan 5-monooxygenase activation protein) family and paranoid
schizophrenia in northern Chinese population. Eur Psychiatry 2004,
19(6):377-379.
45. Molina V, Papiol S, Sanz J, Rosa A, Arias B, Fatjo-Vilas M, Calama J,
Hernandez AI, Becker J, Fananas L: Convergent evidence of the
contribution of TP53 genetic variation (Pro72Arg) to metabolic activity
and white matter volume in the frontal lobe in schizophrenia patients.
Neuroimage 2011, 56(1):45-51.
46. Zhou YD, Zhang EM, Berggreen C, Jing XJ, Osmark P, Lang S, Cilio CM,
Goransson O, Groop L, Renstrom E, et al:Survival of pancreatic beta cells
is partly controlled by a TCF7L2-p53-p53INP1-dependent pathway.
Human Molecular Genetics 2012, 21(1):196-207.
47. Singh RK, Shi J, Zemaitaitis BW, Muma NA: Olanzapine increases RGS7
protein expression via stimulation of the Janus tyrosine kinase-signal
transducer and activator of transcription signaling cascade. J Pharmacol
Exp Ther 2007, 322(1):133-140.
48. Souza RP, Romano-Silva MA, Lieberman JA, Meltzer HY, Wong AH,
Kennedy JL: Association study of GSK3 gene polymorphisms with
schizophrenia and clozapine response. Psychopharmacology (Berl) 2008,
200(2):177-186.
49. Pilot-Storck F, Chopin E, Rual JF, Baudot A, Dobrokhotov P, Robinson-
Rechavi M, Brun C, Cusick ME, Hill DE, Schaeffer L, et al:Interactome
mapping of the phosphatidylinositol 3-kinase-mammalian target of
rapamycin pathway identifies deformed epidermal autoregulatory
factor-1 as a new glycogen synthase kinase-3 interactor. Mol Cell
Proteomics 2010, 9(7):1578-1593.
50. Newcomer JW: Metabolic syndrome and mental illness. Am J Manag Care
2007, 13(7 Suppl):S170-177.
51. Auquier P, Lancon C, Rouillon F, Lader M: Mortality in schizophrenia.
Pharmacoepidemiol Drug Saf 2007, 16(12):1308-1312.
52. Williams-Skipp C, Raman T, Valuck RJ, Watkins H, Palmer BE, Scheinman RI:
Unmasking of a protective tumor necrosis factor receptor I-mediated
signal in the collagen-induced arthritis model. Arthritis and Rheumatism
2009, 60(2):408-418.
53. Brazil DP, Yang ZZ, Hemmings BA: Advances in protein kinase B
signalling: AKTion on multiple fronts. Trends Biochem Sci 2004,
29(5):233-242.
54. Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA: Convergent
evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat
Genet 2004, 36(2):131-137.
55. Kockeritz L, Doble B, Patel S, Woodgett JR: Glycogen synthase kinase-3an
overview of an over-achieving protein kinase. Curr Drug Targets 2006,
7(11):1377-1388.
56. Peineau S, Bradley C, Taghibiglou C, Doherty A, Bortolotto ZA, Wang YT,
Collingridge GL: The role of GSK-3 in synaptic plasticity. Br J Pharmacol
2008, 153(Suppl 1):S428-437.
57. Zhao Z, Ksiezak-Reding H, Riggio S, Haroutunian V, Pasinetti GM: Insulin
receptor deficits in schizophrenia and in cellular and animal models of
insulin receptor dysfunction. Schizophr Res 2006, 84(1):1-14.
58. Thiselton DL, Vladimirov VI, Kuo PH, McClay J, Wormley B, Fanous A,
ONeill FA, Walsh D, Van den Oord EJCG, Kendler KS, et al:AKT1 is
associated with schizophrenia across multiple symptom dimensions in
the Irish study of high density schizophrenia families. Biol Psychiat 2008,
63(5):449-457.
59. Tan HY, Nicodemus KK, Chen Q, Li Z, Brooke JK, Honea R, Kolachana BS,
Straub RE, Meyer-Lindenberg A, Sei Y, et al:Genetic variation in AKT1 is
linked to dopamine-associated prefrontal cortical structure and function
in humans. J Clin Invest 2008, 118(6):2200-2208.
60. Doble BW, Woodgett JR: GSK-3: tricks of the trade for a multi-tasking
kinase. J Cell Sci 2003, 116(Pt 7):1175-1186.
