- 1Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- 2Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
Introduction: The comorbidity of atopic dermatitis (AD) and depression has garnered increased attention in recent years, yet the immunopathological mechanisms underlying this connection remain unclear. To bridge this gap, the study aimed to uncover the immune regulatory networks and identify key genetic markers involved in the comorbidity of depression in AD.
Methods: We performed RNA sequencing on peripheral blood mononuclear cells (PBMCs) collected from 20 AD patients with and without depression. By integrating bioinformatics analyses with machine learning, we conducted weighted gene co-expression network analysis (WGCNA), functional enrichment analysis, and employed machine learning models of least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE). Additionally, validation was carried out in an independent cohort of 20 participants to confirm the expression of the identified potential pivotal gene.
Results: A total of 394 differentially expressed genes (DEGs) were identified in AD patients with depression as compared to those non-depressed counterparts. Weighted gene co-expression network analysis (WGCNA) pinpointed a pink module encompassing 83 genes strongly linked to depressive symptoms. Functional enrichment analysis highlighted biological processes related to neurotransmitter uptake and the negative regulation of T-helper (Th) 17 cell differentiation. Furthermore, machine learning models of least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) consistently identified CHN1 as a potential pivotal gene upregulated in AD patients with depression. The expression level of CHN1 demonstrated positive correlation with Th2 and Th17 cytokine signatures, as well as with the Hospital Anxiety and Depression Scale-Depression (HADS-D) score, and the Eczema Area and Severity Index (EASI). Validation in an independent cohort of 20 participants confirmed the significant upregulation of CHN1 in depressed AD patients.
Discussion: Together, these findings reveal previously unrecognized immunoinflammatory axis underlying AD-associated depression, and shed light on CHN1 as a potential molecular bridge connecting peripheral inflammation and neuropsychiatric manifestations.
1 Introduction
Atopic dermatitis (AD), the most common chronic inflammatory skin disease, affects up to 20% of children and 10% of adults, imposing significant burdens to patients’ quality of life and mental health (1–3). Patients with AD are commonly characterized by eczematous rashes, diffuse xerosis, intense pruritus, with Staphylococcus aureus infections (4). Numerous clinical and epidemiological studies have established a significant association between AD and various psychiatric comorbidities (5–10). Such mental comorbidity not only exacerbates individual suffering, but also imposes substantial socioeconomic burdens, particularly in the long-term management of chronic diseases (11). Among these psychiatric disorders, depression is one of the most prevalent, manifesting as persistent low mood, fatigue, and cognitive impairment (12). Existing research data have revealed a dose-response relationship between AD severity and the risk of developing depression (7).In one cohort of 695 patients, 14.68% exhibited moderate-to-severe depressive symptoms, which closely fluctuated with the severity of AD (13). Despite these observations, the central mechanisms linking AD to depression comorbidity remains poorly defined, impeding progress in early identification and targeted intervention.
Chronic peripheral inflammation is thought to play a critical role in triggering central neuropsychiatric disorders (14). Mounting evidence indicates that peripheral immune dysregulation contributes to the onset and maintenance of AD-associated depressive symptoms (15, 16). In AD, a pronounced T-helper 2 (Th2) cell response, marked by the secretion of Interleukin (IL)-4, IL-5, IL-33, and IL-13, contributes to the formation of a complex cytokine milieu (17–20). This environment can activate peripheral sensory nerves that transmit chronic itching signals to the central nervous system (CNS), thereby affecting neural circuits responsible for pruritus and mood regulation (21). Additionally, peripheral inflammatory factors can compromise blood-brain barrier integrity, and activate microglia and astrocytes, which exacerbates neuroinflammatory cascades, ultimately contributing to depressive phenotypes (22–24). However, researches on these mechanisms remain fragmented, and the interplay between the peripheral immune networks and central neuropsychiatric comorbidities has yet to be systemically explored.
Peripheral blood mononuclear cells (PBMCs) from patients with AD exhibit distinct transcriptional signatures that mirror their systemic inflammation activation, making them an accessible model for studying disease-related inflammation. Integrating transcriptomic data from PBMCs with machine learning approaches allowing comprehensive analysis of high-dimensional transcriptomic datasets, can identify key genes, construct immune regulatory networks, and pinpoint potential biomarkers with unprecedented accuracy (25, 26).
In this study, we conducted transcriptomic analyses of PBMCs from AD patients with or without depression, and employed integrative bioinformatics and machine learning approaches to systemically delineate the molecular mechanisms underlying the depression comorbidity. We further validated the key genes and examined their associations with immune pathways, the disease severity, and depressive symptom scores. These findings provide new insights into the inflammation-depression axis in AD and lay the groundwork for precision diagnostics and targeted interventions in AD-related neuropsychiatric comorbidities. The overall analytical workflow is illustrated in Figure 1.
2 Materials and methods
2.1 Participants
The study enrolled 40 adult patients diagnosed with AD at Wuhan Union Hospital. The inclusion criteria were: (i) a confirmed diagnosis of AD, established by dermatologists according to the guidelines of the American Academy of Allergy, Asthma and Immunology (27), and (ii) being 18 years of age or older. The exclusion criteria included: (i) pregnancy or breastfeeding; (ii) the presence of other known systemic inflammatory diseases, autoimmune disorders, or infections; and (iii) the use of any topical medications within the previous 2 weeks or systemic immunosuppressive or antidepressant therapy within the previous 12 weeks. Among the participants, 19 met the diagnostic criteria for depressive disorder, based on the psychiatrists’ application using the Fifth Edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5), detailed in Supplementary Table 1 (28). All participants provided written informed consent. The study was approved by the Ethics Committee of Wuhan Union Hospital (protocol No. UHCT240584), and conducted in accordance with the Declaration of Helsinki.
