2,146
views
1
recommends
+1 Recommend
2 collections
    0
    shares

      2023 Journal Citation Reports Journal Impact Factor is 0.9. Scopus Citescore 0.8. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Uncovering the Genetic Link between Acute Myocardial Infarction and Ulcerative Colitis Co-Morbidity through a Systems Biology Approach

      Published
      research-article
      Bookmark

            Abstract

            Background: Cardiovascular diseases, particularly acute myocardial infarction, are the leading cause of disability and death. Atherosclerosis, the pathological basis of AMI, can be accelerated by chronic inflammation. Ulcerative colitis (UC), a chronic inflammatory disease associated with immunity, contributes to the risk of AMI development. However, controversy continues to surround the relationship between these two diseases. The present study unravels the pathogenesis of AMI and UC, to provide a new perspective on the clinical management of patients with these comorbidities.

            Methods: Microarray datasets GSE66360 and GSE87473 were downloaded from the Gene Expression Omnibus database. Common differentially expressed genes (co-DEGs) between AMI and UC were identified, and the following analyses were performed: enrichment analysis, protein-protein interaction network construction, hub gene identification and co-expression analysis.

            Results: A total of 267 co-DEGs (233 upregulated and 34 downregulated) were screened for further analysis. GO enrichment analysis suggested important roles of chemokines and cytokines in AMI and UC. In addition, the lipopolysaccharide-mediated signaling pathway was found to be closely associated with both diseases. KEGG enrichment analysis revealed that lipid and atherosclerosis, NF-κB, TNF and IL-17 signaling pathways are the core mechanisms involved in the progression of both diseases. Finally, 11 hub genes were identified with cytoHubba: TNF, IL1B, TLR2, CXCL8, STAT3, MMP9, ITGAX, CCL4, CSF1R, ICAM1 and CXCL1.

            Conclusion: This study reveals a co-pathogenesis mechanism of AMI and UC regulated by specific hub genes, thus providing ideas for further mechanistic studies, and new perspectives on the clinical management of patients with these comorbidities.

            Main article text

            Introduction

            Cardiovascular diseases, particularly acute myocardial infarction (AMI), are the leading cause of disability and death [1]. AMI is a myocardial necrosis caused by acute and persistent coronary artery ischemia and hypoxia [2]. Atherosclerosis, the pathological basis of AMI, can be accelerated by chronic inflammation [3]. Although early treatment with reperfusion after AMI onset can improve clinical outcomes, the risk of recurrence and mortality remains high for as long as 1 year after AMI onset [4]. In addition, reperfusion therapy not only increases damage to ischemic myocardial tissue but also implicates normal myocardial tissue [5]. Reperfusion-induced injury accounts for as much as 50% of overall myocardial injury, and is often associated with serious adverse events [5]. Therefore, preventing AMI is a major priority.

            Ulcerative colitis (UC) is a type of inflammatory bowel disease that causes chronic inflammation of the intestinal tract [6]. Its pathological mechanism is complex and influenced by multiple factors, such as genetic background, intestinal immune status and intestinal microbial balance. The risk of developing colon cancer is 2–3 fold greater in patients with UC than the general population. [7, 8]. The age of onset of UC is bimodal, with peaks at 2–3 years and 50–80 years [9]. UC affects not only the gastrointestinal tract but also the heart, and pericarditis and myocarditis are the most common manifestations [10]. Furthermore, patients with UC are at significantly elevated risk of both venous thromboembolism and mesenteric ischemia [10].

            Although UC has been reported to be a risk factor for AMI progression [11], the relationship between these two diseases remains controversial. Osterman et al. [12] have concluded that patients with UC do not have elevated AMI incidence. However, Choi et al. [13] have reported elevated risk of myocardial infarction in patients with UC and a trend towards a younger incidence of AMI. UC causes systemic inflammatory responses and promotes hypercoagulation in the body [13]. Furthermore, disruption of the intestinal mucosal barrier leads to the transfer of lipopolysaccharides (LPS) and other endotoxins into the bloodstream, thereby inducing the secretion of pro-inflammatory cytokines and leading to endothelial disturbances, atherosclerosis and acute cardiovascular events [14]. Gut microecological dysregulation increases plaque vulnerability by affecting lipid metabolism and the inflammatory response [15]. Azimi et al. [16] have demonstrated that Clostridium difficile, Escherichia coli and Campylobacter are closely associated with the development of UC. Chronic bacterial infections may contribute to the formation of vulnerable plaques through enhanced T-cell activation and inflammatory responses [17]. Therefore, the cardio-intestinal axis may be an important pathway through which UC promotes the development of AMI. Owing to inconsistent research findings regarding the relationship between AMI and UC, no standard of medical care has been established for patients with both conditions. This study therefore explored the pathogenesis of AMI and UC, to provide new perspectives on the clinical management of patients with AMI and UC.

            Matrials and Methods

            Data Source

            Relevant datasets in the Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) database [18] were searched with the keywords “acute myocardial infarction” and “ulcerative colitis.” The inclusion criteria were: 1) Homo sapiens (top organism); 2) expression profiling with an array (study type); 3) total number of samples in a single microarray dataset >50; and 4) single microarray dataset from the same sequencing platform. We downloaded four microarray datasets from GEO: GSE66360-GPL570 [19] and GSE62646-GPL6244 [20] for AMI, and GSE87473-GPL13158 [21] and GSE59071-GPL6244 [22] for UC. The GSE66360 and GSE87473 datasets were used for mechanistic analysis of differentially expressed genes (DEGs), and GSE62646 and GSE59071 were used for further screening of hub genes. GSE66360 contained 49 AMI and 50 normal (NL) samples; GSE62646 included 84 AMI and 14 NL samples; GSE87473 contained 106 UC-active and 21 NL samples; and GSE59071 included 74 UC-active, 23 UC-inactive and 11 NL samples. Datasets were downloaded from the GEO database via the R GEOquery package and pre-processed with Rstudio [23]. Genes were annotated with gene symbols. Because the data used in this study were obtained from public databases, no local ethics committee approval or informed consent was required.

            Identification of DEGs

            We analyzed the GSE66360 and GSE87473 datasets separately with the R limma package [24]. Filtering criteria (|logFC|>0.585 and adjusted P<0.05) were used to determine DEGs between AMI or UC and the corresponding NL groups. Volcano plots of DEGs were constructed with the R ggplot2 package [25, 23]. Among the DEGs, logFC>0.585 or logFC<−0.585 indicated upregulation or downregulation in the disease group, respectively [26, 27]. We used the R VennDiagram package to plot Venn diagrams of up- and downregulated genes between the datasets GSE66360 and GSE87473 to obtain co-DEGs common to both diseases [28].

            Functional Enrichment Analyses of co-DEGs

            Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to evaluate functional annotation of the key module genes. GO terms include biological process, cellular component and molecular function categories [29]. KEGG is a knowledgebase for systematic analysis of gene function, linking genomic information to higher-order functional information [30]. We performed enrichment analysis with the R clusterProfiler package [31].

