Skip to main content
Faculty Reviews logoLink to Faculty Reviews
. 2020 Dec 23;9:30. doi: 10.12703/r/9-30

Cutting-edge genetics in obsessive-compulsive disorder

Leonardo Cardoso Saraiva 1, Carolina Cappi 1, Helen Blair Simpson 2,3,#, Dan J Stein 4,#, Biju Viswanath 5, Odile A van den Heuvel 6, YC Janardhan Reddy 7, Euripedes C Miguel 1, Roseli G Shavitt 1,*
PMCID: PMC7886082  PMID: 33659962

Abstract

This article reviews recent advances in the genetics of obsessive-compulsive disorder (OCD). We cover work on the following: genome-wide association studies, whole-exome sequencing studies, copy number variation studies, gene expression, polygenic risk scores, gene–environment interaction, experimental animal systems, human cell models, imaging genetics, pharmacogenetics, and studies of endophenotypes. Findings from this work underscore the notion that the genetic architecture of OCD is highly complex and shared with other neuropsychiatric disorders. Also, the latest evidence points to the participation of gene networks involved in synaptic transmission, neurodevelopment, and the immune and inflammatory systems in this disorder. We conclude by highlighting that further study of the genetic architecture of OCD, a great part of which remains to be elucidated, could benefit the development of diagnostic and therapeutic approaches based on the biological basis of the disorder. Studies to date revealed that OCD is not a simple homogeneous entity, but rather that the underlying biological pathways are variable and heterogenous. We can expect that translation from bench to bedside, through continuous effort and collaborative work, will ultimately transform our understanding of what causes OCD and thus how best to treat it.

Keywords: obsessive-compulsive disorder, genetics, genomics

Introduction

Obsessive-compulsive disorder (OCD) is a neuropsychiatric disorder characterized by intrusive thoughts (obsessions) and repetitive behaviors (compulsions)1. With a lifetime prevalence of 2–3% and a typically chronic course2, OCD is associated with considerable role impairment3, reduced quality of life4, and morbidity5. Moreover, individuals with OCD have an elevated mortality risk independently of the effects of comorbidities6.

First-line treatments for OCD include both pharmacological and cognitive–behavioral approaches7; alone or combined, these can help about half of patients to achieve minimal symptoms8. The development of more targeted and effective treatment interventions may benefit from further understanding of the underlying etiology of OCD, including underlying genetic mechanisms. Here we review recent advances in OCD genetics research, including genome-wide association studies (GWAS), whole-exome sequencing (WES) studies, copy number variation (CNV) studies; gene expression, polygenic risk score (PRS), gene–environment interactions, experimental animal systems, human cell models, imaging genetics, pharmacogenetics, and studies of endophenotypes. We suggest that recent work converges in pointing out that genes involved in synaptic transmission, neurodevelopment, and the immune and inflammatory systems are involved in the pathophysiology of OCD (Figure 1). Given the genetic overlap between OCD and related disorders, further work is needed to assess the specificity of this involvement.

Figure 1. From genetic architecture to obsessive-compulsive disorder (OCD) symptomatology.

Figure 1.

The genetic architecture of OCD presumably underlies alterations in biological pathways, which in turn lead to disrupted brain circuits and OCD symptoms. CNV, copy number variant; SNP, single nucleotide polymorphism.

From genetic epidemiology to genetic architecture

Family and twin studies are useful for exploring genetic and environmental contributions to a disease. In this context, familiality refers to increased clustering of a disorder amongst families9. Consistent with multiple previous studies1012, a recent family study in Sweden provided further evidence for the familiality of OCD, especially when associated with tics13. Heritability can be defined as the proportion of variance in a phenotypic trait attributable to additive genetic effects (i.e. narrow-sense heritability, h2) or to total genetic effects (i.e. broad-sense heritability, H2)14. Henceforth in this article, narrow-sense heritability (h2) will be referred to as heritability. Past twin studies have estimated the heritability of OCD to range from 27 to 65%15, and a recent heritability estimate of 74% was reported for obsessive-compulsive traits in a pediatric nonclinical twin cohort16.

The heritability of OCD encourages further work on the genetics of this disorder, with the hope that this will, in turn, contribute to the development of more precise diagnostic and therapeutic approaches17. The term “genetic architecture” refers to the overall number, effect size, population frequency, and interactions of genetic variants associated with a phenotype18. Genetic studies that contribute to elucidating genetic architecture include GWAS, WES studies, CNV studies, gene expression analysis, gene–environment interaction studies, experimental models with animal and human cells, imaging genetics studies, pharmacogenetic studies, and studies of endophenotypes. We consider each in turn, presenting a summary of all of the cited studies after the section describing an exploratory model of the genetic architecture of OCD (Table 1).

Table 1. Summary of genetic studies in obsessive-compulsive disorder (OCD) cited throughout this review.