61. van Beveren NJ, Buitendijk GH, Swagemakers S, Krab LC, Roder C, de
Haan L, van der Spek P, Elgersma Y: Marked reduction of AKT1 expression
and deregulation of AKT1-associated pathways in peripheral blood
mononuclear cells of schizophrenia patients. PLoS One 2012, 7(2):e32618.
62. Kraus T, Haack M, Schuld A, Hinze-Selch D, Pollmacher T: Low leptin levels
but normal body mass indices in patients with depression or
schizophrenia. Neuroendocrinology 2001, 73(4):243-247.
63. Melson AK, Selke G, Schweiger J, Farber NB, Newcomer JW: Relationship
between plasma leptin and memory performance in humans with and
without schizophrenia. Schizophrenia Research 2003, 60(1):147-148.
64. Ellingrod VL, Taylor SF, Brook RD, Evans SJ, Zollner SK, Grove TB,
Gardner KM, Bly MJ, Pop-Busui R, Dalack G: Dietary, lifestyle and
pharmacogenetic factors associated with arteriole endothelial-
dependent vasodilatation in schizophrenia patients treated with atypical
antipsychotics (AAPs). Schizophrenia Research 2011, 130(1-3):20-26.
65. Bradley AJ, Dinan TG: A systematic review of hypothalamic-pituitary-
adrenal axis function in schizophrenia: implications for mortality. J
Psychopharmacol 2010, 24(4 Suppl):91-118.
66. Thiemermann C: Corticosteroids and cardioprotection. Nat Med 2002,
8(5):453-455.
67. Zhang L, Yang H, Zhao H, Zhao C: Calcium-related signaling pathways
contributed to dopamine-induced cortical neuron apoptosis. Neurochem
Int 2011, 58(3):281-294.
68. Das UN: Essential Fatty acids - a review. Curr Pharm Biotechnol 2006,
7(6):467-482.
69. Leonard BE, Schwarz M, Myint AM: The metabolic syndrome in
schizophrenia: is inflammation a contributing cause? J Psychopharmacol
2012, 26(5 Suppl):33-41.
70. Li Z, Qi D, Chen J, Zhang C, Yi Z, Yuan C, Wang Z, Hong W, Yu S, Cui D,
et al:Venlafaxine inhibits the upregulation of plasma tumor necrosis
factor-alpha (TNF-alpha) in the Chinese patients with major depressive
disorder: A prospective longitudinal study. Psychoneuroendocrinology
2012.
71. Meyer U, Murray PJ, Urwyler A, Yee BK, Schedlowski M, Feldon J: Adult
behavioral and pharmacological dysfunctions following disruption of the
fetal brain balance between pro-inflammatory and IL-10-mediated anti-
inflammatory signaling. Mol Psychiatr 2008, 13(2):208-221.
72. Babu PVA, Si HW, Fu Z, Zhen W, Liu DM: Genistein Prevents
Hyperglycemia-Induced Monocyte Adhesion to Human Aortic
Endothelial Cells through Preservation of the cAMP Signaling Pathway
and Ameliorates Vascular Inflammation in Obese Diabetic Mice. J Nutr
2012, 142(4):724-730.
73. Pitcher GM, Kalia LV, Ng D, Goodfellow NM, Yee KT, Lambe EK, Salter MW:
Schizophrenia susceptibility pathway neuregulin 1-ErbB4 suppresses Src
upregulation of NMDA receptors. Nat Med 2011, 17(4):470-U111.
74. Huang H, Li L, Wu C, Schibli D, Colwill K, Ma S, Li C, Roy P, Ho K,
Songyang Z, et al:Defining the specificity space of the human SRC
homology 2 domain. Mol Cell Proteomics 2008, 7(4):768-784.
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 13 of 14
75. Dennler S, Itoh S, Vivien D, ten Dijke P, Huet S, Gauthier JM: Direct binding
of Smad3 and Smad4 to critical TGF beta-inducible elements in the
promoter of human plasminogen activator inhibitor-type 1 gene. EMBO J
1998, 17(11):3091-3100.
76. Lan HY, Chung AC: Transforming growth factor-beta and Smads. Contrib
Nephrol 2011, 170:75-82.
77. Wang Y, Symes AJ: Smad3 deficiency reduces neurogenesis in adult
mice. J Mol Neurosci 2010, 41(3):383-396.
78. Sun M, Gewirtz JC, Bofenkamp L, Wickham RJ, Ge H, OConnor MB:
Canonical TGF-beta signaling is required for the balance of excitatory/
inhibitory transmission within the hippocampus and prepulse inhibition
of acoustic startle. Journal of Neuroscience 2010, 30(17):6025-6035.