            PPI Network Construction and Module Analysis

            The STRING (https://cn.string-db.org/) database [32] is designed to integrate all known and predicted associations among proteins. The common DEGs (co-DEGs) were input into the STRING database, “Homo sapiens” was selected, the minimum required interaction score was set to ≥0.4, and the free nodes were hidden. The downloaded results were input into Cytoscape software in tab-separated values format for further analysis [33]. The key modules were obtained with the MCODE plugin in Cytoscape with default settings. Subsequently, enrichment analysis was performed on the module genes.

            Screening and Analysis of Hub Genes

            The cytoHubba plugin in Cytoscape was used to filter hub genes [34]. Here, we used four algorithms (degree, closeness, EPC and MCC) and selected the genes of the first 20 intersections of these four algorithms for further analysis. The co-expression network of these hub genes was constructed via the GeneMANIA (http://genemania.org/) [35] database, with the species set to human.

            Validation of Hub Gene Expression

            We further validated hub gene expression in the GSE62646 and GSE59071 datasets. P-value <0.05 was considered to indicate a significant difference.

            Prediction and Verification of Transcription Factors

            The TRRUST (https://www.grnpedia.org/trrust) database [36] was used to predict transcriptional regulatory networks. The species was set to human, and the co-hub genes stably expressed in the datasets GSE66360, GSE87473, GSE62646 and GSE59071 were imported into the TRRUST database to obtain the associated transcription factors (TFs). TF regulatory networks were constructed in Cytoscape. Subsequently, the expression of these TFs was further validated with the datasets GSE66360 and GSE87473.

            Results

            Identification of DEGs

            Figure 1 shows the diagram of the study design and workflow. After normalization of the microarray data, 670 upregulated and 405 downregulated DEGs in GSE66360, and 1323 upregulated and 1138 downregulated DEGs in GSE87473, were identified (Figure 2A, B). The upregulated and downregulated DEGs were intersected separately to identify co-DEGs for AMI and UC, of which 233 co-DEGs were upregulated, and 34 were downregulated (Figure 2C, D).

            Figure 1

            Schematic Diagram of the Study Design and Workflow.

            Figure 2

            Volcano Plots and Venn Diagrams of DEGs.

            (A) Volcano plot of GSE66360. (B) Volcano plot of GSE87473. Red indicates upregulated genes, and green indicates downregulated genes. Venn diagram of (C) upregulated and (D) downregulated genes in the GSE66360 and GSE87473 datasets.

            Functional Enrichment Analyses of Co-DEGs

            Functional enrichment analyses were performed for 267 co-DEGs (Figure 3A, B). GO enrichment analysis revealed the following biological processes: response to lipopolysaccharide, response to molecule of bacterial origin, positive regulation of cytokine production and leukocyte migration (Figure 3A). KEGG enrichment analysis suggested the importance of osteoclast differentiation, lipid and atherosclerosis, NF-kappa B, TNF and IL-17 signaling pathways (Figure 3B, Table 1). Together, these results suggested that inflammation and the immune response are common core mechanisms of UC and AMI.

            Figure 3

            Functional Enrichment and PPI Network Analyses of Co-DEGs.

            (A) GO. (B) KEGG. (C) STRING. (D) Cytoscape. Red indicates upregulated genes, and green indicates downregulated genes.

            Table 1

            Top 30 Enriched KEGG Pathways.

            IDDescriptionP-valueGeneID
            hsa04380Osteoclast differentiation1.15E-16 IL1B/SOCS3/LILRB2/FOS/FCGR2A/FOSB/JUNB/SIRPA/FCGR2C/LILRA2/LILRB1/IFNGR1/FCGR3B/SPI1/SIRPB1/LILRA3/LILRB3/TNF/CSF1R/NCF2/FCGR1A/IL1A/LILRA1
            hsa04668TNF signaling pathway2.21E-12 MAP3K8/IL1B/CCL20/SOCS3/CXCL2/ICAM1/FOS/JUNB/MMP9/CXCL1/TNFAIP3/CEBPB/CXCL3/TNF/LIF/PTGS2/CREB5/NOD2
            hsa04657IL-17 signaling pathway1.60E-11 IL1B/CCL20/S100A9/CXCL2/FOS/FOSB/MMP9/CXCL1/CXCL8/S100A8/TNFAIP3/CEBPB/CXCL3/TNF/PTGS2/LCN2
            hsa04060Cytokine-cytokine receptor interaction1.15E-09 IL1B/CCL20/IL1RN/CCL4/CXCL2/CCL3L3/IL6ST/CSF3R/CXCL16/CXCL1/CXCL8/OSM/IFNGR1/CXCR1/RELT/TNFRSF10C/CXCL3/TNF/CSF1R/LIF/CSF2RB/IL1A/CXCR2/PPBP
            hsa05140Leishmaniasis1.56E-09 IL1B/TLR2/FOS/FCGR2A/FCGR2C/IFNGR1/FCGR3B/TNF/NCF2/PTGS2/FCGR1A/IL1A/CYBB
            hsa04061Viral protein interaction with cytokine and cytokine receptor4.55E-09 CCL20/CCL4/CXCL2/CCL3L3/IL6ST/CXCL1/CXCL8/CXCR1/TNFRSF10C/CXCL3/TNF/CSF1R/CXCR2/PPBP
            hsa04064NF-kappa B signaling pathway7.67E-09 IL1B/CCL4/CXCL2/ICAM1/BCL2A1/CXCL1/CXCL8/LY96/PLAU/LYN/TNFAIP3/CXCL3/TNF/PTGS2
            hsa05323Rheumatoid arthritis1.53E-07 IL1B/CCL20/TLR2/CXCL2/CCL3L3/ICAM1/FOS/CXCL1/CXCL8/CXCL3/TNF/IL1A
            hsa05417Lipid and atherosclerosis5.60E-07 IL1B/TLR2/CXCL2/CCL3L3/ICAM1/FOS/MMP9/STAT3/CXCL1/CXCL8/LY96/LYN/CXCL3/HSPA6/TNF/NCF2/CYBB
            hsa04062Chemokine signaling pathway3.14E-06 CCL20/CCL4/CXCL2/CCL3L3/CXCL16/STAT3/CXCL1/CXCL8/LYN/CXCR1/FGR/CXCL3/HCK/CXCR2/PPBP
            hsa04610Complement and coagulation cascades4.54E-06 THBD/PLAUR/SERPINA1/CD55/C5AR1/ITGAX/VWF/PLAU/CLU/SERPING1
            hsa05152Tuberculosis7.16E-06 CLEC4E/IL1B/FCER1G/TLR2/FCGR2A/FCGR2C/ITGAX/IFNGR1/FCGR3B/CEBPB/TNF/FCGR1A/IL1A/NOD2
            hsa05134Legionellosis1.03E-05 IL1B/TLR2/CXCL2/CXCL1/CXCL8/CXCL3/HSPA6/TNF
            hsa05167Kaposi sarcoma-associated herpesvirus infection1.69E-05 ZFP36/CXCL2/ICAM1/IL6ST/FOS/MAPKAPK2/STAT3/CXCL1/CXCL8/IFNGR1/LYN/CXCL3/HCK/PTGS2
            hsa04620Toll-like receptor signaling pathway2.50E-05 MAP3K8/IL1B/TLR2/CCL4/CCL3L3/FOS/CXCL8/LY96/TNF/TLR8
            hsa05150 Staphylococcus aureus infection7.84E-05 C5AR1/ICAM1/FCGR2A/FCGR2C/FPR1/FPR2/PTAFR/FCGR3B/FCGR1A
            hsa04933AGE-RAGE signaling pathway in diabetic complications0.000107926 THBD/IL1B/ICAM1/STAT3/CXCL8/TNF/EGR1/IL1A/CYBB
            hsa04625C-type lectin receptor signaling pathway0.000146178 CLEC4E/IL1B/FCER1G/CLEC4D/MAPKAPK2/TNF/EGR2/EGR3/PTGS2
            hsa04662B cell receptor signaling pathway0.000149294 LILRB2/FOS/LILRA2/LILRB1/LYN/LILRA3/LILRB3/LILRA1
            hsa05321Inflammatory bowel disease0.000210338 IL1B/TLR2/STAT3/IFNGR1/TNF/IL1A/NOD2
            hsa04613Neutrophil extracellular trap formation0.000245589 TLR2/C5AR1/AQP9/FCGR2A/FPR1/FPR2/VWF/FCGR3B/TLR8/NCF2/FCGR1A/CYBB
            hsa05202Transcriptional misregulation in cancer0.000283882 NFKBIZ/BCL6/NR4A3/BCL2A1/MMP9/CXCL8/PLAU/SPI1/CEBPB/CSF1R/GZMB/FCGR1A
            hsa05144Malaria0.000332765 IL1B/TLR2/ICAM1/CXCL8/PECAM1/TNF
            hsa05120Epithelial cell signaling in Helicobacter pylori infection0.000334702 CXCL2/CXCL1/CXCL8/LYN/CXCR1/CXCL3/CXCR2
            hsa04640Hematopoietic cell lineage0.000546951 IL1B/CD55/CSF3R/MME/TNF/CSF1R/FCGR1A/IL1A
            hsa05133Pertussis0.000555306 IL1B/FOS/CXCL8/LY96/TNF/SERPING1/IL1A
            hsa05142Chagas disease0.00066795 IL1B/TLR2/CCL3L3/FOS/GNA15/CXCL8/IFNGR1/TNF
            hsa05146Amoebiasis0.00066795 IL1B/TLR2/CXCL2/GNA15/CXCL1/CXCL8/CXCL3/TNF
            hsa05418Fluid shear stress and atherosclerosis0.001243797 THBD/IL1B/ICAM1/FOS/MMP9/PECAM1/TNF/NCF2/IL1A
            hsa05171Coronavirus disease, COVID-190.001456841 IL1B/TLR2/C5AR1/IL6ST/FOS/FCGR2A/STAT3/VWF/CXCL8/TNF/TLR8/CYBB
            PPI Network Construction and Module Analysis