Author Sample Methodology Findings
Brander et al.
(2019)
4,085,367 individuals, including
22,232 OCD probands
Family study Familial clustering of OCD, especially tic-related OCD. Using hazard ratios adjusted for sex
and birth year, the risk of OCD in full siblings of patients with tic-related OCD and patients
with non-tic-related OCD was estimated at 10.63 and 4.52, respectively.
Burton et al.
(2018)
220 pairs of non-clinical twins Twin study Heritability of obsessive-compulsive traits was estimated at 74%. Shared genetic factors
explained most of the variance shared among the obsessive-compulsive trait dimensions
examined in this study, which included cleaning/contamination, symmetry/ordering,
rumination, superstition, counting/checking, and hoarding.
Stewart et al.
(2013)
1,465 OCD probands and 5,557
control subjects
Genome-wide
association study
(GWAS)
No genome-wide significant single nucleotide polymorphism (SNP) was detected in case-
control analysis, which yielded the top SNPs located within DLGAP1. Trio-based analysis
revealed a SNP reaching genome-wide significance located near BTBD3. Combining the
two analyses revealed significant enrichment for regions regulating gene expression and
methylation in the brain.
Mattheisen et al.
(2015)
1,406 OCD probands and 5,061
control subjects
GWAS No genome-wide significant SNP was detected. Top-ranked SNPs were near PTPRD, CDH9,
and CDH10. Two SNPs near GRIK2 and HACE1 detected in the first OCD GWAS were also
among the significant SNPs.
Den Braber
et al. (2016)
Obsessive-compulsive symptoms of
8,267 subjects
GWAS A genome-wide significant SNP was detected at MEF2BNB. Gene-based testing revealed
four genes significantly associated with obsessive-compulsive symptoms located in
the 19p13.11 chromosomal region, which has been associated with brain and immune
processes.
Costas et al.
(2016)
370 OCD probands and 443 control
subjects
GWAS No genome-wide significant SNP was detected. A polygenic risk score (PRS) estimated
using the Psychiatric Genetics Consortium (PGC) schizophrenia (SCZ) meta-analysis sample
significantly predicted OCD case-control status among the study sample.
Guo et al.
(2017)
2,998 OCD case-control samples
and 6,898 autism spectrum disorder
(ASD) case-control samples
GWAS No genome-wide significant SNP was detected. Top-ranked SNP identified is in linkage
disequilibrium (LD) with TUBB3. A PRS estimated using the ASD case-control sample
significantly predicted case-control status in the OCD sample.
PGC OCD
workgroup (2018)
2,688 OCD probands and 7,037
control subjects
GWAS No genome-wide significant SNP was detected. Most significant SNPs were in LD with
GRID2 and KIT. PRSs estimated based on the first and second OCD GWASs significantly
predicted case-control status in the second and the first OCD GWASs case-control samples,
respectively. SNP-based heritability was estimated at 0.28.
Khramtsova et al.
(2018)
PGC-OCD GWAS sample GWAS SNP-based heritability estimates were similar for male and female OCD, and significant
genetic correlation between them was reported. GRID2 and GRP135 were associated only
with female OCD in gene-based tests. SNPs with sex-specific effects were significantly
enriched for regions regulating gene expression in brain and immune tissues.
Smit et al. (2020) PGC-OCD GWAS sample and
compulsive symptoms of 8,267
subjects
GWAS No genome-wide significant SNP was detected. Significant genetic correlation between OCD
and compulsive symptoms was found. WDR7, ADCK1, GRID2, and KIT were detected in
gene-based tests. Top genes detected in gene-based tests were significantly enriched for
genes expressed in the anterior cingulate cortex, nucleus accumbens, and amygdala.
Cross-Disorder
Group of the
PGC (2019)
232,964 probands diagnosed with
anorexia nervosa (AN), attention
deficit hyperactivity disorder (ADHD),
ASD, bipolar disorder (BD), major
depressive disorder (MDD), OCD,
SCZ, or Tourette’s syndrome (TS) and
494,162 controls subjects
GWAS Genetic correlation between OCD and AN was among the highest observed. Exploratory
factor analysis suggested shared genetic liability among OCD, AN, and TS.
Bralten et al.
(2020)
650 non-clinical children and
adolescents
GWAS The individual results from the GWAS of obsessive-compulsive symptoms in the sample of
650 non-clinical children and adolescents were not reported owing to insufficient power.
Nonetheless, PRS analysis revealed shared genetic etiology among OCD, obsessive-
compulsive symptoms, and insulin signaling-related traits.
Cappi et al.
(2016)
17 OCD parent–child trios Whole-exome
sequencing study
20 nonsynonymous de novo single nucleotide variants (SNVs) detected among 17
OCD probands. SNVs were enriched in pathways related to the immune system and
neurodevelopment.
Cappi et al.
(2019)
184 OCD and 777 control parent–
child trios
Whole-exome
sequencing study
A significantly higher prevalence of de novo damaging variants (33.9%) was found among
probands. These variants were estimated to be present in 22.2% of overall OCD cases.
SCUBE1 and CHD8 were identified as high-confidence risk genes for OCD. Genes carrying
these variants significantly overlap among ASD and TS probands and were associated with
immune system-related processes.
Gazzellone et al.
(2016)
307 pediatric OCD probands and
3,861 control subjects
Copy number
variants study
A similar rate of rare copy number variants (CNVs) in early onset OCD probands and controls
was observed. In 5.9% of probands, structural variants were detected in genes associated
with brain function. Particularly, CNVs were found in three targets of fragile X mental
retardation protein (FMRP), including DLGAP1 and PTPRD.
Grünblatt et al.
(2017)
121 pediatric OCD probands and
123 control subjects
Copy number
variants study
Although no significant difference in number and size of rare CNVs between pediatric OCD
patients and controls was detected, the number of CNVs encompassing genes involved in
neurological function was higher among the former.
Zarrei et al.
(2019)
2,691 probands diagnosed with
OCD, SCZ, ASD, or ADHD and 1,769
family members
Copy number
variants study
Clinically relevant CNVs were found in 5.6% of OCD probands. Multiple brain-expressed
genes impacted by CNVs in at least two probands were found. An increased burden of rare
CNVs impacting genes involved in genomic stability was found in probands.
Wang et al.
(2018)
77 OCD, 39 MDD, and 40 SCZ
probands and 67 control subjects
Gene expression
analysis
A blood-based gene expression panel (including FKBP1A, COPS7A, FIBP, TP73-AS1, SDF4,
and GOLGA8A) was able to distinguish OCD probands from MDD and SCZ probands and
healthy controls with 88% sensitivity and 85% specificity.
Piantadosi et al.
(2019)
Postmortem brain samples of 8 OCD
probands and 8 control subjects
Gene expression
analysis
Overall reduced expression of genes related to excitatory synapses in the orbitofrontal cortex
(OFC), but not in striatal regions, of OCD probands. Minor alterations were observed in the
expression of genes related to inhibitory synapses in the OFC of OCD probands.
Nissen et al.
(2016)
21 pediatric and adolescent OCD
probands and 12 female controls
DNA methylation
analysis
No significantly different DNA methylation levels in candidate genes previously implicated in
OCD were found between probands and controls.
Yu et al. (2016) 65 pediatric OCD probands and 96
control subjects
DNA methylation
analysis
Multiple genes with different DNA methylation levels among OCD cases and controls were
found. Further enrichment analysis implicated those genes in cell adhesion- and actin-related
processes.
Alemany-Navarro
et al. (2019)
103 OCD probands Gene–environment
interaction
Although not predicting treatment response, a PRS estimate based on OCD risk variants
predicted pretreatment symptom severity.
Mahjani et al.
(2020)
822,843 individuals, including 7,184
probands diagnosed with OCD
Maternal effects Genetic maternal effects (i.e. the influence of the maternal genotype on the phenotype of
the offspring) accounted for 7.6% of the liability for OCD, whereas additive genetic effects
accounted for 35%.
Trankner et al.
(2019)
Experimental
animal system
Targeted deletion of Hoxb8-lineage microglia led to significant grooming and anxiety-
like behaviors and stress-response among female rodents, which were ameliorated by
suppressing female sex hormones.
Delgado-
Acevedo
et al. (2018)
Experimental
animal system
Overexpression of EAAT3 in the frontal cortex, hippocampus, and striatum increased anxious
and grooming behaviors and prolonged spontaneous recovery of conditioned fear among
rodents. Moreover, alterations in NMDA receptor constitution and corticostriatal synaptic
plasticity were observed.
Zike et al. (2017) Experimental
animal system
Constitutive reduced expression of EAAT3 in rodents caused dampened stereotyped
response following dopaminergic challenges, which was rescued by restoration of EAAT3
expression in the midbrain.
Krabbe et al.
(2017)
Experimental
animal system
Rodent with microglia-restricted progranulin (PGRN) inactivation exhibited increased self-
grooming and marble-burying behaviors, which were normalized with suppression of nuclear
factor κB (NF-κB) signaling.
Ullrich et al.
(2018)
Experimental
animal system
SPRED2 knockout mice displayed severe grooming and anxiety behaviors, dysfunctional
thalamo-amygdala synapses, and altered expression of synaptic proteins in the amygdala.
Inhibition of overactive (TrkB)/ERK-MAPK signaling ameliorated pathologic behavior.
Van de
Vondervoort et al.
(2020)
Experimental
animal system
TALLYHO/JngJ rodents, which are a model of human type 2 diabetes mellitus, exhibited
increased compulsive and anxious behaviors in addition to structural brain abnormalities in
midline corpus callosum, dorsomedial striatum, and superior cerebellar peduncles.
Noh et al. (2017) 592 OCD probands and 560 control
subjects
Functional variants
prioritization
Analysis of 608 genes potentially associated with OCD in human, rodent, and dog studies
revealed significant functional variant burden in four genes (NRXN1, HTR2A, CTTNBP2,
and REEP3) involved in brain pathways implicated in OCD. NRXN1 achieved genome-wide
significance after inclusion of GWAS data from 33,370 controls.
Rodriguez et al.
(2017)
102 OCD early onset OCD probands
and 47 control subjects
Human cell models Higher percentages of total monocytes and CD16+ monocytes in OCD probands as
compared to control subjects. Increased production of proinflammatory cytokines by proband
monocytes after stimulation with lipopolysaccharide.
Özyurt et al.
(2019)
60 drug-naïve adolescent OCD
probands and 128 adolescent
control subjects
Human cell models Significant differences were detected in the neutrophil: lymphocyte ratio and white blood cell,
neutrophil, and platelet counts among OCD probands with comorbid anxiety disorder, OCD
probands with no comorbidities, and control subjects, which remained after controlling for
age and sex.
Hibar et al.
(2018)
First OCD GWAS sample and
neuroimaging GWASs of volume
of eight subcortical brain regions
(13,171 subjects)
Imaging genetics OCD risk SNPs were associated with increased volumes of the nucleus accumbens and the
putamen. Conditional false discovery rate analysis revealed specific SNPs influencing OCD
risk and putamen, amygdala, and thalamus volumes.
Zartaloudi et al.
(2019)
707 unaffected relatives of OCD
probands and 842 control subjects
Meta-analysis of
endophenotype
studies
21 studies were included in the meta-analysis. A significant impairment in global executive
functioning was found among unaffected relatives of OCD probands, with specific deficits in
planning, visuospatial working memory, and verbal fluency.
Qin et al. (2016) 804 OCD probands (514 responders
and 290 non-responders)
Pharmacogenetics A genome-wide significant SNP was detected near the gene DISP1, within a chromosomal
region associated with neurodevelopment. Most significant SNPs were enriched for biological
pathways related to glutamate and serotonin neurotransmission.
Lisoway et al.
(2018)
112 OCD probands Pharmacogenetics No significant associations between a SNP of DISP1 and response to serotonin reuptake
inhibitor treatment.

Genome variants

Early studies on the genetics of OCD focused on candidate gene association studies15. Because of their insufficient power and biased a priori hypotheses, these studies produced rather inconsistent findings and are now considered obsolete19. The development of novel methods capable of assessing the entire genome, coupled with the recruitment of larger samples, allowed the undertaking of unbiased and powerful genetic investigations19. In this regard, cumulative evidence supports the idea that both common and rare genome variants contribute to the polygenic architecture of psychiatric disorders20. Accordingly, distinct methods have been recently used to detect genome variants associated with OCD, as described below.

