Identification of key ferroptosis‑related biomarkers in Kawasaki disease by clinical and experimental validation
- Authors:
- Published online on: November 19, 2024 https://doi.org/10.3892/br.2024.1894
- Article Number: 16
-
Copyright: © Yan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Kawasaki disease (KD; also termed mucocutaneous lymph node syndrome) is an acute febrile illness that primarily occurs in 6-month to 5-year-old children. KD is characterized by systemic non-specific vasculitis as the main pathological change (1) and mainly affects the skin, mucous membranes, blood vessels and lymph nodes. If not treated in time, KD-associated acute vasculitis may progress to coronary artery lesions, which manifest as coronary artery dilation and stenosis, and can lead to coronary artery aneurysm (CAA), myocardial infarction, thrombotic occlusion and sudden death (2,3). CAA occurs in ~25% of children with KD who do not receive timely intervention (4). Moreover, patients with KD who have giant CAAs are predisposed to developing ischemic heart disease (5). In recent years, the incidence of KD has been increasing, and KD has become a common cause of childhood acquired heart disease in developed countries (6,7).
Although >50 years have passed since it was first reported, the etiology of KD remains unclear and the pathological mechanism underlying KD is not well defined. Genetically susceptible individuals become prone to KD when they encounter stimuli from infectious agents and environmental factors that trigger an abnormal immune response in the body (8-10). The preferred treatment for KD involves intravenous immunoglobulin (IVIG) combined with high-dose aspirin administration, and early diagnosis and timely IVIG therapy can reduce the risk of CAA from 25 to 4% (4). Although the incidence of CAA has been greatly reduced, 4% of patients still endure severe sequelae; therefore, it is particularly important to clarify the pathogenesis of this disease and to facilitate early diagnosis and effective treatment. In the absence of precise diagnostic markers, the diagnosis of KD is mainly based on clinical manifestations (11). However, the clinical symptoms of this disease resemble those of other febrile illnesses, which can result in delayed diagnosis or a misdiagnosis (12). If left untreated, CAA-related sequelae may have serious long-term effects on patients. Therefore, it is essential to develop a set of accurate and sensitive laboratory diagnostic markers to assist physicians in the early detection and diagnosis of KD. Moreover, timely treatment can improve the response to IVIG therapy and reduce the incidence of coronary artery injury.
Ferroptosis, a newly discovered iron-dependent form of programmed cell death, is characterized by iron-induced lipid peroxidation and the accumulation of lipophilic reactive oxygen species. The mechanism and regulation of ferroptosis differ from those of cell autophagy, necrosis, apoptosis and other traditional modes of cell death (13,14). Ferroptosis is mainly associated with a variety of physiological and pathological processes, including degenerative disease, infectious disease, coronary artery disease, myocardial injury, kidney damage and sepsis-induced lung injury (15-17). A recent study revealed that serum ferritin could be used to diagnose and predict IVIG resistance in patients with KD (18); however, few studies have assessed the role of ferroptosis-related genes (FRGs) in the diagnosis and progression of KD.
Therefore, in the present study, an integrative bioinformatics approach was used to investigate the role of FRGs in predicting KD by systematically analyzing transcriptome sequencing data from online databases. The findings were then validated using clinical samples. It is expected that these FRGs will serve as biomarkers for the early diagnosis of KD and provide new insights into the pathogenesis of this disease.
Materials and methods
Data collection
In total, three microarray KD gene expression datasets and the corresponding clinical sample information were collected from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/). The GSE73461 dataset (19) was generated based on the GPL10558 platform using 55 normal samples and 77 KD samples. The GSE68004 dataset (GPL10558 platform) contained 76 KD samples and 37 healthy control samples (20). The samples in the GSE73461 and GSE68004 datasets were selected as the training set. The 20 KD samples and 9 normal samples in the GSE18606 dataset (processed on the GPL6480 platform) were used as the independent validation set (21). The training set was used for model construction and marker gene discovery, whereas the validation set was mainly used for marker gene assessment and validation. Inclusion criteria were patients diagnosed with KD who were not treated with IVIG and with complete corresponding transcriptomic information in the dataset. Exclusion criteria were patients with other serious complications that could affect gene expression, such as definite bacterial infections, viral infections and inflammatory diseases. In addition, FRGs were collected from the FerrDb database, and the 259 FRGs are listed in Table SI. In the present study, the Drug Gene Interaction Database (DGIdb; https://www.dgidb.org/) was used to predict drugs that may target these ferroptosis-related markers, and the interaction network was visualized via Cytoscape software (version 3.7.2; https://cytoscape.org/).
