Hostname: page-component-7bb8b95d7b-5mhkq Total loading time: 0 Render date: 2024-10-04T16:05:07.141Z Has data issue: false hasContentIssue false

Genetic and molecular correlates of cortical thickness alterations in adults with obsessive–compulsive disorder: a transcription–neuroimaging association analysis

Published online by Cambridge University Press:  04 October 2024

Da Zhang*
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Changjun Teng
Affiliation:
Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Yinhao Xu
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Lei Tian
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Ping Cao
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Xiao Wang
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Zonghong Li
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Chengbin Guan
Affiliation:
Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Xiao Hu
Affiliation:
Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
*
Corresponding author: Da Zhang; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Background

Although numerous neuroimaging studies have depicted neural alterations in individuals with obsessive–compulsive disorder (OCD), a psychiatric disorder characterized by intrusive cognitions and repetitive behaviors, the molecular mechanisms connecting brain structural changes and gene expression remain poorly understood.

Methods

This study combined the Allen Human Brain Atlas dataset with neuroimaging data from the Meta-Analysis (ENIGMA) consortium and independent cohorts. Later, partial least squares regression and enrichment analysis were performed to probe the correlation between transcription and cortical thickness variation among adults with OCD.

Results

The cortical map of case-control differences in cortical thickness was spatially correlated with cortical expression of a weighted combination of genes enriched for neurobiologically relevant ontology terms preferentially expressed across different cell types and cortical layers. These genes were specifically expressed in brain tissue, spanning all cortical developmental stages. Protein–protein interaction analysis revealed that these genes coded a network of proteins encompassing various highly interactive hubs.

Conclusions

The study findings bridge the gap between neural structure and transcriptome data in OCD, fostering an integrative understanding of the potential biological mechanisms.

Type
Original Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

Obsessive–compulsive disorder (OCD) is a heterogeneous psychiatric condition characterized by persistent intrusive thoughts (obsessions) and compulsive behaviors (Schulze, Kathmann, & Reuter, Reference Schulze, Kathmann and Reuter2018). OCD is estimated to affect 1–3% of the adult population over their lifetime and is related to significant personal impairment and socioeconomic burden (Stein et al., Reference Stein, Costa, Lochner, Miguel, Reddy, Shavitt and Simpson2019). Due to the unsatisfactory response rates of first-line treatments against OCD and its elusive etiology, there is an urgent need for further research into the biological underpinnings of the disorder to identify biomarkers and support prediction, leading to effective treatments (Goodman, Storch, & Sheth, Reference Goodman, Storch and Sheth2021; Robbins, Vaghi, & Banca, Reference Robbins, Vaghi and Banca2019).

The brain, the material foundation for cognition and behavior, undergoes rapid development via a dynamic process with region-specific patterns (Bethlehem et al., Reference Bethlehem, Seidlitz, White, Vogel, Anderson, Adamson and Alexander-Bloch2022; Qiu, Zhang, Kennedy, & Lee, Reference Qiu, Zhang, Kennedy and Lee2021). Neuroimaging studies have revealed that cortical thickness is a valuable measure to assess neuroanatomical patterns associated with neurodegeneration, conferring particular sensitivity for detecting subtle, previously unrecognized anatomical changes (Amlien et al., Reference Amlien, Fjell, Tamnes, Grydeland, Krogsrud, Chaplin and Walhovd2016; Sydnor et al., Reference Sydnor, Larsen, Bassett, Alexander-Bloch, Fair, Liston and Satterthwaite2021; Walhovd, Fjell, Giedd, Dale, & Brown, Reference Walhovd, Fjell, Giedd, Dale and Brown2017). Previous neuroimaging studies on OCD have reported altered cortical thickness, primarily involving the fronto-limbic and fronto-parietal regions (Kühn et al., Reference Kühn, Kaufmann, Simon, Endrass, Gallinat and Kathmann2013; Shin et al., Reference Shin, Yoo, Lee, Ha, Lee, Lee and Kwon2007), associated with cognitive control and emotional regulation. A recent meta-analysis on OCD patients identified altered cortical thickness in the superior and inferior frontal regions, precentral, posterior cingulate, middle temporal, inferior parietal, and precuneus gyri (Fouche et al., Reference Fouche, du Plessis, Hattingh, Roos, Lochner, Soriano-Mas and van den Heuvel2017). These findings indicate the role of cortical abnormalities in OCD pathophysiology and their association with clinical symptoms. However, the molecular mechanisms underlying such brain structural alterations remain unknown.

Genetic factors play a significant role in brain development, with various disease-related imaging-derived phenotypes demonstrating high heritability (Arnatkeviciute, Fulcher, Bellgrove, & Fornito, Reference Arnatkeviciute, Fulcher, Bellgrove and Fornito2021; Fornito, Arnatkevičiūtė, & Fulcher, Reference Fornito, Arnatkevičiūtė and Fulcher2019; Pizzagalli et al., Reference Pizzagalli, Auzias, Yang, Mathias, Faskowitz, Boyd and Jahanshad2020; Thompson et al., Reference Thompson, Cannon, Narr, van Erp, Poutanen, Huttunen and Toga2001). Similarly, OCD is a heritable disorder involving multiple genetic alterations and possibly affecting different molecular pathways, resulting in heterogeneous phenotypes (Alemany-Navarro et al., Reference Alemany-Navarro, Cruz, Real, Segalàs, Bertolín, Rabionet and Alonso2020; Mahjani, Bey, Boberg, & Burton, Reference Mahjani, Bey, Boberg and Burton2021; Monzani, Rijsdijk, Harris, & Mataix-Cols, Reference Monzani, Rijsdijk, Harris and Mataix-Cols2014). Thus, genetic factors can contribute to alterations in molecular pathways that result in the structural abnormalities seen in OCD. While traditional imaging genetics analyses have underscored the substantial influence of genetic factors on a fewer of brain phenotypes associated with OCD, the intermediary mechanisms involved remain undefined (Atmaca et al., Reference Atmaca, Onalan, Yildirim, Yuce, Koc, Korkmaz and Mermi2011; Gassó et al., Reference Gassó, Ortiz, Mas, Morer, Calvo, Bargalló and Lázaro2015; Sinopoli et al., Reference Sinopoli, Erdman, Burton, Easter, Rajendram, Baldwin and Arnold2020; Wu et al., Reference Wu, Hanna, Easter, Kennedy, Rosenberg and Arnold2013). Gene expression constitutes the most essential molecular mechanism by which genes govern the differentiation, development, and functionality of brain cells and tissues (Naumova, Lee, Rychkov, Vlasova, & Grigorenko, Reference Naumova, Lee, Rychkov, Vlasova and Grigorenko2013).

Recent developments in neuroimaging and transcriptomics make it possible to directly investigate the association of neuroimaging-detected alterations with gene expression patterns across distinct brain regions (Atmaca et al., Reference Atmaca, Onalan, Yildirim, Yuce, Koc, Korkmaz and Mermi2011; Grünblatt, Hauser, & Walitza, Reference Grünblatt, Hauser and Walitza2014). The whole-brain atlas of gene expression derived from the Allen Human Brain Atlas (AHBA) database has proven instrumental for delving into disease-related gene expression and the molecular basis of brain abnormalities in various psychiatric disorders (M. Hawrylycz et al., Reference Hawrylycz, Miller, Menon, Feng, Dolbeare, Guillozet-Bongaarts and Lein2015; Li et al., Reference Li, Seidlitz, Suckling, Fan, Ji, Meng and Liao2021; Morgan et al., Reference Morgan, Seidlitz, Whitaker, Romero-Garcia, Clifton, Scarpazza and Bullmore2019; Sun et al., Reference Sun, Huang, Wang, Xu, Zhang, Zhou and Qian2023; Sunkin et al., Reference Sunkin, Ng, Lau, Dolbeare, Gilbert, Thompson and Dang2013; Xue et al., Reference Xue, Guo, Zhu, Liang, Xu, Ma and Liu2023), offering new avenues for deciphering the molecular basis of OCD. Saraiva, Sato, and Cappi (Reference Saraiva, Sato and Cappi2022) illustrate this approach, proposing a model associating connectome alterations in OCD with AHBA expression data to develop personalized therapeutic mechanisms.

