Next Article in Journal
RT-qPCR Testing and Performance Metrics in the COVID-19 Era
Previous Article in Journal
Mining Candidate Genes for Leaf Angle in Brassica napus L. by Combining QTL Mapping and RNA Sequencing Analysis
Previous Article in Special Issue
Next-Generation Sequencing of the Human Aqueous Humour Microbiome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptomic Module Discovery of Diarrhea-Predominant Irritable Bowel Syndrome: A Causal Network Inference Approach

1
Data Science Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte (Ba), Italy
2
Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte (Ba), Italy
3
Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
4
Department of Medicine, Laboratory of Biochemistry, University of Udine P.le Kolbe 4, 33100 Udine, Italy
5
INSERM U1052, CNRS UMR_5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France
6
Institute of Hepatology Lyon (IHL), 69002 Lyon, France
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(17), 9322; https://doi.org/10.3390/ijms25179322
Submission received: 10 July 2024 / Revised: 13 August 2024 / Accepted: 26 August 2024 / Published: 28 August 2024

Abstract

:
Irritable bowel syndrome with diarrhea (IBS-D) is the most prevalent subtype of IBS, characterized by chronic gastrointestinal symptoms in the absence of identifiable pathological findings. This study aims to investigate the molecular mechanisms underlying IBS-D using transcriptomic data. By employing causal network inference methods, we identify key transcriptomic modules associated with IBS-D. Utilizing data from public databases and applying advanced computational techniques, we uncover potential biomarkers and therapeutic targets. Our analysis reveals significant molecular alterations that affect cellular functions, offering new insights into the complex pathophysiology of IBS-D. These findings enhance our understanding of the disease and may foster the development of more effective treatments.

1. Introduction

Functional gastrointestinal disorders (FGIDs) encompass a spectrum of conditions characterized by chronic gastrointestinal (GI) symptoms without identifiable pathological findings on routine diagnostic assessments [1]. Among FGIDs, irritable bowel syndrome (IBS) is the most prevalent subtype [1]. IBS is a multifactorial disorder [2] marked by abdominal pain, altered stool consistency and frequency, and the absence of detectable biochemical or structural anomalies [3]. IBS is classified in four subtypes based on stool patterns: IBS with constipation (IBS-C), IBS with diarrhea (IBS-D), IBS with mixed bowel habit (IBS-M), and IBS unclassified (IBS-U) [4].
This study is primarily focused on IBS-D, which, according to multiple sources, is the most common subtype, followed by IBS-C and IBS-M. One study in an adult population found that IBS-D accounted for approximately 40% of cases, IBS-C for 35%, IBS-M for 23%, and IBS-U for 8% [5]. Interestingly, this functional disorder also affects pediatric populations [6]. Due to the absence of objective diagnostic biomarkers [7], these percentages may vary depending on the study population, the diagnostic criteria used (e.g., Rome III vs. Rome IV), and the geographical region [8]. Nevertheless, IBS-D consistently emerges as the most common subtype across various studies and regions.
Despite its prevalence and clinical impact, the pathophysiology of IBS-D remains poorly understood [9]. Complex diseases like FGIDs are thought to arise from a combination of molecular changes that disrupt cellular function [10]. Advances in omics technologies (e.g., genomics, transcriptomics, and proteomics) [11] have significantly contributed to our understanding of the fundamental mechanisms underlying these conditions [12].
Considerable research has been conducted on IBS and the role of the gut microbiota but our understanding of the biochemical, metabolic, functional, and inflammatory mechanisms underlying IBS-D remains limited [13]. Human studies are often restricted by the limited availability of biopsies for microstructural and molecular analysis [13]. Additionally, there is a lack of robust animal models that fully replicate the pathophysiology of IBS, particularly IBS-D [14]. Thanks to the availability nowadays of well-established experimental protocols and the reduced costs of omics platforms, large quantities of omics data are being generated [14] and made accessible through public databases (e.g., Gene Expression Omnibus) [15]. RNA sequencing (RNA-seq) technology, for example, is a crucial tool for defining the transcriptome landscape from isolated cells, tissue sections, or biopsy samples [16].
In this context, pathway enrichment analysis is essential for identifying active biological processes [17,18,19,20], elucidating disease-associated molecular pathways, devising novel therapeutic strategies, and delineating diagnostic biomarkers [21,22]. Integrating genomic expression data with biomolecular interaction databases, such as Reactome [23] or KEGG [24], offers significant advantages, especially in the big data era. This approach is essential to understand the disease complexity in each patient [12,25]. Consequently, network biology, aimed at enhancing our understanding of complex biological systems, has gained considerable interest in recent years [26]. Identifying pathways that drive disease-specific expression signatures can reveal “hidden nodes,” which, although not differentially expressed, play a crucial role in connecting differentially expressed genes within complex networks [27,28].
In this framework, the modular structure of biological networks provides critical insights into the connections and interactions between elements within a network. A disease module represents a collection of cellular components whose disruption can lead to a disease phenotype. Notably, these modules are identified as clusters of highly interconnected genes that show significant differences in expression between diseased and control cells [27]. Understanding these modules may help to elucidate disease mechanisms and biological functions, particularly during disease progression [29,30]. Furthermore, there is growing interest in employing inference analysis to model and quantify cause–effect relationships within modules, particularly from a multivariate statistical perspective in the context of experimental perturbation [31,32].
Given the complex and multifactorial nature of IBS-D, driven by both genetic and environmental factors, a causal network-based approach utilizing graph theory [33,34] and structural equation modeling [31] could be beneficial. This approach enables transcriptomic interactions, such as active disease modules and their associated communities, to be explored and inferences about their effects drawn. This study aimed to investigate the transcriptomic complexity of IBS-D using a graph theory-SEM integrated approach, applying network model theory to identify an IBS-D active module and its associated communities and explore/infer their effects. This innovative approach may provide new insights into the molecular mechanisms underlying IBS-D.

2. Results

2.1. Preprocessing and Dataset Selection

The gene expression profiles from two RNA-seq datasets, GSE146853 and GSE166869, derived from intestinal biopsies, were obtained from the NCBI Gene Expression Omnibus (GEO) database. A total of 164 raw FastQ files were processed, representing 38 IBS-D patients and 44 healthy controls. The average sequencing coverage per sample was 42.0 million reads (range: 28.3–68.1 million) for the GSE146853 dataset and 25.3 million reads (range: 3.1–44.7 million) for the GSE166869 dataset (Table S1). Additionally, the average ribosomal RNA (rRNA) content was 15% (range: 11.2–21.7%) for GSE146853 and 77.5% (range: 22.7–99.2%) for GSE166869 (Table S1). Due to insufficient quality control, the GSE166869 dataset was excluded from further analysis. The 35 samples collected at time point 1 (T1) from the GSE146853 dataset were subsequently analyzed. The results are summarized in Table 1.

2.2. Differentially Expressed Genes (DEG) and Reactome Enrichment Analyses

Preliminary principal component analysis (PCA) indicated that disease status was not associated with transcriptomic variability, as reflected in the first two principal components, regardless of their complexity (Figure S1). To further explore the transcriptomic complexity, a DEG analysis was conducted as an initial step, comparing the 35 samples (17 IBS-D patients and 18 healthy controls). This analysis identified 54 significant DEGs (p-value < 0.05), 34 being upregulated (log2FoldChange ≥ 1) and 20 downregulated (log2FoldChange ≤ −1). The identified DEGs are detailed in Table 2 and visualized in the volcano plot (Figure 1).
The DEGs detected by their ENSEMBL IDs were used to identify pathways in Reactome analysis tools that include at least one DEG, as detailed in Table S2 and illustrated in Figure 2.

2.3. Statistical Analysis

2.3.1. SEM-Based Gene Set Analysis (SEMgsa)

In the first step, an SEMgsa was performed on notable genes—without considering the newly discovered transcripts by RNA-seq analysis. Table 3 displays the results of the procedure. Notably, the pathways involved in the complement system—including the generation of C4 and C2 activators, classical antibody-mediated complement activation, the complement cascade, and its regulation—show significant inhibition (p < 0.05).
As shown in Table 4, 22 unique DEGs were identified. The pathways complement cascade and regulation of complement cascade included the highest number of DEGs, involving eight and seven genes, respectively. Consequently, these DEGs were also used as seed genes for the Steiner tree (ST) identification procedure.

2.3.2. Analysis of the Active IBS-D Module

Figure 3 shows the ST graph for IBS-D, generated by integrating Reactome pathways [23] and GEO data. The graph consists of 46 nodes and 45 edges. The 22 seed genes corresponding to DEGs are highlighted in aquamarine, and the 24 connector nodes in white. Bidirectional edges are represented in black.
Figure 4 illustrates the active IBS-D module, represented as the optimal IBS-D Directed Acyclic Graph (DAG), derived using a fitting strategy based on Structural Equation Modeling (SEM) and graph theory, as described in the methods section implemented in the R/SEMgraph packages [25]. In this approach, we developed a data-driven model search strategy, relying solely on data without validation against a reference network. Notably, Figure 4 highlights the addition of two edges: LRP6 → CA4 and UBC → SFRP1.
Table 5, Table 6 and Table 7 present the results of the SEM fitting analysis. Notably, both the IBS-D and control models yielded similar findings for the significant CA4 → DUOX2 effect (activation). However, there were contrasting results regarding the significant/suggestive SLC9B1 → CA4 effect—activation observed within IBS-D samples, while no activation was seen in the control samples. Similarly, the SCNN1G → AKAP1 effect showed non-activation in IBS-D samples but activation in control samples (see Table 5).
Regarding perturbation effects, CA4 (upregulated) and SFRP1 (downregulated) showed significant changes, while UBC was upregulated in a suggestive manner (0.05 < p < 0.10) (see Table 6).
Regarding edge differences between groups, the SLC9B1 → CA4 and UBC → SCNN1G effects were significantly upregulated, whereas the SLC9B1 → AKAP1, SLC5A1 → AKAP1, and SCNN1G → AKAP1 effects showed significant downregulation (see Table 7).
To better interpret the results of the SEM analysis, Figure 5 presents the three final graphs of the active IBS-D module: one for IBS-D samples (Figure 5A), one for control samples (Figure 5B), and a combined “node and edge perturbation” model illustrating the differences between the groups (Figure 5C). In these graphs, edges are color-coded based on their significance: significant direct effects (p-value < 0.05) in red (estimate > 0) or blue (estimate < 0), In contrast, non-significant edges are shown in gray. In Figure 5C, nodes are also color-coded, with pink nodes indicating activation and light-blue nodes indicating inhibition. Notably, no changes in causal relationships were observed.