79. Moeschel K, Beck A, Weigert C, Lammers R, Kalbacher H, Voelter W,
Schleicher ED, Haring HU, Lehmann R: Protein kinase C-zeta-induced
phosphorylation of Ser318 in insulin receptor substrate-1 (IRS-1)
attenuates the interaction with the insulin receptor and the tyrosine
phosphorylation of IRS-1. J Biol Chem 2004, 279(24):25157-25163.
80. Jia YB, Yu X, Zhang BY, Yuan YB, Xu Q, Shen YC, Shen Y: An association
study between polymorphisms in three genes of 14-3-3 (tyrosine 3-
monooxygenase/tryptophan 5-monooxygenase activation protein)
family and paranoid schizophrenia in northern Chinese population. Eur
Psychiat 2004, 19(6):377-379.
81. Zhou X, Ren L, Li Y, Zhang M, Yu Y, Yu J: The next-generation sequencing
technology: a technology review and future perspective. Sci China Life Sci
2010, 53(1):44-57.
82. Wu J, Xiao J, Zhang R, Yu J: DNA sequencing leads to genomics progress
in China. Sci China Life Sci 2011, 54(3):290-292.
83. Chen G, Yin K, Wang C, Shi T: De novo transcriptome assembly of RNA-
Seq reads with different strategies. Sci China Life Sci 2011,
54(12):1129-1133.
84. Chen G, Wang C, Shi T: Overview of available methods for diverse RNA-
Seq data analyses. Sci China Life Sci 2011, 54(12):1121-1128.
doi:10.1186/1755-8794-6-S1-S17
Cite this article as: Liu et al.: Exploring the pathogenetic association
between schizophrenia and type 2 diabetes mellitus diseases based on
pathway analysis. BMC Medical Genomics 2013 6(Suppl 1):S17.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Liu et al.BMC Medical Genomics 2013, 6(Suppl 1):S17
http://www.biomedcentral.com/1755-8794/6/S1/S17
Page 14 of 14

Supplementary resources (5)

... For instance, genetic overlap analyses were performed on genomewide summary statistics from two large-scale GWAS of schizophrenia and type II diabetes to discover the shared association between schizophrenia and type II diabetes [6]. Common differential expressed genes (DEGs) were analyzed as the comorbid genes across schizophrenia and type II diabetes and further identified the enriched gene ontology as well as transcription factors with these DEGs [9,10]. Similarly, a set of susceptibility genes for schizophrenia and type II diabetes were retrieved respectively to identify the significant pathways crossing among two syndromes [10]. ...
... Common differential expressed genes (DEGs) were analyzed as the comorbid genes across schizophrenia and type II diabetes and further identified the enriched gene ontology as well as transcription factors with these DEGs [9,10]. Similarly, a set of susceptibility genes for schizophrenia and type II diabetes were retrieved respectively to identify the significant pathways crossing among two syndromes [10]. Mendelian randomization analysis was performed to establish the causal linkage of genetic variants associated with type II diabetes with the risk of schizophrenia using an inverse-variance weighted meta-analysis [11]. ...
Article
Full-text available
The clinical burden of mental illness, in particular schizophrenia and bipolar disorder, are driven by frequent chronic courses and increased mortality, as well as the risk for comorbid conditions such as cardiovascular disease and type 2 diabetes. Evidence suggests an overlap of molecular pathways between psychotic disorders and somatic comorbidities. In this study, we developed a computational framework to perform comorbidity modeling via an improved integrative unsupervised machine learning approach based on multi-rank non-negative matrix factorization (mrNMF). Using this procedure, we extracted molecular signatures potentially explaining shared comorbidity mechanisms. For this, 27 case–control microarray transcriptomic datasets across multiple tissues were collected, covering three main categories of conditions including psychotic disorders, cardiovascular diseases and type II diabetes. We addressed the limitation of normal NMF for parameter selection by introducing multi-rank ensembled NMF to identify signatures under various hierarchical levels simultaneously. Analysis of comorbidity signature pairs was performed to identify several potential mechanisms involving activation of inflammatory response auxiliarily interconnecting angiogenesis, oxidative response and GABAergic neuro-action. Overall, we proposed a general cross-cohorts computing workflow for investigating the comorbid pattern across multiple symptoms, applied it to the real-data comorbidity study on schizophrenia, and further discussed the potential for future application of the approach.