            PPI network analysis of co-DEGs was performed with the STRING database (Figure 3C). The results were imported into Cytoscape in tab-separated values format to construct a PPI network. The PPI network comprised 205 nodes and 1782 edges (Figure 3D). The more connections between nodes and the higher the relevance, the higher the node’s ranking in the PPI network. (Figure 3C, D). In addition, five tightly linked gene modules were screened via the MCODE plugin in Cytoscape (Figure 4A–E). Furthermore, modular genes were integrated, and GO and KEGG analyses were performed (Figure 4F, G). GO enrichment analysis indicated that modular genes were enriched primarily in biological processes, such as response to molecule of bacterial origin, response to lipopolysaccharide and leukocyte migration (Figure 4F). KEGG enrichment analysis revealed that modular genes were involved mainly in osteoclast differentiation, lipid and atherosclerosis, NF-κB, TNF and IL-17 signaling pathways (Figure 4G). Overall, AMI and UC had many common pathogenic mechanisms, which might be mediated by specific hub genes.

            Figure 4

            Significant Gene Module and Enrichment Analysis of the Modular Genes.

            (A-E) Five significant gene clustering modules. Functional enrichment analyses of the modular genes. (F) GO. (G) KEGG.

            Screening and Analysis of Hub Genes

            The top 20 genes were screened in the cytoHubba plugin, with the degree, closeness, EPC and MCC algorithms. (Table 2). The R ggplot2 package was used to plot a Venn diagram to identify hub genes (Figure 5A). Eleven hub genes were screened: TNF, IL1B, TLR2, CXCL8, STAT3, MMP9, ITGAX, CCL4, CSF1R, ICAM1 and CXCL1 (Table 3). The GeneMANIA database was used to structure hub gene co-expression networks [35]. Co-expression network analysis indicated complicated PPI networks with co-expression (72.67%), co-localization (14.25%), physical interactions (5.40%), pathways (2.98%), shared protein domains (2.47%), predicted interactions (1.86%) and genetic interactions (0.37%) (Figure 5B). GO analysis revealed the key associations of hub genes with biological processes such as leukocyte migration, cellular response to lipopolysaccharide, cellular response to molecule of bacterial origin, cytokine-mediated signaling pathway and cytokine receptor binding (Figure 6A). These results suggested the importance of LPS and cytokines in both AMI and UC. In addition, the KEGG results suggested that the hub genes were involved in lipid and atherosclerosis, rheumatoid arthritis (RA), AGE-RAGE signaling pathway in diabetic complications, NF-kappa B, IL-17, and Toll-like receptor signaling pathways (Figure 6B).

            Figure 5

            Venn Diagram and Co-Expression Network of Hub Genes.

            (A) Venn diagram, showing 11 overlapping hub genes identified by four algorithms. (B) Co-expression network of hub genes.

            Figure 6

            Functional Enrichment Analyses of Hub Genes.

            (A) GO. (B) KEGG. The left half-circle represents hub genes significantly enriched in different terms. Different colors on the right side represent different terms. *** P<0.001.

            Table 2

            Top 20 Genes Ranked in cytoHubba.