2.3.3. Gene Communities Detection

Regarding the gene community structure identified through clustering, eight topological clusters (modularity = 0.668) were found in the Steiner tree graph (Figure 6A), and three clusters (modularity = 0.455) were identified in the optimal DAG (Figure 6B). The genes within the eight topological clusters of the Steiner tree are detailed in Table S3.
In the active module, the clusters are organized as follows: the first cluster includes DUOX2, CA4, and LRP6; the second cluster comprises MGAM, SLC5A1, AKAP1, and SLC9B1; and the third cluster consists of SCNN1G, UBC, SFRP1, and FZD1.

2.3.4. Characterization of the Gene Communities

The results of the characterization of the eight ST-related communities are presented in Table 8 and Figure 7. For detailed information on the genes belonging to these eight topological ST clusters, refer to Table S3.
The active IBS-D module’s communities were characterized by inputting the list of genes appertaining to the three communities; in this study, a ‘community number’ was attributed to each community for its identification. Notably, in the active module, the first community contained the DUOX2, CA4, and LRP6 genes; the second the MGAM, SLC5A1, AKAP1, and SLC9B1 genes; and the third one included the SCNN1G, UBC, SFRP1, and FZD1 genes. The results of the enrichment are shown in Figure 8 and Table 9.

3. Discussion

Research on IBS-D remains limited, and the biochemical, metabolic, functional, and inflammatory mechanisms underlying this disorder are still not fully understood. Current guidelines for managing IBS-D patients do not recommend endoscopic investigation with biopsy sampling, which makes it challenging to conduct detailed tissue and molecular investigations [35,36]. This study uses existing RNA-seq data to bridge these gaps and highlights the importance of integrating complementary data using advanced statistical methods. This approach aims to probe the functional and molecular mechanisms of IBS-D pathophysiology and may potentially help to identify molecular targets for developing and validating therapeutic strategies within a comprehensive, personalized treatment framework.
In biology and medicine, a significant challenge is posed by the need to identify crucial relationships or disrupted disease modules. Greedy algorithms are vital to identify the “active” disease module [35]. This study successfully identified and statistically matched the active module of IBS-D through gene network analysis, which revealed changes in molecular activity [35,37].
Using SEM to manage complex systems as multivariate networks, we combined network analysis with causal inference. This approach ensures robustness and reproducibility through a data-driven methodology [25]. Traditional statistical methods such as regression modeling often struggle to simultaneously handle multivariate, inferential, and graph theory aspects, while SEM effectively uncovers subtle gene dysregulations [25].
Our preliminary unsupervised PCA analysis of the 35 samples from GSE146853 indicated that IBS-D status was not associated with transcriptomic variability, suggesting that IBS-D is a functional disorder without significant structural or phenotypic alterations [1]. Topology-based enrichment analysis, a powerful method for identifying enrichment in gene expression data [38], revealed a significant inhibition of seven pathways related to the complement system. Notably, intestinal epithelial cells are the primary source of complement component production in the gastrointestinal tract [39], and the complement system is crucial for systemic microbial defense [40]. This system plays a key role in detecting and eliminating harmful bacteria, maintaining tissue homeostasis, and preventing infections [41]. Several studies have highlighted the interactions between intestinal microbiota dysbiosis and innate immune system activation in IBS [42], leading to inflammation, increased intestinal permeability, and alterations in the neuroendocrine system [43]. Increased levels of β-defensin-2 in IBS patients support the involvement of immune dysfunction in IBS [44,45]. Dysbiosis, an imbalance in gut microbes, is strongly linked to intestinal inflammation and GI disorders [46], particularly in IBS-D [47]. Elevated zonulin levels, indicative of an increased intestinal permeability or “leaky gut”, facilitate bacterial translocation and exacerbate IBS-D symptoms [47].
Although the role of the intestinal complement system in irritable bowel disease (IBD) is well-documented [48], its direct impact on IBS-D remains uncertain. It is hypothesized that, like in IBD, dysregulation of the complement system in IBS-D impairs intestinal barrier function and contributes to dysbiosis. Understanding the interplay among gut microbiota, the complement system, and zonulin activation could provide valuable insights into IBS-D pathophysiology and highlight potential therapeutic targets [49]. Modulating the gut microbiota through probiotics, prebiotics, or dietary interventions, along with targeting the complement system and zonulin pathways, could offer novel strategies for managing IBS-D and improving patient outcomes [50].
Regarding the DEG analysis, classical negative binomial analysis identified DUOX2 and SH2D6 as upregulated DEGs. DUOX2, which encodes dual oxidase 2, is involved in hydrogen peroxide production, which is crucial for mucosal defense and gut microbiota regulation. Elevated DUOX2 expression is associated with inflammatory conditions and microbial imbalances in animal models [51]. SH2D6, or SH2 domain-containing protein 6, plays a role in immune signaling pathways and is a marker for immune CD45+ Tuft-2 cells with antibacterial function [52]. The overexpression of DUOX2 and SH2D6 observed in IBS-D patients aligns with findings related to dysbiosis and complement pathways. The complement system, a component of the innate immune response, is crucial for maintaining gut homeostasis and controlling microbial populations in IBS-D patients, while dysregulation leads to microbial imbalance [41].
In the active IBS-D module, a Steiner tree algorithm was used to discover the transcriptomic network structure through a graph-based data-driven approach. This approach compactly connected “seed” nodes with additional nodes [25], building a more complex and causal network architecture. SEM fitting identified critical gene interactions and regulatory mechanisms, revealing two dysregulated genes: Carbonic Anhydrase 4 (CA4) and Secreted Frizzled-Related Protein 1 (SFRP1). The SEM approach detected these dysregulations, which were not identified using classical negative binomial analysis.
CA4, a zinc metalloenzyme, is involved in numerous physiological functions, including respiration, pH balance, and metabolic processes [53]. In IBD animal models, CA4 inhibition mitigates visceral pain and protects against colon damage [54]. This suggests that CA4 inhibitors could be potential treatments for visceral pain and colon protection in IBS-D.
SFRP1, a Wnt pathway antagonist, is regulated by promoter hypermethylation in colon and gastric tumors [55,56], and similar epigenetic regulation may occur in IBS-D, affecting inflammatory processes [57].
Among the IBS-D active module genes, SLC9B1, SLC5A1, and SCNN1G are involved in proton, electrolyte, and glucose transport. Disruptions in these processes can increase macromolecule permeability [58,59], leading to mucosal inflammation and epithelial barrier disruption, which may trigger innate immune responses and complement activation [50]. This suggests a water and electrolyte absorption disruption in IBS-D, resulting in increased epithelial permeability.
The A-kinase anchoring protein 1 (AKAP1) was involved in three downregulated effects (SLC9B1→ AKAP1, SLC5A1 → AKAP1, SCNN1G → AKAP1). AKAP1 recruits protein kinase A (PKA) and other signaling proteins to the external mitochondrial membrane, playing a crucial role in mitochondrial function, metabolic homeostasis, and disease onset and progression [60,61].
The interaction between SLC9B1 (NHE1) and CA4 suggests potential pH regulation alterations, while the interaction between UBC and SCNN1G involves ubiquitin C and ENaC, affecting sodium ion transport and fluid reabsorption [62,63].
In summary, the reported disease module analysis highlights the complexity of IBS-D pathology and suggests novel therapeutic strategies involving gut microbiota modulation, Wnt pathways, complement system targeting, and the regulation of water and electrolyte absorption. The methodology also identified biological sub-networks through pathway visualization, revealing that three of the eight Steiner tree-related communities are involved in the immune system. Notably, the complement pathway and the third community—enriched in di- and oligosaccharide metabolic processes—were relevant to IBS symptom management [64]. The sixth and seventh communities showed enriched Wnt signaling pathways, with direct correlations to the gut-brain axis [65,66]. The fourth and fifth communities were too small to offer any detailed explanation. Thus, concerning the IBS-D active module, the second and third communities were involved in metabolic processes and gut-brain axis correlations, by confirming the result from the Steiner tree communities analysis. Overall, the two community analyses confirm the IBS-D complexity and the involvement of both immune system and gut–brain axis.

4. Materials and Methods

4.1. Dataset Description and Study Workflow

To select RNA-Seq gene expression datasets from the public NCBI GEO, the query ‘(irritable bowel syndrome) AND (diarrhea OR IBS-D) AND (transcriptome OR RNA-seq) AND biopsy AND Homo Sapiens’ was used (assessed on 13 May 2024). This query yielded two bulk RNA-seq poly-A datasets, identified as GSE146853 and GSE166869. After verifying that these datasets contained experimental case–control data and that the data were generated using the Illumina sequencing platform (Illumina Inc., San Diego, CA, USA), the raw paired-end Fastq files were downloaded. To obtain the IBS-D active module and cluster communities, we followed the workflow depicted in Figure 9.

4.2. RNA-seq Pipeline and Dataset Quality Control Cut-Off

Fastq file coverage, expressed in millions of reads per sample, was assessed using FastQC v0.11.8 [67]. For RNA-seq analysis of complex eukaryotic transcriptomes, achieving 20–30 million reads per sample is crucial for comprehensive coverage, accurate gene expression quantification, and the detection of low-abundance transcripts [68,69]. A quality control cutoff of greater than 30 million reads per dataset was established to exclude datasets with insufficient coverage.
The ribosomal RNA (rRNA) percentage was computed using the Sequence Expression Analyzer (Seal) from BBtools v39.01 [70]. Typically, rRNA constitutes 60–90% of total RNA. A high rRNA content can negatively impact RNA-seq analysis by dominating the sequencing output, reducing the effective depth of mRNA sequencing, and impeding the detection of rare transcripts. Therefore, a quality control cutoff of less than 20% of the average rRNA percentage per dataset was established to exclude datasets with high rRNA content.
Subsequently, to remove sequencing adapters and low-quality bases, Illumina 50 bp paired-end reads from the Fastq files were trimmed using Trim Galore v0.6.10 [71]. The reads were then mapped to the GRCh38 primary assembly, downloaded from GENECODE version 35 [72], using the STAR aligner (version 2.7.0f) [72,73]. The raw gene counts matrix was obtained using Subread feature Counts (v2.0.2) [74], utilizing the GTF file from the primary assembly. The complete RNA-seq pipeline is available at https://github.com/MichelangeloAloisio/mRNAseq_pipeline (accessed on 2 January 2024) and the full matrix containing gene counts for all the 35 samples, is available on request.