2.2 Assessment of clinical characteristics
Clinical characteristics were evaluated including the SCORing of Atopic Dermatitis (SCORAD), Eczema Area and Severity Index (EASI), Hospital Anxiety and Depression Scale-Anxiety (HADS-A) subscale, and HADS-Depression (HADS-D) subscale. SCORAD assesses objective signs (lesion area and severity, 0-80), subjective symptoms (pruritus and sleep disturbance, 0-20), and a visual analog scale (VAS, 0-10) for overall severity, with a total score ranging from 0 to 100 (29). EASI focuses on objective evaluation, including lesion area (weighted by body regions, 0-6) and severity (erythema, infiltration/papulation, excoriation, lichenification, 0-72), with a total score of 0-72 (30). Each HADS subscale uses seven 0–3 Likert items to yield anxiety and depression scores from 0 to 21, with ≥ 8 indicating clinically significant symptoms (31, 32).
2.3 PBMC isolation, RNA extraction and library preparation
Peripheral blood samples were collected from participants in heparin-coated tubes. PBMCs were isolated using Lymphocyte Separation Medium (Corning, Manassas, VA) through density-gradient centrifugation. Cell counts were determined using the Cellometer Auto 2000 (Nexcelom, Lawrence, MA), and cells were cultured at a density of 2 × 106 cells/ml. Total RNA was extracted from PBMCs using TRIzol reagent (Invitrogen, Carlsbad, CA), and RNA quality was assessed using the NanoDrop ND-1000 spectrophotometer (ThermoFisher Scientific, Waltham, MA). Strand-specific RNA-seq libraries were subsequently prepared with the VAHTS Universal V6 RNA-seq Library Prep Kit following the manufacturer’s instructions.
2.4 RNA sequencing and differential gene expression analysis
Libraries were sequenced on an Illumina NovaSeq 6000 platform, generating ~60 million 150-bp paired-end reads per sample. After adapter trimming and quality filtering with fastp, an average of 57.2 million clean reads (Q30 ≥ 93%) remained. These were aligned to the GRCh38 reference genome with HISAT2 (unique-mapping rate 91.8%), and gene-level abundances were quantified as both FPKM (StringTie) and raw counts (HTSeq-count). All raw data have been deposited in the GEO database (accession GSE307177).
Differential gene expression analysis comparing AD patients with and without comorbid depression was performed using the DESeq2 packages in R software. Raw read counts were normalized with the built-in median-of-ratios method, and baseMean values were used to estimate average expression levels. Fold changes were calculated from the normalized counts, and significance was assessed by a negative-binomial Wald test implemented in DESeq2 (33). Differentially expressed genes (DEGs) were identified based on the criteria of adjusted P < 0.05 (Benjamini–Hochberg) and |log2 fold change (log2FC) | > 1 (34). Volcano plots and heatmaps were generated to visualize DEGs using the ‘heatmap’ and ‘ggplot2’ packages.
2.5 Weighted gene co-expression network analysis
WGCNA was performed using the WGCNA R package to identify co-expression gene modules, potentially related to depression in AD. A soft thresholding was applied to establish a scale-free network topology. Modules were identified using hierarchical clustering with a dynamic tree-cut algorithm. Module eigengenes (MEs) were calculated to represent the first principal component of each module, and correlation analysis between MEs and clinical traits was performed to identify depression-related modules (35).
2.6 Pathway enrichment analysis
Pathway enrichment in AD-related depression datasets was evaluated using Gene Set Variation Analysis (GSVA). All hallmark gene sets were obtained from the Molecular Signature Database (MSigDB). An adjusted P-value < 0.05 was considered statistically significant after Benjamini and Hochberg correction (36).
2.7 Functional enrichment analysis
Functional enrichment analysis of DEGs was performed using the Gene Ontology (GO) plot, ReactomePA and clusterProfiler packages in R. GO functional annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, Reactome and WikiPathways enrichment analysis were conducted to explore the biological roles of hub genes. The annotation terms with P-value < 0.05 were considered significantly enriched, and results were visualized in a bubble diagram and heat map (37, 38).
2.8 Machine learning for key genes
Two machine learning algorithms were employed to identify key genes associated with depression in AD patients. Least Absolute Shrinkage and Selection Operator (LASSO) was performed (glmnet R package) to select a subset of genes with the highest predictive power. The model was fitted with 10-fold cross-validation and nlambda = 100, and the λ value was selected to minimize the mean-squared error, which represents the optimal LASSO fit and minimizes the cross-validation error. The gene count at this point is taken as the number of disease-signature genes. Support Vector Machine Recursive Feature Elimination (SVM-RFE) was conducted (caret package) to rank genes based on their importance in predicting depression. Ten-fold cross-validation (with a fixed random seed) was applied to the SVM-RFE pipeline. The average rank of each feature across all folds was computed to determine the optimal feature subset and used plotting functions to visualize the trends in both generalization error and classification accuracy across gene numbers. A Venn diagram was constructed to identify overlapping genes selected by both methods (38–40).
2.9 Receiver operating characteristic curve analysis
ROC curve analysis was performed in R to assess the diagnostic performance of key genes. The area under the curve (AUC) was calculated to validate the diagnostic value of key genes.
2.10 Quantitative reverse transcription-polymerase chain reaction
Total RNA extracted from participants’ PBMCs was reverse—transcribed into cDNA. Following cDNA synthesis, RT-qPCR was conducted using CHN1-specific primers (Table 1), and the relative expression levels were quantified and statistically analyzed using standard ΔΔCt methods and analyzed statistically.