Selection of differentially expressed-FRGs (DE-FRGs)
The limma package in R (version 3.48.3; http://bioinf.wehi.edu.au/limma) was applied to screen differentially expressed genes (DEGs) between healthy individuals and patients with KD in the GSE73461 and GSE68004 datasets. |log [fold change]|≥1 and adjusted P-values (adj. P) <0.05 were set as the screening threshold. A Venn diagram (http://jvenn.toulouse.inra.fr/app/) was then used to display the intersection of the DEGs and FRGs. Furthermore, unsupervised hierarchal clustering analysis of samples based on the expression pattern of selected differentially expressed-FRGs (DE-FRGs) was conducted using the heatmap package (https://hiplot.com.cn/basic/heatmap).
Least absolute shrinkage and selection operator (LASSO) regression and receiver operating characteristic (ROC) analysis
To identify the optimal gene biomarkers for KD, the binomial LASSO regression model, constructed using the glmnet package in R (version 4.1), was applied to build an optimal model with the fewest genes. The penalty parameters (best λ values) were obtained by 10-fold cross-validation with binomial deviance. Based on the optimal λ value, a list of candidate ferroptosis-related markers (named marker genes) for KD was obtained by modifying this model. Then, a logistic regression model, which was built based on 6 ferroptosis-related markers, combined with ROC analysis was used to evaluate the ability of these markers to distinguish patients with KD from healthy controls.
Gene Set Variation Analysis (GSVA)
GSVA can convert gene-level expression data into a pathway-level expression matrix and is used to investigate the changes in biological processes and signaling pathways in different samples. In the present study, the samples from the GSE73461 dataset were divided into the high-expression and low-expression groups according to the expression level of each marker gene. Then, GSVA was conducted using the GSVA package in R to analyze the differences in pathway activation between the high-expression and low-expression groups. The background gene set, ‘c2.kegg.v7.4.symbols’, used in the GSVA package was downloaded from MSigDB (https://www.gsea-msigdb.org/gsea/msigdb). The screening criteria were T-value >2 and P<0.05. Only the top 60 enriched signaling pathways were listed.
Immune cell infiltration analysis
Gene expression data from the GSE73461 and GSE68004 datasets were used to evaluate the immune cell infiltration profiles between patients with KD and healthy individuals. There are several methods for immune infiltration analysis, but their applicability and output results vary in a specific field context. These commonly used methods, ESITMATE, MCP-counter and TIMER, cannot be used to compare the abundance of different cell types in the same sample and are more suitable for analyzing the tumor immune microenvironment. CIBERSORT is able to infer the proportion of different immune cell types in the same sample using transcriptomic data. This meets the needs of the present study. Therefore, the scores of 22 immune cell types were calculated using CIBERSORT to assess the levels of the infiltrating immune cell types between the two groups. CIBERSORT is a deconvolution algorithm that estimates the proportions of specific cell types based on gene expression levels (22). Not all 22 immune cell types could be estimated from the KD datasets; thus, only 18 types were listed in the present study. The correlations between the expression levels of marker genes and the levels of different immune cell types were then analyzed using Pearson's correlation analysis. In addition, the expression levels of immune checkpoint genes between the controls and patients with KD were analyzed, and the correlation between marker genes and immune checkpoint genes were explored using Pearson's correlation analysis.
Clinical sample collection
All peripheral blood samples were obtained from Shenzhen Baoan Women's and Children's Hospital of Jinan University (Shenzhen, China), which included 22 samples from patients with KD and 25 from healthy controls. The age range of the patients was from a minimum of 3 months and 14 days to a maximum of 6 years and 9 months. The average age was 2 years, 5 months and 10 days. These blood samples were used to validate the diagnostic performance of the candidate marker genes. The study procedure was approved (approval no. LLSC-2023-03-09-04-KS) by the Medical Ethics Committee of Shenzhen Baoan Women's and Children's Hospital of Jinan University (Shenzhen, China). Informed consent was acquired from the participants' legal guardian. Validation of marker gene expression was conducted using reverse transcription-quantitative PCR (RT-qPCR).
