Skip to main content
BMJ Open logoLink to BMJ Open
. 2025 Sep 22;15(9):e102876. doi: 10.1136/bmjopen-2025-102876

Mendelian randomisation and single-cell transcriptomic analyses reveal serotonin promotes multiple sclerosis progression by suppressing adenosine deaminase activity

Luofei Huang 1,0, Jian Shi 2,0, Han Li 3, Quanzhi Lin 4,
PMCID: PMC12458839  PMID: 40983575

Abstract

Abstract

Objective

To investigate the causal relationship between serotonin levels, adenosine deaminase (ADA) activity and multiple sclerosis (MS) progression using an integrative multi-omics approach.

Methods and analysis

A two-sample Mendelian randomisation (MR) analysis was performed using inverse variance weighted (IVW) estimation to assess causality between serotonin, ADA and MS risk. Single-cell transcriptomic data from the Gene Expression Omnibus (GSE194078) were analysed to identify ADA-expressing immune cell subpopulations. Moreover, machine learning algorithms (Support Vector Machine-Recursive Feature Elimination, Least Absolute Shrinkage and Selection Operator and random forest) were applied to identify diagnostic biomarkers, following which a nomogram was constructed and validated.

Results

MR analysis revealed that serotonin levels were positively correlated with MS progression (IVW β=0.350, p=3.63E-05), whereas genetically predicted ADA levels were inversely associated with MS risk (IVW β=−0.395, p=2.73E-04). Additionally, serotonin levels exhibited an inverse causal relationship with ADA activity (IVW β=−0.089, p=8.70E-03), with no evidence of reverse causation. Single-cell analysis identified 18 cellular subpopulations and six major immune cell types, with ADA highly expressed in T-NK cells and expressed at lower levels in platelets. Meanwhile, ADA expression was higher in the low immune receptor signalling group. Enrichment analysis indicated that differentially expressed genes were enriched in biological processes such as cytoplasmic translation and RNA splicing, as well as Kyoto Encyclopedia of Genes and Genome pathways such as Ribosome and Neurodegeneration-Multiple Diseases. Three key feature genes (IK, UBA52 and CCDC25) were identified, and the nomogram based on these genes demonstrated high diagnostic accuracy, with an AUC of 1.000 in the training dataset and 0.976 in the validation dataset.

Conclusions

Serotonin promotes MS progression by inhibiting ADA activity, positioning the serotonin-ADA axis as a potential therapeutic target. The identified biomarkers (IK, UBA52 and CCDC25) and the constructed nomogram may enhance diagnostic precision for MS, providing valuable insights for MS management and laying a theoretical reference for future studies.

Keywords: Multiple sclerosis, Chronic Disease, Pathology, GENETICS


STRENGTHS AND LIMITATIONS OF THIS STUDY.

  • This study integrated multi-omics approaches (Mendelian randomisation, single-cell transcriptomics and machine learning) to systematically characterise the serotonin-adenosine deaminase axis in multiple sclerosis.

  • Inverse variance weighted estimation and comprehensive sensitivity analyses ensured robust causal inference, with machine learning identifying core biomarkers.

  • The predominant reliance on European-derived genomic data may introduce population stratification bias, thereby limiting the generalisability of the findings to diverse ethnic groups.

  • While single-cell sequencing provided high-resolution insights into cellular heterogeneity, technical artefacts associated with high-throughput omics and Mendelian randomisation assumptions are methodological constraints.

  • The findings require validation in large-scale, prospective clinical studies to corroborate translational relevance.

Introduction

As is well documented, multiple sclerosis (MS) is a chronic and heterogeneous autoimmune disorder hallmarked by immune-mediated demyelination and neurodegeneration within the central nervous system (CNS),1 2 resulting in a broad spectrum of neurological symptoms and varying degrees of disability.3 4 Its pathogenesis involves genetic susceptibility, environmental factors (such as EB virus infection, vitamin D deficiency, smoking and high sodium diets) and abnormal immune responses.5 Immunologically, autoreactive CD4 T cells attack myelin, with Th1 and Th17 cells playing crucial roles in triggering neuroinflammation and Treg cells exerting immunosuppressive effects.6 B cells contribute to the pathological process through both antibody-dependent and antibody-independent mechanisms, promoting the production of autoantibodies and activating T cells, which further exacerbate the inflammatory response. Prolonged microglial activation results in sustained inflammation and damage to neural tissue.7 Recently, the presence of exhausted T cells, characterised by reduced effector function and persistent expression of inhibitory receptors, has been observed in MS lesions, suggesting a potential role in chronic inflammation and immune dysregulation.8 Although current therapies such as interferon-β and Fingolimod can alleviate symptoms,9 10 side effects and inter-individual variability in treatment responses highlight the pressing need for the development of novel therapeutic targets and strategies.11

Serotonin, a neurotransmitter that participates in mood regulation, also exerts immunomodulatory effects.12 13 Meanwhile, the dysregulation of adenosine deaminase (ADA), an enzyme involved in purine metabolism, has been associated with various immunological disorders.14 15 Thus, the interaction between serotonin levels and ADA activity presents a novel axis with potential therapeutic implications, warranting further mechanistic explorations and clinical investigation in the context of MS.

At present, cutting-edge methodologies such as single-cell sequencing,16 Mendelian randomisation (MR) analysis17 and advanced machine-learning models hold significant promise in elucidating the intricate relationship between serotonin levels, ADA inhibition and MS progression.18 Indeed, MR has been widely applied in biomedical studies19 20 and provides a robust framework for inferring causal relationships between genetic variants, serotonin levels, ADA activity and MS risk.21 Furthermore, the application of sophisticated machine-learning models enables the integration and analysis of complex datasets, revealing intricate biological patterns and interactions underlying disease mechanisms.22 Taken together, these advanced methodologies offer a comprehensive approach to unravelling the role of the serotonin–ADA axis in MS, laying a reference for the development of novel therapeutic strategies and personalised medicine approaches in the management of this debilitating condition.