... Several genetic studies support causal biological associations between PSDs and dysglycemia. For instance, susceptibility genes for schizophrenia and T2D are found in overlapping biological networks, suggesting common underlying mechanisms [12]. Additionally, a prospective study found that genetic predisposition to T2D is associated with an increased risk of psychosis in young adulthood [13]. ...
... We found 221 common DEGs, suggesting that PSDs and early dysglycemia indeed share common gene expression signatures. These findings extend previous work examining the genetic links between PSDs and T2D [12][13][14] by demonstrating that an overlap exists at the gene expression level. The overlapping gene expression signatures between AP-naive FEP and early dysglycemia potentially represent gene expression changes endogenous to PSDs that are responsible for producing dysglycemia independent of AP treatment. ...
Article
Full-text available
Antipsychotic (AP)-naive first-episode psychosis (FEP) patients display early dysglycemia, including insulin resistance and prediabetes. Metabolic dysregulation may therefore be intrinsic to psychosis spectrum disorders (PSDs), independent of the metabolic effects of APs. However, the potential biological pathways that overlap between PSDs and dysglycemic states remain to be identified. Using meta-analytic approaches of transcriptomic datasets, we investigated whether AP-naive FEP patients share overlapping gene expression signatures with non-psychiatrically ill early dysglycemia individuals. We meta-analyzed peripheral transcriptomic datasets of AP-naive FEP patients and non-psychiatrically ill early dysglycemia subjects to identify common gene expression signatures. Common signatures underwent pathway enrichment analysis and were then used to identify potential new pharmacological compounds via Integrative Library of Integrated Network-Based Cellular Signatures (iLINCS). Our search results yielded 5 AP-naive FEP studies and 4 early dysglycemia studies which met inclusion criteria. We discovered that AP-naive FEP and non-psychiatrically ill subjects exhibiting early dysglycemia shared 221 common signatures, which were enriched for pathways related to endoplasmic reticulum stress and abnormal brain energetics. Nine FDA-approved drugs were identified as potential drug treatments, of which the antidiabetic metformin, the first-line treatment for type 2 diabetes, has evidence to attenuate metabolic dysfunction in PSDs. Taken together, our findings support shared gene expression changes and biological pathways associating PSDs with dysglycemic disorders. These data suggest that the pathobiology of PSDs overlaps and potentially contributes to dysglycemia. Finally, we find that metformin may be a potential treatment for early metabolic dysfunction intrinsic to PSDs.
... A recent study reported alterations of plasma metabolic profiles in medication-naïve patients with clinical high-risk for psychosis, a prodromal stage of clinical psychosis, which could discriminate them from healthy controls (Li et al., 2021). These alterations are thought to be intrinsic to the disorder, i.e., molecular pathways underlying both clinical psychosis and metabolic dysregulation (Liu et al., 2013). One such factor may be an altered immune status as there is robust evidence of immune dysregulation in antipsychotic-naïve first-episode clinical psychosis patients, including elevated peripheral levels of adipocytokines interleukin 6 and tumor necrosis factor alpha (Çakici et al., 2020). ...
Article
Background: Metabolic alterations are often found in patients with clinical psychosis early in the course of the disorder. Psychotic-like experiences are observed in the general population, but it is unclear whether these are associated with markers of metabolism. Methods: A population-based cohort of 1890 individuals (mean age 58.0 years; 56.3% women) was included. Metabolic parameters were measured by body-mass index (BMI), concentrations of low-density and high-density lipoprotein cholesterol (LDL-C and HDL-C), total cholesterol, triglycerides, and fasting glucose and insulin in blood. Frequency and distress ratings of psychotic-like experiences from the positive symptom dimension of the Community Assessment of Psychic Experience questionnaire were assessed. Cross-sectional associations were analysed using linear regression analyses. Results: Higher BMI was associated with higher frequency of psychotic-like experiences (adjusted mean difference: 0.04, 95% CI 0.02-0.06) and more distress (adjusted mean difference: 0.02, 95% CI 0.01-0.03). Lower LDL-C was associated with more psychotic-like experiences (adjusted mean difference: − 0.23, 95% CI − 0.40 to − 0.06). When restricting the sample to those not using lipid-lowering medication, the results of BMI and LDL-C remained and an association between lower HDL-C and higher frequency of psychotic-like experiences was found (adjusted mean difference: − 0.37, 95% CI − 0.69 to − 0.05). We observed no significant associations between cholesterol, triglycerides, glucose, insulin or homeostatic model assessment and psychotic-like experiences. Conclusions: In a population-based sample of middle-aged and elderly individuals, higher BMI and lower LDL-C were associated with psychotic-like experiences. This suggests that metabolic markers are associated with psychotic-like experiences across the vulnerability spectrum.