2.11 Immunohistochemistry staining
Skin lesion samples all collected from the limbs were used for IHC. Slides were immunostained for CHN1, and the extent of immunostaining was reviewed and scored by two independent dermatopathologists, blinded to clinical details. Immunostaining scores were calculated by multiplying the percentage of positive cells by staining intensity.
2.12 Statistical analysis
All statistical analyses were performed using R software. Continuous variables were compared using Student’s t-test, and categorical variables were compared using chi-square or Fisher’s exact test. Correlation analysis was performed using Pearson’s correlation coefficients, depending on the normality of the data. A two-tailed P-value <0.05 was considered statistically significant.
3 Results
3.1 Participants characteristics
Skin lesion and PBMC samples were collected from two independent participant cohorts. The discovery cohort included 8 AD patients with depression and 12 without, whose PBMCs were subjected to RNA sequencing and DEG analysis. The validation cohort consisted of 11 AD patients with depression and 9 without, providing clinical samples to validate the identified DEGs (Table 2). All patients met the established diagnostic criteria. No statistically significant differences were observed in demographics or skin severity scores (SCORAD, EASI) between groups. However, anxiety and depression scale (HADS-A, HADS-D) scores were substantially higher in AD patients with depression relative to those without.
Table 2. Characteristics of study participants and data on disease characteristics and comorbid depression.
3.2 DEG analysis of AD patients with and without depression
To identify the molecular features associated with depression in AD, we analyzed PBMC transcriptomes from both groups. Under the criteria of P-adjustment <0.05 and log2 FC >1, a total of 394 DEGs were identified, 257 up-regulated and 137 down-regulated genes, in depressed AD patients compared with non-depressed counterparts. These genes were visualized using a volcano plot (Figures 2A, B), and the top 10 up- and down-regulated DEGs were visualized using a volcano plot (Figure 2C). GSVA analysis revealed that the gene profiles of AD patients with depression were mainly enriched in the T-cell receptor signaling pathway, GnRH signaling pathway, Apelin signaling pathway, and the cGMP-PKG signaling pathway, etc (Figure 2D).
Figure 2. Baseline gene expression analysis in AD patients with and without depression. (A) Volcano plot showing DEGs in depressed AD patients compared with non-depressed counterparts. Blue dots represent downregulated genes and red plots represent upregulated genes. (B) Bar chart showing the number of down- and up-regulated DEGs in AD patients with depression. (C) Top 10 up- and down-regulated DEGs ranked by fold change and Log2FC (P < 0.05). (D) Heatmap displaying GSVA enrichment analysis results, highlighting key pathways associated with depression in AD patients.
3.3 Identification of key modules via WGCNA
WGCNA analysis was constructed to screen out the core genes associated with depression in AD patients As shown in Figure 3A, with a soft threshold to 14 (R2>0.82) and a high average connectivity, 15 modules were identified after merging the strongly associated modules using a height cutoff of 0.25. Supplementary Figure 1 further validated the test results at power 14 and showed the Topological Overlap Matrix (TOM). The module clustering dendrogram showed the primed and merged modules in AD patients with depression (Figure 3B). Correlation analysis of ME values and clinical manifestations revealed a significant association between the pink module and depression (r = 0.48, P < 0.05), while a negative correlation was observed with non-depression (r = −0.48, P < 0.05) (Figure 3C). Genes in the grey module, which lack module assignment, were excluded from further analysis. The scatter plot analysis confirmed the strong association between the pink module and depression (Figure 3D), and 83 genes within this module were selected for further analysis.
Figure 3. Identification of modules highly correlated with depression in AD patients. (A) Topology analysis and mean connectivity assessment across a range of soft threshold powers. (B) Heatmap illustrating the relevance of various modules with depression. (C) Dendrogram of module clustering, with distinct module colors representing different gene module. (D) The pink module was identified as the module most significantly correlated with depression in AD. The scatter plot presenting the correlation between module membership and gene significance within the module.
3.4 Functional enrichment analysis of critical module genes
Functional enrichment analysis within the pink module uncovered the biological roles of DEGs, which most reflected the key signaling pathways activated in AD patients with depression. GO analysis revealed associations with neural and immune pathways, including adaptive immune response, negative regulation of Th17 cell differentiation, T cell activation, positive regulation of chemokine CCL5 production and neurotransmitter uptake (Figure 4A). KEGG enrichment analysis highlighted pathways such as antigen processing and presentation, natural killer cell-mediated cytotoxicity, apoptosis, arachidonic acid metabolism, and the MAPK signaling pathway (Figure 4B). Reactome pathway analysis showed enrichment in immune system interactions, immunoregulatory interactions between a lymphoid and a non-lymphoid cell, IL-17 signaling, etc. (Figure 4C). WikiPathways analysis emphasized high enrichment scores in IL-18 signaling pathway, aryl hydrocarbon receptor pathway, T-cell antigen receptor pathway during staphylococcus aureus infection, and Th17 cell differentiation pathway (Figure 4D).
Figure 4. Functional enrichment analysis of genes within the pink module. (A) GO enrichment analysis in biological processes (BP), (B) KEGG pathway enrichment analysis, (C) Reactome enrichment analysis, and (D) WikiPathways enrichment analysis identifying key signaling pathways within the pink module that are associated with AD patients with depression.
3.5 Identification of key genes by machine learning
Key genes were identified using LASSO regression analysis and SVM-RFE algorithms. Through the rigorous process of LASSO analysis (10-fold CV, mean AUC = 0.81 ± 0.3), 15 feature genes (10 up-regulated and 5 down-regulated) were yielded (Figures 5A, B), while SVM-RFE (10-fold CV, AUC = 0.75 ± 0.17) identified 12 genes (7 upregulated and 5 down-regulated) (Figures 5C, D). The Venn diagram revealed the overlap of key genes identified by the two machine learning methods discussed above (Figure 5E). From the pool of identified genes, CHN1 and HEXD-IT1 emerged as upregulated in AD patients with comorbid depression, whereas CXCR6 was pinpointed as a downregulated gene. ROC analysis illustrated revealed that CHN1 and HEXD-IT1 exhibited high AUC values (> 0.8), indicating a strong association of these two genes with AD-related depression (Figure 5F).