Cell culture and lipopolysaccharide (LPS) stimulation
Human umbilical vein endothelial cells (HUVECs; cat. no. 8000; ScienCell Research Laboratories, Inc.) were cultured in endothelial cell medium (cat. no. 1001; ScienCell Research Laboratories, Inc.) at 37˚C under an atmosphere with 5% CO2. Endothelial cell cultures at passages 5 or 6 were used in all experiments. After reaching confluency, HUVECs were treated with LPS at a final concentration of 1 µg/ml for 2 h. The cells were then harvested and total RNA was isolated using the FastPure Total RNA Isolation Kit (cat. no. RC112; Vazyme Biotech Co., Ltd.) according to the manufacturer's protocol. The cell experiments regarding HUVECs purchased in the present study were approved (approval no. LLSC-2024-03-10-25-KS) by the Medical Ethics Committee of Shenzhen Baoan Women's and Children's Hospital of Jinan University (Shenzhen, China).
RT-qPCR
Total RNA from the peripheral blood samples was isolated using the EasyPure Blood RNA Kit (cat. no. ER401-01; TransGen Biotech Co., Ltd.) following the manufacturer's protocol. A NanoDrop One Microvolume UV-Vis spectrophotometer (Thermo Fisher Scientific, Inc.) was then used to assess the RNA purity (OD260/OD280 >1.9) and concentration. Total RNA from the blood and HUVEC samples was used for cDNA synthesis using HiScript® III All-in-one RT SuperMix (cat. no. R333; Vazyme Biotech Co., Ltd.) according to the manufacturer's protocols. qPCR was then performed in an ABI 7500 real-time PCR machine (Applied Biosystems; Thermo Fisher Scientific, Inc.) using Taq Pro Universal SYBR qPCR Master Mix (cat. no. Q712; Vazyme Biotech Co., Ltd.). The primer sequences are listed in Table I. The PCR cycling protocol was as follows: 95˚C for 30 sec, then 40 cycles of a 2-step amplification at 95˚C for 15 sec and 60˚C for 30 sec. Gene expression was normalized to the expression of the internal reference gene (GAPDH), obtaining the ΔCq value. The mRNA levels of the candidate markers relative to GAPDH were calculated by the 2-ΔΔCq method (23).
Statistical analysis
The sensitivity and specificity of the diagnostic performance of each marker gene in KD vs. the controls were calculated using the ROC analysis and area under the curve (AUC) values. The comparison of gene expression and immune cell infiltration between the healthy controls and patients with KD was performed using an unpaired Student's t-test. P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were conducted using GraphPad Prism Software (version 7.03; Dotmatics).
Results
Selection of DE-FRGs in patients with KD
A flowchart of the study design is shown in Fig. 1. To identify genes associated with KD, DEGs between control and KD samples were analyzed using the limma package in R. Based on the screening thresholds of |log2FC|≥1.0 and adj.P<0.05, 1,857 DEGs were selected in the GSE73461 dataset, which included 700 downregulated genes and 1,157 upregulated genes. In the GSE68004 dataset, 2,488 DEGs were obtained, of which 1,268 were significantly upregulated and 1,220 were downregulated. DEGs in the training sets (from GSE73461 and GSE68004) were visualized using volcano plots (Fig. 2A and B). Then, the intersection of the DEGs and FRGs was screened via a Venn diagram, and 10 overlapping DE-FRGs were identified between the patients with KD and controls (Fig. 2C). Furthermore, an unsupervised clustering analysis was conducted on 132 samples from the GSE73461 dataset using the expression data of the 10 DE-FRGs. As shown in the heatmap, the DE-FRGs were highly expressed in the KD samples and could markedly separate the KD samples from the control samples, suggesting that they were closely associated with KD (Fig. 2D). Therefore, further research was conducted on these 10 genes and used to identify diagnostic markers for KD.
Identification of key ferroptosis-related markers associated with KD
To screen the optimal diagnostic genes for KD, a LASSO regression model was constructed to analyze the DE-FRGs using the glmnet package in R. A 10-fold cross-validation was utilized to select the optimal penalty parameter (λ value) (Fig. 3A). Based on the best λ value, an optimal gene model was developed and 6 key genes associated with the diagnosis of KD were obtained (Fig. 3B). These 6 genes were considered key ferroptosis-related markers (marker genes) and included EPAS1, MAPK14, SLC2A3, PGD, SAT1 and TLR4.