... Subjects with schizophrenia have an inherent genetic risk for insulin resistance [21][22][23][24], which has been substantiated by genome wide association studies [25]. Interestingly, insulin resistance and hyperinsulinemia in schizophrenia subjects have been intriguing and consistent observations for nearly 100 years. ...
Article
A substantial and diverse body of literature suggests that the pathophysiology of schizophrenia is related to deficits of bioenergetic function. While antipsychotics are an effective therapy for the management of positive psychotic symptoms, they are not efficacious for the complete schizophrenia symptom profile, such as the negative and cognitive symptoms. In this review, we discuss the relationship between dysfunction of various metabolic pathways across different brain regions in relation to schizophrenia. We contend that several bioenergetic subprocesses are affected across the brain and such deficits are a core feature of the illness. We provide an overview of central perturbations of insulin signaling, glycolysis, pentose-phosphate pathway, tricarboxylic acid cycle, and oxidative phosphorylation in schizophrenia. Importantly, we discuss pharmacologic and nonpharmacologic interventions that target these pathways and how such interventions may be exploited to improve the symptoms of schizophrenia.
... 61 The PI3K/Akt pathway has roles in insulin sensitivity, neuronal development, dopamine regulation, and the immune system, 62 and has been implicated as a putative mechanism linking schizophrenia and T2D. 63 Fourth, the rs17514846 variant lies in an intron of FURIN, which encodes a protease that processes latent precursor proteins into their biologically active products. FURIN is expressed in neuroendocrine, liver, gut, and brain tissues. ...
Article
Full-text available
Background Schizophrenia commonly co-occurs with cardiometabolic and inflammation-related traits. It is unclear to what extent the comorbidity could be explained by shared genetic aetiology. Methods We used GWAS data to estimate shared genetic aetiology between schizophrenia, cardiometabolic, and inflammation-related traits: fasting insulin (FI), fasting glucose, glycated haemoglobin, glucose tolerance, type 2 diabetes (T2D), lipids, body mass index (BMI), coronary artery disease (CAD), and C-reactive protein (CRP). We examined genome-wide correlation using linkage disequilibrium score regression (LDSC); stratified by minor-allele frequency using genetic covariance analyzer (GNOVA); then refined to locus-level using heritability estimation from summary statistics (ρ-HESS). Regions with local correlation were used in hypothesis prioritization multi-trait colocalization to examine for colocalisation, implying common genetic aetiology. Results We found evidence for weak genome-wide negative correlation of schizophrenia with T2D (rg = −0.07; 95% C.I., −0.03,0.12; P = .002) and BMI (rg = −0.09; 95% C.I., −0.06, −0.12; P = 1.83 × 10−5). We found a trend of evidence for positive genetic correlation between schizophrenia and cardiometabolic traits confined to lower-frequency variants. This was underpinned by 85 regions of locus-level correlation with evidence of opposing mechanisms. Ten loci showed strong evidence of colocalization. Four of those (rs6265 (BDNF); rs8192675 (SLC2A2); rs3800229 (FOXO3); rs17514846 (FURIN)) are implicated in brain-derived neurotrophic factor (BDNF)-related pathways. Conclusions LDSC may lead to downwardly-biased genetic correlation estimates between schizophrenia, cardiometabolic, and inflammation-related traits. Common genetic aetiology for these traits could be confined to lower-frequency common variants and involve opposing mechanisms. Genes related to BDNF and glucose transport amongst others may partly explain the comorbidity between schizophrenia and cardiometabolic disorders.
Article
Full-text available
Background Insulin‐degrading enzyme (IDE) is an important gene in studies of the pathophysiology of type 2 diabetes mellitus (T2DM). Recent studies have suggested a possible link between type 2 diabetes mellitus (T2DM) and the pathophysiology of schizophrenia (SZ). At the same time, significant changes in insulin‐degrading enzyme (IDE) gene expression have been found in the brains of people with schizophrenia. These findings highlight the need to further investigate the role of IDE in schizophrenia pathogenesis. Methods We enrolled 733 participants from the Czech Republic, including 383 patients with schizophrenia and 350 healthy controls. Our study focused on the single nucleotide polymorphism (SNP) rs2421943 in the IDE gene, which has previously been associated with the pathogenesis of Alzheimer's disease. The SNP was analyzed using the PCR‐RFLP method. Results The G allele of the rs2421943 polymorphism was found to significantly increase the risk of developing SZ (p < 0.01) when a gender‐based analysis showed that both AG and GG genotypes were associated with a more than 1.55 times increased risk of SZ in females (p < 0.03) but not in males. Besides, we identified a potential binding site at the G allele locus for has‐miR‐7110‐5p, providing a potential mechanism for the observed association. Conclusion Our results confirm the role of the IDE gene in schizophrenia pathogenesis and suggest that future research should investigate the relationship between miRNA and estrogen influence on IDE expression in schizophrenia pathogenesis.