Figure 5. Identification of key genes in AD patients with and without depression using machine learning methods. (A, B) Identification of hub genes in AD patients with depression using LASSO regression analysis, and (C, D) SVM-RFE algorithms. (E) Venn diagram screening overlapping hub genes identified by both LASSO regression analysis and SVM-RFE. (F) ROC curve analysis of core genes.
3.6 Immune-related interactions of key genes
To delve deeper into the immunological functions of key genes, we investigated their connections with activators across the four principal immune cell families. In the Th1 lineage, IL2 exhibited a substantial positive linkage with CHN1 (Figure 6A). In the Th2 family, markers such as GATA3, CCR3, and IL1RL1 demonstrated significant associations with CHN1 (Figure 6B). Within the Th17 subset, IL17 and IL23 were strongly correlated with the elevated expression of CHN1 and HEXD-IT1, underscoring their significance within the immune response landscape (Figure 6C). Conversely, no significant associations were detected between CXCR6 and the Th1, Th2 or Th17 families. However, in the Treg family, TGFBR2, IL10RB, and TGFB1 were negatively correlated with CHN1 and HEXD-IT1 but positively correlated with CXCR6 (Figure 6D). These findings suggest that the Th2 and Th17 pathways may drive the comorbidity of anxiety and depression in AD, while Treg family might exert a counter-regulatory effect.
Figure 6. Relationship between immune-related active molecules and key genes. Correlation analysis of key genes with members of the (A) Th1, (B) Th2, (C) Th17, and (D) Treg family members. *p < 0.05, *p < 0.01, **p < 0.01, ***p < 0.001.
3.7 Correlations between key genes and clinical features
Subsequently, we investigated the association of CHN1 and CXCR6 with the clinical features of AD patients (Figure 7). CHN1 expression positively correlated with SCORAD (r = 0.53, P = 0.015), EASI (r = 0.47, P = 0.037) and HADS-D (r = 0.56, P = 0.011). However, no significant correlation was observed with HADS-A (r = 0.44, P = 0.053). In contrast, CXCR6 expression showed no significant correlation with SCORAD (r = -0.28, P = 0.238), EASI (r = -0.28, P = 0.230), HADS-A (r = -0.17, P = 0.474) and HADS-D (r = -0.40, P = 0.079). TheX3se findings suggested CHN1 as a potential biomarker of both lesion severity and psychological burden in AD patients with comorbid depression.
Figure 7. Correlation between CHN1 and CXCR6 expression and clinical characteristics. Pearson correlation analysis of CHN1 mRNA expression levels with clinical characteristics, including (A) SCORAD, (B) EASI, (C) HADS-A, and (D) HADS-D. Correlation of CXCR6 mRNA expression levels with (E) SCORAD, (F) EASI, (G) HADS-A, and (H) HADS-D.
3.8 Validation of CHN1 expression in clinical samples
To further verify the correlation between CHN1 and AD-related depression, qPCR and IHC analysis were performed in samples of AD patients with (N = 11) and without depression (N = 9), with cohort demographics and clinical characteristics detailed in Table 2. IHC staining of the skin lesions revealed markedly stronger CHN1 expression in the lesion tissues of AD patients with depression compared to those without (Figures 8A, B). Similarly, qPCR analysis demonstrated significantly higher levels of CHN1 expression in PBMCs of AD patients with depression (Figure 8C). These findings supported the critical role of CHN1 in the pathogenesis of AD-related depression.
Figure 8. Verification of CHN1 expression in clinical samples. (A) IHC staining showing representative CHN1 expression in skin lesions of AD patients. (B) Histograms showing IHC scores of CHN1 expression in AD patients with depression (N = 11) and without depression (N = 9). (C) qPCR analysis of CHN1 mRNA expression levels in PBMCs from AD patients with (N = 11) and without depression (N = 9). **p < 0.01.
4 Discussion
This study firstly combined machine learning techniques with whole-blood transcriptome analysis to explore inflammation-related depression in AD patients. Through these analyses, we identified several key neural and immune pathways enriched in depressed AD, including T-cell receptor signaling, immunoregulatory interactions, negative regulation of Th17 cell differentiation, and neurotransmitter uptake. We further pinpointed CHN1 as a potential biomarker closely linked to both the inflammatory response and depression severity. These findings suggest that depression in AD may be driven by long-lasting peripheral inflammation influencing central nervous system function.
The comorbidity of AD with anxiety and depression has been observed through large-scale epidemiological and neuroimaging studies (8, 41–43). Skin inflammation activates lesional sensory neurons through cytokine receptors (e.g., IL-31R, IL-4R, etc.) and ion channel receptors (e.g., TRPV1, PAR2, etc.), which transmit signals along a three-neuron pathway to the brain, affecting central itch perception and emotional regulation (44, 45). Chronic pruritus activates brain regions involved in emotion regulation, thereby exacerbating anxiety and depression. A DNFB-induced chronic-pruritus mouse model identified a key anxiety-related circuit- the parabrachial nucleus (PBN) – central medial nucleus of the thalamus (CM) – medial prefrontal cortex (mPFC) pathway (46, 47). Additional mechanisms involving the hippocampus and amygdala dysfunction, further contribute to psychiatric comorbidities (48–50). Notably, chronic skin inflammation also affect the nervous system via the bloodstream, leading to comorbidities across organs and tissues, known as the “atopic march”. For example, cutaneous inflammation in AD can gradually promote the progression of allergic colitis through multiple pathways, including the succinate-Tuft cells-IL-25-ILC2s axis, mitochondrial DNA-STING signaling pathway, and the TSLP-eosinophil axis, which all mediate organ-to-organ communication through the circulation (51, 52). Elevated levels of pro-inflammatory factors in the circulation are likewise common in depressed patients, supporting a mechanistic link between peripheral immune activation and the neuropsychiatric disorders (53, 54). Our findings expand on this concept, suggesting that inflammatory mediators transmitted through the blood circulation may contribute to the development of psychiatric comorbidities in AD.
We further identified CHN1 as a key gene significantly upregulated in PBMCs of AD patients with depression. Correlation analysis demonstrated a positive association between CHN1 expression and clinical severity scores, including HADS-D, SCORAD, and EASI scores. CHN1 encodes a2-chimaerin, a regulator of the Rho GTPase - activating protein (GAP), crucial in neurogenesis and axon guidance (55, 56). As a negative regulator of Rac1 in hippocampal neurons, loss of CHN1 disrupts dendritic branching and augments poly - innervated spine formation (57, 58). The deficiency of CHN1 during the embryonic or juvenile period results in a remarkable impact on the cognitive function and behavioral manifestations (59). Moreover, CHN1 has been implicated in neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease (60, 61), and has shown to have differential expression in inflammatory disorders like asthma (62), psoriasis (63), and dermatomyositis (64), suggesting a role in neuro-immune signaling linking peripheral inflammation to the neural function. In our validation cohort, qPCR and IHC confirm CHN1 upregulation in both peripheral blood and skin lesions of AD patients with depression. Given prior evidence of neuro-immune circuit engagement in AD, we propose that neuronal CHN1 elevation may remodel Rac1-dependent axons, heighten pruriceptor excitability, and amplify neuropeptide release that skews cutaneous immunity (65, 66). These results identify CHN1 as a key neuroimmune mediator and potential biomarker for depression risk in AD. Clinically, CHN1 assessment could enable (i) the early identification of high-risk patients, (ii) monitoring of symptom burden and therapeutic response, and (iii) development of CHIN1- targeted interventions once mechanistic pathways are validated. Longitudinal and interventional researches are warranted to establish robust assay platforms, clinically meaningful cutoffs, and correlations with standardized psychiatric outcomes.
Despite these advances, several limitations requiring consideration. The modest size of the validation cohort may restrict the statistical power, increase the risk of overfitting and biological uncertainty, common challenges in small-sample WGCNA analysis and machine-learning analyses that can generate overly optimistic performance estimates. Also, age and SCORAD/EASI scores were not modeled as continuous covariates in the RNA-seq analysis, which may have inflated the number of false-positive DEGs. The limitation may restrict the generalizability of our findings and the robustness of validating CHN1 as a stable biomarker, particularly given the heterogeneity of immune pathway dysregulation among different subgroups of patients with AD and depression comorbidity. Future multi-center studies with larger cohorts are therefore needed to validate the reliability of CHN1 and refine its clinical applicability. Additionally, mechanistic studies should further elucidate the pathways underlying f neuropsychiatric comorbidities in AD, ultimately guiding targeted therapeutic strategies to improve the mental-health outcomes in AD populations.
5 Conclusion
This study advances our understanding of the intricate relationship between AD and depression. By employing bioinformatics and machine learning techniques, we identified CHN1 as a promising biomarker associated with depression in AD patients. Clinical validation confirmed a marked upregulation of CHN1 in peripheral blood and its strong correlation with disease-severity indices. Moreover, increased expression of CHN1 in skin lesions of AD patients with depression underscored CHN1’s potential as a biomarker or therapeutic target. Targeting CHN1 may provide new insights into the neuro-immune mechanisms driving depressive symptoms and pave the way for more precise and effective management of neuropsychiatric comorbid symptoms in AD population.
Data availability statement
The data presented in the study are deposited in the NCBI Gene Expression Omnibus (GEO) repository, accession number GSE307177.
Ethics statement
The studies involving human participants were reviewed and approved by the Ethics Committee of Wuhan Union Hospital (protocol No. UHCT240584). The participants provided their written informed consent to participate in this study.
Author contributions
YW: Conceptualization, Data curation, Writing – original draft, Methodology. YL: Data curation, Writing – original draft, Methodology. MC: Writing – original draft, Data curation. DL: Data curation, Writing – original draft, Supervision. CS: Data curation, Supervision, Writing – review & editing, Conceptualization, Writing – original draft.
Funding
The author(s) declare financial support was received for the research and/or publication of this article. This work was supported by the National Natural Science Foundation (U24A20703) and the Natural Science Foundation of Hubei Province (2023AFB1024).
Acknowledgments
We sincerely thank Professor Adam J. Adler for his significant contributions to review and revise the manuscript. We are also grateful to the Mental Health Center of Tongji Medical College, Huazhong University of Science and Technology (HUST), for sharing the data, and to the School of Public Health at HUST for providing expertise in statistics, genomics, and data science.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
Supplementary material
The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fimmu.2025.1677275/full#supplementary-material
Supplementary Figure 1 | (A). Venn diagram screening overlapping DEGs identified by different analytical model. (B). Determination of soft-thresholding power in WGCNA. (C). Heatmap of the Topological Overlap Matrix for all genes.
References
1. Langan SM, Irvine AD, and Weidinger S. Atopic dermatitis. Lancet. (2020) 396:345–60. doi: 10.1016/S0140-6736(20)31286-1
2. Odhiambo JA, Williams HC, Clayton TO, Robertson CF, and Asher MI. Global variations in prevalence of eczema symptoms in children from ISAAC Phase Three. J Allergy Clin Immunol. (2009) 124:1251–8. doi: 10.1016/j.jaci.2009.10.009
3. Ismail N and Bray N. Atopic dermatitis: economic burden and strategies for high-quality care. Br J Dermatol. (2020) 182:1087–8. doi: 10.1111/bjd.18636
4. Wollenberg A and Bieber T. Atopic dermatitis: from the genes to skin lesions. Allergy. (2000) 55:205–13. doi: 10.1034/j.1398-9995.2000.00115.x
5. Broderick C, Ziehfreund S, van Bart K, Arents B, Eyerich K, Weidinger S, et al. Biomarkers associated with the development of comorbidities in patients with atopic dermatitis: A systematic review. Allergy. (2023) 78:84–120. doi: 10.1111/all.15578
6. Cheng BT and Silverberg JI. Depression and psychological distress in US adults with atopic dermatitis. Ann Allergy Asthma Immunol. (2019) 123:179–85. doi: 10.1016/j.anai.2019.06.002
7. Schonmann Y, Mansfield KE, Hayes JF, Abuabara K, Roberts A, Smeeth L, et al. Atopic eczema in adulthood and risk of depression and anxiety: A population-based cohort study. J Allergy Clin Immunol Pract. (2020) 8:248–57. doi: 10.1016/j.jaip.2019.08.030
8. Rønnstad ATM, Halling-Overgaard A-S, Hamann CR, Skov L, Egeberg A, and Thyssen JP. Association of atopic dermatitis with depression, anxiety, and suicidal ideation in children and adults: A systematic review and meta-analysis. J Am Acad Dermatol. (2018) 79:448–56. doi: 10.1016/j.jaad.2018.03.017
9. Xie Q-W, Dai X, Tang X, Chan CHY, and Chan CLW. Risk of mental disorders in children and adolescents with atopic dermatitis: A systematic review and meta-analysis. Front Psychol. (2019) 10:1773. doi: 10.3389/fpsyg.2019.01773
10. Kern C, Wan J, LeWinn KZ, Ramirez FD, Lee Y, McCulloch CE, et al. Association of atopic dermatitis and mental health outcomes across childhood: A longitudinal cohort study. JAMA Dermatol. (2021) 157:1200–8. doi: 10.1001/jamadermatol.2021.2657
11. Drucker AM, Wang AR, Li W-Q, Sevetson E, Block JK, and Qureshi AA. The burden of atopic dermatitis: summary of a report for the national eczema association. J Invest Dermatol. (2017) 137:26–30. doi: 10.1016/j.jid.2016.07.012
12. Ferrari AJ, Charlson FJ, Norman RE, Patten SB, Freedman G, Murray CJL, et al. Burden of depressive disorders by country, sex, age, and year: findings from the global burden of disease study 2010. PloS Med. (2013) 10:e1001547. doi: 10.1371/journal.pmed.1001547
13. Chatrath S, Lei D, Yousaf M, Chavda R, Gabriel S, and Silverberg JI. Longitudinal course and predictors of depressive symptoms in atopic dermatitis. J Am Acad Dermatol. (2022) 87:582–91. doi: 10.1016/j.jaad.2022.04.061
15. Dantzer R, O'Connor JC, Lawson MA, and Kelley KW. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology. (2011) 36:426–36. doi: 10.1016/j.psyneuen.2010.09.012
16. Howren MB, Lamkin DM, and Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. (2009) 71:171–86. doi: 10.1097/PSY.0b013e3181907c1b
17. Ong PY and Leung DYM. Immune dysregulation in atopic dermatitis. Curr Allergy Asthma Rep. (2006) 6:384–9. doi: 10.1007/s11882-996-0008-5
18. Boguniewicz M and Leung DYM. Atopic dermatitis: a disease of altered skin barrier and immune dysregulation. Immunol Rev. (2011) 242:233–46. doi: 10.1111/j.1600-065X.2011.01027.x
19. Wen H-C, Czarnowicki T, Noda S, Malik K, Pavel AB, Nakajima S, et al. Serum from Asian patients with atopic dermatitis is characterized by TH2/TH22 activation, which is highly correlated with nonlesional skin measures. J Allergy Clin Immunol. (2018) 142:324–8. doi: 10.1016/j.jaci.2018.02.047
20. Ungar B, Garcet S, Gonzalez J, Dhingra N, Correa da Rosa J, Shemer A, et al. An integrated model of atopic dermatitis biomarkers highlights the systemic nature of the disease. J Invest Dermatol. (2017) 137:603–13. doi: 10.1016/j.jid.2016.09.037
21. Chen X-J and Sun Y-G. Central circuit mechanisms of itch. Nat Commun. (2020) 11:3052. doi: 10.1038/s41467-020-16859-5
22. Huang X, Hussain B, and Chang J. Peripheral inflammation and blood-brain barrier disruption: effects and mechanisms. CNS Neurosci Ther. (2021) 27:36–47. doi: 10.1111/cns.13569
23. Elahy M, Jackaman C, Mamo JC, Lam V, Dhaliwal SS, Giles C, et al. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing. (2015) 12:2. doi: 10.1186/s12979-015-0029-9
24. Wen S, Elias PM, Wakefield JS, Mauro TM, and Man MQ. The link between cutaneous inflammation and cognitive impairment. J Eur Acad Dermatol Venereol. (2022) 36:1705–12. doi: 10.1111/jdv.18360
25. Li YJ, Kresock E, Kuplicki R, Savitz J, and McKinney BA. Differential expression of MDGA1 in major depressive disorder. Brain Behav Immun Health. (2022) 26:100534. doi: 10.1016/j.bbih.2022.100534
26. Omrani M, Chiarelli RR, Acquaviva M, Bassani C, Dalla Costa G, Montini F, et al. Machine learning-driven diagnosis of multiple sclerosis from whole blood transcriptomics. Brain Behav Immun. (2024) 121:269–77. doi: 10.1016/j.bbi.2024.07.039
27. Chu DK, Schneider L, Asiniwasis RN, Boguniewicz M, De Benedetto A, Ellison K, et al. Atopic dermatitis (eczema) guidelines: 2023 American Academy of Allergy, Asthma and Immunology/American College of Allergy, Asthma and Immunology Joint Task Force on Practice Parameters GRADE- and Institute of Medicine-based recommendations. Ann Allergy Asthma Immunol. (2024) 132:274–312. doi: 10.1016/j.anai.2023.11.009
29. Atherton DJ, Bieber I, Bonilazi F, Broberg A, Calza A, and Coleman R. Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of the European Task Force on Atopic Dermatitis. Dermatol (Basel Switzerland). (1993) 186:23–31. doi: 10.1159/000247298
30. Hanifin JM, Thurston M, Omoto M, Cherill R, Tofte SJ, and Graeber M. The eczema area and severity index (EASI): assessment of reliability in atopic dermatitis. EASI Evaluator Group Exp Dermatol. (2001) 10:11–8. doi: 10.1034/j.1600-0625.2001.100102.x
31. Watland S, Solberg Nes L, Ekeberg Ø, Rostrup M, Hanson E, Ekstedt M, et al. Effects of The Caregiver Pathway intervention on symptoms of post-intensive care syndrome among family caregivers to critically ill patients: long-term results from a randomized controlled trial. Intensive Care Med. (2025). doi: 10.1007/s00134-025-08139-x
32. Tu Y-H, Huang T-C, Tzeng Y-S, Chen S-P, Wang S-J, and Wang Y-F. Prevalence, clinical correlates, and functional implications of vestibular symptoms in patients with migraine. Neurology. (2025) 105:e214248. doi: 10.1212/WNL.0000000000214248
33. Love MI, Huber W, and Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. (2014) 15:550. doi: 10.1186/s13059-014-0550-8
34. Zhou B, Zhou N, Liu Y, Dong E, Peng L, Wang Y, et al. Identification and validation of CCR5 linking keloid with atopic dermatitis through comprehensive bioinformatics analysis and machine learning. Front Immunol. (2024) 15:1309992. doi: 10.3389/fimmu.2024.1309992
35. Nomiri S, Karami H, Baradaran B, Javadrashid D, Derakhshani A, Nourbakhsh NS, et al. Exploiting systems biology to investigate the gene modules and drugs in ovarian cancer: A hypothesis based on the weighted gene co-expression network analysis. BioMed Pharmacother. (2022) 146:112537. doi: 10.1016/j.biopha.2021.112537
36. Li H, Huang Y, Sharma A, Ming W, Luo K, Gu Z, et al. From cellular infiltration assessment to a functional gene set-based prognostic model for breast cancer. Front Immunol. (2021) 12:751530. doi: 10.3389/fimmu.2021.751530
37. Walter W, Sánchez-Cabo F, and Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics. (2015) 31:2912–4. doi: 10.1093/bioinformatics/btv300
38. Chen D, Liu J, Zang L, Xiao T, Zhang X, Li Z, et al. Integrated machine learning and bioinformatic analyses constructed a novel stemness-related classifier to predict prognosis and immunotherapy responses for hepatocellular carcinoma patients. Int J Biol Sci. (2022) 18:360–73. doi: 10.7150/ijbs.66913
39. Liu Z, Liu L, Weng S, Guo C, Dang Q, Xu H, et al. Machine learning-based integration develops an immune-derived lncRNA signature for improving outcomes in colorectal cancer. Nat Commun. (2022) 13:816. doi: 10.1038/s41467-022-28421-6
40. Reel PS, Reel S, Pearson E, Trucco E, and Jefferson E. Using machine learning approaches for multi-omics data analysis: A review. Biotechnol Adv. (2021) 49:107739. doi: 10.1016/j.bioteChadv.2021.107739
41. Sutaria N, Adawi W, Goldberg R, Roh YS, Choi J, and Kwatra SG. Itch: pathogenesis and treatment. J Am Acad Dermatol. (2022) 86:17–34. doi: 10.1016/j.jaad.2021.07.078
42. Valet M, Pfab F, Sprenger T, Wöller A, Zimmer C, Behrendt H, et al. Cerebral processing of histamine-induced itch using short-term alternating temperature modulation–an FMRI study. J Invest Dermatol. (2008) 128:426–33. doi: 10.1038/sj.jid.5701002
43. Pfab F, Valet M, Sprenger T, Huss-Marp J, Athanasiadis GI, Baurecht HJ, et al. Temperature modulated histamine-itch in lesional and nonlesional skin in atopic eczema - a combined psychophysical and neuroimaging study. Allergy. (2010) 65:84–94. doi: 10.1111/j.1398-9995.2009.02163.x
44. Mack MR and Kim BS. The itch-scratch cycle: A neuroimmune perspective. Trends Immunol. (2018) 39:980–91. doi: 10.1016/j.it.2018.10.001
45. Steinhoff M, Ahmad F, Pandey A, Datsi A, AlHammadi A, Al-Khawaga S, et al. Neuroimmune communication regulating pruritus in atopic dermatitis. J Allergy Clin Immunol. (2022) 149:1875–98. doi: 10.1016/j.jaci.2022.03.010
46. Li J-N, Wu X-M, Zhao L-J, Sun H-X, Hong J, Wu F-L, et al. Central medial thalamic nucleus dynamically participates in acute itch sensation and chronic itch-induced anxiety-like behavior in male mice. Nat Commun. (2023) 14:2539. doi: 10.1038/s41467-023-38264-4
47. Sibener LJ, Mosberger AC, Chen TX, Athalye VR, Murray JM, and Costa RM. Dissociable roles of distinct thalamic circuits in learning reaches to spatial targets. Nat Commun. (2025) 16:2962. doi: 10.1038/s41467-025-58143-4
48. Sanders KM, Sakai K, Henry TD, Hashimoto T, and Akiyama T. A subpopulation of amygdala neurons mediates the affective component of itch. J Neurosci. (2019) 39:3345–56. doi: 10.1523/JNEUROSCI.2759-18.2019
49. Mu D and Sun Y-G. Circuit mechanisms of itch in the brain. J Invest Dermatol. (2022) 142:23–30. doi: 10.1016/j.jid.2021.09.022
50. Chen W, Guo H, Zhou N, Mai Y, Hu T, Xu X, et al. Distinct eLPBChAT projections for methamphetamine withdrawal anxiety and primed reinstatement of conditioned place preference. Theranostics. (2024) 14:2881–96. doi: 10.7150/thno.95383
51. Wang S, Liu B, Huang J, He H, Zhou L, He Y, et al. Succinate and mitochondrial DNA trigger atopic march from atopic dermatitis to intestinal inflammation. J Allergy Clin Immunol. (2023) 151:1050–66. doi: 10.1016/j.jaci.2022.11.026
52. Noti M, Kim BS, Siracusa MC, Rak GD, Kubo M, Moghaddam AE, et al. Exposure to food allergens through inflamed skin promotes intestinal food allergy through the thymic stromal lymphopoietin-basophil axis. J Allergy Clin Immunol. (2014) 133:1390–9. doi: 10.1016/j.jaci.2014.01.021
53. Raison CL, Capuron L, and Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol. (2006) 27:24–31. doi: 10.1016/j.it.2005.11.006
54. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. (2010) 67:446–57. doi: 10.1016/j.biopsych.2009.09.033
55. Recabarren D and Alarcón M. Gene networks in neurodegenerative disorders. Life Sci. (2017) 183:83–97. doi: 10.1016/j.lfs.2017.06.009
56. Zhao H, Wang L, Wang S, Chen X, Liang M, Zhang X, et al. CHN1 promotes epithelial-mesenchymal transition via the Akt/GSK-3β/Snail pathway in cervical carcinoma. J Transl Med. (2021) 19:295. doi: 10.1186/s12967-021-02963-7
57. Valdez CM, Murphy GG, and Beg AA. The Rac-GAP alpha2-chimaerin regulates hippocampal dendrite and spine morphogenesis. Mol Cell Neurosci. (2016) 75:14–26. doi: 10.1016/j.mcn.2016.06.002
58. Zhang H, Ben Zablah Y, Zhang H, and Jia Z. Rho signaling in synaptic plasticity, memory, and brain disorders. Front Cell Dev Biol. (2021) 9:729076. doi: 10.3389/fcell.2021.729076
59. Iwata R, Matsukawa H, Yasuda K, Mizuno H, Itohara S, and Iwasato T. Developmental racGAP α2-chimaerin signaling is a determinant of the morphological features of dendritic spines in adulthood. J Neurosci. (2015) 35:13728–44. doi: 10.1523/JNEUROSCI.0419-15.2015
60. Kato T, Konishi Y, Shimohama S, Beach TG, Akatsu H, and Tooyama I. Alpha1-chimaerin, a Rac1 GTPase-activating protein, is expressed at reduced mRNA levels in the brain of Alzheimer's disease patients. Neurosci Lett. (2015) 591:19–24. doi: 10.1016/j.neulet.2015.02.013
61. Capurro A, Bodea L-G, Schaefer P, Luthi-Carter R, and Perreau VM. Computational deconvolution of genome wide expression data from Parkinson's and Huntington's disease brain tissues using population-specific expression analysis. Front Neurosci. (2014) 8:441. doi: 10.3389/fnins.2014.00441
62. McGeachie MJ, Dahlin A, Qiu W, Croteau-Chonka DC, Savage J, Wu AC, et al. The metabolomics of asthma control: a promising link between genetics and disease. Immun Inflammation Dis. (2015) 3:224–38. doi: 10.1002/iid3.61
63. Liu Y, Wang H, Cook C, Taylor MA, North JP, Hailer A, et al. Defining patient-level molecular heterogeneity in psoriasis vulgaris based on single-cell transcriptomics. Front Immunol. (2022) 13:842651. doi: 10.3389/fimmu.2022.842651
64. Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, et al. Interferon and biologic signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PloS One. (2012) 7:e29161. doi: 10.1371/journal.pone.0029161
65. Feng X, Zhan H, and Sokol CL. Sensory neuronal control of skin barrier immunity. Trends Immunol. (2024) 45:371–80. doi: 10.1016/j.it.2024.03.008
Keywords: atopic dermatitis (AD), depression, CHN1, neuro-immune mechanism, comorbidity, gene
Citation: Wang Y, Liu Y, Chen M, Liu D and Shen C (2025) CHN1 as a potential predictive genetic biomarker for atopic dermatitis-related depression. Front. Immunol. 16:1677275. doi: 10.3389/fimmu.2025.1677275
Received: 31 July 2025; Accepted: 30 October 2025;
Published: 17 November 2025.
Edited by:
Svetlana P. Chapoval, University of Maryland, United StatesReviewed by:
Liborija Lugović-Mihić, University of Zagreb, CroatiaYu Wang, Sichuan University, China
Copyright © 2025 Wang, Liu, Chen, Liu and Shen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Chen Shen, OTA1NjI1NTQ4QHFxLmNvbQ==
†These authors have contributed equally to this work
Miao Chen1,2