4.3. DEGs Analysis and Reactome Enrichment Analysis

To preliminarily assess the raw variability of the data, we performed a principal component analysis (PCA) on log-transformed count data, focusing on the top 500 most variable features [65]. Disease status was also examined as a potential group factor to evaluate its contribution to variation.
Subsequently, a differentially expressed genes (DEG) analysis was conducted comparing IBS-D patients to healthy controls using a negative binomial approach. Based on the median of ratios method, normalized counts were utilized, and genes with ≤10 counts per sample were excluded. A volcano plot was employed to visually identify DEGs, applying a log-fold change threshold of ±2 and a p-value cutoff of <0.05. Selected DEGs, identified by ENSEMBL ID, were then used for pathway enrichment analysis via the Reactome Analysis Tool v88 [23]. The options selected in the Reactome tool included projecting to humans and including interactors. This analysis provided a list of pathways involving at least one DEG, refined by the Reactome enrichment analysis.

4.4. Statistical Analysis

To discover, manage, and fit the IBS-D active module (network), we employed a combination of graph theory and structural equation modeling (SEM) to extract critical relationships (edges) and perturbations (nodes).
Graph theory is widely used to model complex biological networks and has numerous applications in medical and biological literature related to -omics data [33,34]. SEM is a multivariate statistical approach that utilizes a system of simultaneous equations to describe the path relationships that generate the data. In this framework, a given variable (node or gene) can act as an explanatory variable in one or more equations, while serving as an outcome variable in others [31].
In our study, the network was constructed based on notable genes, excluding newly discovered transcripts from the RNA-seq analysis.
The process was carried out in two stages as follows: (i) SEM-based gene set analysis (SEMgsa): we first used SEMgsa [25] to identify differential expression patterns; and (ii) active module fitting: next, we fitted the active IBS-D module (network) [25] based on these patterns.
Graph theory was applied to (i) learn causal architecture using the Reactome [23] data to provide the active IBS-D module and (ii) search for network communities and paths, exploring the structure and relationships within the network.
SEM was used to (i) perform differential expression analysis (SEMgsa) for initial gene set analysis and (ii) fit the model for refining the active IBS-D network module.
It is important to note that graph theory also played a crucial role in the model-searching phase during the model-fitting step.

4.4.1. SEMgsa

SEMgsa was performed to detect differential expression of individual genes between IBS-D cases and controls and to infer their biological structure by pathways interrogation of Reactome [75,76]. In this step, we followed a topology-based approach [77], which incorporated the pathway structure, improving the performance [25]. The core of the methodology is explained in Grassi et al. [25], and is based on the RICF (Residual Iterative Conditional Fitting) algorithm [78,79].
To run this step, normalized gene expression data and the group variable (1 = IBS-D, 0 = control) obtained by the GEO database (https://www.ncbi.nlm.nih.gov/geo/, ID = GSE146853, accessed on 2 January 2024) were provided for the analysis. In this case, the gene expression data were preliminarily transformed by a nonparanormal transformation to complain the normality assumption required by SEM [25]. In addition, a list of pathways was provided involving at least one DEG (differentially expressed gene) returned by the bioinformatic analysis performed by the Reactome database. Briefly, the output returned node-specific group effect p-values, and Brown’s combined p-values of node activation and inhibition. Node-specific p-values were corrected for multiple comparisons (<a = 0.25), by adjusting for the false discovery rate (FDR) used for DEGs identification. The phase of DEGs identification was also carried out in SEM-GSA to account for the topological structure of the considered pathways. The choice to set a = 0.25 was made because this work is explorative, and the aim was to provide and fit an active IBS-D module including as much a priori transcriptomic information as possible. In detail, SEMgsa returned two outputs as follows: a table reporting (i) the number of nodes for each pathway, (ii) the number of differential expression genes (DEGs) within the pathways, after multiple test corrections, (iii) pathway perturbation status (activated vs. inhibited), (iv) Brown’s combined p-value of pathway node activation, (v) Brown’s combined p-value of pathway node inhibition, (vi) the Bonferroni combined p-value of activation and inhibition, expressed as 2× min(activation p-value, inhibition p-value), (vii) the adjusted Bonferroni p-value of pathway perturbation; i.e., min(number of pathways x p-value in [vii]; 1), and (viii) a list with DEG names per pathway.

4.4.2. Analysis of the Active IBS-D Module

We constructed and fitted an SEM-based transcriptomic active IBS-D module using differentially expressed genes (DEGs) identified through SEMgsa analysis. Initially, these DEGs, referred to as seed genes, were mapped onto the Reactome interactome—representing the union of all Reactome pathways—through a graph weighting procedure that utilized Fisher’s r-to-z transformation [80,81]. This approach tested for differences between groups in the correlation coefficients of interacting gene pairs [25].
To generate a perturbed reduced graph, representing the active IBS-D module, we applied a Steiner tree (ST) identification procedure based on Kou’s algorithm [82]. This process connected the seed genes to other genes (referred to as connector genes) by minimizing the total edge distance. For this purpose, the ST procedure transformed the p-values obtained from the r-to-z Fisher transformation by converting them into the inverse of the negative log(p-value), ensuring edge weights were reported within a positive continuous range. In graph theory, the ST is an acyclic graph that connects seed genes to additional nodes in the most compact manner possible [25].
After constructing the transcriptomic active IBS-D module, SEM was applied to the gene expression data to quantify and verify perturbations (status group->gene) and effects (gene-i->gene-j), denoted as β, among the genes within the module, comparing the different groups [25]. The statistical significance of these effects was assessed using two-tailed z-tests, with null hypothesis H0: β = 0, and significance was considered at an adjusted p-value < 0.05 [31,83,84]. SEM fitting was performed using constrained Gaussian graphical modeling (CGGM), incorporating a DAG node-wise Lasso procedure and de-biasing asymptotic inference [85].
Notably, the effects of gene-i or group perturbations on gene-j were represented as regression coefficients, which were conditionally interpretable. This means that the coefficients reflect the expected normalized change in gene-j expression per unit increase in gene-i expression (or the difference between cases and controls), holding other variables constant within the transcriptomic active IBS-D module. Specifically, negative coefficients indicated inhibition (non-activation), while positive coefficients indicated activation in regulatory terms. Similarly, negative group effects signaled downregulation (lower expression levels between cases and controls), whereas positive values indicated upregulation [83].
We fitted three models as follows: (i) Model A: the IBS-D model, evaluating edge effects specifically in IBS-D data; (ii) Model B: the control model, evaluating edge effects in controls; (iii) Model C: the node and edge perturbation model, evaluating both node and edge perturbation effects.
For Model C, a two-step procedure was conducted as follows: (1) In the first step, the group was modeled as an exogenous variable influencing all other graph nodes, and (2) in the second step, the differences in beta coefficients (edges) between the groups were estimated. This was akin to fitting separate models for cases and controls and assessing the significance of edges (direct effects) based on these differences.
Within this framework, indirect effects were also explored to identify potential gene mediators.
To enhance SEM fitting, we applied strategies such as extracting the optimal DAG by balancing model adjustment and graph sparsity and using a de-confounding process to minimize badness-of-fit measures while preserving the strongest perturbation signals present in the original data [25]. Given the exploratory nature of this study, we did not resize the DAG to exclusively reflect the biological structure of the disease based on data alone. However, our goal remained to optimize SEM fitting by considering a balance between model complexity, fitting, and perturbation.
A goodness-of-fit index, such as the standardized root mean-square residual (SRMR), was calculated to assess the model fit of the transcriptomic active IBS-D module to the data [24]. While this study is exploratory, an SRMR of less than 0.10 is generally considered an adequate fit, and values below 0.05 suggest a good fit [86].
Finally, we examined network edge changes to detect alterations in causal relationships between gene regulatory networks under different conditions. This approach is crucial for studying biological systems, as it sheds light on how gene networks shift across conditions, providing a deeper understanding of complex diseases like IBDs [87].

4.4.3. Gene Communities Detection

Subsequently, a clustering procedure was carried out to identify topological gene communities using the walk-trap community detection algorithm (WTC) [25,88]. This algorithm generated the necessary number of clusters to encompass the entire input network. To assess the effectiveness of the clustering, a modularity index was calculated, which indicates the degree of separation between communities; a value close to 1 suggests well-defined community structures. Clustering was applied to both the Steiner tree graph and the optimal directed acyclic graph (DAG) obtained from the previously described fitting strategies, allowing comparison of the outcomes.
Statistical analysis was conducted using R software (version 4.3.3, R Core Team, 2023) with the SEMgraph [25], huge [80], and hgu95av2.db [89] packages (the R code is provided in the Supplemental Materials). The R/SEMgraph package offers tools for modeling complex biological systems as causal multivariate networks. The R/huge package was employed for the non-paranormal transformation of gene expression data, while the R/hgu95av2.db package was used for annotation purposes, specifically to convert Entrez identifiers into gene symbols for graphical representation of the networks.

4.4.4. Communities’ Characterization

Finally, to biologically characterize the communities identified through topological clustering of the Steiner Tree and active IBS-D module, we conducted a functional enrichment analysis using the gProfiler bioinformatics tool [90]. Notably, the g: SCS algorithm [90] was employed to account for multiple testing corrections.

5. Conclusions

In conclusion, this study comprehensively explores the pathophysiology of IBS-D through advanced transcriptomic analysis and network-based bioinformatics approaches. Our findings confirm that IBS-D is primarily a functional disorder characterized by persistent gastrointestinal symptoms without evident structural alterations. We identified dysregulation in the complement system, which plays a crucial role in immune defense and maintaining intestinal homeostasis. This evidence underscores the importance of gut microbiota and epithelial integrity in perpetuating disease symptoms.
This study highlights critical genes, such as DUOX2 and SH2D6, which are involved in mucosal defense and immune signaling, as significant contributors to IBS-D pathophysiology. The network analysis revealed complex gene interactions and specific biological pathways, suggesting potential therapeutic targets for modulating intestinal microbiota, immune responses, and epithelial integrity.
Overall, these results advance our understanding of the molecular mechanisms underlying IBS-D and suggest promising directions for devising targeted therapeutic strategies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25179322/s1.