            RankDegreeClosenessEPCMCC
            1 TNF TNF IL1B TLR2
            2 IL1B IL1B TNF TNF
            3 TLR2 CXCL8 TLR2 IL1B
            4 CXCL8 TLR2 CXCL8 CXCL8
            5 STAT3 STAT3 CCL4 CCL4
            6 MMP9 MMP9 STAT3 STAT3
            7 SPI1 SPI1 ITGAX ICAM1
            8 FCGR3B CCL4 TLR8 CXCL1
            9 TLR8 FCGR3B FCGR3B CXCL2
            10 ITGAX TLR8 SPI1 IL1A
            11 CCL4 ICAM1 MMP9 IL1RN
            12 CSF1R CSF1R CSF1R PTGS2
            13 ICAM1 ITGAX TREM1 CXCL3
            14 PTGS2 PTGS2 CXCL1 MMP9
            15 S100A12 CYBB CYBB CCL20
            16 TREM1 CXCL1 S100A12 CXCR2
            17 CYBB FOS ICAM1 TNFAIP3
            18 FOS S100A12 S100A9 SOCS3
            19 CXCL1 TREM1 LILRB2 ITGAX
            20 FCGR2C FCGR1A FCGR2C CSF1R
            Table 3

            Information on the 11 Hub Genes.

            EntryGene symbolDescription
            P01375 TNF Tumor necrosis factor
            P01584 IL1B Interleukin-1 beta
            O60603 TLR2 Toll-like receptor 2
            P10145 CXCL8 C-X-C motif chemokine 8
            P40763 STAT3 Signal transducer and activator of transcription 3
            P14780 MMP9 Matrix metalloproteinase-9
            P20702 ITGAX Integrin alpha-X
            P13236 CCL4 C-C motif chemokine 4
            P07333 CSF1R Macrophage colony-stimulating factor 1 receptor
            P05362 ICAM1 Intercellular adhesion molecule 1
            P09341 CXCL1 C-X-C motif chemokine 1
            Validation of Hub Gene Expression

            We additionally selected two datasets including AMI and UC and validated the reliability of the 11 hub genes. In the dataset GSE62646, seven hub genes were significantly differentially expressed in the AMI versus NL group: TNF, STAT3, CCL1, ITGAX, CSF1R, ICAM1 and CCL4 (Figure 7). Interestingly, CCL4 expression was downregulated in AMI (GSE62646), contrary to the previous trend of upregulated expression in AMI (GSE66360), possibly because of insufficient sample size, the sequencing technology used, sample variation, etc. The remaining six hub genes with significant differences were all significantly upregulated in the AMI group, in agreement with the previous expression trend. In the GSE59071 dataset, all 11 hub genes were significantly upregulated in the UC-active group compared with the NL group (Figure 8). We defined the six hub genes that were stably upregulated in all datasets as co-hub genes; these genes were TNF, STAT3, ITGAX, CSF1R, ICAM1 and CCL1.

            Figure 7

            Expression of 11 Hub Genes in GSE62646.

            AMI, acute myocardial infarction; NL, normal. *P<0.05; ** P<0.05 and ≥0.001; *** P<0.001.

            Figure 8

            Expression of 11 Hub Genes in GSE59071.

            UC, ulcerative colitis; NL, normal. *P<0.05; ** P<0.05 and ≥0.001; *** P<0.001.

            Prediction and Verification of TFs

            Using the TRRUST database, we identified 11 TFs that might regulate the expression of the co-hub genes (Figure 9 and Table 4). The reliability of the 11 TFs was verified in the GSE66360 and GSE87473 datasets. In GSE66360, eight TFs significantly differed between the AMI and NL groups (Figure 10). Six TFs (SPI1, RELA, NFKB1, JUN, CEBPD and CEBPA) were upregulated, and two TFs (HDAC1 and STAT1) were downregulated in the AMI group compared with the NL group. In GSE87473, six TFs were significantly differentially expressed between the UC-active and NL groups (Figure 11). Five TFs (STAT1, SPI1, RELA, NFKB1 and CEBPD) were upregulated, and one TF (CEBPA) was downregulated in the UC-active group compared with the NL group. Notably, CEBPA and STAT1 showed opposite expression trends in the AMI and UC groups, perhaps because of factors such as disease progression, cell cycle, tissue and sample differences. SPI1, RELA, NFKB1 and CEBPD were stably upregulated in both the AMI and UC-active groups. These TFs may be involved in disease progression through the regulation of TNF, STAT3, ITGAX, ICAM1 and CCL1.

            Figure 9

            Transcriptional Regulatory Network.

            Yellow represents transcription factors, and red represents genes.

            Figure 10

            Expression of 11 TFs in GSE66360.

            AMI, acute myocardial infarction; NL, normal.

            Figure 11

            Expression of 11 TFs in GSE87473.

            UC, ulcerative colitis; NL, normal.

            Table 4

            Key Transcriptional Factors of Co-Hub Genes.

            Key TFDescriptionP-valueGenes
            RELAv-rel reticuloendotheliosis viral oncogene homolog A (avian)5.91E-09 ICAM1, STAT3, ITGAX, TNF, CXCL1
            CEBPACCAAT/enhancer binding protein (C/EBP), alpha3.70E-07 ICAM1, ITGAX, STAT3
            SPI1Spleen focus forming virus (SFFV) proviral integration oncogene spi16.71E-07 STAT3, TNF, ITGAX
            NFKB1Nuclear factor of kappa light polypeptide gene enhancer in B-cells 19.53E-07 ITGAX, ICAM1, TNF, CXCL1
            SP1Sp1 transcription factor5.57E-06 ICAM1, TNF, ITGAX, CXCL1
            CEBPDCCAAT/enhancer binding protein (C/EBP), delta1.14E-05 TNF, CXCL1
            SIRT1Sirtuin 19.44E-05 ICAM1, TNF
            BRCA1Breast cancer 1, early onset0.000133 CXCL1, STAT3
            HDAC1Histone deacetylase 10.000207 STAT3, ICAM1
            STAT1Signal transducer and activator of transcription 1, 91 kDa0.00029 ICAM1, STAT3
            JUNJun proto-oncogene0.00091 ITGAX, TNF

            Discussion

            UC, a type of inflammatory bowel disease, is an immune-associated chronic inflammatory disease affecting the intestines [6]. Notably, active UC not only aggravates the systemic inflammatory response but also contributes to the hypercoagulable state of the body [13]. AMI is a myocardial necrosis caused by acute and persistent coronary artery ischemia and hypoxia [2]. Atherosclerotic plaque rupture is the most common cause of AMI [37]. In addition to accelerating the progression of atherosclerosis, UC can lead to the formation of vulnerable plaques by affecting intestinal microecology [17]. Patients with UC have significantly higher thrombotic events and cardiovascular mortality [38]. Kristensen et al. [39] have similarly concluded that UC contributes to an increased risk of AMI, stroke and cardiovascular death, particularly when UC is active, thus further increasing the incidence of these adverse events. In addition, Ha et al. [40] have suggested that female patients with UC are more likely to develop AMI than their male counterparts. However, Sinh et al. [41] have reported that UC does not increase mortality in patients with AMI. Currently, no consensus exists, and the relationship between AMI and UC remains controversial. The present study explored the pathogenesis of AMI and UC, to provide new perspectives for the clinical management of patients with AMI and UC, and novel ideas for further research on the molecular mechanisms of AMI combined with UC.