Materials and methods

Research and design scheme

In this study, a two-sample MR analysis was performed to elucidate the causal relationships between 91 inflammatory cytokines, 1400 circulating plasma metabolites and MS. Furthermore, the MR analysis was expanded to specifically assess causal links between metabolite levels and MS, thus enabling an evaluation of their potential influence on the observed associations. This approach facilitated an in-depth examination of the intricate relationship between metabolite levels and MS, mediated through the regulation of inflammatory cytokine levels. The validity of MR-based causal inference is predicated on three foundational assumptions: a robust association between genetic variants and exposure variables,23 the absence of potential confounders that influence the relationship between genetic variants and the outcome (ie, MS) and the critical premise that the exposure (metabolite levels) represents the sole pathway through which genetic variants affect the outcome, thereby minimising pleiotropic effects.24 25 This rigorous approach strengthened the reliability of causal inferences, which were subsequently corroborated at the single-cell level, thereby elucidating the intricate interactions between metabolite levels and MS progression through the modulation of inflammatory cytokines (figure 1).

Figure 1. Study flowchart. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; LASSO, Least Absolute Shrinkage and Selection Operator; PCA, principal component analysis; SVM-RFE, Support Vector Machine-Recursive Feature Elimination; UMAP, Uniform Manifold Approximation and Projection.

Figure 1

Sources of exposure and outcome data

Exposure data for inflammatory cytokines were derived from a genome-wide protein quantitative trait loci (pQTL) analysis of 91 plasma proteins measured in 14 824 participants using the Olink Target platform.26 Data on 1091 distinct metabolites and 309 metabolite ratios were retrieved from extensive genome-wide association studies (GWAS) involving 8299 individuals in the Canadian Longitudinal Study on Ageing cohort.27 Both cytokine and metabolite data included summary statistics (ie, effect sizes and P values) for genetic variants associated with their levels. For MS outcomes, GWAS summary statistics (including ORs and p values) for genetic variants linked to MS risk were sourced from the International MS Genetics Consortium, covering 47 429 cases and 68 374 controls of European ancestry.28 All datasets analysed in this study were retrieved from publicly accessible GWAS databases; given their open-access and de-identified nature, the requirement for ethical approval was waived. In the present study, MS progression was indirectly evaluated using cross-sectional GWAS data on MS risk (case-control status), given that longitudinal data on disease progression were not available in the public datasets. While MS risk and progression are related, they represent distinct aspects of the disease. The findings of this study may provide valuable insights into the genetic determinants of MS susceptibility, which may inform mechanisms underlying disease progression. Serotonin levels were estimated using genetic variants associated with serum serotonin metabolites. ADA activity was inferred from genetic variants linked to ADA enzyme activity or gene expression levels in blood samples, as reported in prior GWAS and pQTL studies.

Collection and analysis of single-cell transcriptome sequencing data

The single-cell transcriptomic dataset for MS was obtained from the Gene Expression Omnibus (GEO)29 database, specifically the GSE194078 dataset. Data extraction was conducted using the R programming language in conjunction with the Seurat software package.30 Quality control parameters were established as follows: (1) exclusion of cells expressing fewer than 200 or over 4000 genes and (2) exclusion of cells with mitochondrial gene expression accounting for over 10% of total gene expression. Following preprocessing, gene expression data were normalised. Seurat was employed for quality control of the single-cell dataset. Initially, principal component analysis (PCA) was employed for dimensionality reduction, followed by further reduction using the Uniform Manifold Approximation and Projection (UMAP) algorithm. Based on the results of PCA,31 32 single-cell clustering was visualised via UMAP,33 and subpopulation cell clustering was further refined using t-Distributed Stochastic Neighbour Embedding (t-SNE).34 The MonacoImmuneData tool was used to annotate cellular subpopulations. The subsequent analysis focused on the levels of metabolic receptor signalling (encompassing transport and uptake) and alterations in inflammatory factor expression. Immune cell populations were stratified into two cohorts based on expression levels: one with elevated receptor signal expression and the other with low expression. Stringent selection criteria (p<0.05, Log2 Fold Change >0.25) were applied to identify differentially expressed genes (DEGs) in these cohorts.

Functional enrichment analysis

Functional enrichment analysis was performed to systematically characterise DEGs and identify potential functional targets, as described in previous studies.35,37 Gene Ontology (GO) analysis was performed to annotate genes across three primary domains, namely, molecular function, biological process (BP) and cellular component. Additionally, Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analysis was carried out to identify biological pathways linked with the DEGs.

Selection of signature genes

A systematic analysis of DEGs was performed to identify signature genes associated with MS. Several machine-learning methods, including Random Forest38 and Support Vector Machine-Recursive Feature Elimination (SVM-RFE),39 which are commonly used for key factor selection, were applied. In addition, Least Absolute Shrinkage and Selection Operator (LASSO) regression was employed to construct linear models and identify significant variables,40 as outlined in previous studies.41 42 Finally, a Venn diagram was generated to identify signature genes.

Construction and validation of the diagnostic model

Based on the identified signature genes, a diagnostic nomogram was constructed to predict the risk of MS. The predictive accuracy of the model was evaluated using calibration curves and decision curve analysis (DCA). Finally, ROC curves were plotted using R software to further assess model performance. The nomogram was trained on feature gene expression data from the training set and validated using an independent dataset with no overlapping samples, ensuring an unbiased evaluation of its predictive performance.

Construction of interaction networks for shared feature genes

GeneMANIA (http://www.genemania.org/) was used to construct interaction networks among three shared feature genes. The detailed results of these interaction networks are provided in the online supplemental materials.

Statistical analysis

MR analysis was conducted using the MR software package (version 0.4.3), employing the inverse variance weighted (IVW) method43 and the weighted median method.44 The former was employed to estimate the impact of exposure variables on outcomes based on the validity of the MR assumption. Cochran’s Q test was used to assess residual heterogeneity in the IVW model (p<0.05). Additionally, the MR–Egger intercept test was employed to evaluate potential pleiotropy in causal estimates (p<0.05). Data visualisation encompassed scatter plots, funnel plots and forest plots. Scatter plots were generated to demonstrate the consistency of results and the influence of potential outliers. Funnel plots were constructed to assess the robustness of correlations and verify the absence of heterogeneity. Lastly, forest plots provided a visual representation of the interaction between instrumental variables (single-nucleotide polymorphism (SNP)) and study outcomes. In MR analyses, the beta (β) coefficient represents the expected change in the log-odds ratio of MS risk per SD increase in the exposure (serotonin levels or ADA activity). Instrumental variables (SNPs) for serotonin and ADA were selected based on genome-wide significance (p<5×10⁸) in GWAS datasets. SNPs were excluded if evidence of horizontal pleiotropy was detected (MR–Egger intercept test, p<0.05) or weak instrument bias was present (F-statistic<10).