Author Contributions

Conceptualization, D.G., F.M. and M.A.; Data curation, M.A.; Formal analysis, D.G. and M.A.; Funding acquisition, F.R.; Investigation, D.G., M.A., D.M., B.U., N.G. and M.C.; Methodology, D.G., M.A., B.U., N.G. and M.C.; Resources, F.R.; Software, D.G. and M.A.; Supervision, F.R.; Validation, D.G. and M.A.; Visualization, M.A.; Writing—original draft, D.G., F.M., M.A., D.M., B.U., N.G. and F.R.; Writing—review & editing, F.M., M.A., D.M., B.U. and F.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Italian Ministry of Health RC 2022–2024, Prog. N. 19 (DDG_661_23.12.2022).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Acknowledgments

The authors thank Rossella Donghia, researcher at the National Institute of Gastroenterology IRCCS “Saverio de Bellis”, for the support provided in the visualization contribution. The authors also thank Mary Victoria Pragnell, B.A, for revising the English.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Drossman, D.A.; Hasler, W.L. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. Gastroenterology 2016, 150, 1257–1261. [Google Scholar] [CrossRef] [PubMed]
  2. Vaiopoulou, A.; Karamanolis, G.; Psaltopoulou, T.; Karatzias, G.; Gazouli, M. Molecular basis of the irritable bowel syndrome. World J. Gastroenterol. 2014, 20, 376–383. [Google Scholar] [CrossRef] [PubMed]
  3. Mearin, F.; Lacy, B.E.; Chang, L.; Chey, W.D.; Lembo, A.J.; Simren, M.; Spiller, R. Bowel Disorders. Gastroenterology 2016, 150, 1393–1407.e5. [Google Scholar] [CrossRef]
  4. Longstreth, G.F.; Thompson, W.G.; Chey, W.D.; Houghton, L.A.; Mearin, F.; Spiller, R.C. Functional bowel disorders. Gastroenterology 2006, 130, 1480–1491. [Google Scholar] [CrossRef]
  5. Occhipinti, K.; Smith, J.W. Irritable bowel syndrome: A review and update. Clin. Colon Rectal Surg. 2012, 25, 46–52. [Google Scholar] [CrossRef]
  6. Devanarayana, N.M.; Rajindrajith, S.; Pathmeswaran, A.; Abegunasekara, C.; Gunawardena, N.K.; Benninga, M.A. Epidemiology of irritable bowel syndrome in children and adolescents in Asia. J. Pediatr. Gastroenterol. Nutr. 2015, 60, 792–798. [Google Scholar] [CrossRef]
  7. Hyams, J.S.; Di Lorenzo, C.; Saps, M.; Shulman, R.J.; Staiano, A.; van Tilburg, M. Functional Disorders: Children and Adolescents. Gastroenterology 2016, 150, 1456–1468.e2. [Google Scholar] [CrossRef]
  8. Oka, P.; Parr, H.; Barberio, B.; Black, C.J.; Savarino, E.V.; Ford, A.C. Global prevalence of irritable bowel syndrome according to Rome III or IV criteria: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020, 5, 908–917. [Google Scholar] [CrossRef]
  9. Tang, H.-Y.; Jiang, A.-J.; Wang, X.-Y.; Wang, H.; Guan, Y.-Y.; Li, F.; Shen, G.-M. Uncovering the pathophysiology of irritable bowel syndrome by exploring the gut-brain axis: A narrative review. Ann. Transl. Med. 2021, 9, 1187. [Google Scholar] [CrossRef]
  10. Cho, M.H.; McDonald, M.-L.N.; Zhou, X.; Mattheisen, M.; Castaldi, P.J.; Hersh, C.P.; DeMeo, D.L.; Sylvia, J.S.; Ziniti, J.; Laird, N.M.; et al. Risk loci for chronic obstructive pulmonary disease: A genome-wide association study and meta-analysis. Lancet Respir. Med. 2014, 2, 214–225. [Google Scholar] [CrossRef] [PubMed]
  11. Hasin, Y.; Seldin, M.; Lusis, A. Multi-omics approaches to disease. Genome Biol. 2017, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  12. Mahmud, S.; Biswas, S.; Afrose, S.; Mita, M.A.; Hasan, R.; Shimu, M.S.S.; Paul, G.K.; Chung, S.; Saleh, A.; Alshehri, S.; et al. Use of Next-Generation Sequencing for Identifying Mitochondrial Disorders. Curr. Issues Mol. Biol. 2022, 44, 1127–1148. [Google Scholar] [CrossRef]
  13. James, S.C.; Fraser, K.; Young, W.; McNabb, W.C.; Roy, N.C. Gut Microbial Metabolites and Biochemical Pathways Involved in Irritable Bowel Syndrome: Effects of Diet and Nutrition on the Microbiome. J. Nutr. 2020, 150, 1012–1021. [Google Scholar] [CrossRef]
  14. Dothel, G.; Barbaro, M.R.; Di Vito, A.; Ravegnini, G.; Gorini, F.; Monesmith, S.; Coschina, E.; Benuzzi, E.; Fuschi, D.; Palombo, M.; et al. New insights into irritable bowel syndrome pathophysiological mechanisms: Contribution of epigenetics. J. Gastroenterol. 2023, 58, 605–621. [Google Scholar] [CrossRef] [PubMed]
  15. Edgar, R.; Domrachev, M.; Lash, A.E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002, 30, 207–210. [Google Scholar] [CrossRef] [PubMed]
  16. Thind, A.S.; Thind, A.S.; Monga, I.; Monga, I.; Thakur, P.K.; Thakur, P.K.; Kumari, P.; Kumari, P.; Dindhoria, K.; Dindhoria, K.; et al. Demystifying emerging bulk RNA-Seq applications: The application and utility of bioinformatic methodology. Brief. Bioinform. 2021, 22, bbab259. [Google Scholar] [CrossRef] [PubMed]
  17. Subramanian, A.; Tamayo, P.; Mootha, V.K.; Mukherjee, S.; Ebert, B.L.; Gillette, M.A.; Paulovich, A.; Pomeroy, S.L.; Golub, T.R.; Lander, E.S.; et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 2005, 102, 15545–15550. [Google Scholar] [CrossRef]
  18. Doniger, S.W.; Salomonis, N.; Dahlquist, K.D.; Vranizan, K.; Lawlor, S.C.; Conklin, B.R. MAPPFinder: Using Gene Ontology and GenMAPP to create a global gene-expression profile from microarray data. Genome Biol. 2003, 4, R7. [Google Scholar] [CrossRef]
  19. Maere, S.; Heymans, K.; Kuiper, M. BiNGO: A Cytoscape plugin to assess overrepresentation of Gene Ontology categories in Biological Networks. Bioinformatics 2005, 21, 3448–3449. [Google Scholar] [CrossRef]
  20. Kroes, R.A.; Panksepp, J.; Burgdorf, J.; Otto, N.J.; Moskal, J.R. Modeling depression: Social dominance-submission gene expression patterns in rat neocortex. Neuroscience 2006, 137, 37–49. [Google Scholar] [CrossRef]
  21. Leonard, M.M.; Bai, Y.; Serena, G.; Nickerson, K.P.; Camhi, S.; Sturgeon, C.; Yan, S.; Fiorentino, M.R.; Katz, A.; Nath, B.; et al. RNA sequencing of intestinal mucosa reveals novel pathways functionally linked to celiac disease pathogenesis. PLoS ONE 2019, 14, e0215132. [Google Scholar] [CrossRef] [PubMed]
  22. Akond, Z.; Alam, M.; Mollah, M.N.H. Biomarker Identification from RNA-Seq Data using a Robust Statistical Approach. Bioinformation 2018, 14, 153–163. [Google Scholar] [CrossRef] [PubMed]
  23. Reactome Analysis Tool. Available online: https://reactome.org/PathwayBrowser/#TOOL=AT (accessed on 5 May 2024).
  24. Kanehisa, M.; Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000, 28, 27–30. [Google Scholar] [CrossRef] [PubMed]
  25. Grassi, M.; Palluzzi, F.; Tarantino, B. SEMgraph: An R package for causal network inference of high-throughput data with structural equation models. Bioinformatics 2022, 38, 4829–4830. [Google Scholar] [CrossRef]
  26. Chen, W.; Liu, J.; He, S. Prior knowledge guided active modules identification: An integrated multi-objective approach. BMC Syst. Biol. 2017, 11, 8. [Google Scholar] [CrossRef]
  27. Cho, D.-Y.; Kim, Y.-A.; Przytycka, T.M. Chapter 5: Network biology approach to complex diseases. PLoS Comput. Biol. 2012, 8, e1002820. [Google Scholar] [CrossRef]
  28. Kim, Y.-A.; Przytycka, T.M. Bridging the Gap between Genotype and Phenotype via Network Approaches. Front. Genet. 2013, 3, 31660. [Google Scholar] [CrossRef]
  29. Ideker, T.; Ozier, O.; Schwikowski, B.; Siegel, A.F. Discovering regulatory and signalling circuits in molecular interaction networks. Bioinformatics 2002, 18 (Suppl. S1), S233–S240. [Google Scholar] [CrossRef]
  30. Klammer, M.; Godl, K.; Tebbe, A.; Schaab, C. Identifying differentially regulated subnetworks from phosphoproteomic data. BMC Bioinform. 2010, 11, 351. [Google Scholar] [CrossRef]
  31. Boomsma, A. Book review of K. A. Bollen (1989). In Structural Equations with Latent Variables; John Wiley & Sons: Hoboken, NJ, USA, 2014; Volume 12, pp. 124–131. [Google Scholar]
  32. Dotolo, S.; Abate, R.E.; Roma, C.; Guido, D.; Preziosi, A.; Tropea, B.; Palluzzi, F.; Giacò, L.; Normanno, N. Bioinformatics: From NGS Data to Biological Complexity in Variant Detection and Oncological Clinical Practice. Biomedicines 2022, 10, 2074. [Google Scholar] [CrossRef] [PubMed]
  33. Kohestani, H.; Giuliani, A. Organization principles of biological networks: An explorative study. Biosystems 2016, 141, 31–39. [Google Scholar] [CrossRef] [PubMed]
  34. Chartrand, G.; Zhang, P. Chromatic Graph Theory; Introduction to Graphs; Chapman and Hall/CRC: New York, NY, USA, 2019; pp. 27–52. [Google Scholar]
  35. Barbara, G.; Cremon, C.; Bellini, M.; Corsetti, M.; Di Nardo, G.; Falangone, F.; Fuccio, L.; Galeazzi, F.; Iovino, P.; Sarnelli, G.; et al. Italian guidelines for the management of irritable bowel syndrome: Joint Consensus from the Italian Societies of: Gastroenterology and Endoscopy (SIGE), Neurogastroenterology and Motility (SINGEM), Hospital Gastroenterologists and Endoscopists (AIGO), Digestive Endoscopy (SIED), General Medicine (SIMG), Gastroenterology, Hepatology and Pediatric Nutrition (SIGENP) and Pediatrics (SIP). Dig. Liver Dis. 2023, 55, 187–207. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, D. Methods of integrating data to uncover genotype—Phenotype interactions. Nat. Rev. Genet. 2015, 16, 85–97. [Google Scholar] [CrossRef]