Article
Type 2 diabetes (T2D) is a persistent medical condition characterized by insulin resistance and impaired insulin utilization. T2D is primarily observed in men and is strongly linked to obesity, which is considered a significant contributing factor to its onset. The underlying pathogenesis mechanism of T2D remains uncertain concerning potential sex differences. This study aimed to examine the differentially expressed genes (DEGs) and signaling pathways between males and females with T2D.For analysis of differentially expressed genes in this study, GSE86473 dataset was obtained from the NCBI's Gene Expression Omnibus (GEO). Utilizing Enrichr, we performed pathway enrichment analysis and gene ontology. Protein-protein interactions and hub genes are extracted using Cytoscape. Network-Analyst was employed to investigate transcription factors and microRNAs, protein-drug interactions, and gene-disease relationships.We identified 51 DEGs in men and 237 DEGs in women. We determined the leading 20 pathways and 15 GO terms for each gender. In male subjects, the following 15 hub genes were discovered: LDHA, FLT1, IGFBP3, LILRB4, CSF2, IL1A, HLA-C, UBD, C1QB, HLA-DMA, HLA-DPB1, CFB, HLA-DRB1, HLA-A, and CXCL8. The following genes were identified for female subjects: CPA1, CPB1, CEL, PNLIP, CPA2, CELA3A, SYCN, PRSS3P2, CTRB1, AMY2A, CTRC, PNLIPRP1, PRSS1, HLA-DRB1, and HLA-DQB1. We determined six TF genes shared by both males and females: ATF1, IRF1, MAZ, SSRP1, TFDP1, and MXD3. Mir-124-3p, mir-107, mir-16-5p, mir-23b-3p, mir-27a-3p, mir-103a-3p, mir-155-5p, and mir-1-3p were confirmed as microRNAs shared by males and females in our study. We analyzed protein-drug interactions to investigate the potential effects of a particular drug on the activity of target proteins in the body.The results of these analyses provided insights into the molecular mechanisms underlying the diseases under study and identified potential drug targets for further investigation. The hub genes identified in this study may have varying roles in the disease progression mechanisms of T2D between males and females, and they could serve as potential targets for treatment or prognostic markers for both sexes.
Article
During the last decades the attempt to enlighten the pathobiological substrate of psychosis, from merely focusing on neurotransmitters, has expanded into new areas like the immune and redox systems. Indeed, the inflammatory hypothesis concerning psychosis etiopathology has exponentially grown with findings reflecting dysfunction/aberration of the immune/redox systems' effector components namely cytokines, chemokines, CRP, complement system, antibodies, pro-/anti-oxidants, oxidative stress byproducts just to name a few. Yet, we still lie far from comprehending the underlying cellular mechanisms, their causality directions, and the moderating/mediating parameters affecting these systems; let alone the inter-systemic (between immune and redox) interactions. Findings from preclinical studies on the stress field have provided evidence indicative of multifaceted interactions among the immune and redox components so tightly intertwined as a Gordian knot. Interestingly the literature concerning the interactions between these same systems in the context of psychosis appears minimal (if not absent) and ambiguous. This review attempts to draw a frame of the immune-redox systems' interactions starting from basic research on the stress field and expanding on clinical studies with cohorts with psychosis, hoping to instigate new avenues of research.
Article
The evidence supporting the involvement of a number of systems in the neurobiological etiopathology of psychosis has recently grown exponentially. Indeed, the focus of research has changed from measuring solely neurotransmitters to estimating parameters from fields like immunity, stress/endocrine, redox, and metabolism. Yet, little is known regarding the exact role of each one of these fields on the formation of not only the brain neuropathological substrate in psychosis but also the associated general systemic pathology, in terms of causality directions. Research has shown deviations in the levels and/or function of basic effector molecules of the aforementioned fields namely cytokines, pro-/anti- oxidants, glucocorticoids, catecholamines, glucose, and lipids metabolites as well as kynurenines, in psychosis. Yet the evidence regarding their impact on neurotransmitters is minimal and the findings concerning these systems' interactions in the psychotic context are even more dispersed. The present review aims to draw holistically the frame of the hitherto known "players" in the field of psychosis' cellular pathobiology, with a particular focus on their in-between interactions.