            We identified 267 co-DEGs shared by the two diseases through bioinformatic methods. Finally, six co-hub genes were found to be stably upregulated in both AMI and UC: TNF, STAT3, CCL1, ITGAX, CSF1R and ICAM1. The results of enrichment analysis indicated that UC may promote AMI through inflammatory and immune responses. GO enrichment analysis highlighted the importance of leukocyte migration, cytokine-mediated signaling pathway, cellular response to lipopolysaccharide and positive regulation of NF-kappa B TF activity. Leukocyte migration is an early event in vascular inflammation progression and is closely associated with atherosclerosis [42]. In addition, chemokines and cytokines have been associated with chronic inflammation [43]. Atherosclerosis is a pathological disease characterized by fibroproliferation, chronic inflammation, lipid accumulation and immune disorders of the vessel wall [44]. As atherosclerotic plaques progress to advanced stages, vulnerable plaques rupture, thereby leading to the development of AMI. UC is characterized by a massive accumulation of immune cells, myeloid cells and lymphocytes in the diseased intestines [45]. The continued activation of these cells, together with the production of inflammatory mediators, promotes UC recurrence, thus making complete disease cure difficult [45]. Therefore, blocking the migration of leukocytes to the intestines is the main strategy used to control UC and relieve symptoms [45]. LPS is a potent inducer of inflammation. Intestinal LPS binds LPS-binding protein and celiac particles, and enters the body’s circulation through the lymphatics, thus enhancing the inflammatory process [46]. In addition, disruption of the intestinal mucosal barrier in UC leads to the transfer of LPS and other endotoxins into the bloodstream, thereby inducing a pro-inflammatory cytokine response that leads to endothelial dysfunction, atherosclerosis and acute cardiovascular events [14]. KEGG enrichment analysis highlighted the importance of lipid and atherosclerosis, RA, NF-kappa B, IL-17 and Toll-like receptor signaling pathways. Atherosclerosis is an inflammatory disease characterized by lipid accumulation in the arterial wall [47]. Lipids and atherosclerosis are the pathological basis for the development of AMI. The pathogenesis of RA is associated with chronic inflammatory and immune system disorders [48]. Numerous studies have reported a significantly elevated risk of AMI and UC in patients with RA [49, 50]. NF-κB is involved in immunity, inflammation, cell proliferation and apoptosis [51]. In particular, overactivation of the NF-κB signaling pathway is closely associated with various inflammatory diseases [51].

            IL-17 is expressed by various leukocyte subsets, such as gamma-delta (γδ) T cells, natural killer (NK) cells, NK T cells and neutrophils [52]. IL-17 exacerbates the inflammatory response of plaque tissues, and promotes thrombosis and vulnerable plaque formation [52]. TLRs are clonal transmembrane signaling receptors that link intrinsic and specific immunity [53]. They are found primarily in macrophages, dendritic cells, NK cells and lymphocytes [54]. TLRs are activated by binding damage-associated molecular patterns, microbial-associated molecular patterns and pathogen-associated molecular patterns, thereby regulating inflammation and the immune response [55].

            Eleven TFs that may regulate co-hub gene expression were identified with the TRRUST database. However, colony-stimulating factor 1 receptor (CSF1R) was found to be outside the transcriptional regulatory network. This finding might have been due to a lack of data in the TRRUST database. CSF1R is a type I single-transmembrane protein that is abundantly enriched in myeloid cells. CSF1R binds its endogenous ligands (CSF1 and IL-34) and activates downstream signaling pathways, including PI3K/AKT, JAK/STATs and MAPK, thereby regulating the proliferation, differentiation, migration and activation of target immune cells [56]. Xiang et al.[56] have suggested that CSF-1R is a potential target for regulating inflammatory diseases. Furthermore, through further validation, four TFs (SPI1, RELA, NFKB1 and CEBPD) were found to be stably upregulated in AMI compared with UC. These TFs synergistically regulate co-hub genes (TNF, STAT3, ITGAX, ICAM1 and CCL1).

            TNF acts as a pleiotropic cytokine, which directly induces the expression of inflammation-associated genes, induces cell death, and indirectly drives inflammation and immune responses [57]. Luo et al. [58] have proposed TNF-α as a novel biomarker for predicting plaque rupture in patients with ST-elevation myocardial infarction. Thus, inhibition of TNF-α may decrease the inflammatory load of the body and stabilize plaques, particularly in people with multiple inflammatory diseases. TNF-α antagonists are important drugs in the treatment of UC [59]. However, long-term inhibition of TNF-α increases the risk of opportunistic infections and skin cancer [60]. To overcome such limitations, inhibition of TNF-α downstream inflammation-associated pathways may serve as an alternative. TNF activates caspase protease, JNK and NF-κB signaling pathways, thus regulating apoptosis, inflammation and immune processes [61]. Diminished DNA methylation in diseased tissue during AMI induces SPI1 overexpression and overactivates the TNF-α/NF-κB signaling pathway, thus exacerbating myocardial tissue inflammation [62]. The NF-κB family has five members: NFKB1, NFKB2, RELA, c-REL and RELB [63]. These proteins dimerize and form functional NF-κB. Among them, the NFKB1 gene promoter-94 insertion/deletion ATTG polymorphism is associated with the risk and severity of acute coronary syndromes [64]. Chen et al. [65] have found that inhibition of the inflammatory pathway associated with v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) is an effective target for the treatment of UC. In addition, inhibition of the RELA/TNF-α signaling pathway in AMI protects cardiac function through anti-inflammatory effects [66].

            CCAAT enhancer binding protein delta (CEBPD), a member of the CEBP family, is a TF that regulates many biological processes, particularly inflammation and immune responses [67, 68]. CEBPD is activated by inflammatory factors such as IL-6, IFN-α, IFN-γ and IL1B, and it participates in inflammation and immune regulation [67]. CEBPD is considered an inflammatory enhancer in aortic endothelial cells, and it exacerbates tissue damage in concert with TNF-α [69]. CEBPD aggravates tissue damage by inducing inflammatory gene expression in liver tissue, lung tissue and brain glial cells [70]. Moreover, CEBPD promotes macrophage polarization toward M1 and exacerbates tissue inflammation [71]. CEBPD and TNF have been found to be stably upregulated in patients with AMI and UC. Thus, the CEBPD/TNF axis may be a potential mechanism for the treatment of AMI and UC. However, this possibility must be further verified in in vivo and in vitro models.