  37. Liu, C.; Ma, Y.; Zhao, J.; Nussinov, R.; Zhang, Y.-C.; Cheng, F.; Zhang, Z.-K. Computational network biology: Data, models, and applications. Phys. Rep. 2020, 846, 1–66. [Google Scholar] [CrossRef]
  38. Grassi, M.; Tarantino, B. SEMtree: Tree-based structure learning methods with structural equation models. Bioinformatics 2023, 39, btad377. [Google Scholar] [CrossRef]
  39. Jain, U.; Otley, A.R.; Van Limbergen, J.; Stadnyk, A.W. The complement system in inflammatory bowel disease. Inflamm. Bowel Dis. 2014, 20, 1628–1637. [Google Scholar] [CrossRef]
  40. Andoh, A.; Fujiyama, Y.; Sumiyoshi, K.; Bamba, T. Local secretion of complement C3 in the exocrine pancreas: Ductal epithelial cells as a possible biosynthetic site. Gastroenterology 1996, 110, 1919–1925. [Google Scholar] [CrossRef] [PubMed]
  41. Dunkelberger, J.R.; Song, W.-C. Complement and its role in innate and adaptive immune responses. Cell Res. 2010, 20, 34–50. [Google Scholar] [CrossRef]
  42. McCullough, R.; McCullough, J. Deciphering the pathophysiology of irritable bowel syndrome and functional gastrointestinal disorders-an alternative model for pathogenesis: Cytokine controlled transepithelial multi-feedback loop. Transl. Gastroenterol. Hepatol. 2017, 2, 18. [Google Scholar] [CrossRef]
  43. Wouters, M.M.; Vicario, M.; Santos, J. The role of mast cells in functional GI disorders. Gut 2016, 65, 155–168. [Google Scholar] [CrossRef]
  44. Martin-Viñas, J.J.; Quigley, E.M.M. Immune response in irritable bowel syndrome: A systematic review of systemic and mucosal inflammatory mediators. J. Dig. Dis. 2016, 17, 572–581. [Google Scholar] [CrossRef] [PubMed]
  45. Shulman, R.J.; Devaraj, S.; Heitkemper, M. Activation of the Innate Immune System in Children With Irritable Bowel Syndrome Evidenced by Increased Fecal Human β-Defensin-2. Clin. Gastroenterol. Hepatol. 2021, 19, 2121–2127. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, Y.; Jobin, C. Microbial imbalance and intestinal pathologies: Connections and contributions. Dis. Model. Mech. 2014, 7, 1131–1142. [Google Scholar] [CrossRef] [PubMed]
  47. Linsalata, M.; Riezzo, G.; Orlando, A.; D’attoma, B.; Prospero, L.; Ignazzi, A.; Losurdo, G.; Di Leo, A.; Giannelli, G.; Russo, F. The Role of Intestinal Barrier Function in Overweight Patients with IBS with Diarrhea Undergoing a Long-Term Low Fermentable Oligo-, Di-, and Monosaccharide and Polyol Diet. Nutrients 2023, 15, 4683. [Google Scholar] [CrossRef]
  48. Sina, C.; Kemper, C.; Derer, S. The intestinal complement system in inflammatory bowel disease: Shaping intestinal barrier function. Semin. Immunol. 2018, 37, 66–73. [Google Scholar] [CrossRef]
  49. Veres-Székely, A.; Szász, C.; Pap, D.; Szebeni, B.; Bokrossy, P.; Vannay, Á. Zonulin as a Potential Therapeutic Target in Microbiota-Gut-Brain Axis Disorders: Encouraging Results and Emerging Questions. Int. J. Mol. Sci. 2023, 24, 7548. [Google Scholar] [CrossRef]
  50. Kim, M.-Y.; Choi, S.-W. Dietary modulation of gut microbiota for the relief of irritable bowel syndrome. Nutr. Res. Pract. 2021, 15, 411–430. [Google Scholar] [CrossRef]
  51. Grasberger, H.; Gao, J.; Nagao-Kitamoto, H.; Kitamoto, S.; Zhang, M.; Kamada, N.; Eaton, K.A.; El-Zaatari, M.; Shreiner, A.B.; Merchant, J.L.; et al. Increased Expression of DUOX2 Is an Epithelial Response to Mucosal Dysbiosis Required for Immune Homeostasis in Mouse Intestine. Gastroenterology 2015, 149, 1849–1859. [Google Scholar] [CrossRef]
  52. Xiong, Z.; Zhu, X.; Geng, J.; Xu, Y.; Wu, R.; Li, C.; Fan, D.; Qin, X.; Du, Y.; Tian, Y.; et al. Intestinal Tuft-2 cells exert antimicrobial immunity via sensing bacterial metabolite N-undecanoylglycine. Immunity 2022, 55, 686–700.e7. [Google Scholar] [CrossRef]
  53. Kivelä, A.-J.; Kivelä, J.; Saarnio, J.; Parkkila, S. Carbonic anhydrases in normal gastrointestinal tract and gastrointestinal tumors. World J. Gastroenterol. 2005, 11, 155–163. [Google Scholar] [CrossRef]
  54. Lucarini, E.; Nocentini, A.; Bonardi, A.; Chiaramonte, N.; Parisio, C.; Micheli, L.; Toti, A.; Ferrara, V.; Carrino, D.; Pacini, A.; et al. Carbonic Anhydrase IV Selective Inhibitors Counteract the Development of Colitis-Associated Visceral Pain in Rats. Cells 2021, 10, 2540. [Google Scholar] [CrossRef] [PubMed]
  55. Cheng, Y.Y.; Yu, J.; Wong, Y.P.; Man, E.P.S.; To, K.F.; Jin, V.X.; Li, J.; Tao, Q.; Sung, J.J.Y.; Chan, F.K.L.; et al. Frequent epigenetic inactivation of secreted frizzled-related protein 2 (SFRP2) by promoter methylation in human gastric cancer. Br. J. Cancer 2007, 97, 895–901. [Google Scholar] [CrossRef]
  56. Suzuki, H.; Watkins, D.N.; Jair, K.-W.; E Schuebel, K.; Markowitz, S.D.; Chen, W.D.; Pretlow, T.P.; Yang, B.; Akiyama, Y.; van Engeland, M.; et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat. Genet. 2004, 36, 417–422. [Google Scholar] [CrossRef]
  57. Kim, T.-O.; Han, Y.K.; Yi, J.M. Hypermethylated promoters of tumor suppressor genes were identified in Crohn’s disease patients. Intest. Res. 2020, 18, 297–305. [Google Scholar] [CrossRef] [PubMed]
  58. Al-Shamasi, A.-A.; Elkaffash, R.; Mohamed, M.; Rayan, M.; Al-Khater, D.; Gadeau, A.-P.; Ahmed, R.; Hasan, A.; Eldassouki, H.; Yalcin, H.C.; et al. Crosstalk between Sodium-Glucose Cotransporter Inhibitors and Sodium-Hydrogen Exchanger 1 and 3 in Cardiometabolic Diseases. Int. J. Mol. Sci. 2021, 22, 12677. [Google Scholar] [CrossRef] [PubMed]
  59. Nikolovska, K.; Seidler, U.E.; Stock, C. The Role of Plasma Membrane Sodium/Hydrogen Exchangers in Gastrointestinal Functions: Proliferation and Differentiation, Fluid/Electrolyte Transport and Barrier Integrity. Front. Physiol. 2022, 13, 899286. [Google Scholar] [CrossRef]
  60. Liu, Y.; Merrill, R.A.; Strack, S. A-Kinase Anchoring Protein 1: Emerging Roles in Regulating Mitochondrial Form and Function in Health and Disease. Cells 2020, 9, 298. [Google Scholar] [CrossRef]
  61. Khan, I.; Khan, K. Uncoupling of Carbonic Anhydrase from Na-H exchanger-1 in Experimental Colitis: A Possible Mechanistic Link with Na-H Exchanger. Biomolecules 2019, 9, 700. [Google Scholar] [CrossRef]
  62. Krishnan, D.; Liu, L.; Wiebe, S.A.; Casey, J.R.; Cordat, E.; Alexander, R.T. Carbonic anhydrase II binds to and increases the activity of the epithelial sodium-proton exchanger, NHE3. Am. J. Physiol. Renal Physiol. 2015, 309, F383–F392. [Google Scholar] [CrossRef]
  63. Cai, J.; Culley, M.K.; Zhao, Y.; Zhao, J. The role of ubiquitination and deubiquitination in the regulation of cell junctions. Protein Cell 2018, 9, 754–769. [Google Scholar] [CrossRef]
  64. Prospero, L.; Riezzo, G.; Linsalata, M.; Orlando, A.; D’Attoma, B.; Russo, F. Psychological and Gastrointestinal Symptoms of Patients with Irritable Bowel Syndrome Undergoing a Low-FODMAP Diet: The Role of the Intestinal Barrier. Nutrients 2021, 13, 2469. [Google Scholar] [CrossRef]
  65. Castelo-Branco, G.; Wagner, J.; Rodriguez, F.J.; Kele, J.; Sousa, K.; Rawal, N.; Pasolli, H.A.; Fuchs, E.; Kitajewski, J.; Arenas, E. Differential regulation of midbrain dopaminergic neuron development by Wnt-1, Wnt-3a, and Wnt-5a. Proc. Natl. Acad. Sci. USA 2003, 100, 12747–12752. [Google Scholar] [CrossRef]
  66. Mayer, E.A.; Ryu, H.J.; Bhatt, R.R. The neurobiology of irritable bowel syndrome. Mol. Psychiatry 2023, 28, 1451–1465. [Google Scholar] [CrossRef] [PubMed]
  67. C Babraham Bioinformatics. Available online: https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 2 January 2024).
  68. Hart, S.N.; Therneau, T.M.; Zhang, Y.; Poland, G.A.; Kocher, J.-P. Calculating sample size estimates for RNA sequencing data. J. Comput. Biol. 2013, 20, 970–978. [Google Scholar] [CrossRef]
  69. Kukurba, K.R.; Montgomery, S.B. RNA Sequencing and Analysis. Cold Spring Harb. Protoc. 2015, 2015, 951–969. [Google Scholar] [CrossRef] [PubMed]
  70. Sourceforge. Available online: https://sourceforge.net/projects/bbmap/support (accessed on 2 January 2024).
  71. Babraham Bioinformatics. Available online: https://www.bioinformatics.babraham.ac.uk/projects/trim_galore/ (accessed on 2 January 2024).
  72. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef] [PubMed]
  73. GENCODE. Available online: https://www.gencodegenes.org/human/release_35.html (accessed on 3 January 2024).
  74. Liao, Y.; Smyth, G.K.; Shi, W. featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014, 30, 923–930. [Google Scholar] [CrossRef]
  75. Jassal, B.; Matthews, L.; Viteri, G.; Gong, C.; Lorente, P.; Fabregat, A.; Sidiropoulos, K.; Cook, J.; Gillespie, M.; Haw, R.; et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2020, 48, D498–D503. [Google Scholar] [CrossRef]