Article
Full-text available
In search of interventions targeting brain dysfunction and underlying cognitive impairment in schizophrenia, we look at the brain and beyond to the potential role of dysfunctional systemic metabolism on neural network instability and insulin resistance in serious mental illness. We note that disrupted insulin and cerebral glucose metabolism are seen even in medication-naïve first-episode schizophrenia, suggesting that people with schizophrenia are at risk for Type 2 diabetes and cardiovascular disease, resulting in a shortened life span. Although glucose is the brain's default fuel, ketones are a more efficient fuel for the brain. We highlight evidence that a ketogenic diet can improve both the metabolic and neural stability profiles. Specifically, a ketogenic diet improves mitochondrial metabolism, neurotransmitter function, oxidative stress/inflammation, while also increasing neural network stability and cognitive function. To reverse the neurodegenerative process, increasing the brain's access to ketone bodies may be needed. We describe evidence that metabolic, neuroprotective, and neurochemical benefits of a ketogenic diet potentially provide symptomatic relief to people with schizophrenia while also improving their cardiovascular or metabolic health. We review evidence for KD side effects and note that although high in fat it improves various cardiovascular and metabolic risk markers in overweight/obese individuals. We conclude by calling for controlled clinical trials to confirm or refute the findings from anecdotal and case reports to address the potential beneficial effects of the ketogenic diet in people with serious mental illness.
Article
Full-text available
A genome scan meta-analysis (GSMA) was carried out on 32 independent genome-wide linkage scan analyses that included 3255 pedigrees with 7413 genotyped cases affected with schizophrenia (SCZ) or related disorders. The primary GSMA divided the autosomes into 120 bins, rank-ordered the bins within each study according to the most positive linkage result in each bin, summed these ranks (weighted for study size) for each bin across studies and determined the empirical probability of a given summed rank (P(SR)) by simulation. Suggestive evidence for linkage was observed in two single bins, on chromosomes 5q (142-168 Mb) and 2q (103-134 Mb). Genome-wide evidence for linkage was detected on chromosome 2q (119-152 Mb) when bin boundaries were shifted to the middle of the previous bins. The primary analysis met empirical criteria for 'aggregate' genome-wide significance, indicating that some or all of 10 bins are likely to contain loci linked to SCZ, including regions of chromosomes 1, 2q, 3q, 4q, 5q, 8p and 10q. In a secondary analysis of 22 studies of European-ancestry samples, suggestive evidence for linkage was observed on chromosome 8p (16-33 Mb). Although the newer genome-wide association methodology has greater power to detect weak associations to single common DNA sequence variants, linkage analysis can detect diverse genetic effects that segregate in families, including multiple rare variants within one locus or several weakly associated loci in the same region. Therefore, the regions supported by this meta-analysis deserve close attention in future studies.
Article
Full-text available
Schizophrenia is a severe mental disorder with a lifetime risk of about 1%, characterized by hallucinations, delusions and cognitive deficits, with heritability estimated at up to 80%(1,2). We performed a genome-wide association study of 3,322 European individuals with schizophrenia and 3,587 controls. Here we show, using two analytic approaches, the extent to which common genetic variation underlies the risk of schizophrenia. First, we implicate the major histocompatibility complex. Second, we provide molecular genetic evidence for a substantial polygenic component to the risk of schizophrenia involving thousands of common alleles of very small effect. We show that this component also contributes to the risk of bipolar disorder, but not to several non-psychiatric diseases.
Article
Full-text available
This non-systematic review of the literature summarizes the evidence that inflammation plays a major role in the psychopathology of schizophrenia and in the mechanisms that contribute to physical ill health that is commonly associated with schizophrenia. The impact of prenatal infections on the developing brain, the possible genetic link between the human lymphocyte antigen gene, inflammation, heart disease and diabetes, together with the increase in pro-inflammatory cytokines in the blood and cerebrospinal fluid provide convincing evidence that inflammation is a major factor in the pathology of this disorder. The changes in immune-related markers and specific neurotransmitters associated with the positive symptoms of schizophrenia are described. In addition, the possible mechanism whereby structural changes occur in the brain is associated with the neurotoxic effects of pro-inflammatory cytokines, together with the neurotoxic metabolites from the tryptophan-kynurenine pathway that is activated by pro-inflammatory cytokines, is also discussed. The role of effective antipsychotic drug treatment in attenuating the inflammatory response is described. However, evidence is limited regarding the causal connection between atypical antipsychotic drugs and the changes in glucose and lipid metabolism that could trigger the onset of physical ill health, including diabetes and heart disease. Indeed, there is evidence that there is a metabolic predisposition to diabetes in patients with schizophrenia that is exacerbated by obesity and thereby contributes to cardiovascular disease and other co-morbid illnesses. It is concluded that the effects of inflammatory mediators on the brain causally contribute to the pathology of schizophrenia and the ill health that accompanies the disorder.