Common variants

GWASs employ a case-control design to detect single nucleotide polymorphisms (SNPs) (Figure 2) associated with a disorder21, with significance thresholds set at P = 5 × 10-822. A range of secondary analyses of GWAS data can be conducted to further probe the relevant genetic architecture23. Such analyses include determining the portion of heritability conferred by the SNPs investigated in a GWAS, defined as SNP-based heritability (h2SNP)24, estimating the degree of shared genetic architecture between disorders by determining their genetic correlation25,26, calculating PRSs based on risk variants for a disorder identified in a GWAS27, and undertaking enrichment analysis to assess clustering of detected variants within functionally related genomic regions and biological pathways28.

Figure 2. Variations in the genome.

Figure 2.

Single nucleotide variants (SNVs) constitute positions in the genome comprising a pair of bases for which different alleles (i.e. sequence variants) are found in a population. A SNV can be classified according to the frequency at which its second most common allele is found in a population, termed minor allele frequency (MAF). According to their MAF, SNVs can be classified into common (MAF≥5%), low-frequency (1%≥MAF<5%), and rare (MAF<1%)18. SNVs that occur in at least 1% of the population are called single nucleotide polymorphisms (SNPs)18. When occurring inside the exons (i.e. DNA stretches encoding protein products), SNVs can lead to the production of protein products with a normal amino acid sequence (i.e. synonymous SNVs), with an altered yet full amino acid sequence (i.e. missense SNVs), or with a truncated (i.e. incomplete) amino acid sequence (i.e. nonsense SNVs). The last two types of SNVs are termed nonsynonymous and usually have deleterious biological consequences. Copy number variants (CNVs) comprise genome deletions and duplications.

To date, GWASs of OCD have not revealed any genome-wide significant SNPs associated with this disorder in case-control analyses15. Recently, the Psychiatric Genomics Consortium OCD (PGC-OCD) workgroup combined data from the first two OCD GWASs29,30 to yield a sample of 2,688 OCD cases and 7,037 controls. The top-ranked detected SNP (i.e. strongest association) was in the P = 10-7 significance range31. Nonetheless, SNP-based heritability was estimated at 28% in this sample31, among the highest reported for neuropsychiatric disorders. These findings indicate the need for still larger samples for sufficient statistical power.

The top-ranked SNPs detected in the PGC-OCD GWAS were in linkage disequilibrium with genes previously associated with autism spectrum disorders (ASD)31: GRID2, involved in cerebellar synaptic processes32, and KIT, implicated in neurodevelopment33. Moreover, a sex-stratified GWAS of the PGC-OCD sample revealed that SNPs with sex-specific effects were significantly enriched for genome regions influencing gene expression (i.e. expression quantitative trait loci, eQTLs) in brain and immune tissues34. In line with the findings of an earlier GWAS of obsessive-compulsive symptoms35, a recent meta-analysis of the PGC-OCD GWAS and a GWAS of compulsive symptoms in 8,267 nonclinical subjects revealed the most significant enrichment for genes expressed in brain regions associated with the neurocircuitry of OCD, i.e. the cingulate cortex, nucleus accumbens, and amygdala36. Taken together, the aforementioned GWASs suggest a role for synaptic, neurodevelopmental, and immune processes in the etiology of OCD.

Genetic correlation analyses using data from the PGC-OCD GWAS have suggested a shared genetic architecture between OCD and related conditions. In this respect, two studies found a significant genetic correlation between OCD and anorexia nervosa (AN)37,38, consistent with the hypothesis that obsessive and compulsive traits in these conditions have shared etiology39. Furthermore, PRSs for other neuropsychiatric disorders were able to significantly predict case-control status in previous OCD GWAS samples, indicating a shared genetic liability40. In smaller studies, a PRS for schizophrenia (SCZ) significantly predicted case-control status in a Spanish sample comprising 370 OCD patients and 443 controls41, and a PRS for ASD significantly predicted case-control status in a sample of 2,535 OCD cases and controls42. PRS may, in the future, have applications in clinical practice, ranging from improving diagnostic precision to predicting treatment response20. However, it needs to be emphasized that such applications are not currently possible and that translation of PRSs from a population of one ancestry to a population of different ancestry is problematic43.

Rare variants

WES is a technique employed to detect single nucleotide variants in the exome (i.e. all of the genome exons)20, among which de novo variants (Figure 2) have the most pathogenic effect for neuropsychiatric disorders44. De novo variants refer to genome variants that occur in the proband but not in the parents. WES studies are usually performed in trios composed of healthy parents and an affected child in order to detect de novo variants driving pathology20,44.

Two recent studies reported an increased genome-wide burden of gene-disrupting de novo mutations in patients with OCD. In the first WES study45, 20 nonsynonymous de novo variants were detected among 17 OCD probands at a rate similar to that reported for other neuropsychiatric disorders. Moreover, these variants were enriched for pathways related to the immune system and neurodevelopment.

The second WES study was conducted on a sample of 184 OCD and 777 control parent–child trios46. De novo damaging variants were significantly more frequent among probands, achieving a prevalence of 33.9%. Such findings were used to estimate the rate of these variants among overall OCD cases at 22.2%. Two high-confidence risk genes for OCD were further identified: SCUBE1, potentially involved in endothelial inflammatory responses47, and CHD8, implicated in chromatin metabolism48, neurogenesis49, and synaptic transmission50 and extensively associated with ASD5153. Additionally, genes carrying de novo damaging variants in OCD probands significantly overlapped with genes associated with ASD, Tourette’s syndrome (TS), and other neurodevelopmental disorders and were enriched for immune system pathways. Similar findings were reported in another study which found a significant overlap between genes associated with OCD and genes associated with ASD; in addition, the overlapping genes were significantly enriched for biological pathways related to brain function54.

Copy number variants

The most common type of structural variants in the human genome are CNVs (Figure 2), which comprise duplications and deletions55. The prevalence of CNVs can be assessed across the whole genome using array methods56.

In an earlier CNV study in OCD, a 4.4-fold increase in deletions encompassing loci associated with neurodevelopmental disorders (including 16p13.11) was found among OCD probands57. More recently, two CNV studies were conducted on samples comprising 307 and 121 individuals with pediatric OCD and 3,861 and 124 healthy controls, respectively58,59. The number of CNVs in genes involved in brain function was higher among OCD probands in both studies. In the first study, CNVs were found in three targets of the fragile X mental retardation protein (FMRP). Consistent with FMRP involvement in several processes related to synaptic plasticity60, CNVs encompassing FMRP targets have been associated with SCZ61, ASD62, and attention deficit hyperactivity disorder (ADHD)63. In the second study, CNVs were found in genes implicated in SCZ, ASD, and TS. Finally, a recent study of 2,691 individuals diagnosed with OCD, SCZ, ASD, or ADHD found clinically relevant CNVs in 5.6% of OCD cases and multiple brain-expressed genes impacted by CNVs overlapping across those disorders64.

Taken together, these WES and CNV findings again support a role for synaptic, neurodevelopmental, and immune processes in the etiology of OCD. Moreover, these findings indicate the existence of a shared genetic etiology between OCD and related disorders, suggesting that their current diagnostic categories may not reflect distinct clinical entities. In line with this, a recent study used a machine learning approach to cluster patients with OCD, ASD, and ADHD into homogenous groups based on neuroimaging measures of cortical thickness and behavioral measures and found that those diagnostic categories were not reflected in the groups formed65.

Variation in gene expression

Complex regulatory mechanisms govern the way in which genotype and environment together lead to distinctive patterns of gene expression in different tissues at different developmental stages20,66,67. Methods are now available to explore tissue-specific gene expression and environmental regulatory mechanisms in OCD.

Gene expression in specific tissue

As a proxy of gene expression, the measurement of transcript levels (i.e. mRNA) (Figure 3) in post-mortem brain specimens of patients with psychiatric disorders is useful for genetic investigations68. Furthermore, the discovery of differential transcript levels in the peripheral blood of patients with psychiatric disorders may provide additional insight into mechanisms underlying gene expression69.

Figure 3. Gene expression.

Figure 3.

The gene, composed of exons (protein-coding regions) and introns (non-protein-coding regions), is transcribed into a precursor messenger ribonucleic acid (pre-mRNA), which undergoes alternative splicing to generate a messenger RNA (mRNA), called a primary transcript. Finally, mRNA undergoes translation to generate a protein product. This chain of events enables the flow of genetic information.