  76. Fabregat, A.; Korninger, F.; Viteri, G.; Sidiropoulos, K.; Marin-Garcia, P.; Ping, P.; Wu, G.; Stein, L.; D’eustachio, P.; Hermjakob, H. Reactome graph database: Efficient access to complex pathway data. PLoS Comput. Biol. 2018, 14, e1005968. [Google Scholar] [CrossRef]
  77. Nguyen, T.; Mitrea, C.; Draghici, S. Network-Based Approaches for Pathway Level Analysis. Curr. Protoc. Bioinform. 2018, 61, 8.25.1–8.25.24. [Google Scholar] [CrossRef]
  78. Package ‘ggm’. Available online: https://CRAN.R-project.org/package=ggm (accessed on 3 January 2024).
  79. Rinaldo, A. Computing Maximum Likelihood Estimates in Log-Linear Models; Carnegie Mellon University: Pittsburgh, PA, USA, 2006. [Google Scholar]
  80. Zhao, T.; Roeder, K. The huge Package for High-dimensional Undirected Graph Estimation in R. J. Mach. Learn. Res. 2012, 13, 1059–1062. [Google Scholar] [PubMed]
  81. Fisher, R.A. Frequency Distribution of the Values of the Correlation Coefficient in Samples from an Indefinitely Large Population. Biometrika 1915, 10, 507–521. [Google Scholar] [CrossRef]
  82. Kou, L.; Markowsky, G.; Berman, L. A fast algorithm for Steiner trees. Acta Inform. 1981, 15, 141–145. [Google Scholar] [CrossRef]
  83. Pepe, D.; Grassi, M. Investigating perturbed pathway modules from gene expression data via structural equation models. BMC Bioinform. 2014, 15, 132. [Google Scholar] [CrossRef] [PubMed]
  84. Rondanelli, M.; Perna, S.; Alalwan, T.A.; Cazzola, R.; Gasparri, C.; Infantino, V.; Perdoni, F.; Iannello, G.; Pepe, D.; Guido, D. A structural equation model to assess the pathways of body adiposity and inflammation status on dysmetabolic biomarkers via red cell distribution width and mean corpuscular volume: A cross-sectional study in overweight and obese subjects. Lipids Health Dis. 2020, 19, 154. [Google Scholar] [CrossRef] [PubMed]
  85. Jankov, J. Confidence intervals for high-dimensional inverse covariance estimation. Electron. J. Stat. 2015, 9, 1205–1229. [Google Scholar] [CrossRef]
  86. Hu, L.; Bentler, P.M. Cutoff criteria for fit indexes in covariance structure analysis: Conventional criteria versus new alternatives. Struct. Equ. Model. A Multidiscip. J. 1999, 6, 1–55. [Google Scholar] [CrossRef]
  87. Ideker, T.; Krogan, N.J. Differential network biology. Mol. Syst. Biol. 2012, 8, 565. [Google Scholar] [CrossRef]
  88. Pons, P.; Latapy, M. Computing Communities in Large Networks Using Random Walks. In Computer and Information Sciences-ISCIS 2005: Proceedings of the 20th International Symposium, Istanbul, Turkey, 26–28 October 2005; Springer: Berlin/Heidelberg, Germany, 2005; pp. 284–293. [Google Scholar]
  89. Bioconductor. Available online: https://bioconductor.org/packages/release/data/annotation/html/hgu95av2.db.html (accessed on 4 May 2024).
  90. Kolberg, L.; Raudvere, U.; Kuzmin, I.; Adler, P.; Vilo, J.; Peterson, H. g:Profiler-interoperable web service for functional enrichment analysis and gene identifier mapping (2023 update). Nucleic Acids Res. 2023, 51, W207–W212. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Volcano plot: under-expressed (blue) and over-expressed (red) genes where p value < 0.05. Genes marked with gene name or ENSEMBL ID are the most statistically significant.
Figure 1. Volcano plot: under-expressed (blue) and over-expressed (red) genes where p value < 0.05. Genes marked with gene name or ENSEMBL ID are the most statistically significant.
Ijms 25 09322 g001
Figure 2. Reactome pathways identified by the 54 differentially expressed genes (DEGs) are depicted as dots. The size of each dot represents the number of genes included in the pathway, while the color corresponds to the number of DEGs contained within the gene set of the pathway.
Figure 2. Reactome pathways identified by the 54 differentially expressed genes (DEGs) are depicted as dots. The size of each dot represents the number of genes included in the pathway, while the color corresponds to the number of DEGs contained within the gene set of the pathway.
Ijms 25 09322 g002
Figure 3. Steiner tree graph of IBS-D from Reactome and GEO data. Seed nodes are colored aquamarine, connectors white, and bidirectional edges are colored black. The graph is drawn from DEGs (seed genes) mapped on the Reactome interactome, i.e., the union of all Reactome pathways, in relation to a weighting procedure based on Fisher’s r-to-z transformation.
Figure 3. Steiner tree graph of IBS-D from Reactome and GEO data. Seed nodes are colored aquamarine, connectors white, and bidirectional edges are colored black. The graph is drawn from DEGs (seed genes) mapped on the Reactome interactome, i.e., the union of all Reactome pathways, in relation to a weighting procedure based on Fisher’s r-to-z transformation.
Ijms 25 09322 g003
Figure 4. The active IBS-D module (i.e., the optimal IBS-D DAG was derived using the SEMgraph fitting strategy. Added edges are highlighted in green, while gray edges represent those retained from the original IBS-D graph.
Figure 4. The active IBS-D module (i.e., the optimal IBS-D DAG was derived using the SEMgraph fitting strategy. Added edges are highlighted in green, while gray edges represent those retained from the original IBS-D graph.
Ijms 25 09322 g004
Figure 5. Final active IBS-D modules after SEM fitting. The final active IBS-D modules, derived from structural equation modeling (SEM) fitting, are presented as follows: Nodes and Edges: pink nodes: activated; light blue nodes: inhibited; Edges: red edges: significant direct effects (p-value < 0.05) with a positive effect (b effect > 0). Blue edges: significant direct effects (p-value < 0.05) with a negative effect (b effect < 0). Gray edges: non-significant effects. (A) IBS-D Module. This diagram represents the IBS-D module, including only IBS-D samples. The module was obtained using SEM fitting with constrained Gaussian graphical modeling (CGGM), utilizing a directed acyclic graph (DAG) node-wise Lasso procedure and de-biasing asymptotic inference. In this module, the gene effects are conditional, representing the expected normalized gene expression variation per unit increase of the gene, while keeping other genes fixed. (B) Control module. This diagram illustrates the control module, consisting solely of control samples. It was derived using the same SEM fitting method as for the IBS-D module (CGGM with DAG node-wise Lasso and de-biasing asymptotic inference). (C) Final active IBS-D module (node and edge perturbation). This module shows the final active IBS-D network with node and edge perturbations. It was obtained using SEM fitting with CGGM, incorporating node-wise Lasso and de-biasing asymptotic inference.
Figure 5. Final active IBS-D modules after SEM fitting. The final active IBS-D modules, derived from structural equation modeling (SEM) fitting, are presented as follows: Nodes and Edges: pink nodes: activated; light blue nodes: inhibited; Edges: red edges: significant direct effects (p-value < 0.05) with a positive effect (b effect > 0). Blue edges: significant direct effects (p-value < 0.05) with a negative effect (b effect < 0). Gray edges: non-significant effects. (A) IBS-D Module. This diagram represents the IBS-D module, including only IBS-D samples. The module was obtained using SEM fitting with constrained Gaussian graphical modeling (CGGM), utilizing a directed acyclic graph (DAG) node-wise Lasso procedure and de-biasing asymptotic inference. In this module, the gene effects are conditional, representing the expected normalized gene expression variation per unit increase of the gene, while keeping other genes fixed. (B) Control module. This diagram illustrates the control module, consisting solely of control samples. It was derived using the same SEM fitting method as for the IBS-D module (CGGM with DAG node-wise Lasso and de-biasing asymptotic inference). (C) Final active IBS-D module (node and edge perturbation). This module shows the final active IBS-D network with node and edge perturbations. It was obtained using SEM fitting with CGGM, incorporating node-wise Lasso and de-biasing asymptotic inference.
Ijms 25 09322 g005
Figure 6. The gene community structure obtained through clustering is illustrated as follows: genes are represented by circles, and community clusters are depicted by colored sets. The topological clustering procedure was performed by applying the walk-trap community detection algorithm (WTC), generating as many clusters as needed to cover the whole input network. (A) Clustering performed on the Steiner Tree graph. (B) Clustering performed on the optimal DAG obtained from SEMgraph fitting strategies.