Moreover, logistic regression models and subsequent ROC curve analyses were performed to determine the diagnostic predictive ability of the marker genes in the training and validation sets. The AUC values of the marker genes were 0.9969 in GSE73461 (Fig. 3C), 0.9993 in GES68004 (Fig. 3D), and 1.0000 in GSE18006 (validation set; Fig. 3E). The results indicated that these 6 genes could markedly distinguish patients with KD from healthy controls, suggesting that they could be used as potential diagnostic markers for KD.
GSVA
GSVA was then conducted to explore the differential changes in the signaling pathways between the high-expression and low-expression groups of each marker gene. The data revealed that high SLC2A3 expression was primarily enriched in the Toll-like receptor signaling pathway, carbohydrate metabolism (starch and sucrose metabolism and galactose metabolism), the RIG-I-like receptor signaling pathway, the complement and coagulation cascades and fatty acid metabolism. In comparison, low SLC2A3 expression was associated with adaptive immune responses (primary immunodeficiency, the T-cell receptor signaling pathway, antigen processing and antigen presentation), cardiac muscle contraction and histidine metabolism (Fig. 4A). The upregulation of SAT1 was closely related to innate immune activation (the Toll-like receptor, RIG-I-like receptor and Nod-like receptor signaling pathways), cytosolic DNA sensing, the B-cell receptor signaling pathway, carbohydrate metabolism and fatty acid metabolism. Meanwhile, in the SAT1 low-expression group, the Hedgehog signaling pathway, adaptive immune-related pathways, the Wnt signaling pathway, cell adhesion molecules and cardiac muscle contraction were enriched (Fig. 4B). Notably, in the high and low PGD, EPAS1, TLR4 and MAPK14 gene expression groups, the pathway enrichment patterns were similar to those in the high and low SLC2A3 and SAT1 gene expression groups (Fig. S1A-D). These findings suggested that in the early stage of KD, these marker genes may affect the progression of KD by activating the innate immune-associated pathways, glucose metabolism and fatty acid metabolism, and partially reducing the adaptive immune response-related processes.
Immune cell infiltration landscapes in patients with KD
Considering the GSVA enrichment data was closely associated with immune-related biological processes, immune infiltration analysis was performed using healthy and KD samples by the CIBERSORT algorithm. The immune infiltration scores were visualized using split violin plots (Fig. 5A and B). The infiltration levels of CD8 T cells, CD4 naïve T cells, resting CD4 memory T cells, resting natural killer (NK) cells and activated NK cells were significantly lower in patients with KD than in the controls in the training sets (GSE73461 and GSE68004). By contrast, the infiltration levels of γδ T cells, monocytes, macrophage M0, and neutrophils were significantly higher in patients with KD than in the controls in the training sets (Fig. 5A and B). The levels of memory B cells, macrophage M2 and resting mast cells were reduced in patients with KD in the GSE73461 dataset, and the contents of naïve B cells were reduced in patients with KD in the GSE68004 dataset. Furthermore, the contents of activated CD4 memory T cells and activated mast cells were relatively enhanced in KD group in GSE73461, while the fraction of resting mast cells was significantly higher in patients with KD in GSE68004. In summary, these data indicated that most adaptive immune-related cells are downregulated in patients with KD.
In addition, the CD80, CTLA4, CD274, PDCD1, CD86, CD28, BTLA, LAG3, TIGIT, TNFSF4 and TNFRSF4 genes were selected as immune checkpoint genes, and their expression levels in patients with KD and healthy controls were analyzed using these microarray datasets. The difference in immune levels between the two groups was also presented in the difference in gene expression levels of these immune checkpoints (Fig. 5C). As shown in Fig. 5C, the expression levels of most checkpoint genes were low in patients with KD. By contrast, the CD274 and TNFSF4 genes were highly expressed in KD, while the expression level of CD80 remained unchanged. In summary, most immune checkpoint genes involved in adaptive immunity were downregulated in KD, implying that the adaptive immune response in these patients was suppressed to some extent.
Correlation of marker genes with immune infiltration cells and immune checkpoint genes
Pearson's correlation coefficient was used to analyze the correlation of key markers with immune cells and immune checkpoint genes. The results demonstrated that the key marker genes exhibited a strong negative correlation with CD8 T cells, CD4 naïve T cells, resting NK cells, activated NK cells and macrophage M2, with the strongest correlation noted with CD8 T cells (Fig. 6A). By contrast, these candidate marker genes exhibited a strong positive correlation with activated CD4 memory T cells, γδ T cells, monocytes, macrophage M0 and neutrophils (Fig. 6A).