            STAT3 is an important regulator of cell proliferation, differentiation, apoptosis, angiogenesis, inflammation and the immune response [72]. STAT proteins are activated by a variety of protein kinases, including Janus kinase, growth factor receptor, non-receptor tyrosine kinase and G protein-coupled receptor [73]. Among them, STAT3 is involved in atherosclerosis through the regulation of endothelial cell function, macrophage polarization, inflammation and the immune response [74]. STAT3 has four isoforms with different functions: STAT3-α, STAT3-β, STAT3-γ and STAT3-δ [74]. STAT3-α-mediated activation of the IL-6/JAK2/STAT3 signaling pathway is predominantly pro-inflammatory [74]. Jiang et al. [75] have constructed a UC model in rats and have reported anti-inflammatory protective effects via inhibition of the JAK2/STAT3 signaling pathway, thereby decreasing IL-1β, IL-6 and TNF-α expression. However, STAT3-β promotes the expression of certain anti-inflammatory genes while inhibiting the synthesis of inflammatory factors [74]. These predominant anti-inflammatory effects may be associated with IL-10-mediated activation of STAT3 [76]. Li et al. [77] have found that targeting the activation of the IL-10/STAT3 axis exerts cardioprotective effects by modulating the inflammatory response, myocardial fibrosis and apoptosis. In addition, activation of the IL-10/STAT3 signaling pathway promotes macrophage polarization to the M2 type [78]. STAT3 and SPI1 are overexpressed in patients with ankylosing spondylitis and are closely associated with immune system disorders [79]. Similarly, we found that STAT3 and SPI1 were overexpressed in patients with both AMI and UC, thus suggesting that SPI1/STAT3 may be an important pathway leading to AMI and UC; however, further experimental verification is warranted. Overactivation or inactivation of STAT3 can lead to human disease [80]. Therefore, how to balance the activation of STAT3 must be further explored.

            Integrin alpha-X (ITGAX) is member of the integrin family [81]. ITGAX is considered a potential therapeutic target for various inflammatory and immune-associated diseases, such as periodontitis, atherosclerosis, primary dry syndrome and IgA nephropathy [8284]. Overexpression of ITGAX has been shown to activate the PI3k/Akt axis and promote angiogenesis in ovarian tumor tissues. However, no relevant animal experiments have been reported for ITGAX in AMI or UC.

            ICAM-1 is a transmembrane glycoprotein expressed at low basal levels in immune cells, endothelial cells and epithelial cells [85]. ICAM-1 expression is induced by multiple inflammatory cytokines and shows tissue-specific differences. ICAM-1 in endothelial cells is induced primarily by TNFα or IL-1β, whereas in intestinal epithelial cells, it is induced by IFNγ [86]. Freitas et al. [87] have demonstrated that circulating ICAM-1 levels are positively associated with the risk of coronary artery disease and are a potential marker of cardiovascular disease. The ICAM-1 gene polymorphism is closely associated with the development of UC [88]. ICAM-1 plays a fundamental role in neutrophil crossing of the endothelial cell layer [88]. Yu et al. [89] have found that ICAM-1 accelerates atherosclerosis by promoting leukocyte infiltration. Moreover, upregulation of ICAM-1 is associated with excessive activation of the RELA/NLRP3 axis, and leads to dysfunction of the human umbilical vein endothelium by regulating inflammatory responses and oxidative stress [90]. Similarly, we found that both RELA and ICAM-1 were stably upregulated in the disease group.

            C-C motif chemokine ligand 1 (CCL1), a CC chemokine family member, mediates the migration of monocytes to regions of inflammation [91]. Harpel et al. [92] have suggested that CCL1 and its receptor CCR8 are closely associated with inflammation and atherosclerosis. CCL1 is upregulated in several immune diseases, such as rheumatic heart disease, atopic dermatitis and psoriasis [93, 94]. CCR8 is activated by CCL1 and mediates the recruitment of T helper 2 cells to sites of inflammation [95]. Notably, CCL1 has no significant chemotactic effect on neutrophils [96]. Olsen et al. [95] have demonstrated that elevated circulating CCL1 is strongly associated with prognosis and mortality in colorectal cancer. Current studies on CCL1 in AMI or UC are inadequate. In conclusion, our data indicated that TNF, STAT3, ITGAX, ICAM1 and CCL1 are important targets for the treatment of patients with both UC and AMI.

            Although previous studies have explored the hub genes associated with AMI and UC separately [97, 98], most current animal studies have been limited to a single disease, thus failing to address the clinical coexistence of multiple diseases. Moreover, few studies have explored the common molecular mechanisms between these diseases through advanced bioinformatics methods. We sought to elucidate the molecular mechanisms underlying AMI and UC, to provide novel insight into the clinical management of patients with these comorbidities. Decreasing mortality and improving quality of life in patients with AMI combined with UC may be achieved by further standardizing clinical management strategies. However, this study has several limitations. First, only significant DEGs between disease and normal tissues were analyzed; consequently, certain genes that showed no significant changes in expression but played an important regulatory role were ignored. In addition, animal experiments are necessary to further reveal the causal relationship between these hub genes and the pathogenesis of the two diseases.

            Conclusions

            We used a bioinformatics approach to study patients with AMI combined with UC. We discovered many pathogenic mechanisms shared by AMI and UC and possibly mediated by the specified hub genes. Our research offers novel ideas to further investigate the molecular mechanisms underlying AMI combined with UC.

            Data Availability Statement

            All microarray datasets were obtained from the GEO database (https://www.ncbi.nlm.nih.gov/geo/).

            Ethics Statement

            Not applicable.

            Conflicts of Interest

            No potential conflicts of interest relevant to this article are reported.

            Author Contributions

            QS and QW developed the major research plan. CC and RC analyzed data, constructed graphs and wrote the manuscript. CC helped collect data and references. All authors contributed to the article and approved the submitted version.

            Citation Information

            References

            1. . Association between acute myocardial infarction and periodontitis: a review of the literature. J Int Acad Periodontol 2016;18:23–33.

            2. . Acute myocardial infarction. Reperfusion strategies. Chest 1994;106:1851–66.

            3. , , , , . Inflammation, frailty and cardiovascular disease. Adv Exp Med Biol 2020;1216:55–64.

            4. , , , , . Targeted pharmacotherapy for ischemia reperfusion injury in acute myocardial infarction. Expert Opin Pharmacother 2020;21:1851–65.

            5. , , , . miR-190-5p alleviates myocardial ischemia-reperfusion injury by targeting PHLPP1. Dis Markers 2021;2021:8709298.

            6. , . Epidemiology and pathogenesis of ulcerative colitis. Gastroenterol Clin North Am 2020;49:643–54.

            7. , , , , , , et al. Ulcerative colitis. Nat Rev Dis Primers 2020;6:74.

            8. , , , , , , et al. Colorectal cancer in ulcerative colitis: mechanisms, surveillance and chemoprevention. Curr Oncol 2022;29:6091–114.

            9. , , , , , , et al. A comprehensive review and update on ulcerative colitis. Dis Mon 2019;65:100851.

            10. , , . Acute myocardial infarction complicating active ulcerative colitis: a case report. Case Rep Cardiol 2011;2011:876896.

            11. , , , . Inflammatory bowel disease and the risk of cardiovascular diseases. Gastroenterol Hepatol 2021;44:236–42.

            12. , , , , , . No increased risk of myocardial infarction among patients with ulcerative colitis or Crohn’s disease. Clin Gastroenterol Hepatol 2011;9:875–80.

            13. , , , , , , et al. Patients with inflammatory bowel disease have an increased risk of myocardial infarction: a nationwide study. Aliment Pharmacol Ther 2019;50:769–79.