Figure 6. The gene community structure obtained through clustering is illustrated as follows: genes are represented by circles, and community clusters are depicted by colored sets. The topological clustering procedure was performed by applying the walk-trap community detection algorithm (WTC), generating as many clusters as needed to cover the whole input network. (A) Clustering performed on the Steiner Tree graph. (B) Clustering performed on the optimal DAG obtained from SEMgraph fitting strategies.
Ijms 25 09322 g006
Figure 7. Gene ontology results of ST communities obtained with gprofiler tools. The genes in communities 4 and 5 were not enriched in any GO term pathway. Abbreviations used: BP—biological processes, Term name—GO term name, Term_id—GO term identification, adj p-value—adjusted p-value.
Figure 7. Gene ontology results of ST communities obtained with gprofiler tools. The genes in communities 4 and 5 were not enriched in any GO term pathway. Abbreviations used: BP—biological processes, Term name—GO term name, Term_id—GO term identification, adj p-value—adjusted p-value.
Ijms 25 09322 g007
Figure 8. Gene ontology results of IBS-D module communities obtained with gprofiler tools. The genes in the first community were not enriched in any GO term pathway. Abbreviations used are BP—biological processes, Term_name—GO term name, Term_id—GO term identification, and adj p-value—adjusted p-value.
Figure 8. Gene ontology results of IBS-D module communities obtained with gprofiler tools. The genes in the first community were not enriched in any GO term pathway. Abbreviations used are BP—biological processes, Term_name—GO term name, Term_id—GO term identification, and adj p-value—adjusted p-value.
Ijms 25 09322 g008
Figure 9. The study workflow of the proposed approach. Data collection steps are highlighted in purple, RNA-seq bioinformatic analysis in light blue and the causal network analysis step is highlighted in pink.
Figure 9. The study workflow of the proposed approach. Data collection steps are highlighted in purple, RNA-seq bioinformatic analysis in light blue and the causal network analysis step is highlighted in pink.
Ijms 25 09322 g009
Table 1. Gender, age, and BMI characteristics in IBS-D and healthy patients.
Table 1. Gender, age, and BMI characteristics in IBS-D and healthy patients.
Samples MaleFemaleAge (years)BMI (kg/m2)
Healthy1851323–5927
IBS-D1761126–5927
BMI: Body Mass Index.
Table 2. Negative binomial analysis by DESeq2 identified DEGs, comparing IBS-D versus healthy samples.
Table 2. Negative binomial analysis by DESeq2 identified DEGs, comparing IBS-D versus healthy samples.
Gene_IDENSEMBL_IDGene Descriptionlog2FCp-Value
DUOX2ENSG00000140279dual oxidase 22.520
KLK12ENSG00000186474kallikrein related peptidase 121.380
SH2D6ENSG00000152292SH2 domain containing 61.260
MST1P2ENSG00000186301macrophage stimulating 1 pseudogene −1.840
NoneENSG00000290851novel transcript−2.090
RPL13P12ENSG00000215030ribosomal protein L13 pseudogene 12−2.80
HTR3CENSG000001780845-hydroxytryptamine receptor 3C 1.390.0001
SH2D7ENSG00000183476SH2 domain containing 71.120.0001
HMX3ENSG00000188620H6 family homeobox 31.110.0001
RPSAP15ENSG00000237506ribosomal protein SA pseudogene 151.040.0001
ELFN2ENSG00000166897extracellular leucine-rich repeat and fibronectin type III domain containing 21.010.0003
AGAP7PENSG00000264204ArfGAP with GTPase domain, ankyrin repeat and PH domain 7, pseudogene−1.910.0005
RPL29P4ENSG00000230202ribosomal protein L29 pseudogene 4 1.790.0006
NoneENSG00000277400None1.350.0006
DUOXA2ENSG00000140274dual oxidase maturation factor 220.0007
ACTG1P22ENSG00000271615actin gamma 1 pseudogene 221.160.0008
GTF2H2ENSG00000145736general transcription factor IIH subunit 2 1.030.0008
ATP12AENSG00000075673ATPase H+/K+ transporting non-gastric alpha2 subunit−2.010.0011
HTR3EENSG000001860385-hydroxytryptamine receptor 3E1.160.0012
CHATENSG00000070748choline O-acetyltransferase1.050.0014
MT1HENSG00000205358metallothionein 1H −1.040.0014
HCG4BENSG00000227262HLA complex group 4B−1.120.0016
PKD1P2ENSG00000227827polycystin 1, transient receptor potential channel interacting pseudogene 2−2.080.0017
GATD3ENSG00000160221glutamine amidotransferase class 1 domain containing 31.630.0021
IGHV2-70ENSG00000274576immunoglobulin heavy variable 2–70−1.230.0025
SLC13A3ENSG00000158296solute carrier family 13 member 3 −1.020.0028
IGHV5-78ENSG00000211978immunoglobulin heavy variable 5–78 (pseudogene) 1.40.003
ADGRF1ENSG00000153292adhesion G protein-coupled receptor F11.250.0032
IGKV2-29ENSG00000253998immunoglobulin kappa variable 2–29−1.930.0045
FAM230IENSG00000178248family with sequence similarity of 230 member, I1.320.0048
NoneENSG00000287188novel transcript, antisense to ANXA101.220.0057
ITPRID1ENSG00000180347ITPR interacting domain containing 11.020.0065
PI16ENSG00000164530peptidase inhibitor 16−1.170.0067
MGAM2ENSG00000257743maltase-glucoamylase 2 (putative) 1.050.0078
NoneENSG00000230563novel transcript1.090.0111
CCNOENSG00000152669cyclin O1.040.0117
CHIT1ENSG00000133063chitinase 11.470.0136
AFF2ENSG00000155966ALF transcription elongation factor 2 1.280.0143
HERC2P3ENSG00000290376HERC2 pseudogene 31.120.0144
NoneENSG00000289911novel transcript, antisense to PTP4A1−1.060.0159
SFRP2ENSG00000145423secreted frizzled-related protein 2−1.010.0178
LGSNENSG00000146166lengsin, lens protein with glutamine synthetase domain −1.670.0187
SULT2A1ENSG00000105398sulfotransferase family 2A member 1−2.180.0188
NoneENSG00000274767novel transcript, antisense CCL3L3−1.150.0212
PLA2G10BPENSG00000254609phospholipase A2 group XB, pseudogene1.490.0214
NoneENSG00000289810novel transcript1.30.0244
NoneENSG00000224114ribosomal protein S14 (RPS14) pseudogene−1.310.0249
OLFM4ENSG00000102837olfactomedin 4 1.260.0255
CR2ENSG00000117322complement C3d receptor 2 1.290.0267
LTFENSG00000012223Lactotransferrin1.080.0315
NoneENSG00000213058ribosomal protein S14 (RPS14) pseudogene−1.640.0328
HSD3B2ENSG00000203859hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2 1.230.039
PWP2ENSG00000241945PWP2 small subunit processome component1.090.0406
SVOPLENSG00000157703SVOP like−10.0481
The genes are arranged according to their p-value in ascending order. Abbreviations: Gene_ID: gene identification; ENSEMBL_ID: ENSEMBLE Identification; Log2FC: Log2 fold-change.
Table 3. Results of the SEMgsa.
Table 3. Results of the SEMgsa.
PathwayNo. NodesNo. DEGsPerturbation StatuspNapNipval
Creation of C4 and C2 activators145Down0.9990.0000.001
Formation of the Early Elongation Complex332Up0.0010.9950.003
Classical antibody-mediated complement activation65Down1.0000.0020.004
Complement cascade578Down0.6800.0060.011
Regulation of complement cascade467Down0.6940.0070.014
Initial triggering of complement226Down0.6110.0080.016
Negative regulation of TCF-dependent signaling by WNT ligand antagonists154Down0.7420.0080.016
Digestion of dietary carbohydrate72Down0.3700.1440.288
FCGR activation122Down0.4150.0490.098
Role of phospholipids in phagocytosis240Down0.3060.2310.462
Thyroxine biosynthesis71Up0.2300.6000.460
Scavenging of heme from plasma120Down0.5870.1310.261
Role of LAT2/NTAL/LAB on calcium mobilization120Up0.2980.3720.597
FCERI-mediated Ca+2 mobilization301Up0.1770.4950.353
CD22-mediated BCR regulation50Up0.3680.8440.735
FCGR3A-mediated IL10 synthesis282Up0.3210.3230.642
Antigen activates B-Cell Receptor (BCR), leading to the generation of second messengers290Up0.4140.5900.827
The pathway mineralocorticoid biosynthesis analyzed in SEMgsa has not been included in the table because the RICF algorithm did not converge. Abbreviations: No: number; DEGs: differentially expressed genes; up: activated; down: inhibited; pNa: Brown’s combined p-value of pathway node activation; pNi: Brown’s combined p-value of pathway node inhibition; pval: Bonferroni combined p-value of activation and inhibition, expressed as 2× min (activation p-value, inhibition p-value); ADJP: adjusted Bonferroni p-value of pathway perturbation; i.e., min (number of pathways × p-value; 1).
Table 4. List of differentially expressed genes (DEGs) per pathway.
Table 4. List of differentially expressed genes (DEGs) per pathway.
PathwayDEG 1DEG 2DEG 3DEG 4DEG 5DEG 6DEG 7DEG 8
Creation of C4 and C2 activatorsC1QCC1QBMASP1C1SC1R
Formation of the Early Elongation ComplexGTF2H2SUPT4H1
Classical antibody-mediated complement activationC1QAC1QCC1QBC1SC1R
Complement cascadeC1QCC1QBC4BPAMASP1C6C1SC1RC3AR1
Regulation of complement cascadeC1QCC1QBC4BPAC6C1SC1RC3AR1
Initial triggering of complementC1QCC1QBMASP1C4BC1SC1R
Negative regulation of TCF-dependent signaling by WNT ligand antagonistsLRP5SFRP1LRP5KREMEN1
Digestion of dietary carbohydrateSIMGAM
FCGR activationFGCR2ACD247
Role of phospholipids in phagocytosis
Thyroxine biosynthesisDUOX2
Scavenging of heme from plasma
Role of LAT2/NTAL/LAB on calcium mobilization
FCERI-mediated Ca+2 mobilizationLCP2
CD22-mediated BCR regulation
FCGR3A-mediated IL10 synthesisFGCR2ACD247
Antigen activates B-Cell Receptor (BCR), leading to the generation of second messengers