Based on this innovative landscape, the present study attempts to bridge the gap between cortical thickness alterations within OCD and underlying gene expression patterns, employing a novel transcriptome–neuroimaging spatial correlation analysis to pinpoint genes whose expression levels correlate with cortical thickness changes in OCD. Through comprehensive enrichment analyses, these genes could be mapped to molecular pathways, cellular classifications, cortical layers, and protein–protein interactions (PPIs), providing a multi-faceted view of the pathophysiology of OCD. Figure 1 illustrates our analytical approach, underscoring the potential of integrating neuroimaging and genomic data to elucidate the complex biological mechanisms of psychiatric disorders. This could pave the way for more effective and personalized treatment modalities.

Figure 1. Schematic workflow of the analytical framework. (a) PLS regression was conducted to link cortical thickness abnormalities of OCD with gene expression data, the overlapped genes were identified by taking the intersection of the significant gene list from the first PLS component (PLS1) from both ENIGMA and independent database. (b) Enrichment analyses were performed subsequently on the overlapped genes. AHBA, Allen Human Brain Atlas; CSEA, cell-type-specific expression analysis; OCD, obsessive–compulsive disorder; PPI, protein–protein interaction; PLS, partial least squares; TSEA, tissue-type-specific expression analysis.

ENIGMA participants

For the transcriptional analyses, we used publicly accessible multi-site summary statistics published by the ENIGMA Consortium (available through the ENIGMA Toolbox: https://github.com/MICA-MNI/ENIGMA) (Larivière et al., Reference Larivière, Paquola, Park, Royer, Wang, Benkarim and Bernhardt2021). The ENIGMA-OCD working group comprises 38 datasets from 27 international research institutions, including neuroimaging and clinical data from OCD patients and the healthy controls (HCs) (Boedhoe et al., Reference Boedhoe, Schmaal, Abe, Alonso, Ameis and Anticevic2018). Our analysis was limited to adults to account for age-related variation in cortical structure. We analyzed data from 2934 individuals, including 1498 OCD patients and 1436 HCs. Our analyses were based on covariate-adjusted case-control differences, employing random-effects meta-analyses of z-scored effect sizes (Cohen's d) for cortical thickness. It should be noted that the Desikan–Killiany Atlas (Desikan et al., Reference Desikan, Ségonne, Fischl, Quinn, Dickerson, Blacker and Killiany2006) (consisting of 68 cortical regions) is the sole representation of the ENIGMA datasets. However, we converted the data to the Human Connectome Project (HCP) multi-modal parcellation 1.0 atlas (180 parcellations per hemisphere) for the subsequent analyses (Glasser et al., Reference Glasser, Coalson, Robinson, Hacker, Harwell, Yacoub and Van Essen2016). This atlas enhances our analysis by providing a higher resolution of brain structure, enabling more precise localization of cortical changes associated with OCD. Detailed information regarding magnetic resonance imaging (MRI) acquisition and sample demographics has been previously reported (Boedhoe et al., Reference Boedhoe, Schmaal, Abe, Alonso, Ameis and Anticevic2018).

Independent dataset participants

Cortical thickness effect sizes (Cohen's d) were z-scored and validated within an independent cohort of 57 OCD patients and 57 well-matched HCs. OCD patients were enrolled consecutively from inpatient and outpatient units of the Affiliated Brain Hospital of Nanjing Medical University. HCs with no history of severe medical or neuropsychiatric illnesses were recruited from the local community. This study was approved by the ethics committee of the Affiliated Brain Hospital of Nanjing Medical University, and all the participants provided written informed consent. Demographic and clinical characteristics of the independent sample are presented in online Supplementary Table S1. Details of the inclusion and exclusion criteria, MRI data acquisition parameters, and cortical thickness measurement methods are provided in the online Supplementary materials.

Assessment of regional gene expression

Brain-wide gene expression data were procured from the publicly accessible AHBA database (http://www.brain-map.org) (M. J. Hawrylycz et al., Reference Hawrylycz, Lein, Guillozet-Bongaarts, Shen, Ng, Miller and Jones2012; M. Hawrylycz et al., Reference Hawrylycz, Miller, Menon, Feng, Dolbeare, Guillozet-Bongaarts and Lein2015), which provides comprehensive and high-resolution coverage of normalized microarray expression data across six donated post-mortem brains (online Supplementary Table S2). The original expression data, which encompasses more than 20 000 genes from 3702 spatially distinct brain tissue samples, were processed with the following analytic pipeline (Arnatkeviciute, Fulcher, & Fornito, Reference Arnatkeviciute, Fulcher and Fornito2019): (1) probe-to-gene annotations were confirmed using Re-annotator (Arloth, Bader, Röh, & Altmann, Reference Arloth, Bader, Röh and Altmann2015); (2) probes falling below the background noise intensity in over half of the samples were filtered out; (3) the most RNA-seq-correlated probe for each gene was chosen; (4) samples were assigned to the HCP-360 atlas within a 2 mm Euclidean distance of the parcel (Glasser et al., Reference Glasser, Coalson, Robinson, Hacker, Harwell, Yacoub and Van Essen2016); (5) expression measures were normalized with a scaled robust sigmoid for each participant; and (6) gene set filtering was performed according to differential stability (DS). We recruited the top 50% of genes with the highest DS values to conserve gene expression across subjects for transcription–neuroimaging spatial correlations (M. Hawrylycz et al., Reference Hawrylycz, Miller, Menon, Feng, Dolbeare, Guillozet-Bongaarts and Lein2015). To assess the impact of various DS threshold selections, we performed sensitivity analyses using two alternative DS cutoff thresholds: the upper 40% and 60%. The counts of residual probes and genes at each processing stage are presented in online Supplementary Fig. S1. Finally, the transcriptional level of each gene at each brain region was determined to obtain a matrix (360 regions × 5013 gene expression levels) for subsequent analyses.

Regional cortical thickness alterations and gene expression

The correlation between the transcriptional activity of all 5013 genes and regional changes in cortical thickness (represented by Cohen's d-values from 360 cortical areas) was examined with partial least squares (PLS) regression (Abdi & Williams, Reference Abdi and Williams2013). The z-score normalized gene expression matrix (360 regions × 5013 genes) of this model predicts the z-score normalized Cohen's d-values of cortical thickness variation (response variable). The primary component of the PLS regression result (PLS1) represents a linear combination of gene expression values exhibiting the most significant correlation with regional alterations in cortical thickness. To validate the significance of PLS1 in elucidating the covariance between cortical thickness changes and genome-wide expression, we performed permutation testing using 5000 iterations with spherical rotations to control for spatial autocorrelation (Li et al., Reference Li, Seidlitz, Suckling, Fan, Ji, Meng and Liao2021; Romero-Garcia et al., Reference Romero-Garcia, Seidlitz, Whitaker, Morgan, Fonagy, Dolan and Bullmore2020; Váša et al., Reference Váša, Seidlitz, Romero-Garcia, Whitaker, Rosenthal, Vértes and Bullmore2018). Subsequently, the PLS weight of each gene was evaluated for variability using 5000 bootstrap replications. Then, a z-score was computed for each gene using the ratio of its PLS weight to the standard error from bootstrapping (online Supplementary File 1) (Morgan et al., Reference Morgan, Seidlitz, Whitaker, Romero-Garcia, Clifton, Scarpazza and Bullmore2019). Only genes with significant correlations (Benjamini–Hochberg false-discovery rate [BH-FDR] procedure p < 0.05) were considered for additional analysis (Bigdeli et al., Reference Bigdeli, Lee, Webb, Riley, Vladimirov, Fanous and Bacanu2016). This approach yielded two gene lists: genes depicting a significant positive correlation with regional cortical thickness changes (PLS1+) and another with genes showing a significant negative correlation (PLS1−). Transcriptome–neuroimaging association analysis was performed separately for Cohen's d-values map from ENIGMA and independent datasets.