Recently, reduced expression of genes related to excitatory glutamatergic synapses has been identified in the lateral and frontal regions of the orbitofrontal cortex (OFC) of individuals with OCD70. Speculatively, such findings may be relevant to understanding alterations in OFC volume71 and activity in neuroimaging studies of OCD72.

Gene expression profiling in the peripheral blood has been employed to compare individuals with OCD, major depressive disorder (MDD), and SCZ and healthy controls73. A six-gene panel was able to diagnose OCD cases with 88% sensitivity and 85% specificity73. Among the genes included in the panel, FKBP1A encodes an enzyme involved in immune regulation74 and COPS7A encodes a subunit of a protein complex involved in protein degradation75.

Gene expression and the environment

The reciprocal interactions between gene expression and the environment have been investigated through diverse methods, ranging from basic science to epidemiological approaches76,77. Importantly, epigenetic mechanisms (Figure 3) provide a pathway whereby the environment can modulate gene expression through alterations in chromatin and DNA structure78.

In one recent study, no difference was found in blood DNA methylation levels of candidate genes previously implicated in OCD among 21 pediatric cases and 12 controls79. Conversely, in another study using a genome-wide approach, multiple genes with different methylation levels were detected among 65 pediatric OCD cases and 96 controls80. These findings emphasize the importance of an unbiased approach for studying the genomics of OCD.

A study of gene–environment interactions in a cohort of 103 patients with OCD revealed that neither PRS for OCD nor the presence of a stressful life event at the onset of the disorder predicted treatment response81. Nonetheless, PRS was able to predict illness severity. Moreover, environmental factors, such as perinatal complications82,83 and maternal effects (i.e. the influence of maternal genetic and environmental factors on the phenotype of the offspring)84, have been shown to increase the risk for OCD. Further work is needed to determine whether PRSs, taken together with a range of environmental factors, may be useful in predicting OCD severity and treatment response. As a complement to the gene–environment interaction approach, in-depth environmental research in psychiatry, known as the exposome85, should add to the understanding of the interplay between genetics and the exposure to multiple internal and external stimuli in the prevention and treatment of psychiatric disorders.

Modeling genetic architecture

Laboratory-based experimental models may be used to study genes which appear to be important in clinical studies of the genomics of OCD86. We focus here on work done in animals and in human cells.

Experimental animal systems

Excessive grooming, hoarding behaviors (e.g. marble-burying), and stereotyped behaviors have been studied in rodent models of OCD87. Such work is not intended to replicate the disease in an animal system but may rather provide insight into the biological mechanisms relevant to OCD, which may foster the development of novel treatment approaches88.

Overexpression of the excitatory amino acid transporter 3 (EAAT3), involved in glutamate neurotransmission, in the frontal cortex, hippocampus, and striatum of rodents resulted in increasing grooming and disturbed cortical–striatal excitatory synapse plasticity89. Furthermore, a protective effect against the induction of stereotyped behaviors by dopaminergic challenge was obtained by constitutive EAAT3 reduced expression in mice90.

Immunity- and neuroinflammation-related processes have been consistently associated with OCD91. In this respect, mice with microglia-restricted progranulin inactivation exhibited increased self-grooming and marble-burying, which was normalized after suppression of nuclear factor κB signaling in the microglia92. Also suggesting a potential role for the microglia in the etiology of OCD, targeted deletion of Hoxb8-lineage microglia yielded severe grooming, especially in female mice93. Furthermore, restoring tropomyosin receptor kinase B/ERK-MAPK signaling in mice normalized severe grooming behaviors induced by ablation of SPRED294, which pertain to a family of proteins that has been implicated in neurodevelopment95.

Recent investigations of gene pathway analysis96 and PRSs97 using GWAS data support the association of disturbances in insulin signaling with the pathophysiology of OCD. These findings were further validated in a rodent model of type 2 diabetes mellitus, which revealed increased compulsive-like behaviors and brain abnormalities previously associated with OCD98.

Findings from experimental animal systems may inform clinical studies99. A large set of candidate genes potentially associated with OCD in previous human, rodent, and canine studies was investigated using a target-sequencing approach for the detection of regulatory and coding variants (i.e. functional variants) in 592 OCD cases and 560 controls100. Among the four genes significantly enriched for functional variants in OCD cases (i.e. NRXN1, HTR2A, CTTNBP2, and REEP3), all were involved in brain biological pathways implicated in OCD100. Additionally, NRXN1 achieved genome-wide significance when OCD cases were compared to 33,370 controls100.

Human cell models

Investigations using human-derived cells as models may be useful in studying OCD at the molecular and cellular levels101. Higher ratios of immune cells in the peripheral blood102,103 and abnormal production of inflammatory cytokines upon stimulation by those cells102 have been reported in OCD. Future work on pluripotent stem cells derived from somatic cells of patients with OCD could be useful in extending this preliminary work.

Genetic variations underlying relevant phenotypes

There is growing interest in determining the genetic basis of OCD-associated neuroimaging abnormalities, cognitive dysfunction, and treatment response, as outlined below.

Neuroimaging

The Enhancing Neuroimaging Genetics through Meta-Analysis (ENIGMA) consortium is the largest collaboration working to integrate genetic and neuroimaging findings; it combines findings from sites across the globe104. The combination of data from the first OCD GWAS and the ENIGMA GWAS of subcortical brain structure105 allowed for the detection of a significant overlap between the SNPs associated with increases in the risk for OCD and in the volumes of the putamen, a component of the cortical–striatal–thalamic–cortical circuitry implicated in OCD106, and the nucleus accumbens, a treatment target for deep brain stimulation in treatment-refractory OCD107. Moreover, the SNP associated with both the increased risk for OCD and the increased putamen volume is located near the RSPO4, a gene implicated in pathways related to neurodevelopment108.

Cognitive dysfunction

Cognitive deficits have been frequently explored as OCD endophenotypes109, defined as heritable quantitative traits found at higher rates in unaffected relatives of patients and associated with increased genetic susceptibility to the disorder110,111. The findings of investigations on cognitive deficits as endophenotypes of OCD have been recently meta-analyzed112; this study found significant impairment in global executive functioning among unaffected relatives, with specific deficits in planning, visuospatial working memory, and verbal fluency. Since these endophenotypes may be more proximally related to genetic mechanisms than OCD itself111, further investigation of these deficits may be useful.

Treatment response

Pharmacogenetic studies address the association of genetic variation with drug response113. Although earlier pharmacogenetic studies in psychiatric disorders have focused mainly on candidate genes and on genes related to the cytochrome P450 system114, genome-wide approaches have been recently employed in OCD research.

The association of the response to serotonin reuptake inhibitors (SRIs) treatment with common variants was assessed in a GWAS of 804 OCD cases, including 514 responders and 290 non-responders115. A genome-wide significant SNP was detected near the gene DISP1, within a chromosomal region associated with neurodevelopment. Further enrichment analysis revealed that the most significant SNPs were enriched for biological pathways related to glutamate and serotonin neurotransmission. However, a follow-up study with 112 OCD cases found no association between the SNP located near DISP1 and the response to SRIs treatment116. Further investigation in larger samples is warranted to determine the clinical utility of pharmacogenetic data in OCD.

OCD genetic architecture: an exploratory model

The data described in the previous sections suggest that genes involved in synaptic transmission, neurodevelopment, and the immune and inflammatory systems are involved in the pathophysiology of OCD (Figure 1). Based on these data, an exploratory analysis was undertaken with GeneNets in order to investigate whether genes associated with risk for OCD, identified in studies assessing common and rare variants, would form communities (i.e. clusters of functionally connected genes involved in a particular biological process)117. Three communities were significantly enriched for biological processes: related to glutamate neurotransmission, to cell adhesion, and to the immune system (Figure 4). These findings are consistent with a model in which multiple genes and biological pathways play a role in the pathogenesis of OCD and in which synaptic, neurodevelopmental, and immune pathways may be particularly important.

Figure 4. Gene networks exploratory analysis.

Figure 4.

The GeneNets algorithm was used to ascertain whether a set of obsessive-compulsive disorder (OCD) risk genes were more significantly connected to each other in a functional network as would be expected by chance alone. Accordingly, a total of 204 OCD-associated genes were selected from studies cited throughout the present review (more specifically, seven genome-wide association studies [GWASs]2931,35,36,41,42, two whole-exome sequencing [WES] studies45,46, two copy number variant [CNV] studies58,59, and one functional variants study100) for the GeneNets exploratory analysis. A total of 125 of these genes were included in a significant network, in which three communities were enriched for biological pathways. More specifically, the most significant pathways detected for those communities were the ionotropic activity of kainate receptors (P <10-6), associated with glutamate neurotransmission118 (Figure 4A); the cell adhesion-related processes (P <10-9), involved in synaptic processes119 and neurodevelopment120,121 (Figure 4B); the regulation of kit signaling (P <10-4), associated with immune function122 (Figure 4C); and the activation of GABAA receptors (P <10-12), implicated in neuropsychiatric disorders123. For the pathways displayed in the figure, the type of study from which their respective genes were selected is highlighted in the figure.

OCD genetics: current insights and future prospects

This review shows how several recent developments in genomics have contributed to understanding the genetic architecture of OCD. The use of cutting-edge methods has moved the field from a focus on candidate genes in underpowered samples to unbiased approaches in larger cohorts. At the same time, a great deal more work is needed to fully understand the role of common and rare gene variants in OCD. Work on gene methylation and expression has provided proof-of-principle demonstrations of the value of such studies and deserves expansion. Such work, taken together with careful phenotyping and comparison of individuals with different disorders, may lead to a better understanding of transdiagnostic genetic mechanisms.

The question of how advances in the genomics of OCD will become clinically useful remains to be answered. First-line pharmacological treatments for OCD target mostly serotonergic neurotransmission; biological pathways implicated in recent genomic studies, such as those related to glutamatergic, neurodevelopmental, and the immune and inflammatory systems, may be useful targets in the future. More robust pharmacogenetic evidence may ultimately improve the prediction of response to pharmacological treatments, advancing precision medicine for OCD. Shared genetic liability among OCD, ASD, and SCZ may suggest new approaches to transdiagnostic assessment of patients. That said, to date, genetic studies have not demonstrated clinical utility, suggesting that OCD is not a simple homogeneous entity but rather that the underlying biological pathways are complex, variable, and heterogeneous. We can expect that translation from bench to bedside, through continuous effort and collaborative work, will ultimately transform our understanding of what causes OCD and thus how best to treat it.

The peer reviewers who approve this article are:

  • Adriana Sampaio, Psychological Neuroscience Lab, CIPsi, School of Psychology, University of Minho, Braga, Portugal

  • Christopher Pittenger, OCD Research Clinic, Yale University, New Haven, Connecticut

Funding Statement

DJS has received research grants and/or consultancy Honoraria from Lundbeck and Sun. HBS has received research support for an industry-sponsored clinical trial from Biohaven Pharmaceuticals, royalties from UpToDate Inc., and a stipend from the American Medical Association for her role as Associate Editor of JAMA Psychiatry. RGS has received travel grants from Lundbeck and Libbs. The other authors have no funding information to declare.

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1. American Psychiatric Association: Diagnostic and statistical manual of mental disorders. 5th ed. 2013. 10.1176/appi.books.9780890425596 [DOI] [Google Scholar]
  • 2. Fontenelle LF, Mendlowicz MV, Versiani M: The descriptive epidemiology of obsessive-compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2006; 30(3): 327–37. 10.1016/j.pnpbp.2005.11.001 [DOI] [PubMed] [Google Scholar]
  • 3. Ruscio AM, Stein DJ, Chiu WT, et al. : The epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey Replication. Mol Psychiatry. 2010; 15(1): 53–63. 10.1038/mp.2008.94 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 4. Subramaniam M, Soh P, Vaingankar JA, et al. : Quality of life in obsessive-compulsive disorder: impact of the disorder and of treatment. CNS Drugs. 2013; 27(5): 367–83. 10.1007/s40263-013-0056-z [DOI] [PubMed] [Google Scholar]
  • 5. Torres AR, Prince MJ, Bebbington PE, et al. : Obsessive-compulsive disorder: prevalence, comorbidity, impact, and help-seeking in the British National Psychiatric Morbidity Survey of 2000. Am J Psychiatry. 2006; 163(11): 1978–85. 10.1176/ajp.2006.163.11.1978 [DOI] [PubMed] [Google Scholar]
  • 6. Meier SM, Mattheisen M, Mors O, et al. : Mortality Among Persons With Obsessive-Compulsive Disorder in Denmark. JAMA Psychiatry. 2016; 73(3): 268–74. 10.1001/jamapsychiatry.2015.3105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Hirschtritt ME, Bloch MH, Mathews CA: Obsessive-Compulsive Disorder: Advances in Diagnosis and Treatment. JAMA. 2017; 317(13): 1358–67. 10.1001/jama.2017.2200 [DOI] [PubMed] [Google Scholar]
  • 8. Erzegovesi S, Cavallini MC, Cavedini P, et al. : Clinical predictors of drug response in obsessive-compulsive disorder. J Clin Psychopharmacol. 2001; 21(5): 488–92. 10.1097/00004714-200110000-00006 [DOI] [PubMed] [Google Scholar]
  • 9. Guo SW: Familial aggregation of environmental risk factors and familial aggregation of disease. Am J Epidemiol. 2000; 151(11): 1121–31. 10.1093/oxfordjournals.aje.a010156 [DOI] [PubMed] [Google Scholar]
  • 10. Pauls DL, Alsobrook JP, 2nd, Goodman W, et al. : A family study of obsessive-compulsive disorder. Am J Psychiatry. 1995; 152(1): 76–84. 10.1176/ajp.152.1.76 [DOI] [PubMed] [Google Scholar]
  • 11. do Rosario-Campos MC, Leckman JF, Curi M, et al. : A family study of early-onset obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet. 2005; 136B(1): 92–7. 10.1002/ajmg.b.30149 [DOI] [PubMed] [Google Scholar]
  • 12. Mataix-Cols D, Boman M, Monzani B, et al. : Population-based, multigenerational family clustering study of obsessive-compulsive disorder. JAMA Psychiatry. 2013; 70(7): 709–17. 10.1001/jamapsychiatry.2013.3 [DOI] [PubMed] [Google Scholar]
  • 13. Brander G, Kuja-Halkola R, Rosenqvist MA, et al. : A population-based family clustering study of tic-related obsessive-compulsive disorder. Mol Psychiatry. 2019. 10.1038/s41380-019-0532-z [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 14. Visscher PM, Hill WG, Wray NR: Heritability in the genomics era--concepts and misconceptions. Nat Rev Genet. 2008; 9(4): 255–66. 10.1038/nrg2322 [DOI] [PubMed] [Google Scholar]
  • 15. Zai G, Barta C, Cath D, et al. : New insights and perspectives on the genetics of obsessive-compulsive disorder. Psychiatr Genet. 2019; 29(5): 142–51. 10.1097/YPG.0000000000000230 [DOI] [PubMed] [Google Scholar]
  • 16. Burton CL, Park LS, Corfield EC, et al. : Heritability of obsessive-compulsive trait dimensions in youth from the general population. Transl Psychiatry. 2018; 8(1): 191. 10.1038/s41398-018-0249-9 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 17. Ramamoorthy A, Yee SW, Karnes J: Unveiling the Genetic Architecture of Human Disease for Precision Medicine. Clin Transl Sci. 2019; 12(1): 3–5. 10.1111/cts.12593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Timpson NJ, Greenwood CMT, Soranzo N, et al. : Genetic architecture: the shape of the genetic contribution to human traits and disease. Nat Rev Genet. 2018; 19(2): 110–24. 10.1038/nrg.2017.101 [DOI] [PubMed] [Google Scholar]
  • 19. Duncan LE, Ostacher M, Ballon J: How genome-wide association studies (GWAS) made traditional candidate gene studies obsolete. Neuropsychopharmacology. 2019; 44(9): 1518–23. 10.1038/s41386-019-0389-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Sullivan PF, Geschwind DH: Defining the Genetic, Genomic, Cellular, and Diagnostic Architectures of Psychiatric Disorders. Cell. 2019; 177(1): 162–83. 10.1016/j.cell.2019.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 21. Horwitz T, Lam K, Chen Y, et al. : A decade in psychiatric GWAS research. Mol Psychiatry. 2019; 24(3): 378–89. 10.1038/s41380-018-0055-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Fadista J, Manning AK, Florez JC, et al. : The (in)famous GWAS P-value threshold revisited and updated for low-frequency variants. Eur J Hum Genet. 2016; 24(8): 1202–5. 10.1038/ejhg.2015.269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Collins AL, Sullivan PF: Genome-wide association studies in psychiatry: what have we learned? Br J Psychiatry. 2013; 202(1): 1–4. 10.1192/bjp.bp.112.117002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Yang J, Zeng J, Goddard ME, et al. : Concepts, estimation and interpretation of SNP-based heritability. Nat Genet. 2017; 49(9): 1304–10. 10.1038/ng.3941 [DOI] [PubMed] [Google Scholar]
  • 25. Zhang Z, Ma P, Li Q, et al. : Exploring the Genetic Correlation Between Growth and Immunity Based on Summary Statistics of Genome-Wide Association Studies. Front Genet. 2018; 9: 393. 10.3389/fgene.2018.00393 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 26. van Rheenen W, Peyrot WJ, Schork AJ, et al. : Genetic correlations of polygenic disease traits: from theory to practice. Nat Rev Genet. 2019; 20(10): 567–81. 10.1038/s41576-019-0137-z [DOI] [PubMed] [Google Scholar]
  • 27. Duncan L, Shen H, Gelaye B, et al. : Analysis of polygenic risk score usage and performance in diverse human populations. Nat Commun. 2019; 10(1): 3328. 10.1038/s41467-019-11112-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Reimand J, Isserlin R, Voisin V, et al. : Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc. 2019; 14(2): 482–517. 10.1038/s41596-018-0103-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Stewart SE, Yu D, Scharf JM, et al. : Genome-wide association study of obsessive-compulsive disorder. Mol Psychiatry. 2013; 18(7): 788–98. 10.1038/mp.2012.85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Mattheisen M, Samuels JF, Wang Y, et al. : Genome-wide association study in obsessive-compulsive disorder: results from the OCGAS. Mol Psychiatry. 2015; 20(3): 337–44. 10.1038/mp.2014.43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. International Obsessive Compulsive Disorder Foundation Genetics Collaborative (IOCDF-GC) and OCD Collaborative Genetics Association Studies (OCGAS): Revealing the complex genetic architecture of obsessive-compulsive disorder using meta-analysis. Mol Psychiatry. 2018; 23(5): 1181–8. 10.1038/mp.2017.154 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 32. Yuzaki M: Cerebellar LTD vs. motor learning-lessons learned from studying GluD2. Neural Netw. 2013; 47: 36–41. 10.1016/j.neunet.2012.07.001 [DOI] [PubMed] [Google Scholar]
  • 33. Aoki H, Hara A, Kunisada T: Induced haploinsufficiency of Kit receptor tyrosine kinase impairs brain development. JCI Insight. 2017; 2(19): e94385. 10.1172/jci.insight.94385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Khramtsova EA, Heldman R, Derks EM, et al. : Sex differences in the genetic architecture of obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet. 2019; 180(6): 351–64. 10.1002/ajmg.b.32687 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 35. den Braber A, Zilhão NR, Fedko IO, et al. : Obsessive-compulsive symptoms in a large population-based twin-family sample are predicted by clinically based polygenic scores and by genome-wide SNPs. Transl Psychiatry. 2016; 6(2): e731. 10.1038/tp.2015.223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Smit DJA, Cath D, Zilhão NR, et al. : Genetic meta-analysis of obsessive-compulsive disorder and self-report compulsive symptoms. Am J Med Genet B Neuropsychiatr Genet. 2020; 183(4): 208–216. 10.1002/ajmg.b.32777 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 37. Cross-Disorder Group of the Psychiatric Genomics Consortium: Genomic Relationships, Novel Loci, and Pleiotropic Mechanisms across Eight Psychiatric Disorders. Cell. 2019; 179(7): 1469–1482.e11. 10.1016/j.cell.2019.11.020 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 38. Anttila V, Bulik-Sullivan B, Finucane HK, et al. : Analysis of shared heritability in common disorders of the brain. Science. 2018; 360(6395): eaap8757. 10.1126/science.aap8757 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 39. Allen A, King A, Hollander E: Obsessive-compulsive spectrum disorders. Dialogues Clin Neurosci. 2003; 5(3): 259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. McIntosh AM, Sullivan PF, Lewis CM: Uncovering the Genetic Architecture of Major Depression. Neuron. 2019; 102(1): 91–103. 10.1016/j.neuron.2019.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 41. Costas J, Carrera N, Alonso P, et al. : Exon-focused genome-wide association study of obsessive-compulsive disorder and shared polygenic risk with schizophrenia. Transl Psychiatry. 2016; 6(3): e768. 10.1038/tp.2016.34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Guo W, Samuels JF, Wang Y, et al. : Polygenic risk score and heritability estimates reveals a genetic relationship between ASD and OCD. Eur Neuropsychopharmacol. 2017; 27(7): 657–666. 10.1016/j.euroneuro.2017.03.011 [DOI] [PubMed] [Google Scholar]
  • 43. Martin AR, Gignoux CR, Walters RK, et al. : Human Demographic History Impacts Genetic Risk Prediction across Diverse Populations. Am J Hum Genet. 2017; 100(4): 635–649. 10.1016/j.ajhg.2017.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Wang W, Corominas R, Lin GN: De novo Mutations From Whole Exome Sequencing in Neurodevelopmental and Psychiatric Disorders: From Discovery to Application. Front Genet. 2019; 10: 258. 10.3389/fgene.2019.00258 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 45. Cappi C, Brentani H, Lima L, et al. : Whole-exome sequencing in obsessive-compulsive disorder identifies rare mutations in immunological and neurodevelopmental pathways. Transl Psychiatry. 2016; 6(3): e764. 10.1038/tp.2016.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Cappi C, Oliphant ME, Péter Z, Zai G, et al. : De Novo Damaging DNA Coding Mutations Are Associated With Obsessive-Compulsive Disorder and Overlap With Tourette's Disorder and Autism. Biol Psychiatry. 2020; 87(12): 1035–1044. 10.1016/j.biopsych.2019.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Yang RB, Ng CKD, Wasserman SM, et al. : Identification of a novel family of cell-surface proteins expressed in human vascular endothelium. J Biol Chem. 2002; 277(48): 46364–73. 10.1074/jbc.M207410200 [DOI] [PubMed] [Google Scholar]
  • 48. Ronan JL, Wu W, Crabtree GR: From neural development to cognition: Unexpected roles for chromatin. Nat Rev Genet. 2013; 14(5): 347–59. 10.1038/nrg3413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Durak O, Gao F, Kaeser-Woo YJ, et al. : Chd8 mediates cortical neurogenesis via transcriptional regulation of cell cycle and Wnt signaling. Nat Neurosci. 2016; 19(11): 1477–1488. 10.1038/nn.4400 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 50. Jung H, Park H, Choi Y, et al. : Sexually dimorphic behavior, neuronal activity, and gene expression in Chd8-mutant mice. Nat Neurosci. 2018; 21: 1218–28. 10.1038/s41593-018-0208-z [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 51. Xu Q, Liu YY, Wang X, et al. : Autism-associated CHD8 deficiency impairs axon development and migration of cortical neurons. Mol Autism. 2018; 9: 65. 10.1186/s13229-018-0244-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Yasin H, Gibson WT, Langlois S, et al. : A distinct neurodevelopmental syndrome with intellectual disability, autism spectrum disorder, characteristic facies, and macrocephaly is caused by defects in CHD8. J Hum Genet. 2019; 64(4): 271–80. 10.1038/s10038-019-0561-0 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 53. Kita Y, Katayama Y, Shiraishi T, et al. : The Autism-Related Protein CHD8 Cooperates with C/EBPβ to Regulate Adipogenesis. Cell Rep. 2018; 23(7): 1988–2000. 10.1016/j.celrep.2018.04.050 [DOI] [PubMed] [Google Scholar]
  • 54. Liu D, Cao H, Kural KC, et al. : Integrative analysis of shared genetic pathogenesis by autism spectrum disorder and obsessive-compulsive disorder. Biosci Rep. 2019; 39(12): BSR20191942. 10.1042/BSR20191942 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 55. McCarroll SA, Altshuler DM: Copy-number variation and association studies of human disease. Nat Genet. 2007; 39(7 Suppl): S37–42. 10.1038/ng2080 [DOI] [PubMed] [Google Scholar]
  • 56. Levy RJ, Xu B, Gogos JA, et al. : Copy number variation and psychiatric disease risk. Methods Mol Biol. 2012; 838: 97–113. 10.1007/978-1-61779-507-7_4 [DOI] [PubMed] [Google Scholar]
  • 57. McGrath LM, Yu D, Marshall C, et al. : Copy number variation in obsessive-compulsive disorder and tourette syndrome: A cross-disorder study. J Am Acad Child Adolesc Psychiatry. 2014; 53(8): 910–9. 10.1016/j.jaac.2014.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Gazzellone MJ, Zarrei M, Burton CL, et al. : Uncovering obsessive-compulsive disorder risk genes in a pediatric cohort by high-resolution analysis of copy number variation. J Neurodev Disord. 2016; 8: 36. 10.1186/s11689-016-9170-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Grünblatt E, Oneda B, Ekici AB, et al. : High resolution chromosomal microarray analysis in paediatric obsessive-compulsive disorder. BMC Med Genomics. 2017; 10(1): 68. 10.1186/s12920-017-0299-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Prieto M, Folci A, Martin S: Post-translational modifications of the Fragile X Mental Retardation Protein in neuronal function and dysfunction. Mol Psychiatry. 2020; 25(8): 1688–1703. 10.1038/s41380-019-0629-4 [DOI] [PubMed] [Google Scholar]
  • 61. Szatkiewicz JP, O'Dushlaine C, Chen G, et al. : Copy number variation in schizophrenia in Sweden. Mol Psychiatry. 2014; 19(7): 762–73. 10.1038/mp.2014.40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Pinto D, Delaby E, Merico D, et al. : Convergence of genes and cellular pathways dysregulated in autism spectrum disorders. Am J Hum Genet. 2014; 94: 677–94. 10.1016/j.ajhg.2014.03.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Thapar A, Martin J, Mick E, et al. : Psychiatric gene discoveries shape evidence on ADHD's biology. Mol Psychiatry. 2016; 21(9): 1202–7. 10.1038/mp.2015.163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Zarrei M, Burton CL, Engchuan W, et al. : A large data resource of genomic copy number variation across neurodevelopmental disorders. NPJ Genom Med. 2019; 4: 26. 10.1038/s41525-019-0098-3 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 65. Kushki A, Anagnostou E, Hammill C, et al. : Examining overlap and homogeneity in ASD, ADHD, and OCD: a data-driven, diagnosis-agnostic approach. Transl Psychiatry. 2019; 9(1): 318. 10.1038/s41398-019-0631-2 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 66. Tung J, Gilad Y: Social environmental effects on gene regulation. Cell Mol Life Sci. 2013; 70(22): 4323–39. 10.1007/s00018-013-1357-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Smith M, Flodman PL: Expanded Insights Into Mechanisms of Gene Expression and Disease Related Disruptions. Front Mol Biosci. 2018; 5: 101. 10.3389/fmolb.2018.00101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Guan J, Cai JJ, Ji G, et al. : Commonality in dysregulated expression of gene sets in cortical brains of individuals with autism, schizophrenia, and bipolar disorder. Transl Psychiatry. 2019; 9(1): 152. 10.1038/s41398-019-0488-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 69. Frésard L, Smail C, Ferraro NM, et al. : Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts. Nat Med. 2019; 25(6): 911–919. 10.1038/s41591-019-0457-8 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 70. Piantadosi SC, Chamberlain BL, Glausier JR, et al. : Lower excitatory synaptic gene expression in orbitofrontal cortex and striatum in an initial study of subjects with obsessive compulsive disorder. Mol Psychiatry. 2019. 10.1038/s41380-019-0431-3 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 71. Rotge JY, Guehl D, Dilharreguy B, et al. : Meta-analysis of brain volume changes in obsessive-compulsive disorder. Biol Psychiatry. 2009; 65(1): 75–83. 10.1016/j.biopsych.2008.06.019 [DOI] [PubMed] [Google Scholar]
  • 72. Menzies L, Chamberlain SR, Laird AR, et al. : Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited. Neurosci Biobehav Rev. 2008; 32(3): 525–49. 10.1016/j.neubiorev.2007.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wang Y, Cheng C, Zhang Z, et al. : Blood-based dynamic genomic signature for obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet. 2018; 177(8): 709–16. [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 74. Kang CB, Hong Y, Dhe-Paganon S, et al. : FKBP family proteins: Immunophilins with versatile biological functions. Neurosignals. 2008; 16(4): 318–25. 10.1159/000123041 [DOI] [PubMed] [Google Scholar]
  • 75. Berse M, Bounpheng M, Huang X, et al. : Ubiquitin-dependent degradation of Id1 and Id3 is mediated by the COP9 signalosome. J Mol Biol. 2004; 343(2): 361–70. 10.1016/j.jmb.2004.08.043 [DOI] [PubMed] [Google Scholar]
  • 76. Schmitt A, Malchow B, Hasan A, et al. : The impact of environmental factors in severe psychiatric disorders. Front Neurosci. 2014; 8: 19. 10.3389/fnins.2014.00019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Idaghdour Y, Awadalla P: Exploiting gene expression variation to capture gene-environment interactions for disease. Front Genet. 2012; 3: 228. 10.3389/fgene.2012.00228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Ptak C, Petronis A: Epigenetic approaches to psychiatric disorders. Dialogues Clin Neurosci. 2010; 12(1): 25–35. 10.31887/DCNS.2010.12.1/cptak [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Nissen JB, Hansen CS, Starnawska A, et al. : DNA Methylation at the Neonatal State and at the Time of Diagnosis: Preliminary Support for an Association with the Estrogen Receptor 1, Gamma-Aminobutyric Acid B Receptor 1, and Myelin Oligodendrocyte Glycoprotein in Female Adolescent Patients with OCD. Front Psychiatry. 2016; 7: 35. 10.3389/fpsyt.2016.00035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Yue W, Cheng W, Liu Z, et al. : Genome-wide DNA methylation analysis in obsessive-compulsive disorder patients. Sci Rep. 2016; 6: 31333. 10.1038/srep31333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Alemany-Navarro M, Costas J, Real E, et al. : Do polygenic risk and stressful life events predict pharmacological treatment response in obsessive compulsive disorder? A gene-environment interaction approach. Transl Psychiatry. 2019; 9(1): 70. 10.1038/s41398-019-0410-0 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 82. Brander G, Pérez-Vigil A, Larsson H, et al. : Systematic review of environmental risk factors for Obsessive-Compulsive Disorder: A proposed roadmap from association to causation. Neurosci Biobehav Rev. 2016; 65: 36–62. 10.1016/j.neubiorev.2016.03.011 [DOI] [PubMed] [Google Scholar]
  • 83. Brander G, Rydell M, Kuja-Halkola R, et al. : Association of Perinatal Risk Factors With Obsessive-Compulsive Disorder: A Population-Based Birth Cohort, Sibling Control Study. JAMA Psychiatry. 2016; 73(11): 1135–44. 10.1001/jamapsychiatry.2016.2095 [DOI] [PubMed] [Google Scholar]
  • 84. Mahjani B, Klei L, Hultman CM, et al. : Maternal Effects as Causes of Risk for Obsessive-Compulsive Disorder. Biol Psychiatry. 2020; 87(12): 1045–51. 10.1016/j.biopsych.2020.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 85. Guloksuz S, van Os J, Rutten BPF: The Exposome Paradigm and the Complexities of Environmental Research in Psychiatry. JAMA Psychiatry. 2018; 75(10): 985–6. 10.1001/jamapsychiatry.2018.1211 [DOI] [PubMed] [Google Scholar]
  • 86. Dourlen P, Chapuis J, Lambert JC: Using High-Throughput Animal or Cell-Based Models to Functionally Characterize GWAS Signals. Curr Genet Med Rep. 2018; 6(3): 107–15. 10.1007/s40142-018-0141-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Grados M, Prazak M, Saif A, et al. : A review of animal models of obsessive-compulsive disorder: A focus on developmental, immune, endocrine and behavioral models. Expert Opin Drug Discov. 2016; 11(1): 27–43. 10.1517/17460441.2016.1103225 [DOI] [PubMed] [Google Scholar]
  • 88. Scherma M, Giunti E, Fratta W, et al. : Gene knockout animal models of depression, anxiety and obsessive compulsive disorders. Psychiatr Genet. 2019; 29(5): 191–9. 10.1097/YPG.0000000000000238 [DOI] [PubMed] [Google Scholar]
  • 89. Delgado-Acevedo C, Estay SF, Radke AK, et al. : Behavioral and synaptic alterations relevant to obsessive-compulsive disorder in mice with increased EAAT3 expression. Neuropsychopharmacology. 2019; 44(6): 1163–73. 10.1038/s41386-018-0302-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 90. Zike ID, Chohan MO, Kopelman JM, et al. : OCD candidate gene SLC1A1/EAAT3 impacts basal ganglia-mediated activity and stereotypic behavior. Proc Natl Acad Sci U S A. 2017; 114(22): 5719–24. 10.1073/pnas.1701736114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Gerentes M, Pelissolo A, Rajagopal K, et al. : Obsessive-Compulsive Disorder: Autoimmunity and Neuroinflammation. Curr Psychiatry Rep. 2019; 21(8): 78. 10.1007/s11920-019-1062-8 [DOI] [PubMed] [Google Scholar]
  • 92. Krabbe G, Minami SS, Etchegaray JI, et al. : Microglial NFκB-TNFα hyperactivation induces obsessive-compulsive behavior in mouse models of progranulin-deficient frontotemporal dementia. Proc Natl Acad Sci U S A. 2017; 114(19): 5029–34. 10.1073/pnas.1700477114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Tränkner D, Boulet A, Peden E, et al. : A Microglia Sublineage Protects from Sex-Linked Anxiety Symptoms and Obsessive Compulsion. Cell Rep. 2019; 29(4): 791–799.e3. 10.1016/j.celrep.2019.09.045 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 94. Ullrich M, Weber M, Post AM, et al. : OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency. Mol Psychiatry. 2018; 23(2): 444–58. 10.1038/mp.2016.232 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 95. Phoenix TN, Temple S: Spred1, a negative regulator of Ras-MAPK-ERK, is enriched in CNS germinal zones, dampens NSC proliferation, and maintains ventricular zone structure. Genes Dev. 2010; 24(4): 45–56. 10.1101/gad.1839510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. van de Vondervoort I, Poelmans G, Aschrafi A, et al. : An integrated molecular landscape implicates the regulation of dendritic spine formation through insulin-related signalling in obsessive-compulsive disorder. J Psychiatry Neurosci. 2016; 41(4): 280–5. 10.1503/jpn.140327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Bralten J, Widomska J, Witte WD, et al. : Shared genetic etiology between obsessive-compulsive disorder, obsessive-compulsive symptoms in the population, and insulin signaling. Transl Psychiatry. 2020; 10(1): 121. 10.1038/s41398-020-0793-y [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 98. van de Vondervoort IIGM, Amiri H, Bruchhage MMK, et al. : Converging evidence points towards a role of insulin signaling in regulating compulsive behavior. Transl Psychiatry. 2019; 9(1): 225. 10.1038/s41398-019-0559-6 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 99. Casey BJ, Glatt CE, Lee FS: Treating the Developing versus Developed Brain: Translating Preclinical Mouse and Human Studies. Neuron. 2015; 86(6): 1358–68. 10.1016/j.neuron.2015.05.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Noh HJ, Tang R, Flannick J, et al. : Integrating evolutionary and regulatory information with a multispecies approach implicates genes and pathways in obsessive-compulsive disorder. Nat Commun. 2017; 8(1): 774. 10.1038/s41467-017-00831-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Viswanath B, Rao NP, Narayanaswamy JC, et al. : Discovery biology of neuropsychiatric syndromes (DBNS): A center for integrating clinical medicine and basic science. BMC Psychiatry. 2018; 18(1): 106. 10.1186/s12888-018-1674-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Rodríguez N, Morer A, González-Navarro EA, et al. : Inflammatory dysregulation of monocytes in pediatric patients with obsessive-compulsive disorder. J Neuroinflammation. 2017; 14(1): 261. 10.1186/s12974-017-1042-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Özyurt G, Binici NC: The neutrophil-lymphocyte ratio and platelet-lymphocyte ratio in adolescent obsessive-compulsive disorder: Does comorbid anxiety disorder affect inflammatory response? Psychiatry Res. 2019; 272: 311–5. 10.1016/j.psychres.2018.12.131 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 104. Bearden CE, Thompson PM: Emerging Global Initiatives in Neurogenetics: The Enhancing Neuroimaging Genetics through Meta-analysis (ENIGMA) Consortium. Neuron. 2017; 94(2): 232–6. 10.1016/j.neuron.2017.03.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Hibar DP, Cheung JW, Medland SE, 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. 10.1192/bjp.2018.62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Tang W, Zhu Q, Gong X, et al. : Cortico-striato-thalamo-cortical circuit abnormalities in obsessive-compulsive disorder: A voxel-based morphometric and fMRI study of the whole brain. Behav Brain Res. 2016; 313: 17–22. 10.1016/j.bbr.2016.07.004 [DOI] [PubMed] [Google Scholar]
  • 107. Denys D, Mantione M, Figee M, et al. : Deep brain stimulation of the nucleus accumbens for treatment-refractory obsessive-compulsive disorder. Arch Gen Psychiatry. 2010; 67(10): 1061–8. 10.1001/archgenpsychiatry.2010.122 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 108. Mulligan KA, Cheyette BNR: Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. Mol Neuropsychiatry. 2017; 2(4): 219–46. 10.1159/000453266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Chamberlain SR, Menzies L: Endophenotypes of obsessive-compulsive disorder: Rationale, evidence and future potential. Expert Rev Neurother. 2009; 9(8): 1133–46. 10.1586/ern.09.36 [DOI] [PubMed] [Google Scholar]
  • 110. Gottesman II, Gould TD: The endophenotype concept in psychiatry: Etymology and strategic intentions. Am J Psychiatry. 2003; 160(4): 636–45. 10.1176/appi.ajp.160.4.636 [DOI] [PubMed] [Google Scholar]
  • 111. Iacono WG: Endophenotypes in psychiatric disease: Prospects and challenges. Genome Med. 2018; 10(1): 11. 10.1186/s13073-018-0526-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Zartaloudi E, Laws KR, Bramon E: Endophenotypes of executive functions in obsessive compulsive disorder? A meta-analysis in unaffected relatives. Psychiatr Genet. 2019; 29(6): 211–9. 10.1097/YPG.0000000000000241 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 113. Brown L, Eum S, Haga SB, et al. : Clinical Utilization of Pharmacogenetics in Psychiatry - Perspectives of Pharmacists, Genetic Counselors, Implementation Science, Clinicians, and Industry. Pharmacopsychiatry. 2020; 53(4): 162–73. 10.1055/a-0975-9595 [DOI] [PubMed] [Google Scholar]
  • 114. Zai G, Brandl EJ, Müller DJ, et al. : Pharmacogenetics of antidepressant treatment in obsessive-compulsive disorder: An update and implications for clinicians. Pharmacogenomics. 2014; 15(8): 1147–57. 10.2217/pgs.14.83 [DOI] [PubMed] [Google Scholar]
  • 115. Qin H, Samuels JF, Wang Y, et al. : Whole-genome association analysis of treatment response in obsessive-compulsive disorder. Mol Psychiatry. 2016; 21(2): 270–6. 10.1038/mp.2015.32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lisoway AJ, Zai G, Tiwari AK, et al. : Pharmacogenetic evaluation of a DISP1 gene variant in antidepressant treatment of obsessive-compulsive disorder. Hum Psychopharmacol. 2018; 33(4): e2659. 10.1002/hup.2659 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 117. Li T, Kim A, Rosenbluh J, et al. : GeNets: A unified web platform for network-based genomic analyses. Nat Methods. 2018; 15(7): 543–6. 10.1038/s41592-018-0039-6 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 118. Falcón-Moya R, Losada-Ruiz P, Rodríguez-Moreno A: Kainate Receptor-Mediated Depression of Glutamate Release Involves Protein Kinase A in the Cerebellum. Int J Mol Sci. 2019; 20(17): 4124. 10.3390/ijms20174124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Missler M, Südhof TC, Biederer T: Synaptic cell adhesion. Cold Spring Harb Perspect Biol. 2012; 4(4): a005694. 10.1101/cshperspect.a005694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Solecki DJ: Sticky situations: Recent advances in control of cell adhesion during neuronal migration. Curr Opin Neurobiol. 2012; 22(5): 791–8. 10.1016/j.conb.2012.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Cristino AS, Williams SM, Hawi Z, et al. : Neurodevelopmental and neuropsychiatric disorders represent an interconnected molecular system. Mol Psychiatry. 2014; 19(3): 294–301. 10.1038/mp.2013.16 [DOI] [PubMed] [Google Scholar]
  • 122. Oriss TB, Krishnamoorthy N, Ray P, et al. : Dendritic cell c-kit signaling and adaptive immunity: Implications for the upper airways. Curr Opin Allergy Clin Immunol. 2014; 14(1): 7–12. 10.1097/ACI.0000000000000019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Kolata SM, Nakao K, Jeevakumar V, et al. : Neuropsychiatric Phenotypes Produced by GABA Reduction in Mouse Cortex and Hippocampus. Neuropsychopharmacology. 2018; 43(6): 1445–56. 10.1038/npp.2017.296 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation

Articles from Faculty Reviews are provided here courtesy of Faculty Opinions Ltd.

RESOURCES