The correlations between the marker genes and immune checkpoints are presented in Fig. 6B. In total, 5 key genes (excluding EPAS1) were strongly positively correlated with CD274 and TNFSF4, but negatively correlated with most immune checkpoint genes, such as CTLA4, PDCD1, CD28, BTLA, TIGIT and TNFRSF4. EPAS1 was only positively correlated with CD274 and negatively correlated with CD28 (Fig. 6B). In summary, these results indicated that changes in immune features may be associated with the marker genes, both of which may play a key role in the progression of KD.
Ability of the marker genes to predict KD in the training set
To determine the efficiency of the identified marker genes in predicting KD, their mRNA expression levels in patients with KD and healthy individuals were first examined using the training set. In the GSE73461 and GSE68004 datasets, all 6 ferroptosis-related markers were significantly upregulated in patients with KD (Fig. 7A and B), suggesting that high expression levels of these genes may be involved in the occurrence of KD.
ROC analysis was then conducted to elucidate the classification power of each marker gene. As shown in Fig. 7C and D, the AUC values of MAPK14, SLC2A3, PGD, SAT1, TLR4 and EPAS1 for the prediction of KD were 0.968, 0.986, 0.939, 0.939, 0.863 and 0.692, respectively, in the GSE73461 dataset and 0.978, 0.997, 0.995, 0.960, 0.931 and 0.794, respectively, in the GSE68004 dataset (Fig. 7C and D). Thus, these markers could markedly differentiate patients with KD from healthy controls. Taken together, these findings indicated that the 6 marker genes have notable diagnostic value for predicting KD. Thus, they were considered diagnostic markers for KD.
Diagnostic performance of marker genes in the validation set
The diagnostic applications of these ferroptosis-related markers were evaluated using the validation set (GSE18006). The mRNA expression levels of the 6 genes are exhibited in Fig. 8A, all of which were significantly increased in the KD samples (Fig. 8A). Moreover, the AUC values of the SLC2A3, PGD, MAPK14, SAT1, TLR4 and EPAS1 genes in the validation set were 0.889, 0.956, 0.939, 0.839, 0.856 and 0.839, respectively (Fig. 8B). These data revealed the effectiveness of these markers in the diagnosis of KD and their potential as diagnostic markers for KD.
Drug prediction
DGIdb was applied to predict drugs that may target the identified diagnostic markers. An interaction network was constructed using Cytoscape software and is shown in Fig. 9. The details of the network are also listed in Table SII. A total of 91 drugs were queried, of which 72 drugs targeted MAPK14, 11 targeted TLR4, 2 drugs for PGD, and 3 drugs each targeted SLC2A3 and EPAS1. Furthermore, no drugs targeting SAT1 were detected.
Validation of ferroptosis-related markers in clinical samples and vascular endothelial cells
Blood samples were collected from 22 patients with KD and 25 healthy controls at Shenzhen Baoan Women's and Children's Hospital of Jinan University to further validate the diagnostic value of the candidate FRG markers using RT-qPCR. As demonstrated in Fig. 10A, compared with the healthy controls, the expression levels of MAPK14, SLC2A3 and PGD were significantly increased in patients with KD. However, the expression levels of SAT1, TLR4 and EPAS1 did not differ significantly between the two groups. These clinical validation results suggested that MAPK14, SLC2A3 and PGD have the potential to serve as diagnostic biomarkers for KD. Meanwhile, the present study also provided a set of 6 ferroptosis-related markers for future research.
KD is mainly characterized by acute systemic vasculitis, and the seasonality and prevalence of high incidence areas indicate that KD is caused by infectious factors (9). Therefore, to construct a cellular model to explore the alteration of candidate markers in the early onset of KD, HUVECs were treated with the endotoxin, LPS, to induce an inflammatory response. The expression levels of MAPK14, SLC2A3, PGD, SAT1, TLR4 and EPAS1 in HUVECs were then measured by RT-qPCR. The expression levels of SLC2A3, PGD and SAT1 were significantly upregulated in LPS-stimulated HUVECs compared with the controls (Fig. 10B). These results suggested that SLC2A3, PGD and SAT1 are closely related to the early progression of KD and thus may be potential therapeutic targets.
Discussion
KD is an acute febrile pediatric disease with systemic vasculitis as the main lesion; however, its etiology and pathogenesis are currently unknown. Without timely diagnosis and intervention, 25-30% of patients with KD become susceptible to coronary artery abnormalities (4); therefore, early detection and diagnosis serve a crucial role in the treatment and prognosis of this disease and can improve outcomes. With the rapid development of gene microarray technology, bioinformatics analysis based on altered gene expression data has become a notable and promising approach for discovering key biomarkers in the development and progression of various diseases (24).
In the present study, a comprehensive bioinformatics analysis of KD-associated microarray datasets was employed to identify important ferroptosis-related biomarkers and investigate their diagnostic value in KD. The gene expression profiles of two KD datasets, GSE73461 and GSE68004 were analyzed and 1,148 DEGs, including 10 ferroptosis-related DEGs, between the patients with KD and healthy controls were screened (Fig. 2C). Then, using the LASSO model and ROC curves, 6 candidate ferroptosis-related markers were acquired from 10 FR-DEGs, which had a favorable ability to distinguish KD samples from control samples. All these FRGs were shown to be upregulated in patients with KD. In addition, the immune cell infiltration landscape in the KD samples was further analyzed using the CIBERSORT algorithm and the expression levels of immune checkpoint genes between the two groups were determined. Finally, the diagnostic potential of these key biomarkers for KD were assessed and verified using the training set, validation set, and clinical samples.
In the present study, GSVA revealed that in the early stages of KD, the high-expression group of key marker genes was mainly associated with innate immune signaling pathways (Toll-like receptor, RIG-I-like receptor and Nod-like receptor signaling pathways), carbohydrate metabolism, B-cell receptor signaling pathway and fatty acid metabolism. By contrast, the low-expression group of the marker genes was mainly enriched in adaptive immune response-related processes (such as primary immunodeficiency, T-cell receptor signaling pathway, antigen processing and antigen presentation), cardiac muscle contraction and the Wnt signaling pathway. These findings are similar to those of a previous study (25). It is now generally accepted that multiple viruses, bacteria and fungi as well as certain environmental triggers can induce KD (8,26). Pathogenic microorganisms produce specific molecular motifs, including lipopeptides, peptidoglycans and endotoxins, which are known as pathogen-associated molecular patterns (PAMPs) and microbe-associated molecular patterns (MAMPs). The innate immune system of the host can express pattern recognition receptors (PRRs) (such as TLRs, RLRs and NLRs) to recognize these dangerous PAMPs and MAMPs and thus elicit innate immune responses (27). This may be why the innate immune signaling pathway was shown to be enriched in the present study.
Qian et al (28) reported that the upregulated proteins in patients with KD were mainly associated with certain metabolomics-related processes, such as the pentose phosphate pathway, glycolysis and glyceraldehyde-3-phosphate processes, and that succinic acid, which plays a pivotal role in regulating the tricarboxylic acid (TCA) cycle, was markedly elevated in the serum of patients with KD. The TCA cycle is the central hub and final metabolic pathway of carbohydrate, lipid and amino acid metabolism. This indicates that the metabolic processes of these three major nutrients were also changed in patients with KD. Moreover, previous studies have revealed that T-cell receptor/CD3-induced T-cell proliferation was significantly inhibited (29), and T-cell-related response pathways as well as antigen processing and presentation were significantly downregulated in patients with KD, compared with normal controls (30-32). These findings are in line with those of the present study.
In the present study, the immune cell infiltration profiles in the KD and healthy control samples from two training datasets, GSE73461 and GSE68004, were investigated. The data revealed that the infiltration levels of the innate immunity-associated cells, monocytes, macrophage M0 and neutrophils, were significantly higher in patients with KD than in controls. As aforementioned, invading pathogens enter the body of the host and trigger an inflammatory response through the innate immune cells via PRRs. It is generally accepted that the acute phase in patients with KD is mainly characterized by a marked increase and significant activation of innate immune cells (33). Furthermore, γδ T cells are mainly involved in the innate immune response, and the infiltration of γδ T cells was shown to be significantly elevated in KD in the present study, consistent with previous findings (30). By contrast, the present results revealed that the infiltration scores of most cells related to adaptive immunity, including CD8 T cells, CD4 naïve T cells, resting CD4 memory T cells, resting NK cells and activated NK cells, were notably lower in patients with KD. These findings suggest that the progression of KD is accompanied by abnormalities in adaptive immunity. Popper et al (34) reported that in the acute phase of KD, CD8 cells, T cells and NK cells adhere to the arterial walls and that their expression levels are highly associated with KD progression. In addition, Xie et al (35) noted a marked increase in the abundance of B cells and CD8 T cell subtypes in the coronary arteries of patients with KD. These findings indicate that CD8 T cells, NK cells and B cells are recruited into the arterial walls, particularly the coronary arteries, during the acute phase of KD, which may be why the infiltration levels of these cells were reduced in the peripheral blood mononuclear cells of patients in the present study. Thus, an in-depth understanding of the changes and functions of immune cells, particularly B cells and different subtypes of T cells, in patients with KD is important for the diagnosis and early-stage treatment of KD.
In the present study, 6 ferroptosis-related markers, which are closely associated with immunity and metabolism and may serve an important role in the progression of KD, were screened for future research. These markers showed favorable diagnostic performance in the training and validation sets, and their diagnostic values were also evaluated using RT-qPCR in blood samples from Kawasaki patients and controls. A total of three of the FRG markers (MAPK14, SLC2A3 and PGD) were selected as key biomarkers for the diagnosis of KD based on the clinical sample validation data.
MAPK14 (also known as p38α MAPK) is the most highly expressed isomer out of the four isomers of p38 MAPK, and can be activated by a variety of environmental factors and extracellular stimuli, including LPS, pro-inflammatory cytokines (such as IL-1 and TNF-α), chemokines, endotoxins, oxidative stress, hypoxia and heat shock (36). MAPK14 has been proven to serve a key role in normal inflammatory activation and immune responses and represents a valuable pharmaceutical target for treating chronic and acute inflammatory diseases (37). Additionally, MAPK14 is involved in guiding the cellular response to various extracellular stresses and regulating numerous biological processes, including cell proliferation, differentiation, survival and apoptosis (36). Based on the results of the bioinformatics analysis and clinical validation performed in this study, it is hypothesized that MAPK14 may serve as an early diagnostic marker for KD.
SLC2A3 is a member of the glucose transport protein family, also known as glucose transporter 3. Glucose transport proteins have a crucial role in cellular glucose transport. A recent study has shown that SLC2A3 expression is elevated in patients with inflammation and sepsis (38); however, SLC2A3 expression has rarely been studied in KD. Reddy et al (39) found that upon LPS stimulation, sugar uptake was increased and the SLC2A3 expression levels were significantly elevated in murine macrophages. Moreover, Fu et al (40) reported a significant increase in SLC2A3 protein levels in lymphocytes when the immune system was activated. In the present study, an increase in carbohydrate metabolism that was primarily manifested by elevated glucose utilization during the course of infection and inflammation in patients with KD was noted, which may be the cause of the observed elevated SLC2A3 levels. Therefore, SLC2A3 is likely to be involved in the pathogenesis of KD and may be a potential diagnostic marker for this disease.
PGD is involved in the redox reaction of the substrate, phosphogluconate, and is a key enzyme in the pentose phosphate pathway, a major source of intracellular NADPH. An increasing number of studies have revealed that PGD is upregulated in various cancer types, which may be associated with cancer metastasis and poor prognosis, and plays a role in tumor immunotherapy (41,42). However, to the best of our knowledge, whether PGD has a role in KD has not yet been studied. In the present study, the results of the database and clinical validation analysis revealed that PGD was highly expressed in patients with early-stage KD. Therefore, it is hypothesized that PGD may have a notable role in the development of KD. However, further experiments are needed to verify this hypothesis. In short, the 3 biomarkers identified in the present study, could effectively differentiate between KD and healthy controls, either alone or in combination; therefore, they may serve as potential diagnostic markers for this disease.
In the present study, cross-sectional studies were used to identify the most important FRGs in KD. A cross-sectional study is mainly conducted at the same time or over a shorter period of time. It focuses on comparing different variables of different groups and on revealing the effects of different factors on the subjects. A longitudinal study, also called a tracking study, which is a long-term study that tracks changes in diseases, health conditions, and other factors in the same specific group over time. It is more concerned with identifying patterns of change and trends over time, and the factors that lead to these changes. The two research methods have their own advantages, limitations and applications. In the present study, the differences in gene expression between patients with KD and controls were compared to find the FRG markers that play a key role in the early stage of KD. Cross-sectional studies were therefore the most appropriate choice.
Individual variability in gene expression, including FRGs, may exist in patients with KD. This variability can be caused by a variety of factors, including different genetic backgrounds, different causes of the disease and different environmental factors. All of these may lead to individual differences in gene expression in patients with KD. Individual variability makes it difficult to identify consistent FRGs. Certain genes may be significantly up- or down-regulated in some patients but not in others. This narrows the range of candidate genes. However, individual differences are not unique to KD; they are common to numerous diseases. In the present study, after integrative bioinformatics analysis, consistent ferroptosis-related biomarkers were still found in patients with KD. Strategies to improve this problem are usually to increase the sample size and to improve capturing the variability in gene expression through larger group analyses.