            14. , , , , , , et al. Ischemic heart disease in patients with inflammatory bowel disease: risk factors, mechanisms and prevention. Life (Basel) 2022;12:1113.

            15. , . Role of gut microbiota in atherosclerosis. Nat Rev Cardiol 2017;14:79–87.

            16. , , , , . The role of bacteria in the inflammatory bowel disease development: a narrative review. APMIS 2018;126:275–83.

            17. . Bacterial infections and atherosclerosis. J Investig Med 1998;46:396–402.

            18. , , . Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 2002;30:207–10.

            19. , , , , , , et al. A whole blood molecular signature for acute myocardial infarction. Sci Rep 2017;7:12268.

            20. , , , , , , et al. Altered gene expression pattern in peripheral blood mononuclear cells in patients with acute myocardial infarction. PLoS One 2012;7:e50054.

            21. , , , , , , et al. Molecular comparison of adult and pediatric ulcerative colitis indicates broad similarity of molecular pathways in disease tissue. J Pediatr Gastroenterol Nutr 2018;67:45–52.

            22. , , , , , , et al. Strong upregulation of AIM2 and IFI16 inflammasomes in the mucosa of patients with active inflammatory bowel disease. Inflamm Bowel Dis 2015;21:2673–82.

            23. , . GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics 2007;23:1846–7.

            24. , , , , , , et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43:e47.

            25. . ggplot2: elegant graphics for data analysis. 2nd ed. Computing Reviews; 2017.

            26. , , , , , . A comprehensive analysis of candidate genes and pathways in pancreatic cancer. Tumour Biol 2015;36:1849–57.

            27. , , , , . Weighted gene coexpression network analysis uncovers critical genes and pathways for multiple brain regions in Parkinson’s disease. Biomed Res Int 2021;2021: 6616434.

            28. , . VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R. BMC Bioinform 2011;12:35.

            29. , , , , , , et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res 2004;32:D258–61.

            30. , . KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000;28:27–30.

            31. , , , , , , et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb) 2021;2:100141.

            32. , , , , , , et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res 2021;49:D605–D12.

            33. , , , , , , et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 2003;13:2498–504.

            34. , , , , , . cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol 2014;8(Suppl 4):S11.

            35. , , , , . GeneMANIA: a real-time multiple association network integration algorithm for predicting gene function. Genome Biol 2008;9(Suppl 1):S4.

            36. , , , , , , et al. TRRUST: a reference database of human transcriptional regulatory interactions. Sci Rep 2015;5:11432.

            37. , , , , . Update on acute coronary syndromes: the pathologists’ view. Eur Heart J 2013;34: 719–28.

            38. , , , , , , et al. International consensus on the prevention of venous and arterial thrombotic events in patients with inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2021;18:857–73.

            39. , , , , , , et al. Disease activity in inflammatory bowel disease is associated with increased risk of myocardial infarction, stroke and cardiovascular death--a Danish nationwide cohort study. PLoS One 2013;8:e56944.

            40. , , , , . Risk of arterial thrombotic events in inflammatory bowel disease. Am J Gastroenterol 2009;104:1445–51.

            41. , , , , , , et al. Inflammatory bowel disease does not impact mortality but increases length of hospitalization in patients with acute myocardial infarction. Dig Dis Sci 2021;66:4169–77.

            42. . Monitoring leukocyte migration during atherosclerosis in vivo. Methods Mol Biol 2022;2419:475–9.

            43. , , , , , , et al. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019;115:1266–85.

            44. , , , , , . Targeting epigenetics and non-coding RNAs in atherosclerosis: from mechanisms to therapeutics. Pharmacol Ther 2019;196:15–43.

            45. , , . Controlling leukocyte trafficking in IBD. Pharmacol Res 2020;159: 105050.

            46. , , . The role of gut-derived oxidized lipids and bacterial lipopolysaccharide in systemic inflammation and atherosclerosis. Curr Opin Lipidol 2022;33:277–82.

            47. , , , , , , et al. Interactive effects of interferon-gamma functional single nucleotid polymorphism (+874 T/A) with cardiovascular risk factors in coronary heart disease and early myocardial infarction risk. Mol Biol Rep 2020;47:8397–405.

            48. , , , , , . One year in review 2020: pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol 2020;38:387–97.

            49. , , , , , . Association of acute myocardial infarction with seropositive rheumatoid arthritis in Korea: a nationwide longitudinal cohort study. J Clin Neurosci 2020;78: 97–101.

            50. , . Rheumatoid arthritis and inflammatory bowel disease: a bidirectional two-sample Mendelian randomization study. Semin Arthritis Rheum 2022;55: 151992.

            51. , , , , . The role and therapeutic potential of NF-kappa-B pathway in severe COVID-19 patients. Inflammopharmacology 2021;29: 91–100.

            52. , , , , , , et al. The two faces of interleukin-17A in atherosclerosis. Curr Drug Targets 2017;18:863–73.

            53. , . Toll-like receptors and the control of immunity. Cell 2020;180:1044–66.

            54. , , , , , , et al. Innate immune receptors, key actors in cardiovascular diseases. JACC Basic Transl Sci 2020;5:735–49.

            55. , , . Toll-like receptors: general molecular and structural biology. J Immunol Res 2021;2021: 9914854.

            56. , , . Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2022;187:106566.

            57. , . Death by TNF: a road to inflammation. Nat Rev Immunol 2023;23(5): 289–303.

            58. , , , , . TNF-α is a novel biomarker for predicting plaque rupture in patients with ST-segment elevation myocardial infarction. J Inflamm Res 2022;15:1889–98.

            59. , , , , , . TNF-alpha blockers in inflammatory bowel diseases: practical recommendations and a user’s guide: an update. Digestion 2020;101(Suppl 1):16–26.

            60. , , , . The role of tumor necrosis factor associated factors (TRAFs) in vascular inflammation and atherosclerosis. Front Cardiovasc Med 2022;9:826630.

            61. , , , , , , et al. TNF blockade: an inflammatory issue. Ernst Schering Res Found Workshop 2006;(56):161–86.

            62. , . Upregulation of SPI1 during myocardial infarction aggravates cardiac tissue injury and disease progression through activation of the TLR4/NFκB axis. Am J Transl Res 2022;14:2709–27.

            63. , , , , , . Expression of nuclear factor kappa B in ovine maternal inguinal lymph nodes during early pregnancy. BMC Vet Res 2022;18:266.

            64. , , , , , , et al. NFKB1 gene rs28362491 polymorphism is associated with the susceptibility of acute coronary syndrome. Biosci Rep 2019;39(4):BSR20182292.

            65. , , , , , , et al. Asperuloside suppressing oxidative stress and inflammation in DSS-induced chronic colitis and RAW 264.7 macrophages via Nrf2/HO-1 and NF-κB pathways. Chem Biol Interact 2021;344:109512.