Article
Full-text available
The common approach to the multiplicity problem calls for controlling the familywise error rate (FWER). This approach, though, has faults, and we point out a few. A different approach to problems of multiple significance testing is presented. It calls for controlling the expected proportion of falsely rejected hypotheses – the false discovery rate. This error rate is equivalent to the FWER when all hypotheses are true but is smaller otherwise. Therefore, in problems where the control of the false discovery rate rather than that of the FWER is desired, there is potential for a gain in power. A simple sequential Bonferroni-type procedure is proved to control the false discovery rate for independent test statistics, and a simulation study shows that the gain in power is substantial. The use of the new procedure and the appropriateness of the criterion are illustrated with examples.
Article
Diabetic nephropathy (DN) is a major diabetic complication. Transforming growth factor-beta (TGF-beta) is a key mediator in the development of diabetic complications. It is well known that TGF-beta exerts its biological effects by activating downstream mediators, called Smad2 and Smad3, which is negatively regulated by an inhibitory Smad7. Recent studies also demonstrated that under disease conditions Smads act as signal integrators and interact with other signaling pathways such as the MAPK and NF-kappa B pathways. In addition, Smad2 and Smad3 can reciprocally regulate target genes of TGF-beta signaling. Novel research into microRNA has revealed the complexity of TGF-beta signaling during DN. It has been found that TGF-beta and elevated glucose concentration can positively regulate miR-192 and miR-377, but negatively regulate miR-29a in a diabetic milieu. These microRNAs are found to contribute to DN. Although targeting TGF-beta may exert adverse effects on immune system, therapeutic approach against TGF-beta signaling during DN still draws much attention. Blocking TGF-beta signaling by neutralizing antibody, anti-sense oligonucleotides, and soluble receptors have been tested, but effects are limited. Gene transfer of Smad7 into diseased kidneys demonstrates a prominent inhibition on renal fibrosis and amelioration of renal impairment. Alteration of TGF-beta-regulated microRNA expression in diseased kidneys may provide an alternative therapeutic approach against DN. In conclusion, TGF-beta/Smad signaling plays a critical role in DN. A better understanding of the role of TGF-beta/Smad signaling in the development of DN should provide an effective therapeutic strategy to combat DN. Copyright (C) 2011 S. Karger AG, Basel
Article
Diabetic nephropathy (DN) is a major diabetic complication. Transforming growth factor-β(TGF-β) is a key mediator in the development of diabetic complications. It is well known that TGF-β exerts its biological effects by activating downstream mediators, called Smad2and Smad3, which is negatively regulated by an inhibitory Smad7. Recent studies also demonstrated that under disease conditions Smads act as signal integrators and interact with other signaling pathways such as the MAPK and NF-κB pathways. In addition, Smad2and Smad3 can reciprocally regulate target genes of TGF-β signaling. Novel research into microRNA has revealed the complexity of TGF-β signaling during DN. It has been found that TGF-β and elevated glucose concentration can positively regulate miR-192 and miR-377, but negatively regulate miR-29a in a diabetic milieu. These microRNAs are found to contribute to DN. Although targeting TGF-β may exert adverse effects on immune system, therapeutic approach against TGF-β signaling during DN still draws much attention. Blocking TGF-β signaling by neutralizing antibody, anti-sense oligonucleotides, and soluble receptors have been tested, but effects are limited. Gene transfer of Smad7 into diseased kidneys demonstrates a prominent inhibition on renal fibrosis and amelioration of renal impairment. Alteration of TGF-β-regulated microRNA expression in diseased kidneys may provide an alternative therapeutic approach against DN. In conclusion, TGF-β/Smad signaling plays a critical role in DN. A better understanding of the role of TGF-β/Smad signaling in the development of DN should provide an effective therapeutic strategy to combat DN.