Patient and public involvement

Patients and the public were not involved in the design, conduct, reporting or dissemination of this study. The research used publicly available GWAS datasets and single-cell transcriptomic data from the GEO database, which do not involve direct patient or public participation. No primary data were collected from patients, and the study objectives and outcomes were determined solely by the research team.

Results

MR analysis of serotonin levels regulating MS progression via inhibition of ADA activity

To explore the causal relationship between serotonin-mediated ADA inhibition and MS, a two-sample MR analysis was performed using the IVW method. After selecting and harmonising IVs, eight SNPs were retained for serotonin following the exclusion of palindromic/ambiguous variants, those lacking instrumental utility and SNPs with inconsistent causal directionality (onlinesupplemental tables S1 S2undefined). For ADA, six SNPs were selected (onlinesupplemental tables S3 S4undefined). All retained SNPs exhibited F-statistics exceeding 10, confirming robust instrumental validity. MR results indicated that serotonin promotes MS progression, with statistical significance maintained after false discovery rate (FDR) correction (IVW method:β=0.350, p=3.63E-05, P(FDR)=0.049; online supplemental table S5). In contrast, genetically predicted ADA levels exhibited an inverse association with MS risk, which also remained significant post-FDR adjustment (IVW method: β=−0.395, p=2.73E-04, P(FDR)=0.012; online supplemental table S5). Concurrently, a negative causal relationship was identified between serotonin levels and ADA activity (IVW method: β=−0.089, p=8.70E-03; figure 2A, online supplemental tables S5), with no evidence of reverse causation detected between (IVW method: β=−0.011, p=0.742; online supplemental tables S5). As expected, sensitivity analyses yielded consistent results, with no evidence of pleiotropy-related bias (online supplemental figure S1). The reliability of the findings was further validated using scatter plots (figure 2B–D). Detailed analyses (onlinesupplemental tables S6S9) corroborated the absence of pleiotropic effects. At the same time, Cochran’s Q test revealed the absence of significant heterogeneity (onlinesupplemental tables S10S13). Collectively, these findings suggest that serotonin promotes MS progression by inhibiting ADA activity, supporting the hypothesis of an interaction between serotonin and ADA in the pathogenesis of MS and providing a theoretical foundation for identifying potential therapeutic targets.

Figure 2. Scatter plots illustrating the correlations between serotonin levels, ADA activity and MS progression. (A) Association between ADA activity and MS. (B) Relationship between serotonin levels and MS. (C) Correlation between serotonin levels and ADA activity. (D) Forest plot depicting the causal relationship between serotonin levels and ADA activity. ADA, adenosine deaminase; MR, Mendelian randomisation; MS, multiple sclerosis.; SNP, single-nucleotide polymorphism.

Figure 2

Transcriptomic data reveal divergent cellular constituents in MS

Single-cell transcriptomic analysis, following dimensionality reduction and clustering, yielded 18 distinct cellular subpopulations (figure 3A) and six major immune cell types, namely, T cells, monocytes, NK cells, B cells, common myeloid progenitors and platelets (figure 3B,C). Next, ADA expression levels were quantified and visualised across these cell types (figure 3D), with the highest levels observed in T-NK cells and the lowest in platelets. Consequently, the ensuing analysis focused on T-NK cells to explore the involvement of immune-related metabolic receptor signalling pathways. Immune cells were stratified into two groups according to receptor signalling activity, namely, the high and low expression groups. Interestingly, ADA expression was elevated in the low receptor signalling group (figure 3E,F). Overall, these results suggest that serotonin may modulate MS progression by regulating ADA expression in coordination with immune receptor signalling pathways.

Figure 3. Visualisation of transcriptomic data. (A) UMAP plot displaying 18 clusters. (B) Cell type proportions. (C) ADA levels across different cell types. (D) Distribution of intracellular ADA levels. (E) Proportions of cells with high and low ADA levels in receptor signalling groups. (F) Expression levels of ADA in high and low receptor signalling groups; ADA, adenosine deaminase; UMAP, Uniform Manifold Approximation and Projection.

Figure 3

Enrichment analysis of DEGs

Enrichment analysis revealed that the DEGs were primarily enriched in BPs related to cytoplasmic translation, RNA splicing, ATP synthesis coupled with electron transport and protein–RNA complex organisation (figure 4A,B). The most enriched KEGG pathways included ribosome, amyotrophic lateral sclerosis, pathways of neurodegeneration-multiple diseases and chemical carcinogenesis-reactive oxygen species (figure 4C,D). These findings offer insights into the molecular mechanisms underlying related diseases and may assist in the identification of potential biomarkers or therapeutic targets, contributing to the development of future treatment strategies.

Figure 4. Functional enrichment analysis of differentially expressed genes. (A)Gene Ontology (GO) bar chart. (B) GO bubble chart. (C) Kyoto Encyclopedia of Genes and Genome (KEGG) bar chart. (D) KEGG bubble chart.

Figure 4

Selection of feature genes

Initially, 16 key feature variables were identified using SVM-RFE (figure 5A,B). Logistic regression analysis subsequently identified 15 related genes with statistically significant associations (figure 5C,D). A random forest model, integrated with feature selection techniques, was then applied to evaluate classification error, resulting in the identification of 30 relatively important genes (figure 5E,F). Thereafter, a Venn diagram was plotted to visualise overlaps among the gene sets acquired from these methods (figure 5G), ultimately revealing three shared feature genes, namely, IK, UBA52 and CCDC25.

Figure 5. Workflow for Identifying multiple sclerosis-related feature genes; (A, B) Selection and validation of biomarker genes using the Support Vector Machine-Recursive Feature Elimination method. (C) Feature selection using the Least Absolute Shrinkage and Selection Operator regression model. (D) Optimisation of feature selection under minimum absolute shrinkage criteria. (E) Relationship between the number of classification trees and error rates in the random forest model. (F) Relevant feature genes with Millennium Development Goal scores. (H) Screening of feature genes using Venn diagrams. MS, multiple sclerosis.