            66. , , , , , , et al. [Effects of electroacupuncture on inflammatory response of cardiac muscle tissue in mice with acute myocardial ischemia]. Zhongguo Zhen Jiu 2018;38:5133–8.

            67. , , , , , , et al. Expression and regulatory characteristics of peripheral blood immune cells in primary Sjögren’s syndrome patients using single-cell transcriptomic. iScience 2022;25:105509.

            68. , , , , . Functional role of NF-IL6beta and its sumoylation and acetylation modifications in promoter activation of cyclooxygenase 2 gene. Nucleic Acids Res 2006;34:217–31.

            69. , , , , , , et al. A hierarchical and collaborative BRD4/CEBPD partnership governs vascular smooth muscle cell inflammation. Mol Ther Methods Clin Dev 2021;21:54–66.

            70. , , , , , , et al. An anti-inflammatory role for C/EBPδ in human brain pericytes. Sci Rep 2015;5:12132.

            71. , . The many faces of C/EBPδ and their relevance for inflammation and cancer. Int J Biol Sci 2013;9:917–33.

            72. , , . Highlighted STAT3 as a potential drug target for cancer therapy. BMB Rep 2019;52:415–23.

            73. , . Multiple roles of STAT3 in cardiovascular inflammatory responses. Prog Mol Biol Transl Sci 2012;106:63–73.

            74. , , , , , , et al. Targeted inhibition of STAT3 as a potential treatment strategy for atherosclerosis. Theranostics 2019;9:6424–42.

            75. , , , , , , et al. Retardant effect of dihydroartemisinin on ulcerative colitis in a JAK2/STAT3-dependent manner. Acta Biochim Biophys Sin (Shanghai) 2021;53:1113–23.

            76. , . The IL-10/STAT3 axis: contributions to immune tolerance by thymus and peripherally derived regulatory T-cells. Eur J Immunol 2017;47:1256–65.

            77. , , , , , , et al. YQWY decoction improves myocardial remodeling via activating the IL-10/Stat3 signaling pathway. Evid Based Complement Alternat Med 2020;2020:7532892.

            78. , , , , , , et al. Peripheral blood-derived mesenchymal stem cells modulate macrophage plasticity through the IL-10/STAT3 pathway. Stem Cells Int 2022;2022:5181241.

            79. , , , , , , et al. STAT3 and SPI1, may lead to the immune system dysregulation and heterotopic ossification in ankylosing spondylitis. BMC Immunol 2022;23:3.

            80. , , , . STAT3 signaling in immunity. Cytokine Growth Factor Rev 2016;31:1–15.

            81. , , , , . Integrin alpha x stimulates cancer angiogenesis through PI3K/Akt signaling-mediated VEGFR2/VEGF-A overexpression in blood vessel endothelial cells. J Cell Biochem 2019;120:1807–18.

            82. , , . Integrated analysis and exploration of potential shared gene signatures between carotid atherosclerosis and periodontitis. BMC Med Genomics 2022;15:227.

            83. , , , , , , et al. Gene expression profiling of epithelium-associated FcRL4(+) B cells in primary Sjögren’s syndrome reveals a pathogenic signature. J Autoimmun 2020;109: 102439.

            84. , , , , , , et al. Formalin-fixed paraffin-embedded renal biopsy tissues: an underexploited biospecimen resource for gene expression profiling in IgA nephropathy. Sci Rep 2020; 10:15164.

            85. , , , . Intercellular adhesion molecule-1 as a drug target in asthma and rhinitis. Respirology 2014;19:508–13.

            86. , , . ICAM-1: a master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol 2020;108:787–99.

            87. , , , , , , et al. Novel biomarkers in the prognosis of patients with atherosclerotic coronary artery disease. Rev Port Cardiol (Engl Ed) 2020;39:667–72.

            88. . Intercellular adhesion molecule-1 (ICAM-1) in ulcerative colitis: presence, visualization, and significance. Inflamm Res 2005;54:313–27.

            89. , , , . Serum VCAM-1 and ICAM-1 measurement assists for MACE risk estimation in ST-segment elevation myocardial infarction patients. J Clin Lab Anal 2022;36:e24685.

            90. , , , , , , et al. Salidroside ameliorates endothelial inflammation and oxidative stress by regulating the AMPK/NF-κB/NLRP3 signaling pathway in AGEs-induced HUVECs. Eur J Pharmacol 2020;867:172797.

            91. , , , , , , et al. Genetic polymorphisms of CCL1 rs2072069 G/A and TLR2 rs3804099 T/C in pulmonary or meningeal tuberculosis patients. Int J Clin Exp Pathol 2015;8:12608–20.

            92. , . Chemokine receptor-8: potential role in atherogenesis. Isr Med Assoc J 2002;4:1025–7.

            93. , . Rheumatic heart disease: molecules involved in valve tissue inflammation leading to the autoimmune process and anti-S. pyogenes vaccine. Front Immunol 2013;4:352.

            94. , , , , , . Expression of CCL1 and CCL18 in atopic dermatitis and psoriasis. Clin Exp Dermatol 2012;37:521–6.

            95. , , , , . Circulating inflammatory factors associated with worse long-term prognosis in colorectal cancer. World J Gastroenterol 2017;23:6212–9.

            96. , . The human cytokine I-309 is a monocyte chemoattractant. Proc Natl Acad Sci U S A 1992;89: 2950–4.

            97. , , , , , , et al. The identification and validation of hub genes associated with acute myocardial infarction using weighted gene co-expression network analysis. J Cardiovasc Dev Dis 2022;9:30.

            98. , , , , , , et al. Identification of immune-related gene signature and prediction of CeRNA network in active ulcerative colitis. Front Immunol 2022;13:855645.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            14 June 2023
            : 8
            : 1
            : e978
            Affiliations
            [1] 1Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
            [2] 2Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
            [3] 3Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
            [4] 4Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing 100853, China
            Author notes
            Correspondence: Qiang Su, PhD, Professor, Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin 541000, Guangxi Zhuang Autonomous Region, China, E-mail: suqiang1983@ 123456foxmail.com ; and Qiang Wu, Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China, E-mail: wuqiang@ 123456jgc301.com
            Article
            cvia.2023.0034
            10.15212/CVIA.2023.0034
            2bcfc09e-ea52-4965-a0d2-4f0ec01b7cff
            Copyright © 2023 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 17 January 2023
            : 07 May 2023
            : 17 May 2023
            Page count
            Figures: 11, Tables: 4, References: 98, Pages: 20
            Funding
            Funded by: Guangxi Natural Science Foundation
            Award ID: 2020GXNSFDA238007
            Funded by: Key Research and Development Program of Guangxi
            Award ID: AB20159005
            This work was supported by the Guangxi Natural Science Foundation (2020GXNSFDA238007) and the Key Research and Development Program of Guangxi (AB20159005).
            Categories
            Research Article

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            hub genes,bioinformatics,ulcerative colitis,differentially expressed genes,acute myocardial infarction

            Comments

            Comment on this article