Enrichment analyses

Various enrichment analyses were performed for significant PLS+/− genes associated with OCD pathogenesis. First, functional annotation was performed using WebGestalt (https://www.webgestalt.org/) (Liao, Wang, Jaehnig, Shi, & Zhang, Reference Liao, Wang, Jaehnig, Shi and Zhang2019). Gene ontology (GO) was used to ascertain biological functions, encompassing molecular functions (MFs), biological processes (BPs), and cellular components (CCs) (Thomas et al., Reference Thomas, Ebert, Muruganujan, Mushayahama, Albou and Mi2022). The Kyoto Encyclopedia of Genes and Genomes (KEGG) helped identify relevant biological pathways (Kanehisa, Furumichi, Tanabe, Sato, & Morishima, Reference Kanehisa, Furumichi, Tanabe, Sato and Morishima2017). Second, online tools for Tissue-type-Specific Expression Analysis (TSEA) and Cell-type-Specific Expression Analysis (CSEA) (http://genetics.wustl.edu/jdlab/tsea/ and http://genetics.wustl.edu/jdlab/csea-tool-3/) helped conduct specialized analyses for tissue and temporal expression (Dougherty, Schmidt, Nakajima, & Heintz, Reference Dougherty, Schmidt, Nakajima and Heintz2010; Xu, Wells, O'Brien, Nehorai, & Dougherty, Reference Xu, Wells, O'Brien, Nehorai and Dougherty2014). Third, cell-type-specific expression was analyzed to determine the specific cell types associated with the gene set. Gene expression data for specific cell types in the human brain, including neurons, astrocytes, oligodendrocytes, microglia, and macrophages, were obtained from the CellMarker database (Zhang et al., Reference Zhang, Lan, Xu, Quan, Zhao, Deng and Xiao2019). Cortical layer enrichment was executed with marker genes from a previous transcriptomic study (He et al., Reference He, Han, Efimova, Guijarro, Yu, Oleksiak and Khaitovich2017). Fourth, PPI analysis was performed using STRING v12.0 (https://string-db.org/) (Szklarczyk et al., Reference Szklarczyk, Kirsch, Koutrouli, Nastou, Mehryary, Hachilif and von Mering2023). Lastly, we examined the overlap between genes associated with alterations in cortical thickness in OCD patients identified in the present study and OCD-associated genes within the MalaCards (https://www.malacards.org/) and Obsessive–Compulsive Disorder databases (http://alpha.dmi.unict.it/ocdb/) (Privitera et al., Reference Privitera, Distefano, Wefer, Ferro, Pulvirenti and Giugno2015; Rappaport et al., Reference Rappaport, Twik, Plaschkes, Nudel, Iny Stein, Levitt and Lancet2017). Further details of these enrichment analyses are provided in the online Supplementary materials.

Validation analysis

The primary analysis focused on genes in the top 50% of the highest differential stability scores to obtain consistent expression patterns across six donors. To ensure robustness in processing brain gene expression data, we also explored the effects of varying DS cutoff thresholds, the top 40% and 60% (Chen et al., Reference Chen, Zhang, Wang, Jiang, Cai, Zhao and Yu2022; Sun et al., Reference Sun, Huang, Wang, Xu, Zhang, Zhou and Qian2023). Gene selection was based on the intersection of findings from two independent transcriptome–neuroimaging association analyses. This intersection approach strengthens the confidence in the identified genes, combining results from different analyses, thereby enhancing the reliability and robustness of the findings. Considering the potential confounding effects of spatial autocorrelation, a spin based method permutation tests (5000 times) were performed to preserve the spatial covariance structure of the data (https://github.com/frantisekvasa/rotate_parcellation) (Váša et al., Reference Váša, Seidlitz, Romero-Garcia, Whitaker, Rosenthal, Vértes and Bullmore2018).

Results

Genes associated with cortical thickness alterations in OCD patients

The study possessed sufficient statistical power to identify subtle cortical abnormalities in OCD. The Cohen's d-values map, produced via the ENIGMA Toolbox (Larivière et al., Reference Larivière, Paquola, Park, Royer, Wang, Benkarim and Bernhardt2021), illustrates case-control differences in cortical thickness across databases (Fig. 2a). The spatial correlation analysis revealed a slight negative correlation (Pearson's r (358) = −0.13, p spin = 0.042, Fig. 2b) between the Cohen's d-value map of case-control cortical thickness differences within the two datasets. In the PLS regression analysis, gene expression in brain regions demonstrated significant correlations with inter-group differences in cortical thickness (ENIGMA dataset, Pearson's r (358) = 0.44, p spin < 0.0001; independent dataset, Pearson's r (358) = 0.28, p spin < 0.0001, Fig. 3a, b), utilizing a DS threshold of 50%. By ranking the normalized weights of the first PLS component using univariate z-tests (p < 0.05, FDR-corrected), we identified 1922 genes significantly connected with case-control cortical thickness changes in the ENIGMA dataset (PLS1 explained 28.3% of the variance) and 1045 in the independent dataset (PLS1 explained 22.5% of the variance), of which 609 genes overlapped (Fig. 3c, online Supplementary File 1). Significant intersections were also observed between the genes in the principal analysis and those identified using the two alternative DS thresholds of 40% (with an overlap ratio of 98.93%) and 60% (with an overlap ratio of 82.65%) (online Supplementary File 2).

Figure 2. Cortical thickness alterations between patients with OCD and HCs. (a) Cohen's d maps indicating case-control differences in cortical thickness. (b) Region-wise Pearson's correlation between Cohen's d maps from the two datasets (Pearson's r = −0.13, p spin = 0.042). HC, healthy control; OCD, obsessive–compulsive disorder.

Figure 3. Gene expression profiles related to case-control differences in cortical thickness. (a) Cortical map of regional PLS1 scores. (b) Scatterplot of regional PLS1 scores (weighted sum of 5013 gene expression scores) v. case-control differences in regional cortical thickness. (c) Ranked PLS1 gene expression weights and the overlapped genes of the two datasets (609 genes are overlapped).

Enrichment for biological functions

The GO and KEGG pathways were aligned with the overlapped gene list using WebGestalt (Liao et al., Reference Liao, Wang, Jaehnig, Shi and Zhang2019). The most significant GO biological function and KEGG pathway enrichment terms are presented in Fig. 4. The genes associated with cortical thickness alterations were enriched for BPs, including regulation of ion transport across cell membranes, cellular responses to environmental stimuli, regulation of metal ion transport, neuron ensheathment, amyloid precursor protein metabolism, glutamate receptor signaling, glutamatergic synaptic transmission, and sodium ion transport. MFs enriched among these genes comprised of cell adhesion mediator activity, while the enriched CCs included synaptic membranes, axon segments, pigment granules, cell–cell junctions, and neuronal cell bodies. KEGG pathway analysis indicated significant cortical thickness alteration-related gene enrichment in the phospholipase D signaling pathway.

Figure 4. Biological function analysis the genes associated with cortical thickness alterations in OCD patients. Only items passed the BH-FDR-corrected threshold (p < 0.05) are displayed in the bubble chart. The x-axis shows the gene ratio of each item, the y-axis shows GO terms or KEGG pathway, the bubble shapes representing different categories, the bubble size indicates the intersecting number of genes in the category, and the color bar represents the −log10(P). BH-FDR, Benjamini–Hochberg false-discovery rate; BP, biological process; CC, cellular component; GO, gene ontology; KEGG, Kyoto Encyclopedia of genes and genomes; MF, molecular function; OCD, obsessive–compulsive disorder.

Tissue-specific and temporal-specific expression analyses

TSEA demonstrated that the 609 genes expressed specific brain tissue expression (p = 8.00 × 10−14, BH-FDR-corrected) under a pSI threshold of 0.05 (Fig. 5a and online Supplementary File 3). We conducted a temporal-specific expression analysis using the CSEA tool (Xu et al., Reference Xu, Wells, O'Brien, Nehorai and Dougherty2014), comparing the selected gene list to developmental enrichment profiles. Developmental gene expression analysis indicated that these genes were expressed in the brain from early/mid-fetal development onward and across several brain regions comprising the cortex and subcortex (such as the thalamus, striatum, amygdala, and hippocampus). Expression remained stable in the cortex from early-/mid-fetal stages through young adulthood. However, expression was reduced in the hippocampus, striatum, and thalamus during early adulthood (Fig. 5b and online Supplementary File 3).

Figure 5. Specific expression analysis of the genes associated with cortical thickness alterations in OCD patients. (a) Tissue-type-specific expression analysis. (b) Cell-type-specific expression analysis. (c) Cortical layer enrichment analysis. (d) Developmental gene expression enrichment analysis. Ado, adolescence; EC, early childhood; EF, early fetal; EMF, early and mid-fetal; LF, late fetal; LI, late infancy; LMF, late and mid-fetal; MLC, middle and late childhood; NEI, neonatal and early infancy; OCD, obsessive–compulsive disorder; YA, young adulthood.

Cell-type-specific expression and cortical layer enrichment analyses

The CSEA highlighted gene set overexpression in three distinct human brain cell types. Specifically, genes related to alterations in cortical thickness depicted significant enrichment in neurons (corrected p-value = 6.33 × 10−7), oligodendrocytes (corrected p-value = 5.21 × 10−5), and astrocytes (corrected p-value = 0.0012, Fig. 5c). Furthermore, the cortical layer enrichment analysis depicted that these genes were significantly enriched across layers I, II, III, IV, and VI of the cortices, with particularly substantial enrichment within layers II and VI (Fig. 5d).

PPI analysis and hub genes

Under an interaction confidence score of 0.4, a network comprising 598 nodes and 1137 edges was constructed. This network contained 809 edges, significantly more than expected by chance (p = 1.0 × 10−16, Fig. 6a). The top three hub genes in the PPI network were identified with the maximal clique centrality method in CytoHubba (Chin et al., Reference Chin, Chen, Wu, Ho, Ko and Lin2014; Shannon et al., Reference Shannon, Markiel, Ozier, Baliga, Wang, Ramage and Ideker2003): secreted phosphoprotein 1 (SPP1), integrin subunit beta 1 (ITGB1), and collagen type I alpha 2 chain (COL1A2). The spatiotemporal expression trajectories of these three hub genes were delineated, highlighting how their expression patterns change across brain regions and developmental stages (Fig. 6b).

Figure 6. Protein–protein interaction networks and expression patterns of hub genes. (a) The whole PPI network with 598 genes and 1137 edges. Each node represents a protein, and each edge represents an interaction between two proteins. The p value denotes the statistical significance of how likely the proteins encoded by the input genes are connected to construct a network. (b) Spatial–temporal expression curves of three hub genes (i.e. SPP1, ITGB1, and COL1A2). COL1A2, collagen type I alpha 2 chain; PPI, protein–protein interaction; ITGB1, integrin subunit beta 1; SPP1; secreted phosphoprotein 1.

Genetic overlap with OCD-related genes

Fisher's exact test indicated significant overlap between the 609 cortical thickness-related genes identified in this study and the 173 OCD-associated genes derived from the intersection of 153 genes from the Obsessive–Compulsive Disorder and 64 genes from the MalaCards databases (Privitera et al., Reference Privitera, Distefano, Wefer, Ferro, Pulvirenti and Giugno2015; Rappaport et al., Reference Rappaport, Twik, Plaschkes, Nudel, Iny Stein, Levitt and Lancet2017). Thirteen genes overlapped between the two gene sets (odds ratio = 3.19, p = 4.33 × 10−4).

Discussion

Although OCD is a highly heritable disorder, genome-wide association studies of OCD have identified only one genetic locus reaching genome-wide significance because of limited statistical power to detect genetic variants with subtle effects (Strom et al., Reference Strom, Yu, Gerring, Halvorsen, Abdellaoui, Rodriguez-Fontenla and Mattheisen2021). Therefore, the genes implicated in OCD-related brain structure alterations remain obscure. Hence, the present integrative analysis linking cortical thickness changes and transcriptomic data provides novel insights into the genetic and molecular factors of brain abnormalities in OCD. Through spatial correlation analysis, over 1000 differentially expressed genes linked with OCD cortical alterations were identified across two datasets. Further investigation of the 609 overlapping genes highlighted enriched BPs, developmental trajectories, cell-type specificities, and protein interactions contributing to OCD pathogenesis.

The significant overlap of genes between the datasets is a notable discovery, mainly because only a modest spatial correlation of cortical thickness changes was observed between these datasets. The multi-site ENIGMA study is the most extensive investigation of structural morphometry in OCD and provides robust evidence for syndrome-specific cortical thickness abnormalities (Boedhoe et al., Reference Boedhoe, Schmaal, Abe, Alonso, Ameis and Anticevic2018). Challenges such as small sample sizes in independent datasets, the confounding effects of medication, and variations in disease severity among participants contribute to this low-spatial correlation. Nonetheless, the consistency of gene association with OCD cortical pathology across different cohorts underscores the robustness of these implicated genes despite the varying patterns of alterations.

It is well-established that genetic factors significantly influence cortical thickness (Jha et al., Reference Jha, Xia, Schmitt, Ahn, Girault, Murphy and Gilmore2018; Panizzon et al., Reference Panizzon, Fennema-Notestine, Eyler, Jernigan, Prom-Wormley, Neale and Kremen2009). These genetic underpinnings intersect with various cellular processes to orchestrate the dynamic development of the cerebral cortex (Grasby et al., Reference Grasby, Jahanshad, Painter, Colodro-Conde, Bralten, Hibar and Medland2020; Rakic, Reference Rakic2009). The observed gene overlap indicates that macroscale heterogeneity in OCD neuroimaging findings may converge onto shared molecular pathways. The biological functions and pathways enriched in the gene set provide further insight into the potential mechanisms underlying OCD-related cortical thickness alterations. Notably, the enrichment of genes involved in ion transport regulation, responses to environmental stimuli, and metal ion transport regulation suggests the roles of these processes in OCD pathophysiology. Enrichment in glutamate signaling pathways aligns with evidence that genetic disruptions can lead to circuit-level excitatory/inhibitory imbalance (Ahmari, Risbrough, Geyer, & Simpson, Reference Ahmari, Risbrough, Geyer and Simpson2012; Hoenig, Hochrein, Quednow, Maier, & Wagner, Reference Hoenig, Hochrein, Quednow, Maier and Wagner2005). Glutamate-mediated excitotoxicity may lead to the structural and functional cortical disturbances observed in OCD (Robbins et al., Reference Robbins, Vaghi and Banca2019). Genes involved in synaptic structure and transmission were also overrepresented, consistent with synaptic dysfunction in OCD (Piantadosi et al., Reference Piantadosi, McClain, Klei, Wang, Chamberlain, Springer and Ahmari2021; Wang, Kavalali, & Monteggia, Reference Wang, Kavalali and Monteggia2022). The enriched phospholipase D signaling pathway regulates synaptic function and plasticity (Bruntz, Lindsley, & Brown, Reference Bruntz, Lindsley and Brown2014). Despite the distinct spatial patterns, this convergence emphasizes the need to connect multi-scale perspectives to elucidate the complex OCD origins.

The tissue-specific and temporal-specific expression analyses of the identified genes highlight roles in brain development and function, supporting the observed transcriptome–neuroimaging relationships. The specific expression of these genes in brain tissue and their expression from early-/mid-fetal development onward underscore their neurodevelopmental process relevance. Stable expression in the cortex during young adulthood but variable expression in other brain regions provides a potential link between the genes and maturation of brain regions implicated in OCD. Enriched expression of cortical thickness-related genes in neurons, oligodendrocytes, and astrocytes connects these specific cell types in OCD pathogenesis (Soto et al., Reference Soto, Jami-Alahmadi, Chacon, Moye, Diaz-Castro, Wohlschlegel and Khakh2023; Writing Committee for the Attention-Deficit/Hyperactivity Disorder et al., Reference Paus2021). The findings of layer-specific enrichment (particularly layers II and VI of the cortices) suggest a potential role in neural circuitry and connectivity. Deciphering how these genes affect the architecture of cortical layers may provide insights into neural circuits linked with OCD symptomatology (Goodman et al., Reference Goodman, Storch and Sheth2021; Maia, Cooney, & Peterson, Reference Maia, Cooney and Peterson2008).

The PPI network revealed interconnections among the genes. Identifying hub genes, viz., SPP1, ITGB1, and COL1A2, underscores their centrality in the genetic network linked with cortical thickness alterations in OCD. The spatiotemporal expression trajectories of these hub genes provide insights into their dynamic roles across brain regions and developmental stages. Notably, hub gene SPP1 depicts declining expression from infancy onward, consistent with its hypothesized role in neurodevelopment (Khodosevich & Sellgren, Reference Khodosevich and Sellgren2023). Type I collagen, produced by genes like COL1A2, contributes to the structural integrity of perineuronal net (Fawcett, Oohashi, & Pizzorusso, Reference Fawcett, Oohashi and Pizzorusso2019). These structures are essential for maintaining the physical and biochemical milieu around neurons, offering protection against oxidative stress and regulating synaptic functions (Fawcett et al., Reference Fawcett, Oohashi and Pizzorusso2019). Alterations in COL1A2 expression or activity could potentially contribute to structural or functional changes in the brain relevant to OCD. The significant overlap between the genes related to cortical thickness alterations and the genes associated with OCD (13 overlapping genes) highlights a potential genetic link between the observed structural changes in OCD and the disorder itself. The SHANK2 gene encodes a protein that regulates synaptic architecture and is linked with neurodevelopmental processes (Lim et al., Reference Lim, Naisbitt, Yoon, Hwang, Suh, Sheng and Kim1999). Dysfunctional synaptic connectivity and signaling, potentially arising from SHANK2 variations, could underlie the morphological brain changes observed within OCD patients, particularly in regions linked with OCD symptomatology (Monteiro & Feng, Reference Monteiro and Feng2017; Radua, van den Heuvel, Surguladze, & Mataix-Cols, Reference Radua, van den Heuvel, Surguladze and Mataix-Cols2010). Understanding the shared genetic basis between cortical thickness alterations and OCD could provide more targeted and personalized treatment approaches.

Lastly, some limitations of this study must be noted. First, the gene expression data used in AHBA were obtained from six adult donors and not from psychiatric patients with OCD symptoms. Thus, data from OCD patients must be collected to improve our understanding of psychiatric disorders at the molecular level. Second, most OCD patients in the ENIGMA and independent datasets were chronic cases taking medication, leading to confounding variables related to disease profile and drug use. Future research should include drug-naïve first-episode patients to eliminate these confounding factors and corroborate the initial findings. Moreover, our analysis lacks longitudinal data to determine causality between the implicated genes and cortical changes.

Conclusion

This study identified over 600 genes related to cortical thickness changes in OCD by integrating neuroimaging and transcriptional data. Subsequent analyses provided insights into the BPs, cell types, and pathways implicated in OCD cortical pathology. These findings bridge the gap between macroscopic modifications in brain structure in OCD and specific microscopic molecular events, thereby providing unique insights into genetic and molecular factors that may contribute to cortical abnormalities in OCD. These findings provide the groundwork for future work to elucidate OCD etiology, develop molecular biomarkers, and identify therapeutic targets.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0033291724001909.

Data availability statement

The findings of this study are supported by a range of valuable resources and tools. Human gene expression data are available from the Allen Brain Atlas (https://human.brain-map.org/static/download) and gene expression analysis was conducted using code available on GitHub (https://github.com/BMHLab/AHBAprocessing). Probe-to-gene annotations were obtained using the Re-annotator toolkit (http://sourceforge.net/projects/reannotator), and preprocessing of neuroimaging data was performed using freely available software (http://rfmri.org/DPABISurf). The code used for PLS analysis is openly available on GitHub (https://github.com/SarahMorgan/Morphometric_Similarity_SZ). Biological function enrichment analysis was performed using the WebGestalt online tool (https://www.webgestalt.org/). Tissue-specific and temporal-specific expression analyses can be conducted using specific websites (http://genetics.wustl.edu/jdlab/tsea/ and http://genetics.wustl.edu/jdlab/csea). Cell-specific gene expression data can be downloaded from the CellMarker database (http://xteam.xbio.top/CellMarker/). Layer markers were obtained from a dataset by He et al. (https://static-content.springer.com/esm/art%3A10.1038%2Fnn.4548/MediaObjects/41593_2017_BFnn4548_MOESM255_ESM.xlsx). Construction of the PPI network utilized the STRING database version 12.0 (https://string-db.org/).

Acknowledgments

This study was supported by the National Natural Science Foundation of China (NSFC) (No. 81701671). We appreciate MogoEdit (https://www.mogoedit.com) for its English editing during the preparation of this manuscript. We appreciate the founders and staff of the Allen Institute for Brain Science for providing the brain expression data. Furthermore, we are grateful to the ENIGMA Consortium for sharing publicly accessible data and toolbox that made this work possible.

Author contributions

D. Z., C. G., and X. H. designed research; C. T., Y. X., L. T., P. C., X. W., and Z. L. contributed to data acquisition; L. T., C. G., and X. H. provided guidance and advice; D. Z. created the figures and wrote the manuscript; X. H. revised the manuscript. All authors reviewed and commented on the manuscript.

Competing interests

None.

References

Abdi, H., & Williams, L. J. (2013). Partial least squares methods: Partial least squares correlation and partial least square regression. Methods in Molecular Biology (Clifton, N.J.), 930, 549579. https://doi.org/10.1007/978-1-62703-059-5_23CrossRefGoogle ScholarPubMed
Ahmari, S. E., Risbrough, V. B., Geyer, M. A., & Simpson, H. B. (2012). Impaired sensorimotor gating in unmedicated adults with obsessive–compulsive disorder. Neuropsychopharmacology, 37(5), 12161223. https://doi.org/10.1038/npp.2011.308CrossRefGoogle ScholarPubMed
Alemany-Navarro, M., Cruz, R., Real, E., Segalàs, C., Bertolín, S., Rabionet, R., … Alonso, P. (2020). Looking into the genetic bases of OCD dimensions: A pilot genome-wide association study. Translational Psychiatry, 10(1), 151. https://doi.org/10.1038/s41398-020-0804-zCrossRefGoogle ScholarPubMed
Amlien, I. K., Fjell, A. M., Tamnes, C. K., Grydeland, H., Krogsrud, S. K., Chaplin, T. A., … Walhovd, K. B. (2016). Organizing principles of human cortical development – Thickness and area from 4 to 30 years: Insights from comparative primate neuroanatomy. Cerebral Cortex (New York, N.Y.: 1991), 26(1), 257267. https://doi.org/10.1093/cercor/bhu214CrossRefGoogle ScholarPubMed
Arloth, J., Bader, D. M., Röh, S., & Altmann, A. (2015). Re-Annotator: Annotation pipeline for microarray probe sequences. PLoS ONE, 10(10), e0139516. https://doi.org/10.1371/journal.pone.0139516CrossRefGoogle ScholarPubMed
Arnatkeviciute, A., Fulcher, B. D., & Fornito, A. (2019). A practical guide to linking brain-wide gene expression and neuroimaging data. NeuroImage, 189, 353367. https://doi.org/10.1016/j.neuroimage.2019.01.011CrossRefGoogle ScholarPubMed
Arnatkeviciute, A., Fulcher, B. D., Bellgrove, M. A., & Fornito, A. (2021). Where the genome meets the connectome: Understanding how genes shape human brain connectivity. NeuroImage, 244, 118570. https://doi.org/10.1016/j.neuroimage.2021.118570CrossRefGoogle ScholarPubMed
Atmaca, M., Onalan, E., Yildirim, H., Yuce, H., Koc, M., Korkmaz, S., & Mermi, O. (2011). Serotonin transporter gene polymorphism implicates reduced orbito-frontal cortex in obsessive–compulsive disorder. Journal of Anxiety Disorders, 25(5), 680685. https://doi.org/10.1016/j.janxdis.2011.03.002CrossRefGoogle ScholarPubMed
Bethlehem, R. a. I., Seidlitz, J., White, S. R., Vogel, J. W., Anderson, K. M., Adamson, C., … Alexander-Bloch, A. F. (2022). Brain charts for the human lifespan. Nature, 604(7906), 525533. https://doi.org/10.1038/s41586-022-04554-yCrossRefGoogle ScholarPubMed
Bigdeli, T. B., Lee, D., Webb, B. T., Riley, B. P., Vladimirov, V. I., Fanous, A. H., … Bacanu, S.-A. (2016). A simple yet accurate correction for winner's curse can predict signals discovered in much larger genome scans. Bioinformatics (Oxford, England), 32(17), 25982603. https://doi.org/10.1093/bioinformatics/btw303Google ScholarPubMed
Bruntz, R. C., Lindsley, C. W., & Brown, H. A. (2014). Phospholipase D signaling pathways and phosphatidic acid as therapeutic targets in cancer. Pharmacological Reviews, 66(4), 10331079. https://doi.org/10.1124/pr.114.009217CrossRefGoogle ScholarPubMed
Chen, J., Zhang, C., Wang, R., Jiang, P., Cai, H., Zhao, W., … Yu, Y. (2022). Molecular basis underlying functional connectivity of fusiform gyrus subregions: A transcriptome–neuroimaging spatial correlation study. Cortex; a Journal Devoted to the Study of the Nervous System and Behavior, 152, 5973. https://doi.org/10.1016/j.cortex.2022.03.016CrossRefGoogle ScholarPubMed
Chin, C.-H., Chen, S.-H., Wu, H.-H., Ho, C.-W., Ko, M.-T., & Lin, C.-Y. (2014). Cytohubba: Identifying hub objects and sub-networks from complex interactome. BMC Systems Biology, 8(Suppl 4), S11. https://doi.org/10.1186/1752-0509-8-S4-S11CrossRefGoogle ScholarPubMed
Desikan, R. S., Ségonne, F., Fischl, B., Quinn, B. T., Dickerson, B. C., Blacker, D., … Killiany, R. J. (2006). An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. NeuroImage, 31(3), 968980. https://doi.org/10.1016/j.neuroimage.2006.01.021CrossRefGoogle ScholarPubMed
Dougherty, J. D., Schmidt, E. F., Nakajima, M., & Heintz, N. (2010). Analytical approaches to RNA profiling data for the identification of genes enriched in specific cells. Nucleic Acids Research, 38(13), 42184230. https://doi.org/10.1093/nar/gkq130CrossRefGoogle ScholarPubMed
Boedhoe, P. S. W., Schmaal, L., Abe, Y., Alonso, P., Ameis, S. H., & Anticevic, A., … ENIGMA OCD Working Group. (2018). Cortical abnormalities associated with pediatric and adult obsessive–compulsive disorder: Findings from the ENIGMA obsessive–compulsive disorder working group. The American Journal of Psychiatry, 175(5), 453462. https://doi.org/10.1176/appi.ajp.2017.17050485CrossRefGoogle ScholarPubMed
Fawcett, J. W., Oohashi, T., & Pizzorusso, T. (2019). The roles of perineuronal nets and the perinodal extracellular matrix in neuronal function. Nature Reviews. Neuroscience, 20(8), 451465. https://doi.org/10.1038/s41583-019-0196-3CrossRefGoogle ScholarPubMed
Fornito, A., Arnatkevičiūtė, A., & Fulcher, B. D. (2019). Bridging the gap between connectome and transcriptome. Trends in Cognitive Sciences, 23(1), 3450. https://doi.org/10.1016/j.tics.2018.10.005CrossRefGoogle ScholarPubMed
Fouche, J.-P., du Plessis, S., Hattingh, C., Roos, A., Lochner, C., Soriano-Mas, C., … van den Heuvel, O. A. (2017). Cortical thickness in obsessive–compulsive disorder: Multisite mega-analysis of 780 brain scans from six centres. The British Journal of Psychiatry: The Journal of Mental Science, 210(1), 6774. https://doi.org/10.1192/bjp.bp.115.164020CrossRefGoogle ScholarPubMed
Gassó, P., Ortiz, A. E., Mas, S., Morer, A., Calvo, A., Bargalló, N., … Lázaro, L. (2015). Association between genetic variants related to glutamatergic, dopaminergic and neurodevelopment pathways and white matter microstructure in child and adolescent patients with obsessive–compulsive disorder. Journal of Affective Disorders, 186, 284292. https://doi.org/10.1016/j.jad.2015.07.035CrossRefGoogle ScholarPubMed
Glasser, M. F., Coalson, T. S., Robinson, E. C., Hacker, C. D., Harwell, J., Yacoub, E., … Van Essen, D. C. (2016). A multi-modal parcellation of human cerebral cortex. Nature, 536(7615), 171178. https://doi.org/10.1038/nature18933CrossRefGoogle ScholarPubMed
Goodman, W. K., Storch, E. A., & Sheth, S. A. (2021). Harmonizing the neurobiology and treatment of obsessive–compulsive disorder. The American Journal of Psychiatry, 178(1), 1729. https://doi.org/10.1176/appi.ajp.2020.20111601CrossRefGoogle ScholarPubMed
Grasby, K. L., Jahanshad, N., Painter, J. N., Colodro-Conde, L., Bralten, J., Hibar, D. P., … Medland, S. E. (2020). The genetic architecture of the human cerebral cortex. Science (New York, N.Y.), 367(6484), eaay6690. https://doi.org/10.1126/science.aay6690CrossRefGoogle ScholarPubMed
Grünblatt, E., Hauser, T. U., & Walitza, S. (2014). Imaging genetics in obsessive–compulsive disorder: Linking genetic variations to alterations in neuroimaging. Progress in Neurobiology, 121, 114124. https://doi.org/10.1016/j.pneurobio.2014.07.003CrossRefGoogle ScholarPubMed
Hawrylycz, M. J., Lein, E. S., Guillozet-Bongaarts, A. L., Shen, E. H., Ng, L., Miller, J. A., … Jones, A. R. (2012). An anatomically comprehensive atlas of the adult human brain transcriptome. Nature, 489(7416), 391399. https://doi.org/10.1038/nature11405CrossRefGoogle ScholarPubMed
Hawrylycz, M., Miller, J. A., Menon, V., Feng, D., Dolbeare, T., Guillozet-Bongaarts, A. L., … Lein, E. (2015). Canonical genetic signatures of the adult human brain. Nature Neuroscience, 18(12), 18321844. https://doi.org/10.1038/nn.4171CrossRefGoogle ScholarPubMed
He, Z., Han, D., Efimova, O., Guijarro, P., Yu, Q., Oleksiak, A., … Khaitovich, P. (2017). Comprehensive transcriptome analysis of neocortical layers in humans, chimpanzees and macaques. Nature Neuroscience, 20(6), 886895. https://doi.org/10.1038/nn.4548CrossRefGoogle ScholarPubMed
Hoenig, K., Hochrein, A., Quednow, B. B., Maier, W., & Wagner, M. (2005). Impaired prepulse inhibition of acoustic startle in obsessive–compulsive disorder. Biological Psychiatry, 57(10), 11531158. https://doi.org/10.1016/j.biopsych.2005.01.040CrossRefGoogle ScholarPubMed
Jha, S. C., Xia, K., Schmitt, J. E., Ahn, M., Girault, J. B., Murphy, V. A., … Gilmore, J. H. (2018). Genetic influences on neonatal cortical thickness and surface area. Human Brain Mapping, 39(12), 49985013. https://doi.org/10.1002/hbm.24340CrossRefGoogle ScholarPubMed
Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y., & Morishima, K. (2017). KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Research, 45(D1), D353D361. https://doi.org/10.1093/nar/gkw1092CrossRefGoogle ScholarPubMed
Khodosevich, K., & Sellgren, C. M. (2023). Neurodevelopmental disorders – High-resolution rethinking of disease modeling. Molecular Psychiatry, 28(1), 3443. https://doi.org/10.1038/s41380-022-01876-1CrossRefGoogle ScholarPubMed
Kühn, S., Kaufmann, C., Simon, D., Endrass, T., Gallinat, J., & Kathmann, N. (2013). Reduced thickness of anterior cingulate cortex in obsessive–compulsive disorder. Cortex; a Journal Devoted to the Study of the Nervous System and Behavior, 49(8), 21782185. https://doi.org/10.1016/j.cortex.2012.09.001CrossRefGoogle ScholarPubMed
Larivière, S., Paquola, C., Park, B.-Y., Royer, J., Wang, Y., Benkarim, O., … Bernhardt, B. C. (2021). The ENIGMA Toolbox: Multiscale neural contextualization of multisite neuroimaging datasets. Nature Methods, 18(7), 698700. https://doi.org/10.1038/s41592-021-01186-4CrossRefGoogle ScholarPubMed
Li, J., Seidlitz, J., Suckling, J., Fan, F., Ji, G.-J., Meng, Y., … Liao, W. (2021). Cortical structural differences in major depressive disorder correlate with cell type-specific transcriptional signatures. Nature Communications, 12(1), 1647. https://doi.org/10.1038/s41467-021-21943-5CrossRefGoogle ScholarPubMed
Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z., & Zhang, B. (2019). WebGestalt 2019: Gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Research, 47(W1), W199W205. https://doi.org/10.1093/nar/gkz401CrossRefGoogle ScholarPubMed
Lim, S., Naisbitt, S., Yoon, J., Hwang, J. I., Suh, P. G., Sheng, M., & Kim, E. (1999). Characterization of the Shank family of synaptic proteins. Multiple genes, alternative splicing, and differential expression in brain and development. The Journal of Biological Chemistry, 274(41), 2951029518. https://doi.org/10.1074/jbc.274.41.29510CrossRefGoogle ScholarPubMed
Mahjani, B., Bey, K., Boberg, J., & Burton, C. (2021). Genetics of obsessive–compulsive disorder. Psychological Medicine, 51(13), 22472259. https://doi.org/10.1017/S0033291721001744CrossRefGoogle ScholarPubMed
Maia, T. V., Cooney, R. E., & Peterson, B. S. (2008). The neural bases of obsessive–compulsive disorder in children and adults. Development and Psychopathology, 20(4), 12511283. https://doi.org/10.1017/S0954579408000606CrossRefGoogle ScholarPubMed
Monteiro, P., & Feng, G. (2017). SHANK proteins: Roles at the synapse and in autism spectrum disorder. Nature Reviews. Neuroscience, 18(3), 147157. https://doi.org/10.1038/nrn.2016.183CrossRefGoogle ScholarPubMed
Monzani, B., Rijsdijk, F., Harris, J., & Mataix-Cols, D. (2014). The structure of genetic and environmental risk factors for dimensional representations of DSM-5 obsessive–compulsive spectrum disorders. JAMA Psychiatry, 71(2), 182189. https://doi.org/10.1001/jamapsychiatry.2013.3524CrossRefGoogle ScholarPubMed
Morgan, S. E., Seidlitz, J., Whitaker, K. J., Romero-Garcia, R., Clifton, N. E., Scarpazza, C., … Bullmore, E. T. (2019). Cortical patterning of abnormal morphometric similarity in psychosis is associated with brain expression of schizophrenia-related genes. Proceedings of the National Academy of Sciences of the United States of America, 116(19), 96049609. https://doi.org/10.1073/pnas.1820754116CrossRefGoogle ScholarPubMed
Naumova, O. Y., Lee, M., Rychkov, S. Y., Vlasova, N. V., & Grigorenko, E. L. (2013). Gene expression in the human brain: The current state of the study of specificity and spatiotemporal dynamics. Child Development, 84(1), 7688. https://doi.org/10.1111/cdev.12014CrossRefGoogle Scholar
Panizzon, M. S., Fennema-Notestine, C., Eyler, L. T., Jernigan, T. L., Prom-Wormley, E., Neale, M., … Kremen, W. S. (2009). Distinct genetic influences on cortical surface area and cortical thickness. Cerebral Cortex (New York, N.Y.: 1991), 19(11), 27282735. https://doi.org/10.1093/cercor/bhp026CrossRefGoogle ScholarPubMed
Piantadosi, S. C., McClain, L. L., Klei, L., Wang, J., Chamberlain, B. L., Springer, S. A., … Ahmari, S. E. (2021). Transcriptome alterations are enriched for synapse-associated genes in the striatum of subjects with obsessive–compulsive disorder. Translational Psychiatry, 11(1), 171. https://doi.org/10.1038/s41398-021-01290-1CrossRefGoogle ScholarPubMed
Pizzagalli, F., Auzias, G., Yang, Q., Mathias, S. R., Faskowitz, J., Boyd, J. D., … Jahanshad, N. (2020). The reliability and heritability of cortical folds and their genetic correlations across hemispheres. Communications Biology, 3(1), 510. https://doi.org/10.1038/s42003-020-01163-1CrossRefGoogle ScholarPubMed
Privitera, A. P., Distefano, R., Wefer, H. A., Ferro, A., Pulvirenti, A., & Giugno, R. (2015). OCDB: A database collecting genes, miRNAs and drugs for obsessive–compulsive disorder. Database: The Journal of Biological Databases and Curation, 2015, bav069. https://doi.org/10.1093/database/bav069CrossRefGoogle ScholarPubMed
Qiu, A., Zhang, H., Kennedy, B. K., & Lee, A. (2021). Spatio-temporal correlates of gene expression and cortical morphology across lifespan and aging. NeuroImage, 224, 117426. https://doi.org/10.1016/j.neuroimage.2020.117426CrossRefGoogle ScholarPubMed
Radua, J., van den Heuvel, O. A., Surguladze, S., & Mataix-Cols, D. (2010). Meta-analytical comparison of voxel-based morphometry studies in obsessive–compulsive disorder vs other anxiety disorders. Archives of General Psychiatry, 67(7), 701711. https://doi.org/10.1001/archgenpsychiatry.2010.70CrossRefGoogle ScholarPubMed
Rakic, P. (2009). Evolution of the neocortex: A perspective from developmental biology. Nature Reviews. Neuroscience, 10(10), 724735. https://doi.org/10.1038/nrn2719CrossRefGoogle ScholarPubMed
Rappaport, N., Twik, M., Plaschkes, I., Nudel, R., Iny Stein, T., Levitt, J., … Lancet, D. (2017). MalaCards: An amalgamated human disease compendium with diverse clinical and genetic annotation and structured search. Nucleic Acids Research, 45(D1), D877D887. https://doi.org/10.1093/nar/gkw1012CrossRefGoogle ScholarPubMed
Robbins, T. W., Vaghi, M. M., & Banca, P. (2019). Obsessive–compulsive disorder: Puzzles and prospects. Neuron, 102(1), 2747. https://doi.org/10.1016/j.neuron.2019.01.046CrossRefGoogle ScholarPubMed
Romero-Garcia, R., Seidlitz, J., Whitaker, K. J., Morgan, S. E., Fonagy, P., Dolan, R. J., … Bullmore, E. T. (2020). Schizotypy-related magnetization of cortex in healthy adolescence is colocated with expression of schizophrenia-related genes. Biological Psychiatry, 88(3), 248259. https://doi.org/10.1016/j.biopsych.2019.12.005CrossRefGoogle ScholarPubMed
Saraiva, L. C., Sato, J. R., & Cappi, C. (2022). Probing the genetic and molecular correlates of connectome alterations in obsessive-compulsive disorder. Molecular Psychiatry, 27(9), 35583559. https://doi.org/10.1038/s41380-022-01590-yCrossRefGoogle ScholarPubMed
Schulze, D., Kathmann, N., & Reuter, B. (2018). Getting it just right: A reevaluation of OCD symptom dimensions integrating traditional and Bayesian approaches. Journal of Anxiety Disorders, 56, 6373. https://doi.org/10.1016/j.janxdis.2018.04.003CrossRefGoogle ScholarPubMed
Shannon, P., Markiel, A., Ozier, O., Baliga, N. S., Wang, J. T., Ramage, D., … Ideker, T. (2003). Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Research, 13(11), 24982504. https://doi.org/10.1101/gr.1239303CrossRefGoogle ScholarPubMed
Shin, Y.-W., Yoo, S. Y., Lee, J. K., Ha, T. H., Lee, K. J., Lee, J. M., … Kwon, J. S. (2007). Cortical thinning in obsessive compulsive disorder. Human Brain Mapping, 28(11), 11281135. https://doi.org/10.1002/hbm.20338CrossRefGoogle ScholarPubMed
Sinopoli, V. M., Erdman, L., Burton, C. L., Easter, P., Rajendram, R., Baldwin, G., … Arnold, P. D. (2020). Serotonin system gene variants and regional brain volume differences in pediatric OCD. Brain Imaging and Behavior, 14(5), 16121625. https://doi.org/10.1007/s11682-019-00092-wCrossRefGoogle ScholarPubMed
Soto, J. S., Jami-Alahmadi, Y., Chacon, J., Moye, S. L., Diaz-Castro, B., Wohlschlegel, J. A., & Khakh, B. S. (2023). Astrocyte–neuron subproteomes and obsessive–compulsive disorder mechanisms. Nature, 616(7958), 764773. https://doi.org/10.1038/s41586-023-05927-7CrossRefGoogle ScholarPubMed
Stein, D. J., Costa, D. L. C., Lochner, C., Miguel, E. C., Reddy, Y. C. J., Shavitt, R. G., … Simpson, H. B. (2019). Obsessive–compulsive disorder. Nature Reviews. Disease Primers, 5(1), 52. https://doi.org/10.1038/s41572-019-0102-3CrossRefGoogle ScholarPubMed
Strom, N. I., Yu, D., Gerring, Z. F., Halvorsen, M. W., Abdellaoui, A., Rodriguez-Fontenla, C., … Mattheisen, M. (2021, October 23). Genome-wide association study identifies new locus associated with OCD (p. 2021.10.13.21261078). medRxiv. https://doi.org/10.1101/2021.10.13.21261078CrossRefGoogle Scholar
Sun, X., Huang, W., Wang, J., Xu, R., Zhang, X., Zhou, J., … Qian, Y. (2023). Cerebral blood flow changes and their genetic mechanisms in major depressive disorder: A combined neuroimaging and transcriptome study. Psychological Medicine, 113. https://doi.org/10.1017/S0033291722003750Google ScholarPubMed
Sunkin, S. M., Ng, L., Lau, C., Dolbeare, T., Gilbert, T. L., Thompson, C. L., … Dang, C. (2013). Allen Brain Atlas: An integrated spatio-temporal portal for exploring the central nervous system. Nucleic Acids Research, 41(Database issue), D996D1008. https://doi.org/10.1093/nar/gks1042CrossRefGoogle ScholarPubMed
Sydnor, V. J., Larsen, B., Bassett, D. S., Alexander-Bloch, A., Fair, D. A., Liston, C., … Satterthwaite, T. D. (2021). Neurodevelopment of the association cortices: Patterns, mechanisms, and implications for psychopathology. Neuron, 109(18), 28202846. https://doi.org/10.1016/j.neuron.2021.06.016CrossRefGoogle ScholarPubMed
Szklarczyk, D., Kirsch, R., Koutrouli, M., Nastou, K., Mehryary, F., Hachilif, R., … von Mering, C. (2023). The STRING database in 2023: Protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Research, 51(D1), D638D646. https://doi.org/10.1093/nar/gkac1000CrossRefGoogle ScholarPubMed
Thomas, P. D., Ebert, D., Muruganujan, A., Mushayahama, T., Albou, L., & Mi, H. (2022). PANTHER: Making genome-scale phylogenetics accessible to all. Protein Science: A Publication of the Protein Society, 31(1), 822. https://doi.org/10.1002/pro.4218CrossRefGoogle Scholar
Thompson, P. M., Cannon, T. D., Narr, K. L., van Erp, T., Poutanen, V. P., Huttunen, M., … Toga, A. W. (2001). Genetic influences on brain structure. Nature Neuroscience, 4(12), 12531258. https://doi.org/10.1038/nn758CrossRefGoogle ScholarPubMed
Váša, F., Seidlitz, J., Romero-Garcia, R., Whitaker, K. J., Rosenthal, G., Vértes, P. E., … Bullmore, E. T. (2018). Adolescent tuning of association cortex in human structural brain networks. Cerebral Cortex (New York, N.Y.: 1991), 28(1), 281294. https://doi.org/10.1093/cercor/bhx249CrossRefGoogle ScholarPubMed
Walhovd, K. B., Fjell, A. M., Giedd, J., Dale, A. M., & Brown, T. T. (2017). Through thick and thin: A need to reconcile contradictory results on trajectories in human cortical development. Cerebral Cortex (New York, N.Y.: 1991), 27(2), 14721481. https://doi.org/10.1093/cercor/bhv301Google Scholar
Wang, C. S., Kavalali, E. T., & Monteggia, L. M. (2022). BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell, 185(1), 6276. https://doi.org/10.1016/j.cell.2021.12.003CrossRefGoogle ScholarPubMed
Writing Committee for the Attention-Deficit/Hyperactivity Disorder, Autism Spectrum Disorder, Bipolar Disorder, Major Depressive Disorder, Obsessive–Compulsive Disorder, and Schizophrenia ENIGMA Working Groups, … Paus, T. (2021). Virtual histology of cortical thickness and shared neurobiology in 6 psychiatric disorders. JAMA Psychiatry, 78(1), 4763. https://doi.org/10.1001/jamapsychiatry.2020.2694Google Scholar
Wu, K., Hanna, G. L., Easter, P., Kennedy, J. L., Rosenberg, D. R., & Arnold, P. D. (2013). Glutamate system genes and brain volume alterations in pediatric obsessive–compulsive disorder: A preliminary study. Psychiatry Research, 211(3), 214220. https://doi.org/10.1016/j.pscychresns.2012.07.003CrossRefGoogle ScholarPubMed
Xu, X., Wells, A. B., O'Brien, D. R., Nehorai, A., & Dougherty, J. D. (2014). Cell type-specific expression analysis to identify putative cellular mechanisms for neurogenetic disorders. The Journal of Neuroscience, 34(4), 14201431. https://doi.org/10.1523/JNEUROSCI.4488-13.2014CrossRefGoogle ScholarPubMed
Xue, K., Guo, L., Zhu, W., Liang, S., Xu, Q., Ma, L., … Liu, F. (2023). Transcriptional signatures of the cortical morphometric similarity network gradient in first-episode, treatment-naive major depressive disorder. Neuropsychopharmacology, 48(3), 518528. https://doi.org/10.1038/s41386-022-01474-3CrossRefGoogle ScholarPubMed
Zhang, X., Lan, Y., Xu, J., Quan, F., Zhao, E., Deng, C., … Xiao, Y. (2019). CellMarker: A manually curated resource of cell markers in human and mouse. Nucleic Acids Research, 47(D1), D721D728. https://doi.org/10.1093/nar/gky900CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Schematic workflow of the analytical framework. (a) PLS regression was conducted to link cortical thickness abnormalities of OCD with gene expression data, the overlapped genes were identified by taking the intersection of the significant gene list from the first PLS component (PLS1) from both ENIGMA and independent database. (b) Enrichment analyses were performed subsequently on the overlapped genes. AHBA, Allen Human Brain Atlas; CSEA, cell-type-specific expression analysis; OCD, obsessive–compulsive disorder; PPI, protein–protein interaction; PLS, partial least squares; TSEA, tissue-type-specific expression analysis.

Figure 1

Figure 2. Cortical thickness alterations between patients with OCD and HCs. (a) Cohen's d maps indicating case-control differences in cortical thickness. (b) Region-wise Pearson's correlation between Cohen's d maps from the two datasets (Pearson's r = −0.13, pspin = 0.042). HC, healthy control; OCD, obsessive–compulsive disorder.

Figure 2

Figure 3. Gene expression profiles related to case-control differences in cortical thickness. (a) Cortical map of regional PLS1 scores. (b) Scatterplot of regional PLS1 scores (weighted sum of 5013 gene expression scores) v. case-control differences in regional cortical thickness. (c) Ranked PLS1 gene expression weights and the overlapped genes of the two datasets (609 genes are overlapped).

Figure 3

Figure 4. Biological function analysis the genes associated with cortical thickness alterations in OCD patients. Only items passed the BH-FDR-corrected threshold (p < 0.05) are displayed in the bubble chart. The x-axis shows the gene ratio of each item, the y-axis shows GO terms or KEGG pathway, the bubble shapes representing different categories, the bubble size indicates the intersecting number of genes in the category, and the color bar represents the −log10(P). BH-FDR, Benjamini–Hochberg false-discovery rate; BP, biological process; CC, cellular component; GO, gene ontology; KEGG, Kyoto Encyclopedia of genes and genomes; MF, molecular function; OCD, obsessive–compulsive disorder.

Figure 4

Figure 5. Specific expression analysis of the genes associated with cortical thickness alterations in OCD patients. (a) Tissue-type-specific expression analysis. (b) Cell-type-specific expression analysis. (c) Cortical layer enrichment analysis. (d) Developmental gene expression enrichment analysis. Ado, adolescence; EC, early childhood; EF, early fetal; EMF, early and mid-fetal; LF, late fetal; LI, late infancy; LMF, late and mid-fetal; MLC, middle and late childhood; NEI, neonatal and early infancy; OCD, obsessive–compulsive disorder; YA, young adulthood.

Figure 5

Figure 6. Protein–protein interaction networks and expression patterns of hub genes. (a) The whole PPI network with 598 genes and 1137 edges. Each node represents a protein, and each edge represents an interaction between two proteins. The p value denotes the statistical significance of how likely the proteins encoded by the input genes are connected to construct a network. (b) Spatial–temporal expression curves of three hub genes (i.e. SPP1, ITGB1, and COL1A2). COL1A2, collagen type I alpha 2 chain; PPI, protein–protein interaction; ITGB1, integrin subunit beta 1; SPP1; secreted phosphoprotein 1.

Supplementary material: File

Zhang et al. supplementary material 1

Zhang et al. supplementary material
Download Zhang et al. supplementary material 1(File)
File 129.3 KB
Supplementary material: File

Zhang et al. supplementary material 2

Zhang et al. supplementary material
Download Zhang et al. supplementary material 2(File)
File 33.9 KB
Supplementary material: File

Zhang et al. supplementary material 3

Zhang et al. supplementary material
Download Zhang et al. supplementary material 3(File)
File 12.8 KB
Supplementary material: File

Zhang et al. supplementary material 4

Zhang et al. supplementary material
Download Zhang et al. supplementary material 4(File)
File 129.9 KB