Figure 5

Construction of an MS diagnostic model based on feature genes

Furthermore, a diagnostic nomogram for MS was constructed based on the feature genes IK, UBA52 and CCDC25. The nomogram was visualised (figure 6A), and its performance was evaluated using a calibration curve (figure 6B), and DCA (figure 6C) in R software. The calibration curve illustrated a strong agreement between predicted and observed outcomes, with the ‘Number high risk’ curve closely overlapping the ‘Number high risk with event’ curve. These findings collectively signal that the nomogram exhibits robust predictive performance and high accuracy for MS diagnosis (figure 6D).

Figure 6. Construction and validation of the diagnostic model for MS. (A) Nomogram for MS. (B) Calibration curve of the model. (C) Decision curve analysis. (D) Clinical impact curve; MS, multiple sclerosis.

Figure 6

Evaluation of the nomogram using training and validation datasets

The diagnostic performance of the nomogram constructed from the three shared feature genes (IK, UBA52 and CCDC25) was evaluated using ROC curve analysis. In the training dataset (GSE13551), the individual AUCs for IK, UBA52 and CCDC25 were 0.985, 0.969 and 0.988, respectively (figure 7A). Notably, the combined nomogram achieved perfect classification performance, with an AUC of 1.000 (95% CI: 1.000 to 1.000) (figure 7B). In the validation dataset (GSE21942), the corresponding AUCs for the three genes were 0.798, 0.867 and 0.819 (figure 7C), with the nomogram maintaining high accuracy with an AUC of 0.976 (95% CI: 0.914 to 1.000) (figure 7D). These results conjointly demonstrate that IK, UBA52 and CCDC25, when integrated into the nomogram, may serve as effective biomarkers for the diagnosis of MS.

Figure 7. Validation of the nomogram using receiver operating characteristic (ROC) curves: (A) ROC curve and area under the curve (AUC) for each common feature gene in GSE13551. (B) ROC curve and AUC for the nomogram in GSE13551. (C) ROC curve and AUC for each common feature gene in GSE21942. (D) ROC curve and AUC for the nomogram in GSE21942.

Figure 7

Interaction network of shared feature genes and their co-expressed genes

Subsequently, a comprehensive analysis was carried out to elucidate the interaction network and functional associations of the shared feature genes. The results revealed a complex interaction network, with physical interactions comprising the largest proportion (77.64%). Additional interaction types included co-expression (8.01%), predicted interactions (5.37%), co-localisation (3.63%), genetic interactions (2.87%), pathway-related associations (1.88%) and shared protein domain interactions (0.60%). Functionally, the network was primarily involved in processes such as protein targeting to the endoplasmic reticulum, establishment of protein localisation to the endoplasmic reticulum, protein-membrane targeting, cotranslational protein targeting to membranes and cytosolic ribosome activity (figure 8 and Supplementary Materials S1).

Figure 8. Analysis of shared feature genes and their co-expression using GeneMANIA.

Figure 8

Discussion

This comprehensive analysis highlights the intricate interplay between serotonin levels, ADA activity and MS progression. By integrating MR analysis, single-cell transcriptomic profiling and validation through advanced machine-learning models, this study provides novel insights into the molecular mechanisms underlying MS pathogenesis and the identification of potential therapeutic targets.

MR analysis revealed a dual role for serotonin and ADA in MS progression. Specifically, a positive causal relationship was observed between serotonin levels and MS risk, suggesting that elevated serotonin may increase MS susceptibility. In contrast, genetically predicted ADA levels were negatively correlated with MS risk, indicating a potential protective effect against MS susceptibility. Of note, the inverse causal relationship between serotonin and ADA supports the hypothesis that serotonin may partially contribute to MS susceptibility by inhibiting ADA activity. These findings emphasise the potential of targeting the serotonin–ADA axis as a therapeutic strategy for MS. Serotonin,45 traditionally recognised for its role in mood regulation,46 also exerts immunomodulatory effects, thereby impacting susceptibility to autoimmune disorders such as MS through a complex array of mechanisms.47

By interacting with a broad spectrum of 5-hydroxytryptamine (5-HT) receptors across various immune cell types, serotonin can either drive pro-inflammatory cytokine release or exert anti-inflammatory effects, thereby influencing inflammatory processes central to MS pathogenesis.48,50 Additionally, its capacity to alter the permeability of the blood–brain barrier (BBB) facilitates immune cell infiltration into the CNS, further contributing to demyelination and neuronal damage.12 51 52 Within the CNS, serotonin exerts neuroimmunomodulatory effects and affects microglial and astrocyte activity, thereby fostering a neuroinflammatory environment.53 Molecularly, its impact is mediated via diverse signalling pathways activated by its receptors, including cyclic adenosine monophosphate (cAMP), phosphoinositide and mitogen-activated protein kinase (MAPK) pathways, which collectively modulate gene expression, cytokine production and cell survival. This intricate interplay of immunological, neuroinflammatory and molecular mechanisms highlights its significant yet complex role in MS progression and its potential as a therapeutic target. A key aspect of serotonin’s action involves its interaction with ADA, where elevated serotonin levels suppress ADA activity, leading to adenosine dysregulation and potentially aggravating inflammatory responses.54 55 The relationship between ADA and MS encompasses several mechanisms, including the immunomodulatory role of adenosine, which can influence immune cell behaviour and cytokine profiles, thereby affecting MS pathogenesis.56 57 Dysregulated ADA activity contributes to neuroinflammation within the CNS, exacerbating demyelination and neuronal damage, which are hallmarks of MS.58 Furthermore, the effect of ADA on BBB integrity and lymphocyte function demonstrates its role in immune cell infiltration into the CNS and the autoimmune response characteristic of MS.15 By affecting adenosine levels, ADA indirectly mediates glial cell activation and the inflammatory cascade in the CNS, emphasising its multifaceted involvement in the pathophysiology of MS. Single-cell transcriptomic analysis provided additional mechanistic insights by identifying cellular subpopulations and signalling pathways implicated in MS pathogenesis.31 59 Besides, single-cell transcriptomic data offer profound insights into the characterisation of cell subpopulations and signalling pathways; however, some limitations, such as technical and batch effects, merit acknowledgement.60 61

Differential ADA expression was observed across various immune cell types, with significantly high levels in T-NK cells. Furthermore, the analysis unveiled a correlation between immune receptor signalling and ADA expression, suggesting a potential regulatory mechanism through which serotonin may modulate ADA activity via immune cell signalling pathways. Serotonin exerts its effects by binding to 5-HT receptors on immune cells, thereby activating downstream signalling cascades such as the cAMP, phosphatidylinositol and MAPK pathways. These pathways, in turn, regulate gene transcription, cytokine production and immune cell survival. In MS, serotonin may influence immune cell function and inflammatory responses by inhibiting ADA activity, leading to disruptions in adenosine metabolism. Specifically, reduced ADA activity results in elevated extracellular levels of adenosine, which binds to its receptors on immune cells and modulates cytokine secretion and immune activation. This adenosine-mediated immunoregulation may play a decisive role in MS progression. In addition, enrichment analysis of DEGs provided further mechanistic insight. The enrichment of BPs such as cytoplasmic translation, RNA splicing and ATP synthesis coupled with electron transport implies that MS is associated with impairments in core cellular functions. More importantly, KEGG pathway analysis identified links to neurodegenerative diseases, including amyotrophic lateral sclerosis, indicating potential shared mechanisms between MS and other neurodegenerative disorders. Taken together, these findings signal that serotonin-mediated ADA suppression may contribute not only to immune dysregulation but also to neurodegenerative processes in MS, offering a more integrated perspective of disease progression and potential targets for therapeutic intervention.

The identification of three feature genes, IK, UBA52 and CCDC25, through machine-learning-based feature selection methods further expands our understanding of key molecular contributors to MS. These genes, involved in immune regulation and protein synthesis, may represent novel biomarkers for MS diagnosis and progression. As anticipated, the nomogram developed based on these genes demonstrated excellent predictive accuracy in both the training and validation datasets, indicating its potential as a valuable diagnostic tool for clinical application.

The IK genes, particularly IKKα, IKKβ and IKKγ, are critical regulatory factors in the NF-κB signalling pathway and are closely related to the pathogenesis of MS. NF-κB plays a vital role in immune and inflammatory processes by phosphorylating the IkB protein, facilitating the entry of NF-κB into the nucleus and up-regulating the expression of pro-inflammatory genes. In patients with MS, aberrant NF-κB pathway activation leads to an overactive immune response that damages myelin.62 The IKK complex plays an integral role in the activation of T cells and B cells63 by promoting the production of Th1 and Th17 cells and anti-myelin antibodies, thereby enhancing immune responses.64 Additionally, the IK genes promote the release of inflammatory mediators through the NF-κB pathway, leading to excessive activation of glial cells, which in turn exacerbates damage to myelin and neurons. Therefore, inhibiting IKK activity may be a potential therapeutic strategy for MS, aiming to attenuate inflammation and myelin damage. UBA52 may influence immune cell protein homeostasis via ubiquitination, while CCDC25 can modulate cytoskeletal dynamics during immune cell migration. These novel associations require functional exploration in MS models.

Besides, the findings indicate that these three genes may play an essential role in the pathogenesis of MS, warranting further exploration and analysis in future studies. Furthermore, the interaction network analysis of these shared feature genes illustrated extensive co-expression and physical interaction patterns, with protein targeting the endoplasmic reticulum emerging as a central biological function. This suggests that disruptions in protein transport and localisation may contribute to MS pathogenesis. The relationships between serotonin levels, ADA activity and protein targeting pathways offer therapeutic opportunities for exploring targeted therapies aimed at correcting these dysregulated processes. Noteworthily, a recent study using Clustered Regularly Interspaced Short Palindromic Repeats-based screening and cell line IC50 data has identified novel key genes implicated in drug resistance, emphasising the broader relevance of gene-based approaches in uncovering disease mechanisms and therapeutic vulnerabilities.65 These findings suggest that serotonin contributes to the progression of multiple sclerosis by inhibiting ADA activity, highlighting a potential therapeutic axis for intervention. Targeting ADA may aid in suppressing inflammatory responses and promoting neuroprotection, thereby mitigating disease progression.

Conclusion

This study reveals that serotonin promotes MS progression by inhibiting ADA activity. Genetic and single-cell analyses linked serotonin to increased MS risk and ADA to protective effects. Three key genes (IK, UBA52 and CCDC25) were identified as potential diagnostic biomarkers. These findings highlight the serotonin–ADA axis as a therapeutic target and provide insights for future MS research.

Supplementary material

online supplemental file 1
bmjopen-15-9-s001.docx (14KB, docx)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 2
bmjopen-15-9-s002.pdf (1.9MB, pdf)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 3
bmjopen-15-9-s003.pdf (332KB, pdf)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 4
bmjopen-15-9-s004.csv (1.9KB, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 5
bmjopen-15-9-s005.csv (2.1KB, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 6
bmjopen-15-9-s006.csv (3.5KB, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 7
bmjopen-15-9-s007.csv (5.1KB, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 8
bmjopen-15-9-s008.csv (280B, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 9
bmjopen-15-9-s009.csv (147B, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 10
bmjopen-15-9-s010.xls (19KB, xls)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 11
bmjopen-15-9-s011.xls (18.5KB, xls)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 12
bmjopen-15-9-s012.xls (18.5KB, xls)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 13
bmjopen-15-9-s013.csv (240B, csv)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 14
bmjopen-15-9-s014.xls (19KB, xls)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 15
bmjopen-15-9-s015.xls (19KB, xls)
DOI: 10.1136/bmjopen-2025-102876
online supplemental file 16
bmjopen-15-9-s016.xls (19KB, xls)
DOI: 10.1136/bmjopen-2025-102876

Acknowledgements

We express our sincere gratitude to the Medicine Collaborative Group for their invaluable support.

Footnotes

Funding: This study received no specific grant from any funding agency, commercial or not-for-profit sectors. QL serves as the guarantor and assumes full responsibility for research conduct and publication decisions.

Prepublication history and additional supplemental material for this paper are available online. To view these files, please visit the journal online (https://doi.org/10.1136/bmjopen-2025-102876 ).

Provenance and peer review: Not commissioned; externally peer reviewed.

Patient consent for publication: Not applicable.

Ethics approval: This study was based on an analysis of publicly available datasets, including single-cell transcriptomic data from the Gene Expression Omnibus (GEO) database (accession numbers: GSE194078, GSE13551 and GSE21942) and genome-wide association study (GWAS) data from the International MS Genetics Consortium, Zhao et al (2023), and Chen et al (2023) cohorts. All datasets are de-identified and publicly accessible, and thus, the requirements for informed consent and ethical approval were waived in accordance with local institutional guidelines and regulations.For inquiries related to research integrity and ethical compliance of this study, please contact the Research Management Office of the corresponding author's institution: The First Affiliated Hospital of Guangxi University of Science and Technology, Email: kjklyyfy@163.com.

Data availability free text: All data used in the study can be accessed via the GEO digital repository, the IEUOpen GWAS project database (https://gwas.mrcieu.ac.uk/datasets/) and the IEUOpen Unlimited download GWAS project database (https://gwas.mrcieu.ac.uk/datasets/).

Map disclaimer: The inclusion of any map (including the depiction of any boundaries therein), or of any geographic or locational reference, does not imply the expression of any opinion whatsoever on the part of BMJ concerning the legal status of any country, territory, jurisdiction or area or of its authorities. Any such expression remains solely that of the relevant source and is not endorsed by BMJ. Maps are provided without any warranty of any kind, either express or implied.

Patient and public involvement: Patients and/or the public were not involved in the design, conduct, reporting or dissemination plans of this research.

Data availability statement

Data are available in a public, open access repository.

References

  • 1.Koch-Henriksen N, Magyari M. Apparent changes in the epidemiology and severity of multiple sclerosis. Nat Rev Neurol. 2021;17:676–88. doi: 10.1038/s41582-021-00556-y. [DOI] [PubMed] [Google Scholar]
  • 2.Hafler DA. Multiple sclerosis. J Clin Invest. 2004;113:788–94. doi: 10.1172/JCI21357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Solaro C, Gamberini G, Masuccio FG. Depression in Multiple Sclerosis: Epidemiology, Aetiology, Diagnosis and Treatment. CNS Drugs. 2018;32:117–33. doi: 10.1007/s40263-018-0489-5. [DOI] [PubMed] [Google Scholar]
  • 4.Doshi A, Chataway J. Multiple sclerosis, a treatable disease. Clin Med (Lond) 2016;16:s53–9. doi: 10.7861/clinmedicine.16-6-s53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yamout BI, Alroughani R. Multiple Sclerosis. Semin Neurol. 2018;38:212–25. doi: 10.1055/s-0038-1649502. [DOI] [PubMed] [Google Scholar]
  • 6.Kaskow BJ, Baecher-Allan C. Effector T Cells in Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018;8:a029025. doi: 10.1101/cshperspect.a029025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wanleenuwat P, Iwanowski P. Role of B cells and antibodies in multiple sclerosis. Mult Scler Relat Disord. 2019;36:101416. doi: 10.1016/j.msard.2019.101416. [DOI] [PubMed] [Google Scholar]
  • 8.Liu H, Dong A, Rasteh AM, et al. Identification of the novel exhausted T cell CD8 + markers in breast cancer. Sci Rep. 2024;14:19142. doi: 10.1038/s41598-024-70184-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jakimovski D, Kolb C, Ramanathan M, et al. Interferon β for Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018;8:11.:a032003. doi: 10.1101/cshperspect.a032003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Brinkmann V, Davis MD, Heise CE, et al. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J Biol Chem. 2002;277:21453–7. doi: 10.1074/jbc.C200176200. [DOI] [PubMed] [Google Scholar]
  • 11.Oh J, Vidal-Jordana A, Montalban X. Multiple sclerosis: clinical aspects. Curr Opin Neurol. 2018;31:752–9. doi: 10.1097/WCO.0000000000000622. [DOI] [PubMed] [Google Scholar]
  • 12.Malinova TS, Dijkstra CD, de Vries HE. Serotonin: A mediator of the gut-brain axis in multiple sclerosis. Mult Scler. 2018;24:1144–50. doi: 10.1177/1352458517739975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sagonas I, Daoussis D. Serotonin and systemic sclerosis. An emerging player in pathogenesis. Joint Bone Spine. 2022;89:S1297-319X(21)00182-2. doi: 10.1016/j.jbspin.2021.105309. [DOI] [PubMed] [Google Scholar]
  • 14.Kutryb-Zajac B, Kawecka A, Caratis F, et al. The impaired distribution of adenosine deaminase isoenzymes in multiple sclerosis plasma and cerebrospinal fluid. Front Mol Neurosci. 2022;15:998023. doi: 10.3389/fnmol.2022.998023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Samuraki M, Sakai K, Odake Y, et al. Multiple sclerosis showing elevation of adenosine deaminase levels in the cerebrospinal fluid. Mult Scler Relat Disord. 2017;13:44–6. doi: 10.1016/j.msard.2017.02.005. [DOI] [PubMed] [Google Scholar]
  • 16.Jovic D, Liang X, Zeng H, et al. Single-cell RNA sequencing technologies and applications: A brief overview. Clin Transl Med. 2022;12:e694. doi: 10.1002/ctm2.694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Emdin CA, Khera AV, Kathiresan S. Mendelian Randomization. JAMA. 2017;318:1925–6. doi: 10.1001/jama.2017.17219. [DOI] [PubMed] [Google Scholar]
  • 18.Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018;50:1–14. doi: 10.1038/s12276-018-0071-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu H, Xie R, Dai Q, et al. Exploring the mechanism underlying hyperuricemia using comprehensive research on multi-omics. Sci Rep. 2023;13:7161. doi: 10.1038/s41598-023-34426-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liu H. Association between sleep duration and depression: A Mendelian randomization analysis. J Affect Disord. 2023;335:152–4. doi: 10.1016/j.jad.2023.05.020. [DOI] [PubMed] [Google Scholar]
  • 21.Birney E. Mendelian Randomization. Cold Spring Harb Perspect Med. 2022;12:a041302. doi: 10.1101/cshperspect.a041302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Aslam N, Khan IU, Bashamakh A, et al. Multiple Sclerosis Diagnosis Using Machine Learning and Deep Learning: Challenges and Opportunities. Sensors (Basel) 2022;22:7856. doi: 10.3390/s22207856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sleiman PMA, Grant SFA. Mendelian randomization in the era of genomewide association studies. Clin Chem. 2010;56:723–8. doi: 10.1373/clinchem.2009.141564. [DOI] [PubMed] [Google Scholar]
  • 24.Verduijn M, Siegerink B, Jager KJ, et al. Mendelian randomization: use of genetics to enable causal inference in observational studies. Nephrol Dial Transplant. 2010;25:1394–8. doi: 10.1093/ndt/gfq098. [DOI] [PubMed] [Google Scholar]
  • 25.Khasawneh LQ, Al-Mahayri ZN, Ali BR. Mendelian randomization in pharmacogenomics: The unforeseen potentials. Biomed Pharmacother. 2022;150:S0753-3322(22)00341-9. doi: 10.1016/j.biopha.2022.112952. [DOI] [PubMed] [Google Scholar]
  • 26.Zhao JH, Stacey D, Eriksson N, et al. Genetics of circulating inflammatory proteins identifies drivers of immune-mediated disease risk and therapeutic targets. Nat Immunol. 2023;24:1540–51. doi: 10.1038/s41590-023-01588-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen Y, Lu T, Pettersson-Kymmer U, et al. Genomic atlas of the plasma metabolome prioritizes metabolites implicated in human diseases. Nat Genet. 2023;55:44–53. doi: 10.1038/s41588-022-01270-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science. 2019;365 doi: 10.1126/science.aav7188. https://gwas.mrcieu.ac.uk/datasets/ Available. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Barrett T, Wilhite SE, Ledoux P, et al. NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids Res. 2013;41:D991–5. doi: 10.1093/nar/gks1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Satija R, Farrell JA, Gennert D, et al. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33:495–502. doi: 10.1038/nbt.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rodríguez-Lorenzo S, van Olst L, Rodriguez-Mogeda C, et al. Single-cell profiling reveals periventricular CD56bright NK cell accumulation in multiple sclerosis. Elife. 2022;11:e73849. doi: 10.7554/eLife.73849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liu Z. Visualizing Single-Cell RNA-seq Data with Semisupervised Principal Component Analysis. IJMS. 2020;21:5797. doi: 10.3390/ijms21165797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Becht E, McInnes L, Healy J, et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. 2018 doi: 10.1038/nbt.4314. [DOI] [PubMed] [Google Scholar]
  • 34.Kobak D, Berens P. The art of using t-SNE for single-cell transcriptomics. Nat Commun. 2019;10:5416. doi: 10.1038/s41467-019-13056-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liu H, Weng J. A comprehensive bioinformatic analysis of cyclin-dependent kinase 2 (CDK2) in glioma. Gene. 2022;822:S0378-1119(22)00144-5. doi: 10.1016/j.gene.2022.146325. [DOI] [PubMed] [Google Scholar]
  • 36.Li Y, Liu H. Clinical powers of Aminoacyl tRNA Synthetase Complex Interacting Multifunctional Protein 1 (AIMP1) for head-neck squamous cell carcinoma. Cancer Biomark. 2022;34:359–74. doi: 10.3233/CBM-210340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Liu H, Li Y. Potential roles of Cornichon Family AMPA Receptor Auxiliary Protein 4 (CNIH4) in head and neck squamous cell carcinoma. Cancer Biomark. 2022;35:439–50. doi: 10.3233/CBM-220143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Becker T, Rousseau A-J, Geubbelmans M, et al. Decision trees and random forests. Am J Orthod Dentofacial Orthop. 2023;164:894–7. doi: 10.1016/j.ajodo.2023.09.011. [DOI] [PubMed] [Google Scholar]
  • 39.Wu X, Qin K, Iroegbu CD, et al. Genetic analysis of potential biomarkers and therapeutic targets in ferroptosis from coronary artery disease. J Cell Mol Med. 2022;26:2177–90. doi: 10.1111/jcmm.17239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wang Q, Qiao W, Zhang H, et al. Nomogram established on account of Lasso-Cox regression for predicting recurrence in patients with early-stage hepatocellular carcinoma. Front Immunol. 2022;13:1019638. doi: 10.3389/fimmu.2022.1019638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Liu H, Tang T. A bioinformatic study of IGFBPs in glioma regarding their diagnostic, prognostic, and therapeutic prediction value. Am J Transl Res. 2023;15:2140–55. [PMC free article] [PubMed] [Google Scholar]
  • 42.Liu H, Tang T. MAPK signaling pathway-based glioma subtypes, machine-learning risk model, and key hub proteins identification. Sci Rep. 2023;13:19055. doi: 10.1038/s41598-023-45774-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Burgess S, Small DS, Thompson SG. A review of instrumental variable estimators for Mendelian randomization. Stat Methods Med Res. 2017;26:2333–55. doi: 10.1177/0962280215597579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bowden J, Davey Smith G, Haycock PC, et al. Consistent Estimation in Mendelian Randomization with Some Invalid Instruments Using a Weighted Median Estimator. Genet Epidemiol. 2016;40:304–14. doi: 10.1002/gepi.21965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mohammad-Zadeh LF, Moses L, Gwaltney-Brant SM. Serotonin: a review. J Vet Pharmacol Ther. 2008;31:187–99. doi: 10.1111/j.1365-2885.2008.00944.x. [DOI] [PubMed] [Google Scholar]
  • 46.MacLean MR, Fanburg B, Hill N, et al. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol. 2022;12:4103–18. doi: 10.1002/cphy.c220004. [DOI] [PubMed] [Google Scholar]
  • 47.Foley P, Lawler A, Chandran S, et al. Potential disease-modifying effects of selective serotonin reuptake inhibitors in multiple sclerosis: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2014;85:709–10. doi: 10.1136/jnnp-2013-306829. [DOI] [PubMed] [Google Scholar]
  • 48.Reverchon F, Guillard C, Mollet L, et al. T Lymphocyte Serotonin 5-HT7 Receptor Is Dysregulated in Natalizumab-Treated Multiple Sclerosis Patients. Biomedicines. 2022;10:2418. doi: 10.3390/biomedicines10102418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sales MC, Kasahara TM, Sacramento PM, et al. Selective serotonin reuptake inhibitor attenuates the hyperresponsiveness of TLR2+ and TLR4+ Th17/Tc17-like cells in multiple sclerosis patients with major depression. Immunology. 2021;162:290–305. doi: 10.1111/imm.13281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sacramento PM, Monteiro C, Dias ASO, et al. Serotonin decreases the production of Th1/Th17 cytokines and elevates the frequency of regulatory CD4+ T-cell subsets in multiple sclerosis patients. Eur J Immunol. 2018;48:1376–88. doi: 10.1002/eji.201847525. [DOI] [PubMed] [Google Scholar]
  • 51.Melnikov M, Sviridova A, Rogovskii V, et al. Serotoninergic system targeting in multiple sclerosis: the prospective for pathogenetic therapy. Mult Scler Relat Disord. 2021;51:S2211-0348(21)00155-3. doi: 10.1016/j.msard.2021.102888. [DOI] [PubMed] [Google Scholar]
  • 52.Sandyk R. Serotonergic neuronal sprouting as a potential mechanism of recovery in multiple sclerosis. Int J Neurosci. 1999;97:131–8. doi: 10.3109/00207459908994307. [DOI] [PubMed] [Google Scholar]
  • 53.El Oussini H, Bayer H, Scekic-Zahirovic J, et al. Serotonin 2B receptor slows disease progression and prevents degeneration of spinal cord mononuclear phagocytes in amyotrophic lateral sclerosis. Acta Neuropathol. 2016;131:465–80. doi: 10.1007/s00401-016-1534-4. [DOI] [PubMed] [Google Scholar]
  • 54.Duarte-Silva E, Ulrich H, Oliveira-Giacomelli Á, et al. The adenosinergic signaling in the pathogenesis and treatment of multiple sclerosis. Front Immunol. 2022;13:946698. doi: 10.3389/fimmu.2022.946698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Safarzadeh E, Jadidi-Niaragh F, Motallebnezhad M, et al. The role of adenosine and adenosine receptors in the immunopathogenesis of multiple sclerosis. Inflamm Res. 2016;65:511–20. doi: 10.1007/s00011-016-0936-z. [DOI] [PubMed] [Google Scholar]
  • 56.Spanevello RM, Mazzanti CM, Schmatz R, et al. The activity and expression of NTPDase is altered in lymphocytes of multiple sclerosis patients. Clin Chim Acta. 2010;411:210–4. doi: 10.1016/j.cca.2009.11.005. [DOI] [PubMed] [Google Scholar]
  • 57.Vivekanandhan S, Soundararajan CC, Tripathi M, et al. Adenosine deaminase and 5’nucleotidase activities in peripheral blood T cells of multiple sclerosis patients. Neurochem Res. 2005;30:453–6. doi: 10.1007/s11064-005-2680-6. [DOI] [PubMed] [Google Scholar]
  • 58.Polachini CRN, Spanevello RM, Casali EA, et al. Alterations in the cholinesterase and adenosine deaminase activities and inflammation biomarker levels in patients with multiple sclerosis. Neuroscience. 2014;266:266–74. doi: 10.1016/j.neuroscience.2014.01.048. [DOI] [PubMed] [Google Scholar]
  • 59.Jagadeesh KA, Dey KK, Montoro DT, et al. Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat Genet. 2022;54:1479–92. doi: 10.1038/s41588-022-01187-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu H, Guo Z, Wang P. Genetic expression in cancer research: Challenges and complexity. Gene Rep. 2024;37:102042. doi: 10.1016/j.genrep.2024.102042. [DOI] [Google Scholar]
  • 61.Liu H, Li Y, Karsidag M, et al. Technical and Biological Biases in Bulk Transcriptomic Data Mining for Cancer Research. J Cancer. 2025;16:34–43. doi: 10.7150/jca.100922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yan J, McCombe PA, Pender MP, et al. Reduced IκB-α Protein Levels in Peripheral Blood Cells of Patients with Multiple Sclerosis-A Possible Cause of Constitutive NF-κB Activation. J Clin Med. 2020;9:2534. doi: 10.3390/jcm9082534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pontoriero M, Fiume G, Vecchio E, et al. Activation of NF-κB in B cell receptor signaling through Bruton’s tyrosine kinase-dependent phosphorylation of IκB-α. J Mol Med (Berl) 2019;97:675–90. doi: 10.1007/s00109-019-01777-x. [DOI] [PubMed] [Google Scholar]
  • 64.Park H-L, Lee S-M, Min J-K, et al. IK acts as an immunoregulator of inflammatory arthritis by suppressing TH17 cell differentiation and macrophage activation. Sci Rep. 2017;7:40280. doi: 10.1038/srep40280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Liu H, Wang P. CRISPR screening and cell line IC50 data reveal novel key genes for trametinib resistance. Clin Exp Med. 2024;25:21. doi: 10.1007/s10238-024-01538-2. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    online supplemental file 1
    bmjopen-15-9-s001.docx (14KB, docx)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 2
    bmjopen-15-9-s002.pdf (1.9MB, pdf)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 3
    bmjopen-15-9-s003.pdf (332KB, pdf)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 4
    bmjopen-15-9-s004.csv (1.9KB, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 5
    bmjopen-15-9-s005.csv (2.1KB, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 6
    bmjopen-15-9-s006.csv (3.5KB, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 7
    bmjopen-15-9-s007.csv (5.1KB, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 8
    bmjopen-15-9-s008.csv (280B, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 9
    bmjopen-15-9-s009.csv (147B, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 10
    bmjopen-15-9-s010.xls (19KB, xls)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 11
    bmjopen-15-9-s011.xls (18.5KB, xls)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 12
    bmjopen-15-9-s012.xls (18.5KB, xls)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 13
    bmjopen-15-9-s013.csv (240B, csv)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 14
    bmjopen-15-9-s014.xls (19KB, xls)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 15
    bmjopen-15-9-s015.xls (19KB, xls)
    DOI: 10.1136/bmjopen-2025-102876
    online supplemental file 16
    bmjopen-15-9-s016.xls (19KB, xls)
    DOI: 10.1136/bmjopen-2025-102876

    Data Availability Statement

    Data are available in a public, open access repository.


    Articles from BMJ Open are provided here courtesy of BMJ Publishing Group

    RESOURCES