Table 5. Results of the SEM fitting: gene effects in the IBS-D and control group.
Table 5. Results of the SEM fitting: gene effects in the IBS-D and control group.
GenePathGeneIBS-D ModelControl Model
βp-Value95%CI inf95%CI SupA/NAβp-Value95%CI inf95%CI SupA/NA
SLC9B1CA40.4810.0160.0890.874A−0.3430.084−0.7320.046NA
LRP6CA4−0.2510.21−0.6440.141NA−0.3830.054−0.7720.006NA
MGAMSLC5A10.0230.926−0.4530.498A0.3860.076−0.0410.812A
UBCSCNN1G0.81900.5461.092A−0.0680.772−0.5290.393NA
UBCSFRP1−0.3310.149−0.7790.118NA0.0780.74−0.3830.539A
CA4DUOX20.4570.0340.0340.88A0.80800.5361.08A
SLC9B1AKAP1−0.6020−0.875−0.33NA0.2270.167−0.0950.548A
SLC5A1AKAP10.0930.468−0.1590.345A0.61900.280.959A
SCNN1GAKAP1−0.4090.004−0.687−0.131NA0.3040.085−0.0420.649A
SFRP1FZD10.5330.0090.1310.935A0.3560.106−0.0760.787A
A: Activated; NA: not activated. Significant results (p < 0.05) are shown in bold, and suggestive results (0.05 < p < 0.10) in italics. β: Effect. The standardized root mean-squared residual (SRMR) for the IBS-D model was 0.212, compared to 0.224 for the control model.
Table 6. Results of the SEM fitting: gene differences (i.e., node perturbation by “node and edge perturbation” model) between the IBS-D and control group.
Table 6. Results of the SEM fitting: gene differences (i.e., node perturbation by “node and edge perturbation” model) between the IBS-D and control group.
SourcePathGenebp-Value95%CI inf95%CI supUp/Down
GroupSLC9B1−0.1770.288−0.5030.149down-expressed
GroupLRP60.1010.547−0.2280.431upregulated
GroupMGAM0.2660.102−0.0530.586upregulated
GroupUBC0.2710.096−0.0480.589upregulated
GroupCA40.4740.0010.1920.755upregulated
GroupSLC5A10.2210.185−0.1060.547upregulated
GroupSCNN1G−0.2470.11−0.5490.056down-expressed
GroupSFRP1−0.4050.009−0.71−0.1down-expressed
GroupDUOX20.0980.471−0.1690.366upregulated
GroupAKAP1−0.0390.788−0.3240.246down-expressed
GroupFZD1−0.0810.62−0.4020.24down-expressed
In bold, significant (p < 0.05) effects, and in italics suggestive results (0.05 < p < 0.10). b: effect. 95%CI: 95% confidence interval. The SRMR of the model was equal to 0.218.
Table 7. Results of the SEM fitting: gene effect differences between the IBS-D and control group (i.e., edge perturbation, from “node and edge perturbation” model).
Table 7. Results of the SEM fitting: gene effect differences between the IBS-D and control group (i.e., edge perturbation, from “node and edge perturbation” model).
GenePathGenedbp Value95%CI inf95%CI SupUp/Down
SLC9B1CA40.8250.0030.2721.377upregulated
LRP6CA40.1320.640−0.4210.684upregulated
MGAMSLC5A1−0.3630.265−1.0010.275downregulated
UBCSCNN1G0.8870.0010.3521.423upregulated
UBCSFRP1−0.4090.213−1.0520.234downregulated
CA4DUOX2−0.3510.171−0.8540.151downregulated
SLC9B1AKAP1−0.8290.000−1.250−0.407downregulated
SLC5A1AKAP1−0.5260.015−0.949−0.103downregulated
SCNN1GAKAP1−0.7130.002−1.156−0.269downregulated
SFRP1FZD10.1780.555−0.4120.768upregulated
In bold, the significant (p < 0.05) effects. Abbreviations: db: effect difference between IBS-D and control samples. 95%CI: 95% confidence interval. The SRMR of the model was equal to 0.218.
Table 8. Gene ontology results of ST communities. Abbreviations: BP: biological processes, Term_name: GO term name, Term_id: GO term identification, adj p-value: adjusted p-value.
Table 8. Gene ontology results of ST communities. Abbreviations: BP: biological processes, Term_name: GO term name, Term_id: GO term identification, adj p-value: adjusted p-value.
CommunitySourceTerm NameTerm idadj_p-Value
1GO:BPcomplement activationGO:00069561.38 × 10−2
2GO:BPimmune response-regulating cell surface receptor signaling pathwayGO:00027682.48 × 10−4
GO:BPregulation of immune responseGO:00507761.15 × 10−3
GO:BPregulation of immune system processGO:00026821.15 × 10−3
GO:BPimmune response-activating signaling pathwayGO:00027571.42 × 10−3
GO:BPimmune response-regulating signaling pathwayGO:00027641.87 × 10−3
GO:BPactivation of immune responseGO:00022532.72 × 10−3
GO:BPpositive regulation of immune system processGO:00026843.28 × 10−3
GO:BPimmune system processGO:00023763.82 × 10−3
GO:BPtransmembrane receptor protein tyrosine kinase signaling pathwayGO:00071695.63 × 10−3
3GO:BPdisaccharide catabolic processGO:00463524.11 × 10−5
GO:BPdisaccharide metabolic processGO:00059844.52 × 10−4
GO:BPoligosaccharide catabolic processGO:00093137.19 × 10−4
GO:BPoligosaccharide metabolic processGO:00093111.51 × 10−2
4----
5----
6GO:BPmidbrain dopaminergic neuron differentiationGO:19049487.82 × 10−7
GO:BPdopaminergic neuron differentiationGO:00715421.83 × 10−5
GO:BPcanonical Wnt signaling pathwayGO:00600702.89 × 10−5
GO:BPWnt signaling pathway involved in somitogenesisGO:00902441.09 × 10−4
GO:BPWnt signaling pathwayGO:00160551.40 × 10−4
GO:BPcell-cell signaling by wntGO:01987381.43 × 10−4
GO:BPmidbrain developmentGO:00309012.15 × 10−4
GO:BPnegative regulation of canonical Wnt signaling pathwayGO:00900907.02 × 10−4
GO:BPplanar cell polarity pathway involved in neural tube closureGO:00901799.94 × 10−4
GO:BPregulation of establishment of planar polarity involved in neural tube closureGO:00901781.19 × 10−3
7GO:BPvisual system developmentGO:01500633.51 × 10−4
GO:BPsensory system developmentGO:00488803.78 × 10−4
GO:BPtube morphogenesisGO:00352394.28 × 10−4
GO:BPNorrin signaling pathwayGO:01101351.11 × 10−3
GO:BPtube developmentGO:00352951.60 × 10−3
GO:BPextracellular matrix-cell signalingGO:00354261.85 × 10−3
GO:BPWnt signaling pathway, calcium modulating pathwayGO:00072231.85 × 10−3
GO:BPretinal blood vessel morphogenesisGO:00613042.77 × 10−3
GO:BPblood vessel morphogenesisGO:00485142.96 × 10−3
GO:BPcellular response to retinoic acidGO:00713003.32 × 10−3
8GO:BPcomplement activationGO:00069561.47 × 10−19
GO:BPhumoral immune responseGO:00069595.60 × 10−14
GO:BPcomplement activation, classical pathwayGO:00069589.30 × 10−12
GO:BPhumoral immune response mediated by circulating immunoglobulinGO:00024555.90 × 10−11
GO:BPimmune effector processGO:00022525.40 × 10−10
GO:BPpositive regulation of immune responseGO:00507786.93 × 10−10
GO:BPregulation of immune responseGO:00507764.37 × 10−9
GO:BPinnate immune responseGO:00450879.93 × 10−9
GO:BPpositive regulation of immune system processGO:00026842.19 × 10−8
GO:BPsynapse pruningGO:009988833.30 × 10−5
Table 9. Gene ontology results of IBS-D module. Abbreviations: ‘community number’, BP: biological processes, Term_name: GO term name, Term_id: GO term identification, adj_p-value: adjusted -value.
Table 9. Gene ontology results of IBS-D module. Abbreviations: ‘community number’, BP: biological processes, Term_name: GO term name, Term_id: GO term identification, adj_p-value: adjusted -value.
CommunitiesSourceTerm_NameTerm_idadj_p_Value
1----
2GO:BPsodium ion transmembrane transportGO:00357253.20 × 10−2
GO:BPmaltose catabolic processesGO:00901784.95 × 10−2
GO:BPstarch metabolic processesGO:00901774.95 × 10−2
GO:BPstarch catabolic processesGO:00901754.95 × 10−2
GO:BPdextrin catabolic processesGO:19049484.95 × 10−2
3GO:BPplanar cell polarity pathway involved in neural tube closureGO:00901799.94 × 10−4
GO:BPregulation of establishment of planar polarity involved in neural tube closureGO:00901781.19 × 10−3
GO:BPestablishment of planar polarity involved in neural tube closureGO:00901771.41 × 10−3
GO:BPregulation of establishment of planar polarityGO:00901751.90 × 10−3
GO:BPmidbrain dopaminergic neuron differentiationGO:19049481.90 × 10−3
GO:BPestablishment of planar polarity of embryonic epitheliumGO:00422491.90 × 10−3
GO:BPestablishment of tissue polarityGO:00071641.27 × 10−2
GO:BPestablishment of planar polarityGO:00017361.27 × 10−2
GO:BPdopaminergic neuron differentiationGO:00715421.48 × 10−2
GO:BPWnt signaling pathway, planar cell polarity pathwayGO:00600712.21 × 10−2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guido, D.; Maqoud, F.; Aloisio, M.; Mallardi, D.; Ura, B.; Gualandi, N.; Cocca, M.; Russo, F. Transcriptomic Module Discovery of Diarrhea-Predominant Irritable Bowel Syndrome: A Causal Network Inference Approach. Int. J. Mol. Sci. 2024, 25, 9322. https://doi.org/10.3390/ijms25179322

AMA Style

Guido D, Maqoud F, Aloisio M, Mallardi D, Ura B, Gualandi N, Cocca M, Russo F. Transcriptomic Module Discovery of Diarrhea-Predominant Irritable Bowel Syndrome: A Causal Network Inference Approach. International Journal of Molecular Sciences. 2024; 25(17):9322. https://doi.org/10.3390/ijms25179322

Chicago/Turabian Style

Guido, Davide, Fatima Maqoud, Michelangelo Aloisio, Domenica Mallardi, Blendi Ura, Nicolò Gualandi, Massimiliano Cocca, and Francesco Russo. 2024. "Transcriptomic Module Discovery of Diarrhea-Predominant Irritable Bowel Syndrome: A Causal Network Inference Approach" International Journal of Molecular Sciences 25, no. 17: 9322. https://doi.org/10.3390/ijms25179322

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop