Skip to main content
Erschienen in: BMC Psychiatry 1/2023

Open Access 01.12.2023 | Research

Disclosing common biological signatures and predicting new therapeutic targets in schizophrenia and obsessive–compulsive disorder by integrated bioinformatics analysis

verfasst von: Abdolhakim Ghanbarzehi, Ali Sepehrinezhad, Nazanin Hashemi, Minoo Karimi, Ali Shahbazi

Erschienen in: BMC Psychiatry | Ausgabe 1/2023

Abstract

Schizophrenia (SCZ) is a severe mental illness mainly characterized by a number of psychiatric symptoms. Obsessive–compulsive disorder (OCD) is a long-lasting and devastating mental disorder. SCZ has high co-occurrence with OCD resulting in the emergence of a concept entitled “schizo-obsessive disorder” as a new specific clinical entity with more severe psychiatric symptoms. Many studies have been done on SCZ and OCD, but the common pathogenesis between them is not clear yet. Therefore, this study aimed to identify shared genetic basis, potential biomarkers and therapeutic targets between these two disorders. Gene sets were extracted from the Geneweaver and Harmonizome databases for each disorder. Interestingly, the combination of both sets revealed 89 common genes between SCZ and OCD, the most important of which were BDNF, SLC6A4, GAD1, HTR2A, GRIN2B, DRD2, SLC6A3, COMT, TH and DLG4. Then, we conducted a comprehensive bioinformatics analysis of the common genes. Receptor activity as the molecular functions, neuron projection and synapse as the cellular components as well as serotonergic synapse, dopaminergic synapse and alcoholism as the pathways were the most significant commonalities in enrichment analyses. In addition, transcription factor (TFs) analysis predicted significant TFs such as HMGA1, MAPK14, HINFP and TEAD2. Hsa-miR-3121-3p and hsa-miR-495-3p were the most important microRNAs (miRNAs) associated with both disorders. Finally, our study predicted 19 existing drugs (importantly, Haloperidol, Fluoxetine and Melatonin) that may have a potential influence on this co-occurrence. To summarize, this study may help us to better understand and handle the co-occurrence of SCZ and OCD by identifying potential biomarkers and therapeutic targets.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12888-023-04543-z.
Abdolhakim Ghanbarzehi and Ali Sepehrinezhad contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Schizophrenia (SCZ) is a complex and severe mental illness mainly characterized by a combination of positive symptoms (delusions and hallucinations), negative symptoms (social withdrawal, paucity of spontaneous speech and amotivation) and debilitating cognitive deficits [1]. Overall, the prevalence of SCZ in both sexes is approximately equal, but with earlier onset and greater severity in males than in females [2]. Environmental and social factors such as childhood trauma and social isolation predispose individuals to SCZ [3, 4]. However, it is highly heritable (~ 80%) with complex polygenic architecture which both common and rare genetic variants contribute to its etiology [5]. Emerging evidence has suggested that SCZ has a common molecular etiology with other psychiatric disorders such as obsessive–compulsive disorder (OCD), autism spectrum disorders (ASD) and bipolar disorder (BD), despite their unique clinical characteristics [6].
OCD, with the prevalence of about 1–3% in the general population, is a long-lasting and devastating mental disorder characterized by recurrent, persistent and unwanted thoughts, images or impulses called obsessions and repetitive behaviors called compulsions [7]. The World Health Organization has ranked it as one of the most debilitating disorders worldwide as it results in poor quality of life and can substantially impair the patient’s occupational, marital, emotional and social functioning [8, 9]. Moreover, plenty of epidemiological studies demonstrated that co-occurrence rates in OCD are generally higher than rates of other comorbid disorders [1012].
In the past years, the categorizing of psychiatric disorders has been rearranged through the increasing a lot of studies that emphasize co-occurring and/or comorbid disorders [13]. Relating to SCZ, the specific co-occurrence of obsessive–compulsive symptoms (OCS) and SCZ has been revealed for more than a century (in 1878) [14]. A meta-analysis study indicated that nearly 38.3% of SCZ patients experience anxiety disorders during their illnesses. Also, the prevalence of OCD in these patients was reported at 12.1% [15]. In the same way, another meta-analysis reported that the prevalence of OCS and OCD in SCZ patients is 30.3% and 12.3%, respectively [16]. These results along with abundant research that focus on the presence of OCD and OCS among the SCZ patients emerged the concept of “schizo-obsessive disorder” as a new specific clinical entity [1719]. Meanwhile, the accurate examination of these patients manifested that they have more severe psychotic and depressive symptoms, lower social functioning and higher suicidality [13]. Although the major psychiatric disorders are very debilitating, early diagnosis and treatment can substantially reduce the unfavorable outcomes of them [7]. Therefore, identifying more possible biomarkers and effective drugs can be essential for reducing the severity of these disorders.
There are two hypotheses for the co-occurrence of OCS during the course of SCZ. One assumes that second-generation antipsychotics, especially clozapine, might exacerbate or generate second-onset OCS. The second is an important role of genetic risk factors that dispose of patients with SCZ to develop OCS [20]. In the past decade, studies using both gene data and genome-wide association study (GWAS) have debated that some genes may be responsible for the co-occurrence of SCZ and OCS/OCD. [2022]. However, as far as we know, there has been no bioinformatic study performed with a special focus on the common genes between these disorders. In a recent study, O’Connell et al. displayed common genetic etiology for SCZ, BD, ASD and OCD. They proposed that more research on shared components of these disorders is needed to obtain actionable and translatable results in order to combat psychiatric disorders [6].
Although there have been many studies done on SCZ and OCD, the common pathogenesis of them is not been well identified at the molecular level until now. Based on this point, we assume that the co-occurrence of SCZ and OCD is rooted in the genetic similarities and it may reveal the shared genetic basis, potential biomarkers and therapeutic targets between the two disorders. In this regard, we conducted a comprehensive bioinformatics analysis to identify common genes, molecular functions, cellular components and biological pathways along with predicting transcription factors (TFs) and posttranscriptional regulator microRNAs (miRNAs) as well as to repurpose candidate drugs for both SCZ and OCD.

Material and Methods

At the beginning of conducting the study, the existing data were used in this way that all genes related to SCZ and OCD were extracted from GeneWeaver (https://​www.​geneweaver.​org/​) and Harmonizome (https://​maayanlab.​cloud/​Harmonizome/​) databases [23, 24]. GeneWeaver is an available web-based tool for conducting integrative functional genomics on our target genes in combination with large gene sets from different databases [25]. Harmonizome is a collection of processed datasets that provides comprehensive information about genes and proteins concerning diseases [26] and contained 71,597,788 associations between 295,485 attributes and 56,720 genes from 112 datasets provided by 65 resources. Extracted gene sets were taken from genome-wide association studies (GWAS) and other genetic association studies in Harmonizome. After that, each separated gene set was pasted into an excel file and then their common genes were saved for bioinformatics analyses. All shared genes were uploaded into the STRING (https://​string-db.​org/​) to obtain gene–gene interactions file [27]. Finally, we used windows version of Cytoscape 3.7.0 to visualize and interpret gene–gene interactions and reconstruct the genetic network for shared genes between SCZ and OCD [28]. Besides, Network Analyzer Tool was applied to calculate and identify network parameters such as degree and betweenness centrality. In all conducted analyses P-value < 0.05 or false discovery rate (FDR) < 0.05 were considered as significantly level.

Gene Ontology and pathway enrichment analysis

In order to investigate the possible mechanisms of OCD in SCZ, we conducted a gene enrichment analysis through ToppGene (https://​toppgene.​cchmc.​org/​). To achieve this goal, we uploaded common genes into the ToppGene through ToppFun section and then selected Gene Ontology (GO) to proceed the enrichment analysis for Molecular Function and Cellular Component of target genes. Also, our target genes were submitted into the KEEG pathway section of ToppGene database to investigate the possible disrupted pathways in both SCZ and OCD [29, 30]. Statistical significance (p < 0.05) was considered by a likelihood-ratio test with correction for FDR using Benjamini–Hochberg method to show multiple comparison.

Prediction of transcription factors and microRNAs besides drug repurposing

We used Enrichr (https://​maayanlab.​cloud/​Enrichr/​) to predict some significant TFs for common genes between SCZ and OCD. This goal was achieved through TRANSFAC and JASPAR PWMs panel of Enrichr [31]. In addition, miRDB database (http://​mirdb.​org/​index.​html) was used to predict main target miRNAs that represented both SCZ and OCD [32]. miRDB database uses constructive machine learning procedures to find target prediction score between 50–100 throughout datasets. Higher target prediction score represented more validated miRNA. All human miRNAs with target score of more than 95, were selected and uploaded to Cytoscape software to visualize gene-miRNAs network.
Finally, drug repurposing was conducted through Stitch and CTD section of ToppGene database. Cytoscape was used to visualize gene-pathways, gene-miRNAs, gene-drug interaction networks and determine the degree parameter in each network.

Results

Identification of SCZ-OCD shared genes and finding hub genes from reconstructed genetic network

At first, with the exploration of existing data in Geneweaver and Harmonizome, we identified 914 and 197 genes in association with SCZ and OCD respectively (Supplementary Tables 1 and 2). Interestingly, the combination of both gene sets revealed that 89 genes were shared between these two disorders (Fig. 1 and Supplementary Table 3). The following analyzing of common genetic network manifested that ten genes such as brain-derived neurotrophic factor (BDNF; Degree:63; Betweenness centrality:0.104), sodium-dependent serotonin transporter and solute carrier family 6 member 4 (SLC6A4; D:46; B:0.050), glutamate decarboxylase 1 (GAD1; D:46; B:0.026), 5-hydroxytryptamine receptor 2A (HTR2A; D:45; B:0.059), glutamate ionotropic receptor NMDA type subunit 2B (GRIN2B; D:45; B:0.027), dopamine receptor D2 (DRD2; D:44; B:0.024), solute carrier family 6 member 3 (SLC6A3; D:43; B:0.020), catechol-O-methyltransferase (COMT; D:40; B;0.032), tyrosine hydroxylase (TH; D:40; B:0.022) and discs large MAGUK scaffold protein 4 (DLG4; D:38; B:0.033) were the most significant genes (hub genes) according to their network parameters (Fig. 1). To note, all subsequent bioinformatic analyses were conducted on the common genes between SCZ and OCD.

Gene Ontology and pathway analysis for common genes

GO enrichment analyses revealed some significant molecular functions such as neurotransmitter receptor activity, transmitter-gated ion channel activity, signaling receptor activity, molecular transducer activity, postsynaptic neurotransmitter receptor, transmembrane signaling receptor activity, glutamate binding, ligand-gated ion channel activity and dopamine binding that may involve in both SCZ and OCD (Table.1). These analyses also resulted that neuron projection, synapse, somatodendritic compartment, neuronal cell body, dendritic tree, intrinsic component of synaptic membrane, synapse, neuron spine and distal axon were the most disrupted cellular components in these two disorders (Table.1). In addition, KEGG pathway analysis indicated that some pathways such as serotonergic synapse, cocaine addiction, dopaminergic synapse, amphetamine addiction, alcoholism, taste transduction, glutamatergic synapse, tyrosine metabolism and estrogen signaling pathway may contribute to the co-occurrence of OCD in SCZ patients. Reconstructed pathway-gene interaction network indicated that serotonergic synapse (D:18), dopaminergic synapse (D:16), alcoholism (D:14) and cocaine addiction were more connected nodes (Fig. 2).
Table 1
Result of molecular function and cellular component enrichment analysis for SCZ-OCD common genes
Name
q-value FDR B&H
Involved Genes
GO: Molecular Functions
Neurotransmitter receptor activity
8.42E-27
GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4,HTR1B,HTR2A,HTR3A,HTR7,HTR3E,
GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,GRM5,GABBR1,GABRA1
Transmitter-gated ion channel activity
6.02E-15
GABRG2,HTR3A,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,GABRA1
Signaling receptor activity
6.02E-15
GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4,HTR1B,NRXN3,HTR2A,OXTR,
HTR3A,NRXN1,HTR7,NTRK1,NTRK2,ESR1,HLA-DRB1,NTRK3,ESR2,NPSR1,
HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,NR3C1,GRM5,
FAS,NPY,GABBR1,GABRA1,OPRM1
Molecular transducer activity
6.02E-15
GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4,HTR1B,NRXN3,HTR2A,OXTR,
HTR3A,NRXN1,HTR7,NTRK1,NTRK2,ESR1,HLA-DRB1,NTRK3,ESR2,NPSR1,HTR3E,
GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,NR3C1,
GRM5,FAS,NPY,GABBR1,GABRA1,OPRM1
Postsynaptic neurotransmitter receptor activity
6.02E-15
GABRG2,DRD1,DRD2,DRD3,DRD4,GRIA2,GRIA3,GRIK2,GRIK3,GRIN2B,
GRM5,GABBR1,GABRA1
Transmembrane signaling receptor activity
8.26E-14
GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4,HTR1B,HTR2A,OXTR,HTR3A,
NRXN1,HTR7,NTRK1,NTRK2,HLA-DRB1,NTRK3,NPSR1,HTR3E,GRIA2,
GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,GRM5,
FAS,NPY,GABBR1,GABRA1,OPRM1
Glutamate binding
2.09E-12
GAD1,GAD2,GRIA2,GRIA3,GRIK2,GRIK3,GRIN2B,GRM5,SLC1A1
Ligand-gated ion channel activity
6.77E-12
GABRG2,HTR3A,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,
GRIN2B,HTR3B,GABRA1
Dopamine binding
8.19E-12
SLC6A3,TH,DRD1,DRD2,DRD3,DRD4
Transmembrane transporter activity
8.19E-12
SLC6A3,SLC6A4,GABRG2,DRD4,HTR1B,NRXN3,HTR3A,NRXN1,SLC18A1,SLC18A2,KCNH2,UCP2,
HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,SLC1A1,SLC1A2,NPY,GABRA1,
ABCB1,OPRM1,KCNH5
Passive transmembrane transporter activity
8.19E-12
GABRG2,DRD4,HTR1B,NRXN3,HTR3A,NRXN1,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,
GRIK2,GRIK3,GRIN2B,HTR3B,SLC1A1,NPY,GABRA1,OPRM1,KCNH5
Serotonin-gated cation-selective channel activity
5.90E-11
HTR3A,HTR3E,HTR3C,HTR3D,HTR3B
Inorganic molecular entity transmembrane transporter activity
6.33E-11
SLC6A3,SLC6A4,GABRG2,DRD4,HTR1B,HTR3A,SLC18A1,SLC18A2,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,
HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,SLC1A1,SLC1A2,GABRA1,ABCB1,OPRM1,KCNH5
Ligand-gated cation channel activity
1.04E-10
HTR3A,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B
Monooxygenase activity
1.09E-10
TH,CYP2C19,CYP2D6,CYP2E1,CYP3A4,CYP3A5,ESR1,TPH2,AKT1,DBH,TPH1
serotonin binding
1.18E-10
SLC6A4,HTR1A,DRD4,HTR1B,HTR2A,HTR3A,MAOA,HTR7
Amine binding
1.71E-10
SLC6A4,HTR1A,DRD4,HTR1B,HTR2A,HTR3A,MAOA,HTR7
Cation transmembrane transporter activity
1.74E-10
SLC6A3,SLC6A4,DRD4,HTR1B,HTR3A,SLC18A1,SLC18A2,KCNH2,HTR3E,GRIA2,
GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,SLC1A1,SLC1A2,OPRM1,KCNH5
Amino acid binding
2.37E-10
GAD1,GAD2,TH,GRIA2,GRIA3,GRIK2,GRIK3,GRIN2B,GRM5,SLC1A1
Ion channel activity
3.41E-10
GABRG2,DRD4,HTR1B,HTR3A,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,
GRIN2B,HTR3B,SLC1A1,GABRA1,OPRM1,KCNH5
Catecholamine binding
4.35E-10
SLC6A3,TH,DRD1,DRD2,DRD3,DRD4
Ion transmembrane Transporter activity
4.95E-10
SLC6A3,SLC6A4,GABRG2,DRD4,HTR1B,HTR3A,SLC18A1,SLC18A2,KCNH2,HTR3E,GRIA2,
GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,SLC1A1,SLC1A2,GABRA1,ABCB1,OPRM1,KCNH5
Neurotransmitter receptor activity involved in regulation of postsynaptic membrane potential
2.58E-09
GABRG2,GRIA2,GRIA3,GRIK2,GRIK3,GRIN2B,GABBR1,GABRA1
Cation channel activity
9.12E-09
DRD4,HTR1B,HTR3A,KCNH2,HTR3E,GRIA2,GRIA3,HTR3C,HTR3D,GRIK2,GRIK3,GRIN2B,HTR3B,OPRM1,KCNH5
Oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen
9.29E-09
TH,CYP2C19,CYP2D6,CYP2E1,CYP3A4,CYP3A5,ESR1,TPH2,AKT1,DBH,TPH1
Dopamine neurotransmitter receptor activity
3.34E-08
DRD1,DRD2,DRD3,DRD4
Glutamate receptor activity
3.39E-08
GRIA2,GRIA3,GRIK2,GRIK3,GRIN2B,GRM5
GO: Cellular Components
Neuron projection
1.89E-38
SLC6A3,SLC6A4,GABRG2,GAD1,GAD2,TH,DRD1,DRD2,HTR1A,DRD4,HTR1B,HTR2A,HTR3A,COMT,NRCAM,NRXN1, HTR7,MAP2,SLC18A1,MTOR,SLC18A2,NTRK1,NTRK2,ESR1,NTRK3,ESR2,HTR3E,TPH2,CNTNAP2,ARC,SYP,
GRIA2, GRIA3,DLG4,HTR3C,HTR3D,GRIK2,GRIK3,DBH,GRIN2B,HTR3B,CTNNB1,NR3C1,GRM5,FAS,CREB1,
SLC1A1, SLC1A2, AR,HOMER1,CRH,BDNF,NPY,GABBR1,SOD1,GABRA1,OPRM1,TPH1
Synapse
6.03E-35
SLC6A3,SLC6A4,GABRG2,GAD1,GAD2,TH,DRD1,DRD2,DRD3,HTR1A,DRD4,HTR1B,NRXN3,HTR2A,HTR3A,
COMT,NRCAM,NRXN1,HTR7,MAP2,SLC18A1,MTOR,SLC18A2,NTRK2,ESR1,NTRK3,HTR3E,CNTNAP2,
ARC,SYP,GRIA2,GRIA3,DLG4,HTR3C,HTR3D,AKT1,GRIK2,GRIK3,DBH,GRIN2B,HTR3B,CTNNB1,NR3C1,GRM5,
SLC1A1,SLC1A2,HOMER1,CRH,BDNF,NPY,GABBR1,NOS1AP,GABRA1,OPRM1
Somatodendritic compartment
1.10E-30
SLC6A3,GABRG2,TH,DRD1,DRD2,HTR1A,DRD4,HTR1B,HTR2A, HTR3A,COMT,NRXN1,HTR7,MAP2,
MTOR,SLC18A2,NTRK1, NTRK2,ESR1,ESR2,CNTNAP2,ARC,GRIA2,GRIA3,DLG4,GRIK2,
GRIK3,DBH,GRIN2B,HTR3B,CTNNB1,NR3C1,GRM5,FAS,SLC1A1,SLC1A2,AR,HOMER1,CRH,BDNF,NPY,
GABBR1,SOD1,GABRA1,OPRM1
Neuronal cell body
1.85E-25
SLC6A3,TH,DRD1,DRD2,HTR1A,DRD4,HTR2A,HTR3A,NRXN1,HTR7, MAP2,MTOR,SLC18A2,NTRK1,NTRK2,ESR1,
ESR2, CNTNAP2,ARC,GRIA2,GRIA3,GRIK2,GRIK3, DBH,GRIN2B,HTR3B,FAS,SLC1A1, HOMER1,CRH,
BDNF,NPY,GABBR1,SOD1,OPRM1
Dendritic tree
2.07E-25
GABRG2,TH,DRD1,DRD2,HTR1A,DRD4,HTR1B,HTR2A,COMT,HTR7, MAP2,MTOR,NTRK1,NTRK2,CNTNAP2,
ARC,GRIA2,GRIA3,DLG4, GRIK2,GRIK3,DBH,GRIN2B,CTNNB1,NR3C1,GRM5,FAS,SLC1A1,
SLC1A2,AR,HOMER1,BDNF,GABBR1,SOD1,GABRA1,OPRM1
Intrinsic component of synaptic membrane
1.23E-24
SLC6A3,SLC6A4,GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4, HTR1B,HTR2A,HTR3A,NRCAM,NRXN1,
HTR7,NTRK3,GRIA2,GRIA3,DLG4,GRIN2B,GRM5,SLC1A2,GABBR1,GABRA1,OPRM1
glutamatergic synapse
2.06E-18
GABRG2,DRD1,DRD2,DRD3,DRD4,HTR2A,HTR3A,NRCAM,NRXN1, MTOR,NTRK2,NTRK3,ARC,GRIA2,GRIA3,
DLG4,GRIK2,GRIK3, GRIN2B,NR3C1,GRM5,SLC1A2,HOMER1,GABBR1,NOS1AP
Neuron spine
5.57E-18
DRD1,DRD2,DRD4,COMT,NTRK2,CNTNAP2,ARC,SYP,GRIA2, GRIA3,DLG4,GRIK2,GRIN2B,
NR3C1,GRM5,SLC1A1,SLC1A2, HOMER1,GABBR1,OPRM1
Distal axon
2.31E-17
GAD1,TH,DRD1,DRD2,DRD4,HTR1B,NRXN1,HTR7,MAP2,SLC18A1, SLC18A2,NTRK2,ESR1,SYP,
GRIA2,GRIA3,GRIK2,GRIK3, DBH,GRIN2B,SLC1A1,BDNF,NPY
Terminal bouton
4.14E-16
TH,DRD4,SLC18A1,SLC18A2,NTRK2,ESR1,SYP,GRIA2, GRIA3,GRIK2,GRIK3,DBH,
GRIN2B,BDNF,NPY
Dendritic spine
1.68E-15
DRD1,DRD2,DRD4,COMT,NTRK2,ARC,GRIA2,GRIA3,DLG4,GRIK2,GRIN2B,NR3C1,GRM5,SLC1A1,SLC1A2,
HOMER1,GABBR1,OPRM1
Dendritic shaft
1.46E-13
DRD1,HTR2A,MAP2,GRIA2,GRIA3,CTNNB1,GRM5,SLC1A1,SLC1A2,HOMER1,GABBR1
Perikaryon
2.04E-13
TH,DRD2,NTRK2,ESR1,ESR2,CNTNAP2,GRIA2,GRIA3,GRIK2, GRIK3,SLC1A1,CRH,BDNF,NPY,OPRM1
Asymmetric synapse
6.82E-12
DRD1,DRD2,DRD3,NRCAM,MAP2,NTRK2,ARC,SYP, GRIA2,GRIA3,DLG4,GRIK2,GRIN2B,CTNNB1,
NR3C1,GRM5,SLC1A1,HOMER1
Neuron to neuron synapse
1.87E-11
DRD1,DRD2,DRD3,NRCAM,MAP2,NTRK2,ARC,SYP,GRIA2, GRIA3,DLG4,GRIK2,GRIN2B,CTNNB1,
NR3C1,GRM5,SLC1A1,HOMER1
Receptor complex
3.58E-10
GABRG2,HTR1B,HTR2A,HTR3A,NTRK1,NTRK2,NTRK3,GRIA2,GRIA3,DLG4,GRIK2,GRIK3, GRIN2B,HTR3B,
GDNF,IL6,GABBR1,GABRA1
GABA-ergic synapse
5.15E-10
GABRG2,DRD1,DRD2,DRD3,HTR1A,DRD4,NRXN3,NRXN1,GABBR1,GABRA1

Predicted transcription factors and microRNAs for target genes

As regards gene expression patterns strongly regulate at transcriptional and posttranscriptional levels, the importance of the next steps becomes apparent. TF analysis predicted 15 significant TFs, namely HMGA1, MAPK14, HINFP, TEAD2, JUND, NFIA, SAMD9L, NFKB1, REL, PRDM1, HOXA5, GATA1, NKX3-1, VDR and STAT3 (Table.2). We also predicted 121 miRNAs with target score of more than 95 through miRDB database for our target genes (Fig. 3). Reconstructed gene-miRNAs interaction network showed that two miRNAs hsa-miR-3121-3p and hsa-miR-495-3p represented three interactions with their target genes meanwhile hsa-miR-369-3p, hsa-miR-186-5p, hsa-miR-218-5p, hsa-miR-22-3p, hsa-miR-330-3p, hsa-miR-543, hsa-miR-1271-5p, hsa-miR-96-5p, hsa-miR-148b-3p, hsa-miR-152-3p, hsa-miR-148a-3p, hsa-miR-19b-3p and hsa-miR-19a-3p interacted with two different targeted genes (Fig. 3). Furthermore, hsa-miR-144-3p, hsa-miR-22-3p, hsa-miR-221-3p, hsa-miR-3121-3p, hsa-miR-19a-3p, hsa-miR-200c-3p, hsa-miR-429, hsa-miR-381-3p, hsa-miR-126-5p, hsa-miR-200b-3p, hsa-miR-222-3p, and hsa-miR-488-3p had target prediction scores more than 99.
Table 2
Result of transcription factor prediction for SCZ-OCD shared genes
Name
P-value
Involved Genes
HMGA1
0.0001071
GRIA2, GABRA1, ABCB1, BDNF, NRXN1, GSTP1, NRXN3, HTR3D, HTR3A, PRL,OPRM1,NR3C1, RGS4, NOS1AP, UCP2,DRD2,GRIA3
MAPK14
0.0002271
GABRA1, KCNH5, SLC1A1, OPRM1, NR3C1, ESR1, RGS4,AR, NOS1AP,CRH, DRD1, ANKK1, SLC18A1, DRD3,SLC18A2
HINFP
0.0002323
CNTNAP2, MAOB, COMT, NR3C1, SLC6A3, RGS4, AKT1, NRCAM, DRD2,SLC18A2, KCNH2, NTRK1,NTRK2,EGR1,GABRA1,BDNF,NTRK3,OLIG2,OPRM1,HTR3B,ESR1,AR, ARC,NOS1AP,CRH,CYP2E1,HLA-DRB1
TEAD2
0.0007907
HOMER1, BDNF, NTRK3, GRIK3, HTR2A,COMT, NR3C1, SLC6A3, LMX1A,TH, GDNF, DLG4, UCP2, CTNNB1, ANKK1, SLC18A2,
JUND
0.001069
GRIA2, GABRA1, MAOB, BDNF, NRXN1, NRXN3, PRL,NR3C1, CYP3A5,GABRG2, HTR7, NPY, CRH, NRCAM, SLC18A1
NFIA
0.001384
MAOB, PRL,NR3C1, CYP3A4, ESR1, ESR2, SOD1, AR, GRM5, LMX1A, TH, CYP2D6, DLG4, CYP2E1, DRD3
SAMD9L
0.003779
KCNH2, NPSR1,TPH2,GRIA2,EGR1,MOG,HTR3D, GAD2, HTR3A,NR3C1,CYP3A5,ESR1,DRD1,DRD3
NFKB1
0.004332
GSTP1, SLC1A1,NRXN3,COMT, SLC6A3, SLC6A4, AKT1, EGR1, EGR2, KCNH5, BDNF, NTRK3, GAD1, GAD2, OPRM1, GRIN2B, ESR1, GABRG2, MTOR, ESR2,ARC,IL6, CREB1, GDNF, NOS1AP,ANKK1
REL
0.00493
KCNH5, LMX1A, BDNF, NTRK3,PRL,SLC6A4
PRDM1
0.005229
NTRK2, GABRA1, BDNF, NFKBIL1, NRXN1, NRXN3, DBH, OPRM1, COMT, NR3C1, RGS4, CREB1, GDNF, HLA-DRB1
HOXA5
0.005306
CNTNAP2, GRIA2, GABRA1, GABBR1, EGR2, MAOB, BDNF, SLC1A1, SLC1A2,NRXN3, HTR1A, HTR3D, GAD2, HTR3A, OLIG2, OPRM1, COMT, NR3C1, SLC6A3, GDNF, MAP2, NRCAM, DRD3
GATA1
0.006189
NPSR1, NTRK2, EGR1, ABCB1, BDNF, NRXN3, PRL, OPRM1, SYP, CYP3A4, ESR1, RGS4, GRM5, HTR7, MAP2, DLG4, NPY, CRH, FAS, DRD3, SLC18A2
NKX3-1
0.006509
KCNH5, NRXN3, ESR1, MTOR
VDR
0.007118
BDNF, CYP3A4, CYP3A5
STAT3
0.008629
GRIA2, TPH1, GABBR1, EGR2, ABCB1, MOG, BDNF, NFKBIL1, NTRK3, GAD1, DBH, OPRM1, CYP3A5, ESR1, SLC6A4, RGS4, AR, IL6, MAP2, NRCAM, HLA-DRB1

Repurposed drug and gene-drug interaction network for SCZ-OCD associated genes

Based on results from ToppGene database, we repurposed 19 potential significant drugs, namely Haloperidol, Clozapine, Desipramine, Fluoxetine, Nicorette, Pseudococaine, Amitriptyline, Amphetamine, Risperidone, Clomipramine, Reboxetine, Imipramine, Reserpine, Citalopram, Levodopa, Pargyline, Melatonin, Olanzapine and Buspirone for SCZ-OCD-associated common genes (Fig. 4). In addition, the gene-drug interaction network revealed that four drugs Haloperidol, Clozapine, Fluoxetine and Melatonin were more connected drugs according to their degrees (Fig. 4).

Discussion

Previous studies have shown that SCZ is closely related to OCD [13, 1519]. In the present study, we used globally accessible databases (Geneweaver and Harmonizome) for exploiting related genes of SCZ and OCD to survey (by conducting bioinformatic analyses) the hypothesis that these two mental disorders display prospective significant shared genetic basis, potential biomarkers and therapeutic targets in terms of common genes. Initially, we found a big common gene set (with 89 genes) between SCZ and OCD which can be evidence for the common pathogenesis and co-occurrence of these two disorders. The results of enrichment analysis on the common genes showed that ten genes (BDNF, SLC6A4, GAD1, HTR2A, GRIN2B, DRD2, SLC6A3, COMT, TH and DLG4) are the most central genes in associated with both disorders.
BDNF was found as the most central shared gene (Fig. 1). This gene encodes BDNF protein which is a member of a large family of neuronal growth factors called neurotrophins and plays a pivotal role in neurogenesis, differentiation and cell survival [33]. Meanwhile, it contributes to the transcription and translation of proteins involved in the synaptogenesis, development and stability of synapses [34]. BDNF has been widely studied in psychiatric disorders and its possible roles in the pathophysiology of them were discussed in the last two decades. It is released at the synapse and affects synaptic plasticity, subsequently induces critical changes in cognitive functions, learning and memory [35, 36]. While, a defect in the regulation of BDNF release can cause abnormalities in the underlying brain processes and cognitive dysfunctions in the psychiatric disorders [37]. BDNF signaling may substantially promote the structure and functioning of several neural circuits involved in the modulation of various neurotransmitter systems, including the dopaminergic [38], serotoninergic [39] and GABAergic [40] systems, all closely related to SCZ. In this regard, the normal development of these systems may disturb by dysfunction of BDNF-TrkB signaling during critical developmental periods, consequently leading to physiological dysregulation and vulnerability to SCZ[41]. Relating to OCD, it has been demonstrated that sequence variants in the BDNF gene are strongly associated with OCD [42]. For example, it was reported that the Val66Met BDNF gene variant contributes to the OCD pathobiology [43, 44]. In addition, more studies have shown that this variant lead to functional differences and consequently decrease the activity of the BDNF system, which can be a risk factor for OCD [45].
Furthermore, the interface between SCZ and OCD can be explained through the serotoninergic neurotransmission as two important common genes are SLC6A4 and HTR2A which have critical functions in the serotoninergic pathway. SLC6A4 by the encoding of serotonin transporter (5-HTT) has a crucial role in the regulation of serotonin via reuptake of it from synaptic clefts [46]. Because this gene is involved in the pathogenesis of SCZ, it is considered as a candidate gene for SCZ [47]. It has been reported that mRNA and protein levels of 5-HTT were changed in SCZ patients compared with healthy subjects [48, 49]. Likewise, there is a lot of evidence reporting that serotonergic system is implicated in the pathophysiology of OCD, in particular well-known anti-obsessional efficacy of selective serotonin reuptake inhibitors (SSRIs) have suggested an important role for 5-HTT in the etiology of OCD [50]. Zitterl et al. showed a significant reduction in 5-HTT availability in the brain regions of OCD patients [51]. Clearly, several studies demonstrated that there is a significant association between the SLC6A4 polymorphisms and OCD susceptibility [52]. The HTR2A gene encodes the serotonin receptor type 2A (5-HTR2A) and is abundantly expressed in the glutamatergic neurons and GABA-ergic interneurons in the prefrontal cortex and hippocampal regions which both neurotransmission systems are well known to be associated with SCZ [53]. It has been shown that 5-HTR2A is involved in the pathogenesis of SCZ as its activation regulates both dopaminergic and glutamatergic transmission in the brain [54]. Relating to OCD, findings from candidate gene studies demonstrated that HTR2A is a most important gene for development of the disorder [55]. Also, recent meta-analytic evidence showed that polymorphic variants within this gene are significantly associated with OCD pathogenesis [56, 57].
GAD1 is one of the other important genes in both SCZ and OCD. It encodes the glutamic acid decarboxylase-67 (GAD67) enzyme in multiple cortical regions (particularly in the prefrontal cortex and hippocampus) which is responsible for most cortical γ-Aminobutyric acid (GABA) synthesis [58]. This gene with a lower expression level in the SCZ subjects and consequently lower levels of GAD67 mRNA and protein is a well-known biomarker for SCZ [59]. In OCD patients, a recent clinical study reported that GABA abnormalities can be found within the anterior cingulate cortex [60]. Besides, Zhang et al. indicated that GABA concentration in the prefrontal cortex contributes to the psychopathology of OCD [61].
According to our results, another candidate gene for co-occurrence of SCZ and OCD is GRIN2B encodes the NR2B subunit of N-methyl D-aspartate (NMDA) glutamate receptors and may play an important role in synaptic plasticity, circuit formation and brain development [62]. Recently, it has been reported that variations in GRIN2B can be associated with SCZ which may be due to abnormalities of the NR2B subunit and consequently altered function of NMDA receptors [63]. In addition, hypofunction of NMDA receptors has been suggested as a mechanism in the pathogenesis of SCZ, based on the point that noncompetitive antagonists of the NMDA receptors, such as ketamine and phencyclidine, induce SCZ-like symptoms [64]. In contrast, it is proposed that neuronal excitotoxicity resulting from hyperactive NMDA receptors has an important role in SCZ [65]. Clinical and preclinical evidence have suggested that dysregulation of glutamatergic system contributes to the etiology of OCD [66]. In imaging studies of OCD, it has been shown that a hyperglutamatergic dysfunction may lead to abnormalities in the cortico–striatal–thalamo–cortical circuits [67]. In addition, it is demonstrated that the GRIN2B gene is implicated in OCD as mutations of it have been associated with the disorder in males [68] and the presence of contamination obsessions and cleaning compulsions [69].
The DRD2 gene, coding for dopamine D2 receptor, is an attractive candidate gene for SCZ due to its role in dopaminergic system [70]. Several polymorphisms of this gene have been identified related to SCZ, hence it is well considered as a causative factor in SCZ [71]. Besides, DRD2 likely contributes to the OCD pathology since it has been demonstrated that the dopaminergic system is implicated in inducing or aggravating the symptoms of OCD [72] and in particular it is reported that the DRD2 A2 allele is significantly higher in male OCD patients compared to controls[73].
Other important shared genes between SCZ and OCD are SLC6A3, COMT, TH and DLG4. The SLC6A3 gene encodes the dopamine transporter and can be determinative in the regulation of dopamine in the synaptic cleft. It has been well indicated that its polymorphisms are risk factors for SCZ [74]. Also, the manifestation of OCD symptoms can be associated with SLC6A3 [75]. Since the COMT enzyme has a crucial role in the metabolism of dopamine, the COMT gene is considered as an important factor in the etiology of SCZ [76]. Higher dopamine levels are likely involved in OCD, hence COMT can be a suitable candidate for OCD [77]. Tyrosine hydroxylase (TH), encoded by TH gene, is a rate-limiting enzyme that produces dopamine in the brain and can be related to SCZ [78]. Dopamine neurotransmission is likely associated with OCD [77]. Therefore, the TH gene may be involved in OCD. Post-synaptic density protein 95 (PSD95), encoded by the DLG4 gene, has an essential rolein regulating NMDA receptor activity and altered expression of DLG4 has been revealed in the post-mortem brain of subjects with SCZ [79]. In OCD pathogenesis, since glutamatergic system can be disrupted [80], DLG4 may be an interference factor in this system.
In our study, the significant molecular functions exhibit receptor activity as the most common of them associated with SCZ and OCD. As recently reviewed, multiple serotonin receptors (5-HTR) are implicated in SCZ, including 5-HT1AR, 5-HT2CR, 5-HT3R, 5-HT6R, 5-HT7R and several studies link 5-HT2AR to the pathogenesis of SCZ as its activation regulates both dopaminergic and glutamatergic transmission [54]. It is worth mentioning in SCZ that the more-focused and well-known receptors are dopamine D2 receptors involved in the mesolimbic dopamine pathway, and their hyperactivity is responsible for the cause of the positive symptoms of SCZ, with all antipsychotics acting to block them [81]. Also, Dopamine D2 receptors likely play a part in the OCD pathology as decreased striatal D2/D3 and D1 receptors were reported in patients with OCD [82]. Positron emission tomography (PET) showed a significant reduction of 5-HT2AR availability in OCD patients compared to healthy subjects [83].
Based on cellular component enrichment analysis, the cellular components involved in the transition of the signals, namely, neuron projection and synapse were the most important cellular components in both SCZ and OCD. As mentioned, serotonergic synapse markers (5-HTT and 5-HT2AR) contributed to the development of OCD. Besides, the neurocircuitry of OCD is located between different brain regions which are densely innervated by neuron projections of the serotonergic and dopaminergic neurotransmitter systems [84]. Also, Rosoklija et al. reported the structural abnormalities of dendrites in SCZ and mood disorders [85]. In addition, Faludi and Mirnics showed that synaptic disturbances are important and integral part of SCZ pathophysiology [86].
KEGG pathway analysis indicated that the identified common genes mostly contributed to serotonergic synapse, cocaine addiction, dopaminergic synapse, amphetamine addiction, alcoholism, taste transduction, glutamatergic synapse, tyrosine metabolism and estrogen signaling pathways. As discussed above, several studies have shown that serotonergic pathway has a substantial role in the pathophysiology of SCZ and OCD. In addition to the dopamine and glutamate theory of psychosis, there is also the serotonin theory states that cortical 5-HT2AR hyperfunction can also result in psychosis [81].
TF analysis predicted that HMGA1, MAPK14, HINFP and TEAD2 are the more relevant TF biomarkers associated with both SCZ and OCD. According to the gene cards database (www.​genecards.​org); HMGA1 is associated with Type 2 Diabetes Mellitus and Multiple Lipomatosis. Likewise, Diseases associated with MAPK14 include Chlamydia and Alexander Disease. HINFP show an association with Cerebro-oculo-facio-skeletal Syndrome 2 and Fetal Alcohol Spectrum Disorder. Diseases associated with TEAD2 include Sveinsson Chorioretinal Atrophy and Multiple Acyl-CoA Dehydrogenase Deficiency [87].
This study predicted 121 significant miRNAs for common genes through miRDB database (Fig. 3). Among them, hsa-miR-3121-3p, hsa-miR-495-3p, hsa-miR-369-3p, hsa-miR-186-5p, hsa-miR-218-5p, hsa-miR-22-3p, hsa-miR-330-3p, hsa-miR-543, hsa-miR-1271-5p, hsa-miR-96-5p, hsa-miR-148b-3p, hsa-miR-152-3p, hsa-miR-148a-3p, hsa-miR-19b-3p and hsa-miR-19a-3p were more relevant identified biomarkers in associated to both SCZ and OCD. In human, almost 70% of miRNAs express in the nervous system [88] and they have a role in the regulation of neural structure and function such as formation of dendrites and dendritic spines, axon growth, neural developments and the maturation process [89]. Also, abnormal expressions of miRNAs could be important as they are involved in the occurrence of neuropsychiatric disorders [90]. Therefore, considering them as potential biomarkers for co-occurrence of SCZ and OCD can be useful for more diagnostic and therapeutic targets.
A class of non-coding RNA molecules as miRNAs act as negative regulators of post-transcriptional processes that mediate gene expression profile and subsequent biological functions. In the present study, we predicted 121 miRNAs with target predicting score of more than 95 concerning SCZ and OCD. Pan et al., showed that hsa-miR-144-3p is associated with schizophrenia through a decrease in the expression of ATPase Na+/K+ transporting subunit beta 2 (ATP1B2) and PI3K/Akt/mTOR signalling in rat hippocampus [91]. It has been reported that miR-3121-3p suppressed RAP1 GTPase activating protein (Rap1GAP) [92], while this protein is crucial for development of dentate gyrus and prevention of SCZ-like behavior in mice [93]. Importantly next-generation sequencing and real-time quantitative polymerase chain reaction (qRT-PCR) on peripheral blood cells in patients with SCZ, suggested hsa-miR-22-3p as a biomarker for these patients [94]. Interestingly, the level of hsa-miR-22-3p also significantly increased in patients with OCD compared to healthy subjects [95]. A bioinformatics study on microarray dataset from brain post-mortem samples of SCZ patients identified hsa-miR-26a-5p as a differentially expressed miRNA compared to control samples [96]. Also, comparison of expression pattern of several miRNAs in peripheral blood cells by qRT-PCR revealed a significant increase in the expression of miR-26a-5p in OCD patients compared to control subjects [97].
Finally, current research identified drugs (particularly Haloperidol, Fluoxetine, Clozapine and Melatonin) that may have a potential influence on the co-occurrence of SCZ and OCD. Haloperidol, a highly effective first-generation antipsychotic (FGA), is one of the most used antipsychotic drugs, and it has very high anti-dopaminergic activity in the mesolimbic dopamine pathway [98] hence it is very efficient for the treatment of SCZ [99]. However, like other FGAs, it is associated with severe extrapyramidal side effects [100]. Haloperidol is considered as second in line with antipsychotic augmenting agents in OCD patients who showed an inadequate response to SSRIs but better response with antipsychotic augmentation [101]. Fluoxetine is one of the oldest SSRIs and is applied as a first-line drug for the treatment of some mental illnesses such as major depressive disorder, premenstrual dysphoric disorder, panic disorder and bulimia nervosa [102]. Furthermore, it is widely used in OCD patients due to its good therapeutic response, good compliance and low side effects [103, 104]. Also, in treating SCZ, it has been indicated that the use of antidepressants like fluoxetine as an add-on therapy to antipsychotics can improve the negative symptoms in patients with chronic SCZ [105]. Clozapine, a dibenzodiazepine developed in 1961, is an antipsychotic approved in treating resistant SCZ [106]. It has been indicated that clozapine is more effective than any other antipsychotic drug (first or second-generation) in the treatment of resistant SCZ [107]. However, the anti-serotonergic effects of clozapine in cortico-striatal serotoninergic circuits may induce OCD-like behavior in mice and generate OCS in patients treated with it [108, 109]. Therefore, it is hypothesized that decrease plasma concentration of clozapine may alleviate OCD, but it can exacerbate the severity of SCZ [20]. Clinicians should be aware of the exacerbation of OCD and OCS after chronic prescription of clozapine in treatment of SCZ. However, prescription of SSRIs along with antipsychotic medications such as clozapine and olanzapine might be a proposed alternative treatment in co-occurrence of SCZ and OCD. As, Stryjer et al. reported that administration of 20 mg/day escitalopram in patients with SCZ and OCD that were treated with antipsychotic drugs (i.e. clozapine, risperidone and quetiapine), decreased the total Yale Brown Obsessive–Compulsive Scale scores (Y-BOCS, P = 0.001) and Positive and Negative Syndrome Scale scores (PANSS, P = 0.03) [110]. Melatonin, the endogenous hormone that regulates circadian rhythms, is used exogenously for the treatment of sleep disorders [111]. Available evidence has suggested that melatonin is linked to SCZ. Sleep disorder is a usual feature of SCZ [112] and appear to be caused by abnormal melatonin functions and abnormal circadian implicated in the pathophysiology of SCZ [113]. Additionally, decreased melatonin level in SCZ patients was reported [114116]. A recent systematic review of melatonin use for SCZ showed that adjunctive melatonin therapy can be useful for sleep, metabolic profile and tardive dyskinesia in SCZ patients [117]. Sleep disorders are also prevalent among OCD patients, as up to 48% of them report these disorders [118]. Furthermore, Monteleone et al. showed that overall plasma concentrations of melatonin are lower in patients with OCD compared with normal controls [119]. Hence, melatonin therapy may be helpful for OCD. Our study utilized a bioinformatic method to identify common molecular and cellular mechanisms and predict new therapeutic drugs for SCZ and OCD. However, the present study has a number of limitations. One limitation was the present study failed to verify main findings with experimental results due to lack of confirmatory experimental animal studies that mimic SCZ and OCD-like behavior. Another limitation is lack of an integrated comprehensive database to support genetic basis of OCD in human subjects. Furthermore, the function of most predicted miRNAs have yet to be determined. Therefore, validation of predicted results requires further in vitro, in vivo, and especially clinical future experimental researches with large sample size.

Conclusion

Obsessive–compulsive behavior is a common comorbid condition with schizophrenia. Herein, we conducted a comprehensive enrichment analysis on the common genetic basis of SCZ and OCD. Regarding comorbid disorders, bioinformatics studies may help us to better understand the common pathophysiology of these disorders by identifying possible biomarkers and underlying mechanisms involved in them. Furthermore, finding more potential therapeutic options for disorders can be another implication of such analyses and future experimental studies.

Acknowledgements

Not applicable.

Declarations

Competing interests

The authors declare no competing interests.
Not applicable.
Not applicable.

Competing interest

The authors declare no conflicts of interest regarding this article.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Merikangas AK, Shelly M, Knighton A, Kotler N, Tanenbaum N, Almasy L. What genes are differentially expressed in individuals with schizophrenia? A systematic review. Molecular Psychiatry. 2022;27:1373–1383. Merikangas AK, Shelly M, Knighton A, Kotler N, Tanenbaum N, Almasy L. What genes are differentially expressed in individuals with schizophrenia? A systematic review. Molecular Psychiatry. 2022;27:1373–1383.
2.
Zurück zum Zitat Merikangas AK, Almasy L. Using the tools of genetic epidemiology to understand sex differences in neuropsychiatric disorders. Genes Brain Behav. 2020;19(6): e12660.CrossRef Merikangas AK, Almasy L. Using the tools of genetic epidemiology to understand sex differences in neuropsychiatric disorders. Genes Brain Behav. 2020;19(6): e12660.CrossRef
3.
Zurück zum Zitat Bozorgmehr A, Sadeghi B, Zavari EST, Bahrami E, Zamani F, Shahbazi A. An integrative gene network-based approach to uncover the cellular and molecular infrastructures of schizophrenia. Life Sci. 2020;260: 118345.CrossRef Bozorgmehr A, Sadeghi B, Zavari EST, Bahrami E, Zamani F, Shahbazi A. An integrative gene network-based approach to uncover the cellular and molecular infrastructures of schizophrenia. Life Sci. 2020;260: 118345.CrossRef
4.
Zurück zum Zitat Patel KR, Cherian J, Gohil K, Atkinson D. Schizophrenia: overview and treatment options. Pharmacy and Therapeutics. 2014;39(9):638. Patel KR, Cherian J, Gohil K, Atkinson D. Schizophrenia: overview and treatment options. Pharmacy and Therapeutics. 2014;39(9):638.
5.
Zurück zum Zitat Owen M, Sawa A, Mortensen P. Schizophrenia Lancet Lond Engl. 2016;388:86–97.CrossRef Owen M, Sawa A, Mortensen P. Schizophrenia Lancet Lond Engl. 2016;388:86–97.CrossRef
6.
Zurück zum Zitat O’Connell KS, McGregor NW, Lochner C, Emsley R, Warnich L. The genetic architecture of schizophrenia, bipolar disorder, obsessive-compulsive disorder and autism spectrum disorder. Mol Cell Neurosci. 2018;88:300–7.CrossRef O’Connell KS, McGregor NW, Lochner C, Emsley R, Warnich L. The genetic architecture of schizophrenia, bipolar disorder, obsessive-compulsive disorder and autism spectrum disorder. Mol Cell Neurosci. 2018;88:300–7.CrossRef
7.
Zurück zum Zitat Hamidian S, Pourshahbaz A, Bozorgmehr A, Ananloo ES, Dolatshahi B, Ohadi M. How obsessive–compulsive and bipolar disorders meet each other? An integrative gene-based enrichment approach. Ann Gen Psychiatry. 2020;19(1):1–10.CrossRef Hamidian S, Pourshahbaz A, Bozorgmehr A, Ananloo ES, Dolatshahi B, Ohadi M. How obsessive–compulsive and bipolar disorders meet each other? An integrative gene-based enrichment approach. Ann Gen Psychiatry. 2020;19(1):1–10.CrossRef
8.
Zurück zum Zitat Singh A, Beniwal RP, Bhatia T, Deshpande SN. Schizophrenia with and without obsessive-compulsive symptoms: a comparative analysis of performance on trail making test and disability on WHODAS. General psychiatry. 2020;33(6):e100237. Singh A, Beniwal RP, Bhatia T, Deshpande SN. Schizophrenia with and without obsessive-compulsive symptoms: a comparative analysis of performance on trail making test and disability on WHODAS. General psychiatry. 2020;33(6):e100237.
9.
Zurück zum Zitat Strom NI, Grove J, Meier SM, Bækvad-Hansen M, Nissen JB, Als TD, et al. Polygenic heterogeneity across obsessive-compulsive disorder subgroups defined by a comorbid diagnosis. Frontiers in Genetics. 2021;12:711624. Strom NI, Grove J, Meier SM, Bækvad-Hansen M, Nissen JB, Als TD, et al. Polygenic heterogeneity across obsessive-compulsive disorder subgroups defined by a comorbid diagnosis. Frontiers in Genetics. 2021;12:711624.
10.
Zurück zum Zitat Miguel EC, Ferrão YA, Rosário MCd, Mathis MAd, Torres AR, Fontenelle LF, et al. The Brazilian Research Consortium on Obsessive-Compulsive Spectrum Disorders: recruitment, assessment instruments, methods for the development of multicenter collaborative studies and preliminary results. Braz J Psychiatry. 2008;30(3):185–96.CrossRef Miguel EC, Ferrão YA, Rosário MCd, Mathis MAd, Torres AR, Fontenelle LF, et al. The Brazilian Research Consortium on Obsessive-Compulsive Spectrum Disorders: recruitment, assessment instruments, methods for the development of multicenter collaborative studies and preliminary results. Braz J Psychiatry. 2008;30(3):185–96.CrossRef
11.
Zurück zum Zitat Pinto A, Mancebo MC, Eisen JL, Pagano ME, Rasmussen SA. The Brown Longitudinal Obsessive Compulsive Study: clinical features and symptoms of the sample at intake. J Clin Psychiatry. 2006;67(5):2146.CrossRef Pinto A, Mancebo MC, Eisen JL, Pagano ME, Rasmussen SA. The Brown Longitudinal Obsessive Compulsive Study: clinical features and symptoms of the sample at intake. J Clin Psychiatry. 2006;67(5):2146.CrossRef
12.
Zurück zum Zitat van Oudheusden LJ, van de Schoot R, Hoogendoorn A, van Oppen P, Kaarsemaker M, Meynen G, et al. Classification of comorbidity in obsessive–compulsive disorder: A latent class analysis. Brain and behavior. 2020;10(7): e01641.CrossRef van Oudheusden LJ, van de Schoot R, Hoogendoorn A, van Oppen P, Kaarsemaker M, Meynen G, et al. Classification of comorbidity in obsessive–compulsive disorder: A latent class analysis. Brain and behavior. 2020;10(7): e01641.CrossRef
13.
Zurück zum Zitat Tezenas du Montcel C, Pelissolo A, Schürhoff F, Pignon B. Obsessive-compulsive symptoms in schizophrenia: an up-to-date review of literature. Curr Psychiatry Rep. 2019;21(8):64.CrossRef Tezenas du Montcel C, Pelissolo A, Schürhoff F, Pignon B. Obsessive-compulsive symptoms in schizophrenia: an up-to-date review of literature. Curr Psychiatry Rep. 2019;21(8):64.CrossRef
14.
Zurück zum Zitat Attademo L, Bernardini F, Paolini E, Quartesan R. History and conceptual problems of the relationship between obsessions and hallucinations. Harv Rev Psychiatry. 2015;23(1):19–27.CrossRef Attademo L, Bernardini F, Paolini E, Quartesan R. History and conceptual problems of the relationship between obsessions and hallucinations. Harv Rev Psychiatry. 2015;23(1):19–27.CrossRef
15.
Zurück zum Zitat Achim AM, Maziade M, Raymond É, Olivier D, Mérette C, Roy M-A. How prevalent are anxiety disorders in schizophrenia? A meta-analysis and critical review on a significant association. Schizophr Bull. 2011;37(4):811–21.CrossRef Achim AM, Maziade M, Raymond É, Olivier D, Mérette C, Roy M-A. How prevalent are anxiety disorders in schizophrenia? A meta-analysis and critical review on a significant association. Schizophr Bull. 2011;37(4):811–21.CrossRef
16.
Zurück zum Zitat Swets M, Dekker J, van Emmerik-van OK, Smid GE, Smit F, de Haan L, et al. The obsessive compulsive spectrum in schizophrenia, a meta-analysis and meta-regression exploring prevalence rates. Schizophr Res. 2014;152(2–3):458–68.CrossRef Swets M, Dekker J, van Emmerik-van OK, Smid GE, Smit F, de Haan L, et al. The obsessive compulsive spectrum in schizophrenia, a meta-analysis and meta-regression exploring prevalence rates. Schizophr Res. 2014;152(2–3):458–68.CrossRef
17.
Zurück zum Zitat Attademo L, De Giorgio G, Quartesan R, Moretti P. Schizophrenia and obsessive-compulsive disorder: from comorbidity to schizo-obsessive disorder. Riv Psichiatr. 2012;47(2):106–15. Attademo L, De Giorgio G, Quartesan R, Moretti P. Schizophrenia and obsessive-compulsive disorder: from comorbidity to schizo-obsessive disorder. Riv Psichiatr. 2012;47(2):106–15.
18.
Zurück zum Zitat Bottas A, Cooke RG, Richter MA. Comorbidity and pathophysiology of obsessive–compulsive disorder in schizophrenia: is there evidence for a schizo-obsessive subtype of schizophrenia? J Psychiatry Neurosci. 2005;30(3):187–95. Bottas A, Cooke RG, Richter MA. Comorbidity and pathophysiology of obsessive–compulsive disorder in schizophrenia: is there evidence for a schizo-obsessive subtype of schizophrenia? J Psychiatry Neurosci. 2005;30(3):187–95.
19.
Zurück zum Zitat Poyurovsky M, Weizman A, Weizman R. Obsessive-compulsive disorder in schizophrenia. CNS Drugs. 2004;18(14):989–1010.CrossRef Poyurovsky M, Weizman A, Weizman R. Obsessive-compulsive disorder in schizophrenia. CNS Drugs. 2004;18(14):989–1010.CrossRef
20.
Zurück zum Zitat Schirmbeck F, Zink M. Comorbid obsessive-compulsive symptoms in schizophrenia: contributions of pharmacological and genetic factors. Front Pharmacol. 2013;4:99.CrossRef Schirmbeck F, Zink M. Comorbid obsessive-compulsive symptoms in schizophrenia: contributions of pharmacological and genetic factors. Front Pharmacol. 2013;4:99.CrossRef
21.
Zurück zum Zitat Costas J, Carrera N, Alonso P, Gurriarán X, Segalàs C, Real E, et al. Exon-focused genome-wide association study of obsessive-compulsive disorder and shared polygenic risk with schizophrenia. Translational psychiatry. 2016;6(3):e768.CrossRef Costas J, Carrera N, Alonso P, Gurriarán X, Segalàs C, Real E, et al. Exon-focused genome-wide association study of obsessive-compulsive disorder and shared polygenic risk with schizophrenia. Translational psychiatry. 2016;6(3):e768.CrossRef
22.
Zurück zum Zitat Zink M. Comorbid obsessive-compulsive symptoms in schizophrenia: insight into pathomechanisms facilitates treatment. Advances in Medicine. 2014;2014:317980. Zink M. Comorbid obsessive-compulsive symptoms in schizophrenia: insight into pathomechanisms facilitates treatment. Advances in Medicine. 2014;2014:317980.
23.
Zurück zum Zitat Kropiwnicki E, Binder JL, Yang JJ, Holmes J, Lachmann A, Clarke DJ, et al. Getting Started with the IDG KMC Datasets and Tools. Current Protocols. 2022;2(1): e355.CrossRef Kropiwnicki E, Binder JL, Yang JJ, Holmes J, Lachmann A, Clarke DJ, et al. Getting Started with the IDG KMC Datasets and Tools. Current Protocols. 2022;2(1): e355.CrossRef
24.
Zurück zum Zitat Sepehrinezhad A, Rezaeitalab F, Shahbazi A, Sahab-Negah S. A computational-based drug repurposing method targeting SARS-CoV-2 and its neurological manifestations genes and signaling pathways. Bioinform Biol Insights. 2021;15:11779322211026728.CrossRef Sepehrinezhad A, Rezaeitalab F, Shahbazi A, Sahab-Negah S. A computational-based drug repurposing method targeting SARS-CoV-2 and its neurological manifestations genes and signaling pathways. Bioinform Biol Insights. 2021;15:11779322211026728.CrossRef
25.
Zurück zum Zitat Baker EJ, Jay JJ, Bubier JA, Langston MA, Chesler EJ. GeneWeaver: a web-based system for integrative functional genomics. Nucleic Acids Res. 2012;40(D1):D1067–76.CrossRef Baker EJ, Jay JJ, Bubier JA, Langston MA, Chesler EJ. GeneWeaver: a web-based system for integrative functional genomics. Nucleic Acids Res. 2012;40(D1):D1067–76.CrossRef
26.
Zurück zum Zitat Rouillard AD, Gundersen GW, Fernandez NF, Wang Z, Monteiro CD, McDermott MG, et al. The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database: the journal of biological databases and curation. 2016;2016:baw100: 1-16. Rouillard AD, Gundersen GW, Fernandez NF, Wang Z, Monteiro CD, McDermott MG, et al. The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database: the journal of biological databases and curation. 2016;2016:baw100: 1-16.
27.
Zurück zum Zitat Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–12.CrossRef Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–12.CrossRef
28.
Zurück zum Zitat Ragueneau E, Shrivastava A, Morris JH, Del-Toro N, Hermjakob H, Porras P. IntAct App: a Cytoscape application for molecular interaction network visualization and analysis. Bioinformatics. 2021;37(20):3684–5.CrossRef Ragueneau E, Shrivastava A, Morris JH, Del-Toro N, Hermjakob H, Porras P. IntAct App: a Cytoscape application for molecular interaction network visualization and analysis. Bioinformatics. 2021;37(20):3684–5.CrossRef
29.
Zurück zum Zitat Weng S, Wang B, Li M, Chao S, Lin R, Zheng R, et al. Genome-wide association study of antipsychotic-induced sinus bradycardia in Chinese schizophrenia patients. PLoS ONE. 2021;16(4): e0249997.CrossRef Weng S, Wang B, Li M, Chao S, Lin R, Zheng R, et al. Genome-wide association study of antipsychotic-induced sinus bradycardia in Chinese schizophrenia patients. PLoS ONE. 2021;16(4): e0249997.CrossRef
30.
Zurück zum Zitat Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.CrossRef Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.CrossRef
31.
Zurück zum Zitat Xie Z, Bailey A, Kuleshov MV, Clarke DJ, Evangelista JE, Jenkins SL, et al. Gene set knowledge discovery with enrichr. Current protocols. 2021;1(3): e90.CrossRef Xie Z, Bailey A, Kuleshov MV, Clarke DJ, Evangelista JE, Jenkins SL, et al. Gene set knowledge discovery with enrichr. Current protocols. 2021;1(3): e90.CrossRef
32.
Zurück zum Zitat Wan J, Liu B. Construction of lncRNA-related ceRNA regulatory network in diabetic subdermal endothelial cells. Bioengineered. 2021;12(1):2592–602.CrossRef Wan J, Liu B. Construction of lncRNA-related ceRNA regulatory network in diabetic subdermal endothelial cells. Bioengineered. 2021;12(1):2592–602.CrossRef
33.
Zurück zum Zitat Dincheva I, Lynch NB, Lee FS. The role of BDNF in the development of fear learning. Depress Anxiety. 2016;33(10):907–16.CrossRef Dincheva I, Lynch NB, Lee FS. The role of BDNF in the development of fear learning. Depress Anxiety. 2016;33(10):907–16.CrossRef
34.
Zurück zum Zitat Cattaneo A, Cattane N, Begni V, Pariante CM, Riva M. The human BDNF gene: peripheral gene expression and protein levels as biomarkers for psychiatric disorders. Translational psychiatry. 2016;6(11):e958.CrossRef Cattaneo A, Cattane N, Begni V, Pariante CM, Riva M. The human BDNF gene: peripheral gene expression and protein levels as biomarkers for psychiatric disorders. Translational psychiatry. 2016;6(11):e958.CrossRef
35.
Zurück zum Zitat Lu B, Martinowich K, editors. Cell biology of BDNF and its relevance to schizophrenia. Novartis Foundation Symposium; 2008:289:119-29; discussion 129-35, 193-5; NIH Public Access. Lu B, Martinowich K, editors. Cell biology of BDNF and its relevance to schizophrenia. Novartis Foundation Symposium; 2008:289:119-29; discussion 129-35, 193-5; NIH Public Access.
36.
Zurück zum Zitat Nieto R, Kukuljan M, Silva H. BDNF and schizophrenia: from neurodevelopment to neuronal plasticity, learning, and memory. Front Psych. 2013;4:45. Nieto R, Kukuljan M, Silva H. BDNF and schizophrenia: from neurodevelopment to neuronal plasticity, learning, and memory. Front Psych. 2013;4:45.
37.
Zurück zum Zitat Di Carlo P, Punzi G, Ursini G. BDNF and Schizophrenia. Psychiatr Genet. 2019;29(5):200. Di Carlo P, Punzi G, Ursini G. BDNF and Schizophrenia. Psychiatr Genet. 2019;29(5):200.
38.
Zurück zum Zitat Krebs M, Guillin O, Bourdel M, Schwartz J, Olie J, Poirier M, et al. Brain derived neurotrophic factor (BDNF) gene variants association with age at onset and therapeutic response in schizophrenia. Mol Psychiatry. 2000;5(5):558–62.CrossRef Krebs M, Guillin O, Bourdel M, Schwartz J, Olie J, Poirier M, et al. Brain derived neurotrophic factor (BDNF) gene variants association with age at onset and therapeutic response in schizophrenia. Mol Psychiatry. 2000;5(5):558–62.CrossRef
39.
Zurück zum Zitat Martinowich K, Lu B. Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology. 2008;33(1):73–83.CrossRef Martinowich K, Lu B. Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology. 2008;33(1):73–83.CrossRef
40.
Zurück zum Zitat BruÈnig I, Penschuck S, Berninger B, Benson J, Fritschy JM. BDNF reduces miniature inhibitory postsynaptic currents by rapid downregulation of GABAA receptor surface expression. Eur J Neurosci. 2001;13(7):1320–8.CrossRef BruÈnig I, Penschuck S, Berninger B, Benson J, Fritschy JM. BDNF reduces miniature inhibitory postsynaptic currents by rapid downregulation of GABAA receptor surface expression. Eur J Neurosci. 2001;13(7):1320–8.CrossRef
41.
Zurück zum Zitat Buckley PF, Mahadik S, Pillai A, Terry A Jr. Neurotrophins and schizophrenia. Schizophr Res. 2007;94(1–3):1–11. Buckley PF, Mahadik S, Pillai A, Terry A Jr. Neurotrophins and schizophrenia. Schizophr Res. 2007;94(1–3):1–11.
42.
Zurück zum Zitat Hall D, Dhilla A, Charalambous A, Gogos JA, Karayiorgou M. Sequence variants of the brain-derived neurotrophic factor (BDNF) gene are strongly associated with obsessive-compulsive disorder. The American Journal of Human Genetics. 2003;73(2):370–6.CrossRef Hall D, Dhilla A, Charalambous A, Gogos JA, Karayiorgou M. Sequence variants of the brain-derived neurotrophic factor (BDNF) gene are strongly associated with obsessive-compulsive disorder. The American Journal of Human Genetics. 2003;73(2):370–6.CrossRef
43.
Zurück zum Zitat Hemmings SM, Kinnear CJ, Van Der Merwe L, Lochner C, Corfield VA, Moolman-Smook JC, et al. Investigating the role of the brain-derived neurotrophic factor (BDNF) val66met variant in obsessive-compulsive disorder (OCD). The World Journal of Biological Psychiatry. 2008;9(2):126–34.CrossRef Hemmings SM, Kinnear CJ, Van Der Merwe L, Lochner C, Corfield VA, Moolman-Smook JC, et al. Investigating the role of the brain-derived neurotrophic factor (BDNF) val66met variant in obsessive-compulsive disorder (OCD). The World Journal of Biological Psychiatry. 2008;9(2):126–34.CrossRef
44.
Zurück zum Zitat Katerberg H, Lochner C, Cath DC, de Jonge P, Bochdanovits Z, Moolman-Smook JC, et al. The role of the brain-derived neurotrophic factor (BDNF) val66met variant in the phenotypic expression of obsessive-compulsive disorder (OCD). Am J Med Genet B Neuropsychiatr Genet. 2009;150(8):1050–62.CrossRef Katerberg H, Lochner C, Cath DC, de Jonge P, Bochdanovits Z, Moolman-Smook JC, et al. The role of the brain-derived neurotrophic factor (BDNF) val66met variant in the phenotypic expression of obsessive-compulsive disorder (OCD). Am J Med Genet B Neuropsychiatr Genet. 2009;150(8):1050–62.CrossRef
45.
Zurück zum Zitat Wang S, Xu X, Yan P, Song M, Li J, Wang S. Is Brain-Derived Neurotrophic Factor (BDNF) Val66met polymorphism associated with obsessive-compulsive disorder? A meta-analysis Psychiatria Danubina. 2019;31(2):141–7.CrossRef Wang S, Xu X, Yan P, Song M, Li J, Wang S. Is Brain-Derived Neurotrophic Factor (BDNF) Val66met polymorphism associated with obsessive-compulsive disorder? A meta-analysis Psychiatria Danubina. 2019;31(2):141–7.CrossRef
46.
Zurück zum Zitat Ghamari R, Yazarlou F, Khosravizadeh Z, Moradkhani A, Abdollahi E, Alizadeh F. Serotonin transporter functional polymorphisms potentially increase risk of schizophrenia separately and as a haplotype. Sci Rep. 2022;12(1):1–9.CrossRef Ghamari R, Yazarlou F, Khosravizadeh Z, Moradkhani A, Abdollahi E, Alizadeh F. Serotonin transporter functional polymorphisms potentially increase risk of schizophrenia separately and as a haplotype. Sci Rep. 2022;12(1):1–9.CrossRef
47.
Zurück zum Zitat Xu F-l, Wang B-j, Yao J. Association between the SLC6A4 gene and schizophrenia: an updated meta-analysis. Neuropsychiatric Disease and Treatment. 2018;15:143-155. Xu F-l, Wang B-j, Yao J. Association between the SLC6A4 gene and schizophrenia: an updated meta-analysis. Neuropsychiatric Disease and Treatment. 2018;15:143-155.
48.
Zurück zum Zitat Kim J-H, Kim J-H, Son Y-D, Joo Y-H, Lee S-Y, Kim H-K, et al. Altered interregional correlations between serotonin transporter availability and cerebral glucose metabolism in schizophrenia: a high-resolution PET study using [11C] DASB and [18F] FDG. Schizophr Res. 2017;182:55–65.CrossRef Kim J-H, Kim J-H, Son Y-D, Joo Y-H, Lee S-Y, Kim H-K, et al. Altered interregional correlations between serotonin transporter availability and cerebral glucose metabolism in schizophrenia: a high-resolution PET study using [11C] DASB and [18F] FDG. Schizophr Res. 2017;182:55–65.CrossRef
49.
Zurück zum Zitat Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res. 2018;265:25–38.CrossRef Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res. 2018;265:25–38.CrossRef
50.
Zurück zum Zitat Gomes CKF, Vieira-Fonseca T, Melo-Felippe FB, de Salles Andrade JB, Fontenelle LF, Kohlrausch FB. Association analysis of SLC6A4 and HTR2A genes with obsessive-compulsive disorder: Influence of the STin2 polymorphism. Compr Psychiatry. 2018;82:1–6.CrossRef Gomes CKF, Vieira-Fonseca T, Melo-Felippe FB, de Salles Andrade JB, Fontenelle LF, Kohlrausch FB. Association analysis of SLC6A4 and HTR2A genes with obsessive-compulsive disorder: Influence of the STin2 polymorphism. Compr Psychiatry. 2018;82:1–6.CrossRef
51.
Zurück zum Zitat Zitterl W, Aigner M, Stompe T, Zitterl-Eglseer K, Gutierrez-Lobos K, Wenzel T, et al. Changes in thalamus–hypothalamus serotonin transporter availability during clomipramine administration in patients with obsessive–compulsive disorder. Neuropsychopharmacology. 2008;33(13):3126–34.CrossRef Zitterl W, Aigner M, Stompe T, Zitterl-Eglseer K, Gutierrez-Lobos K, Wenzel T, et al. Changes in thalamus–hypothalamus serotonin transporter availability during clomipramine administration in patients with obsessive–compulsive disorder. Neuropsychopharmacology. 2008;33(13):3126–34.CrossRef
52.
Zurück zum Zitat Rashidi FS, Ahmadipour E, Shiravand S, Ahmadiani A, Asadi S, Shams J. Association of the functional serotonin transporter haplotype with familial form of obsessive compulsive disorder in Iranian patients. Int J Psychiatry Clin Pract. 2018;22(1):47–53.CrossRef Rashidi FS, Ahmadipour E, Shiravand S, Ahmadiani A, Asadi S, Shams J. Association of the functional serotonin transporter haplotype with familial form of obsessive compulsive disorder in Iranian patients. Int J Psychiatry Clin Pract. 2018;22(1):47–53.CrossRef
53.
Zurück zum Zitat Kim Y-K, Yoon H-K. Effect of serotonin-related gene polymorphisms on pathogenesis and treatment response in Korean schizophrenic patients. Behav Genet. 2011;41(5):709–15.CrossRef Kim Y-K, Yoon H-K. Effect of serotonin-related gene polymorphisms on pathogenesis and treatment response in Korean schizophrenic patients. Behav Genet. 2011;41(5):709–15.CrossRef
54.
Zurück zum Zitat Kantrowitz JT. Targeting serotonin 5-HT2A receptors to better treat schizophrenia: rationale and current approaches. CNS Drugs. 2020;34(9):947–59.CrossRef Kantrowitz JT. Targeting serotonin 5-HT2A receptors to better treat schizophrenia: rationale and current approaches. CNS Drugs. 2020;34(9):947–59.CrossRef
55.
Zurück zum Zitat Sinopoli VM, Burton CL, Kronenberg S, Arnold PD. A review of the role of serotonin system genes in obsessive-compulsive disorder. Neurosci Biobehav Rev. 2017;80:372–81.CrossRef Sinopoli VM, Burton CL, Kronenberg S, Arnold PD. A review of the role of serotonin system genes in obsessive-compulsive disorder. Neurosci Biobehav Rev. 2017;80:372–81.CrossRef
56.
Zurück zum Zitat Mattina GF, Samaan Z, Hall GB, Steiner M. The association of HTR2A polymorphisms with obsessive-compulsive disorder and its subtypes: a meta-analysis. J Affect Disord. 2020;275:278–89.CrossRef Mattina GF, Samaan Z, Hall GB, Steiner M. The association of HTR2A polymorphisms with obsessive-compulsive disorder and its subtypes: a meta-analysis. J Affect Disord. 2020;275:278–89.CrossRef
57.
Zurück zum Zitat Taylor S. Molecular genetics of obsessive–compulsive disorder: a comprehensive meta-analysis of genetic association studies. Mol Psychiatry. 2013;18(7):799–805.CrossRef Taylor S. Molecular genetics of obsessive–compulsive disorder: a comprehensive meta-analysis of genetic association studies. Mol Psychiatry. 2013;18(7):799–805.CrossRef
58.
Zurück zum Zitat Yee JY, Nurjono M, Teo SR, Lee T-S, Lee J. GAD1 gene expression in blood of patients with first-episode psychosis. PLoS ONE. 2017;12(1): e0170805.CrossRef Yee JY, Nurjono M, Teo SR, Lee T-S, Lee J. GAD1 gene expression in blood of patients with first-episode psychosis. PLoS ONE. 2017;12(1): e0170805.CrossRef
59.
Zurück zum Zitat Curley AA, Eggan SM, Lazarus MS, Huang ZJ, Volk DW, Lewis DA. Role of glutamic acid decarboxylase 67 in regulating cortical parvalbumin and GABA membrane transporter 1 expression: implications for schizophrenia. Neurobiol Dis. 2013;50:179–86.CrossRef Curley AA, Eggan SM, Lazarus MS, Huang ZJ, Volk DW, Lewis DA. Role of glutamic acid decarboxylase 67 in regulating cortical parvalbumin and GABA membrane transporter 1 expression: implications for schizophrenia. Neurobiol Dis. 2013;50:179–86.CrossRef
60.
Zurück zum Zitat Li Y, Zhang CC, Weidacker K, Zhang Y, He N, Jin H, et al. Investigation of anterior cingulate cortex gamma-aminobutyric acid and glutamate-glutamine levels in obsessive-compulsive disorder using magnetic resonance spectroscopy. BMC Psychiatry. 2019;19(1):1–9.CrossRef Li Y, Zhang CC, Weidacker K, Zhang Y, He N, Jin H, et al. Investigation of anterior cingulate cortex gamma-aminobutyric acid and glutamate-glutamine levels in obsessive-compulsive disorder using magnetic resonance spectroscopy. BMC Psychiatry. 2019;19(1):1–9.CrossRef
61.
Zurück zum Zitat Zhang Z, Fan Q, Bai Y, Wang Z, Zhang H, Xiao Z. Brain Gamma-Aminobutyric Acid (GABA) concentration of the prefrontal lobe in unmedicated patients with obsessive-compulsive disorder: a research of magnetic resonance spectroscopy. Shanghai Arch Psychiatry. 2016;28(5):263. Zhang Z, Fan Q, Bai Y, Wang Z, Zhang H, Xiao Z. Brain Gamma-Aminobutyric Acid (GABA) concentration of the prefrontal lobe in unmedicated patients with obsessive-compulsive disorder: a research of magnetic resonance spectroscopy. Shanghai Arch Psychiatry. 2016;28(5):263.
62.
Zurück zum Zitat Hu C, Chen W, Myers SJ, Yuan H, Traynelis SF. Human GRIN2B variants in neurodevelopmental disorders. J Pharmacol Sci. 2016;132(2):115–21.CrossRef Hu C, Chen W, Myers SJ, Yuan H, Traynelis SF. Human GRIN2B variants in neurodevelopmental disorders. J Pharmacol Sci. 2016;132(2):115–21.CrossRef
63.
Zurück zum Zitat Guo Z, Niu W, Bi Y, Zhang R, Ren D, Hu J, et al. A study of single nucleotide polymorphisms of GRIN2B in schizophrenia from Chinese Han population. Neurosci Lett. 2016;630:132–5.CrossRef Guo Z, Niu W, Bi Y, Zhang R, Ren D, Hu J, et al. A study of single nucleotide polymorphisms of GRIN2B in schizophrenia from Chinese Han population. Neurosci Lett. 2016;630:132–5.CrossRef
64.
Zurück zum Zitat Uno Y, Coyle JT. Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci. 2019;73(5):204–15.CrossRef Uno Y, Coyle JT. Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci. 2019;73(5):204–15.CrossRef
65.
Zurück zum Zitat Poltavskaya EG, Fedorenko OY, Kornetova EG, Loonen AJ, Kornetov AN, Bokhan NA, et al. Study of early onset schizophrenia: Associations of GRIN2A and GRIN2B polymorphisms. Life. 2021;11(10):997.CrossRef Poltavskaya EG, Fedorenko OY, Kornetova EG, Loonen AJ, Kornetov AN, Bokhan NA, et al. Study of early onset schizophrenia: Associations of GRIN2A and GRIN2B polymorphisms. Life. 2021;11(10):997.CrossRef
66.
Zurück zum Zitat Ting JT, Feng G. Glutamatergic synaptic dysfunction and obsessive-compulsive disorder. Current chemical genomics. 2008;2:62.CrossRef Ting JT, Feng G. Glutamatergic synaptic dysfunction and obsessive-compulsive disorder. Current chemical genomics. 2008;2:62.CrossRef
67.
Zurück zum Zitat Saxena S, Rauch SL. Functional neuroimaging and the neuroanatomy of obsessive-compulsive disorder. Psychiatr Clin North Am. 2000;23(3):563–86.CrossRef Saxena S, Rauch SL. Functional neuroimaging and the neuroanatomy of obsessive-compulsive disorder. Psychiatr Clin North Am. 2000;23(3):563–86.CrossRef
68.
Zurück zum Zitat Stewart S, Mayerfeld C, Arnold P, Crane J, O’dushlaine C, Fagerness J, et al. Meta-analysis of association between obsessive-compulsive disorder and the 3′ region of neuronal glutamate transporter gene SLC1A1. Am J Med Genet B Neuropsychiatr Genet. 2013;162(4):367–79.CrossRef Stewart S, Mayerfeld C, Arnold P, Crane J, O’dushlaine C, Fagerness J, et al. Meta-analysis of association between obsessive-compulsive disorder and the 3′ region of neuronal glutamate transporter gene SLC1A1. Am J Med Genet B Neuropsychiatr Genet. 2013;162(4):367–79.CrossRef
69.
Zurück zum Zitat Alonso P, Gratacós M, Segalàs C, Escaramís G, Real E, Bayés M, et al. Association between the NMDA glutamate receptor GRIN2B gene and obsessive–compulsive disorder. J Psychiatry Neurosci. 2012;37(4):273–81.CrossRef Alonso P, Gratacós M, Segalàs C, Escaramís G, Real E, Bayés M, et al. Association between the NMDA glutamate receptor GRIN2B gene and obsessive–compulsive disorder. J Psychiatry Neurosci. 2012;37(4):273–81.CrossRef
70.
Zurück zum Zitat Hussain MS, Siddiqui SA, Mondal S, Millat MS, Marzan S, Uddin MG, et al. Association of DRD2 gene polymorphisms with schizophrenia in the young Bangladeshi population: A pilot study. Heliyon. 2020;6(10): e05125.CrossRef Hussain MS, Siddiqui SA, Mondal S, Millat MS, Marzan S, Uddin MG, et al. Association of DRD2 gene polymorphisms with schizophrenia in the young Bangladeshi population: A pilot study. Heliyon. 2020;6(10): e05125.CrossRef
71.
Zurück zum Zitat Prananjaya A, Saleh I, Parisa N. DRD2 Gene Polymorphisms in Schizophrenia Patients. 2021. Prananjaya A, Saleh I, Parisa N. DRD2 Gene Polymorphisms in Schizophrenia Patients. 2021.
72.
Zurück zum Zitat Koo M-S, Kim E-J, Roh D, Kim C-H. Role of dopamine in the pathophysiology and treatment of obsessive–compulsive disorder. Expert Rev Neurother. 2010;10(2):275–90.CrossRef Koo M-S, Kim E-J, Roh D, Kim C-H. Role of dopamine in the pathophysiology and treatment of obsessive–compulsive disorder. Expert Rev Neurother. 2010;10(2):275–90.CrossRef
73.
Zurück zum Zitat Denys D, Van Nieuwerburgh F, Deforce D, Westenberg H. Association between the dopamine D2 receptor TaqI A2 allele and low activity COMT allele with obsessive–compulsive disorder in males. Eur Neuropsychopharmacol. 2006;16(6):446–50.CrossRef Denys D, Van Nieuwerburgh F, Deforce D, Westenberg H. Association between the dopamine D2 receptor TaqI A2 allele and low activity COMT allele with obsessive–compulsive disorder in males. Eur Neuropsychopharmacol. 2006;16(6):446–50.CrossRef
74.
Zurück zum Zitat Xu FI, Ding M, Wu X, Liu Yp, Xia X, Yao J, et al. A meta-analysis of the association between SLC6A3 gene polymorphisms and schizophrenia. J Mol Neurosci. 2020;70(2):155–66.CrossRef Xu FI, Ding M, Wu X, Liu Yp, Xia X, Yao J, et al. A meta-analysis of the association between SLC6A3 gene polymorphisms and schizophrenia. J Mol Neurosci. 2020;70(2):155–66.CrossRef
75.
Zurück zum Zitat Warshaneyan SS, Srivastava P. Analysis and subsequent molecular docking of selected phytochemicals with SLC6A3 and SLC6A4 as potential therapeutic agents for Obsessive-Compulsive Disorder (OCD). bioRxiv. 2019:776922. https://doi.org/10.1101/776922. Warshaneyan SS, Srivastava P. Analysis and subsequent molecular docking of selected phytochemicals with SLC6A3 and SLC6A4 as potential therapeutic agents for Obsessive-Compulsive Disorder (OCD). bioRxiv. 2019:776922. https://​doi.​org/​10.​1101/​776922.
76.
Zurück zum Zitat Escamilla R, Camarena B, Saracco-Alvarez R, Fresán A, Hernández S, Aguilar-García A. Association study between COMT, DRD2, and DRD3 gene variants and antipsychotic treatment response in Mexican patients with schizophrenia. Neuropsychiatr Dis Treat. 2018;14:2981.CrossRef Escamilla R, Camarena B, Saracco-Alvarez R, Fresán A, Hernández S, Aguilar-García A. Association study between COMT, DRD2, and DRD3 gene variants and antipsychotic treatment response in Mexican patients with schizophrenia. Neuropsychiatr Dis Treat. 2018;14:2981.CrossRef
77.
Zurück zum Zitat Kumar P, Rai V. Catechol-O-methyltransferase gene Val158Met polymorphism and obsessive compulsive disorder susceptibility: a meta-analysis. Metab Brain Dis. 2020;35(2):241–51.CrossRef Kumar P, Rai V. Catechol-O-methyltransferase gene Val158Met polymorphism and obsessive compulsive disorder susceptibility: a meta-analysis. Metab Brain Dis. 2020;35(2):241–51.CrossRef
78.
Zurück zum Zitat Horiguchi M, Ohi K, Hashimoto R, Hao Q, Yasuda Y, Yamamori H, et al. Functional polymorphism (C-824T) of the tyrosine hydroxylase gene affects IQ in schizophrenia. Psychiatry Clin Neurosci. 2014;68(6):456–62.CrossRef Horiguchi M, Ohi K, Hashimoto R, Hao Q, Yasuda Y, Yamamori H, et al. Functional polymorphism (C-824T) of the tyrosine hydroxylase gene affects IQ in schizophrenia. Psychiatry Clin Neurosci. 2014;68(6):456–62.CrossRef
79.
Zurück zum Zitat Cheng M-C, Lu C-L, Luu S-U, Tsai H-M, Hsu S-H, Chen T-T, et al. Genetic and functional analysis of the DLG4 gene encoding the post-synaptic density protein 95 in schizophrenia. PLoS ONE. 2010;5(12): e15107.CrossRef Cheng M-C, Lu C-L, Luu S-U, Tsai H-M, Hsu S-H, Chen T-T, et al. Genetic and functional analysis of the DLG4 gene encoding the post-synaptic density protein 95 in schizophrenia. PLoS ONE. 2010;5(12): e15107.CrossRef
80.
Zurück zum Zitat Hibar DP, Cheung JW, Medland SE, Mufford MS, Jahanshad N, Dalvie S, et al. Significant concordance of genetic variation that increases both the risk for obsessive–compulsive disorder and the volumes of the nucleus accumbens and putamen. Br J Psychiatry. 2018;213(1):430–6.CrossRef Hibar DP, Cheung JW, Medland SE, Mufford MS, Jahanshad N, Dalvie S, et al. Significant concordance of genetic variation that increases both the risk for obsessive–compulsive disorder and the volumes of the nucleus accumbens and putamen. Br J Psychiatry. 2018;213(1):430–6.CrossRef
81.
Zurück zum Zitat Stahl SM. Beyond the dopamine hypothesis of schizophrenia to three neural networks of psychosis: dopamine, serotonin, and glutamate. CNS Spectr. 2018;23(3):187–91.CrossRef Stahl SM. Beyond the dopamine hypothesis of schizophrenia to three neural networks of psychosis: dopamine, serotonin, and glutamate. CNS Spectr. 2018;23(3):187–91.CrossRef
82.
Zurück zum Zitat Asl MA, Asgari P, Bakhti Z. Treatment approaches based on neuroscientific data in patients with obsessive-compulsive disorder. International Clinical Neuroscience Journal. 2021;8(3):107–17.CrossRef Asl MA, Asgari P, Bakhti Z. Treatment approaches based on neuroscientific data in patients with obsessive-compulsive disorder. International Clinical Neuroscience Journal. 2021;8(3):107–17.CrossRef
83.
Zurück zum Zitat Perani D, Garibotto V, Gorini A, Moresco RM, Henin M, Panzacchi A, et al. In vivo PET study of 5HT2A serotonin and D2 dopamine dysfunction in drug-naive obsessive-compulsive disorder. Neuroimage. 2008;42(1):306–14.CrossRef Perani D, Garibotto V, Gorini A, Moresco RM, Henin M, Panzacchi A, et al. In vivo PET study of 5HT2A serotonin and D2 dopamine dysfunction in drug-naive obsessive-compulsive disorder. Neuroimage. 2008;42(1):306–14.CrossRef
84.
Zurück zum Zitat Vermeire S, Audenaert K, De Meester R, Vandermeulen E, Waelbers T, De Spiegeleer B, et al. Serotonin 2A receptor, serotonin transporter and dopamine transporter alterations in dogs with compulsive behaviour as a promising model for human obsessive-compulsive disorder. Psychiatry Research: Neuroimaging. 2012;201(1):78–87.CrossRef Vermeire S, Audenaert K, De Meester R, Vandermeulen E, Waelbers T, De Spiegeleer B, et al. Serotonin 2A receptor, serotonin transporter and dopamine transporter alterations in dogs with compulsive behaviour as a promising model for human obsessive-compulsive disorder. Psychiatry Research: Neuroimaging. 2012;201(1):78–87.CrossRef
85.
Zurück zum Zitat Rosoklija G, Toomayan G, Ellis SP, Keilp J, Mann JJ, Latov N, et al. Structural abnormalities of subicular dendrites in subjects with schizophrenia and mood disorders: preliminary findings. Arch Gen Psychiatry. 2000;57(4):349–56.CrossRef Rosoklija G, Toomayan G, Ellis SP, Keilp J, Mann JJ, Latov N, et al. Structural abnormalities of subicular dendrites in subjects with schizophrenia and mood disorders: preliminary findings. Arch Gen Psychiatry. 2000;57(4):349–56.CrossRef
86.
Zurück zum Zitat Faludi G, Mirnics K. Synaptic changes in the brain of subjects with schizophrenia. Int J Dev Neurosci. 2011;29(3):305–9.CrossRef Faludi G, Mirnics K. Synaptic changes in the brain of subjects with schizophrenia. Int J Dev Neurosci. 2011;29(3):305–9.CrossRef
88.
Zurück zum Zitat Nowak JS, Michlewski G. miRNAs in development and pathogenesis of the nervous system. Biochemical Society transactions. 2013;41(4):815–820. Nowak JS, Michlewski G. miRNAs in development and pathogenesis of the nervous system. Biochemical Society transactions. 2013;41(4):815–820.
89.
Zurück zum Zitat Wang J, Wang Y, Yang J, Huang Y. microRNAs as novel biomarkers of schizophrenia. Exp Ther Med. 2014;8(6):1671–6.CrossRef Wang J, Wang Y, Yang J, Huang Y. microRNAs as novel biomarkers of schizophrenia. Exp Ther Med. 2014;8(6):1671–6.CrossRef
90.
Zurück zum Zitat He K, Guo C, He L, Shi Y. MiRNAs of peripheral blood as the biomarker of schizophrenia. Hereditas. 2018;155(1):1–5.CrossRef He K, Guo C, He L, Shi Y. MiRNAs of peripheral blood as the biomarker of schizophrenia. Hereditas. 2018;155(1):1–5.CrossRef
91.
Zurück zum Zitat Pan B, Wang Y, Shi Y, Yang Q, Han B, Zhu X, et al. Altered expression levels of miR-144–3p and ATP1B2 are associated with schizophrenia. The world journal of biological psychiatry : the official journal of the World Federation of Societies of Biological Psychiatry. 2022:23(9):666-676. Pan B, Wang Y, Shi Y, Yang Q, Han B, Zhu X, et al. Altered expression levels of miR-144–3p and ATP1B2 are associated with schizophrenia. The world journal of biological psychiatry : the official journal of the World Federation of Societies of Biological Psychiatry. 2022:23(9):666-676.
92.
Zurück zum Zitat Xu M, Zhou J, Zhang Q, Le K, Xi Z, Yi P, et al. MiR-3121-3p promotes tumor invasion and metastasis by suppressing Rap1GAP in papillary thyroid cancer in vitro. Annals of translational medicine. 2020;8(19):1229.CrossRef Xu M, Zhou J, Zhang Q, Le K, Xi Z, Yi P, et al. MiR-3121-3p promotes tumor invasion and metastasis by suppressing Rap1GAP in papillary thyroid cancer in vitro. Annals of translational medicine. 2020;8(19):1229.CrossRef
93.
Zurück zum Zitat Zhao XF, Kohen R, Parent R, Duan Y, Fisher GL, Korn MJ, et al. PlexinA2 Forward Signaling through Rap1 GTPases Regulates Dentate Gyrus Development and Schizophrenia-like Behaviors. Cell Rep. 2018;22(2):456–70.CrossRef Zhao XF, Kohen R, Parent R, Duan Y, Fisher GL, Korn MJ, et al. PlexinA2 Forward Signaling through Rap1 GTPases Regulates Dentate Gyrus Development and Schizophrenia-like Behaviors. Cell Rep. 2018;22(2):456–70.CrossRef
94.
Zurück zum Zitat Ma J, Shang S, Wang J, Zhang T, Nie F, Song X, et al. Identification of miR-22-3p, miR-92a-3p, and miR-137 in peripheral blood as biomarker for schizophrenia. Psychiatry Res. 2018;265:70–6.CrossRef Ma J, Shang S, Wang J, Zhang T, Nie F, Song X, et al. Identification of miR-22-3p, miR-92a-3p, and miR-137 in peripheral blood as biomarker for schizophrenia. Psychiatry Res. 2018;265:70–6.CrossRef
95.
Zurück zum Zitat Kandemir H, Erdal ME, Selek S, İzci Ay Ö, Karababa İF, Ay ME, et al. Microribonucleic acid dysregulations in children and adolescents with obsessive-compulsive disorder. Neuropsychiatr Dis Treat. 2015;11:1695–701.CrossRef Kandemir H, Erdal ME, Selek S, İzci Ay Ö, Karababa İF, Ay ME, et al. Microribonucleic acid dysregulations in children and adolescents with obsessive-compulsive disorder. Neuropsychiatr Dis Treat. 2015;11:1695–701.CrossRef
96.
Zurück zum Zitat Sabaie H, Gholipour M, Asadi MR, Abed S, Sharifi-Bonab M, Taheri M, et al. Identification of key long non-coding RNA-associated competing endogenous RNA axes in Brodmann Area 10 brain region of schizophrenia patients. Front Psychiatry. 2022;13:1010977.CrossRef Sabaie H, Gholipour M, Asadi MR, Abed S, Sharifi-Bonab M, Taheri M, et al. Identification of key long non-coding RNA-associated competing endogenous RNA axes in Brodmann Area 10 brain region of schizophrenia patients. Front Psychiatry. 2022;13:1010977.CrossRef
97.
Zurück zum Zitat Aydın EP, Alsaadoni H, Beğenen AG, Özer ÖA, Karamustafalıoğlu KO, Pençe S. Can miRNA Expression Levels Predict Treatment Resistance to Serotonin Reuptake Inhibitors in Patients with Obsessive-Compulsive Disorder? PSYCHIATRY AND CLINICAL PSYCHOPHARMACOLOGY. 2022;32(2):98–106.CrossRef Aydın EP, Alsaadoni H, Beğenen AG, Özer ÖA, Karamustafalıoğlu KO, Pençe S. Can miRNA Expression Levels Predict Treatment Resistance to Serotonin Reuptake Inhibitors in Patients with Obsessive-Compulsive Disorder? PSYCHIATRY AND CLINICAL PSYCHOPHARMACOLOGY. 2022;32(2):98–106.CrossRef
98.
Zurück zum Zitat Grubor M, Zivkovic M, Sagud M, Nikolac Perkovic M, Mihaljevic-Peles A, Pivac N, et al. HTR1A, HTR1B, HTR2A, HTR2C and HTR6 gene polymorphisms and extrapyramidal side effects in haloperidol-treated patients with schizophrenia. Int J Mol Sci. 2020;21(7):2345.CrossRef Grubor M, Zivkovic M, Sagud M, Nikolac Perkovic M, Mihaljevic-Peles A, Pivac N, et al. HTR1A, HTR1B, HTR2A, HTR2C and HTR6 gene polymorphisms and extrapyramidal side effects in haloperidol-treated patients with schizophrenia. Int J Mol Sci. 2020;21(7):2345.CrossRef
99.
Zurück zum Zitat Essali A, Turkmani K, Aboudamaah S, AbouDamaah A, Aldeen MRD, Marwa ME, et al. Haloperidol discontinuation for people with schizophrenia. Cochrane Database of Systematic Reviews. 2019;4(4):CD011408. Essali A, Turkmani K, Aboudamaah S, AbouDamaah A, Aldeen MRD, Marwa ME, et al. Haloperidol discontinuation for people with schizophrenia. Cochrane Database of Systematic Reviews. 2019;4(4):CD011408.
100.
Zurück zum Zitat Leucht S, Cipriani A, Spineli L, Mavridis D, Örey D, Richter F, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. The Lancet. 2013;382(9896):951–62.CrossRef Leucht S, Cipriani A, Spineli L, Mavridis D, Örey D, Richter F, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. The Lancet. 2013;382(9896):951–62.CrossRef
101.
Zurück zum Zitat Thamby A, Jaisoorya T. Antipsychotic augmentation in the treatment of obsessive-compulsive disorder. Indian Journal of Psychiatry. 2019;61(Suppl 1):S51. Thamby A, Jaisoorya T. Antipsychotic augmentation in the treatment of obsessive-compulsive disorder. Indian Journal of Psychiatry. 2019;61(Suppl 1):S51.
102.
Zurück zum Zitat Chatterjee SS, Mitra S, Mallik N. Emerging hyperprolactinemic galactorrhea in obsessive compulsive disorder with a stable dose of fluoxetine. Clinical Psychopharmacology and Neuroscience. 2015;13(3):316.CrossRef Chatterjee SS, Mitra S, Mallik N. Emerging hyperprolactinemic galactorrhea in obsessive compulsive disorder with a stable dose of fluoxetine. Clinical Psychopharmacology and Neuroscience. 2015;13(3):316.CrossRef
103.
Zurück zum Zitat Chakraborty S, Dasgupta A, Das HN, Singh OP, Mandal AK, Mandal N. Study of oxidative stress in obsessive compulsive disorder in response to treatment with Fluoxetine. Indian J Clin Biochem. 2009;24(2):194–7.CrossRef Chakraborty S, Dasgupta A, Das HN, Singh OP, Mandal AK, Mandal N. Study of oxidative stress in obsessive compulsive disorder in response to treatment with Fluoxetine. Indian J Clin Biochem. 2009;24(2):194–7.CrossRef
104.
Zurück zum Zitat Pigott TA, Seay SM. A review of the efficacy of selective serotonin reuptake inhibitors in obsessive-compulsive disorder. J Clin Psychiatry. 1999;60(2):18284.CrossRef Pigott TA, Seay SM. A review of the efficacy of selective serotonin reuptake inhibitors in obsessive-compulsive disorder. J Clin Psychiatry. 1999;60(2):18284.CrossRef
105.
Zurück zum Zitat Mao Y-M, Zhang M-D. Augmentation with antidepressants in schizophrenia treatment: benefit or risk. Neuropsychiatr Dis Treat. 2015;11:701. Mao Y-M, Zhang M-D. Augmentation with antidepressants in schizophrenia treatment: benefit or risk. Neuropsychiatr Dis Treat. 2015;11:701.
106.
Zurück zum Zitat De Berardis D, Rapini G, Olivieri L, Di Nicola D, Tomasetti C, Valchera A, et al. Safety of antipsychotics for the treatment of schizophrenia: a focus on the adverse effects of clozapine. Therapeutic advances in drug safety. 2018;9(5):237–56.CrossRef De Berardis D, Rapini G, Olivieri L, Di Nicola D, Tomasetti C, Valchera A, et al. Safety of antipsychotics for the treatment of schizophrenia: a focus on the adverse effects of clozapine. Therapeutic advances in drug safety. 2018;9(5):237–56.CrossRef
107.
Zurück zum Zitat Siskind D, McCartney L, Goldschlager R, Kisely S. Clozapine v. first-and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry. 2016;209(5):385–92.CrossRef Siskind D, McCartney L, Goldschlager R, Kisely S. Clozapine v. first-and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry. 2016;209(5):385–92.CrossRef
108.
Zurück zum Zitat Kim DD, Barr AM, Lu C, Stewart SE, White RF, Honer WG, et al. Clozapine-associated obsessive-compulsive symptoms and their management: a systematic review and analysis of 107 reported cases. Psychother Psychosom. 2020;89(3):151–60.CrossRef Kim DD, Barr AM, Lu C, Stewart SE, White RF, Honer WG, et al. Clozapine-associated obsessive-compulsive symptoms and their management: a systematic review and analysis of 107 reported cases. Psychother Psychosom. 2020;89(3):151–60.CrossRef
109.
Zurück zum Zitat Kang S, Noh HJ, Bae SH, Kim Y-S, Lew H, Lim J, et al. Clozapine generates obsessive compulsive disorder-like behavior in mice. Mol Brain. 2020;13(1):84.CrossRef Kang S, Noh HJ, Bae SH, Kim Y-S, Lew H, Lim J, et al. Clozapine generates obsessive compulsive disorder-like behavior in mice. Mol Brain. 2020;13(1):84.CrossRef
110.
Zurück zum Zitat Stryjer R, Dambinsky Y, Timinsky I, Green T, Kotler M, Weizman A, et al. Escitalopram in the treatment of patients with schizophrenia and obsessive-compulsive disorder: an open-label, prospective study. Int Clin Psychopharmacol. 2013;28(2):96–8.CrossRef Stryjer R, Dambinsky Y, Timinsky I, Green T, Kotler M, Weizman A, et al. Escitalopram in the treatment of patients with schizophrenia and obsessive-compulsive disorder: an open-label, prospective study. Int Clin Psychopharmacol. 2013;28(2):96–8.CrossRef
111.
Zurück zum Zitat Starr KE, Burns K, Demler TL. Pharmacological and philosophical considerations for the around-the-clock use of scheduled melatonin to promote sedation and reduce aggression in individuals with serious mental illness: a case report. Int Clin Psychopharmacol. 2021;36(6):296–304.CrossRef Starr KE, Burns K, Demler TL. Pharmacological and philosophical considerations for the around-the-clock use of scheduled melatonin to promote sedation and reduce aggression in individuals with serious mental illness: a case report. Int Clin Psychopharmacol. 2021;36(6):296–304.CrossRef
112.
Zurück zum Zitat Monti JM, BaHammam AS, Pandi-Perumal SR, Bromundt V, Spence DW, Cardinali DP, et al. Sleep and circadian rhythm dysregulation in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:209–16.CrossRef Monti JM, BaHammam AS, Pandi-Perumal SR, Bromundt V, Spence DW, Cardinali DP, et al. Sleep and circadian rhythm dysregulation in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:209–16.CrossRef
113.
Zurück zum Zitat Maiti R, Mishra BR, Jena M, Mishra A, Nath S. Effect of haloperidol and risperidone on serum melatonin and GAP-43 in patients with schizophrenia: A prospective cohort study. Clinical Psychopharmacology and Neuroscience. 2021;19(1):125.CrossRef Maiti R, Mishra BR, Jena M, Mishra A, Nath S. Effect of haloperidol and risperidone on serum melatonin and GAP-43 in patients with schizophrenia: A prospective cohort study. Clinical Psychopharmacology and Neuroscience. 2021;19(1):125.CrossRef
114.
Zurück zum Zitat Fanget F, Claustrat B, Dalery J, Brun J, Terra J-L, Marie-Cardine M, et al. Nocturnal plasma melatonin levels in schizophrenic patients. Biological Psychiatry. 1989;25(4):499-501. Fanget F, Claustrat B, Dalery J, Brun J, Terra J-L, Marie-Cardine M, et al. Nocturnal plasma melatonin levels in schizophrenic patients. Biological Psychiatry. 1989;25(4):499-501.
115.
Zurück zum Zitat Monteleone P, Natale M, La Rocca A, Maj M. Decreased nocturnal secretion of melatonin in drug-free schizophrenics: no change after subchronic treatment with antipsychotics. Neuropsychobiology. 1997;36(4):159–63.CrossRef Monteleone P, Natale M, La Rocca A, Maj M. Decreased nocturnal secretion of melatonin in drug-free schizophrenics: no change after subchronic treatment with antipsychotics. Neuropsychobiology. 1997;36(4):159–63.CrossRef
116.
Zurück zum Zitat Robinson S, Rosca P, Durst R, Shai U, Ghinea C, Schmidt U, et al. Serum melatonin levels in schizophrenic and schizoaffective hospitalized patients. Acta Psychiatr Scand. 1991;84(3):221–4.CrossRef Robinson S, Rosca P, Durst R, Shai U, Ghinea C, Schmidt U, et al. Serum melatonin levels in schizophrenic and schizoaffective hospitalized patients. Acta Psychiatr Scand. 1991;84(3):221–4.CrossRef
117.
Zurück zum Zitat Duan C, Jenkins ZM, Castle D. Therapeutic use of melatonin in schizophrenia: A systematic review. World Journal of Psychiatry. 2021;11(8):463.CrossRef Duan C, Jenkins ZM, Castle D. Therapeutic use of melatonin in schizophrenia: A systematic review. World Journal of Psychiatry. 2021;11(8):463.CrossRef
118.
Zurück zum Zitat Paterson JL, Reynolds AC, Ferguson SA, Dawson D. Sleep and obsessive-compulsive disorder (OCD). Sleep Med Rev. 2013;17(6):465–74.CrossRef Paterson JL, Reynolds AC, Ferguson SA, Dawson D. Sleep and obsessive-compulsive disorder (OCD). Sleep Med Rev. 2013;17(6):465–74.CrossRef
119.
Zurück zum Zitat Monteleone P, Catapano F, Del Buono G, Maj M. Circadian rhythms of melatonin, cortisol and prolactin in patients with obsessive-compulsive disorder. Acta Psychiatr Scand. 1994;89(6):411–5.CrossRef Monteleone P, Catapano F, Del Buono G, Maj M. Circadian rhythms of melatonin, cortisol and prolactin in patients with obsessive-compulsive disorder. Acta Psychiatr Scand. 1994;89(6):411–5.CrossRef
Metadaten
Titel
Disclosing common biological signatures and predicting new therapeutic targets in schizophrenia and obsessive–compulsive disorder by integrated bioinformatics analysis
verfasst von
Abdolhakim Ghanbarzehi
Ali Sepehrinezhad
Nazanin Hashemi
Minoo Karimi
Ali Shahbazi
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Psychiatry / Ausgabe 1/2023
Elektronische ISSN: 1471-244X
DOI
https://doi.org/10.1186/s12888-023-04543-z

Weitere Artikel der Ausgabe 1/2023

BMC Psychiatry 1/2023 Zur Ausgabe

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Klinikreform soll zehntausende Menschenleben retten

15.05.2024 Klinik aktuell Nachrichten

Gesundheitsminister Lauterbach hat die vom Bundeskabinett beschlossene Klinikreform verteidigt. Kritik an den Plänen kommt vom Marburger Bund. Und in den Ländern wird über den Gang zum Vermittlungsausschuss spekuliert.

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.