The present study still has certain limitations. Firstly, there are very few KD datasets in public databases, and even fewer datasets that meet our research needs and in which patients have not been treated with IVIG as a therapy. The authors were only able to find 3 KD datasets that met the requirements of the present study, and the sample size was relatively small. Small sample sizes may affect the statistical power of a study. This is mainly because small sample sizes may lead to a reduced ability to detect effect size, which can increase the likelihood of false negatives. Therefore, large sample sizes are needed in future studies to increase the generalizability of the study. Secondly, although the diagnostic ability of the marker genes identified by the comprehensive analysis was clinically validated, the clinical sample size was small. Hence, future prospective research with a larger sample size is needed for further verification to obtain more convincing findings. Furthermore, studies on the biological roles and mechanisms of the key genes were lacking. To gain a deeper insight to the exact roles of these biomarkers in the pathogenesis of KD and their associated regulatory mechanisms, future experiments should focus on understanding their functions and signaling pathways. In addition, since the LASSO model also has certain limitations, other machine learning algorithms or multi-model comparisons could be used in future studies to more comprehensively identify potential key biomarkers in KD. Last, a small number of patients who were unsure about the use of medication or who had other underlying medical conditions may not have been excluded from the cohort. As such relevant information was not easy to find in the datasets, the authors were unable to make adjustments during the analysis. Future studies should consider other potential medical conditions or medications that may affect biomarker levels, and find effective adjustment methods in the analysis process.
In conclusion, the infiltration landscapes of immune cells and the expression of immune checkpoint genes in patients with KD were investigated in the present study. Using integrated bioinformatics analyses, 6 important FRGs that are closely associated with KD were identified. Of these genes, MAPK14, SLC2A3 and PGD could effectively discriminate patients with KD from the controls, as demonstrated through clinical validation, and may serve as significant biomarkers for KD diagnosis. These findings not only indicate the importance of FRGs in the early prediction and treatment of KD, but also provide new insights into the pathogenesis underlying this disease.
Supplementary Material
GSVA of the signaling pathways of the ferroptosis-related markers. (A-D) Differences in the Kyoto Encyclopedia of Genes and Genomes pathways identified by GSVA between the high and low expression group of (A) MAPK14, (B) PGD, (C) TLR4 and (D) EPAS1. The T-values shown are from a linear model. The red and green columns indicate the signaling pathways enriched in the high and low gene expression groups, respectively. GSVA, gene set variation analysis.
Ferroptosis-related genes
Prediction of drugs targeting ferroptosis-related markers.
Acknowledgements
The authors would like to thank Mr Dede Xu (Haining Annealing Clinical Testing Laboratory Company, LTD.) for his help in bioinformatics analysis.
Funding
Funding: The present study was supported by the research fund of National Natural Science Foundation of China (grant no. 82101804), the Basic research project of Shenzhen Science and Technology Innovation Program (grant no. JCYJ20220530160015034) and the High-level Medical Team Project in Baoan, Shenzhen (grant no. 202404).
Availability of data and materials
The data generated in the present study may be found in the GEO under accession numbers GSE73461, GSE68004 and GSE18606 or at the following URL: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE73461; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE68004; and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE18606. The data generated in the present study are included in the figures and/or tables of this article.
Authors' contributions
TZ and RY conceived the idea and devised the study. RY performed the bioinformatics analysis and conducted laboratory experiments. SC performed the data analyses and prepared figures and/or tables. XL acquired patient samples and collected patient data. JL conducted the statistical analyses and interpreted the findings. RY and TZ wrote and revised the manuscript. SC and TZ confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
The study procedure was approved (approval. no. LLSC-2023-03-09-04-KS) by the Medical Ethics Committee of Shenzhen Baoan Women's and Children's Hospital of Jinan University (Shenzhen, China). Informed consent was acquired from the participants' legal guardian.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Ramphul K and Mejias SG: Kawasaki disease: A comprehensive review. Arch Med Sci Atheroscler Dis. 3:e41–e45. 2018.PubMed/NCBI View Article : Google Scholar | |
Senzaki H: Long-term outcome of Kawasaki disease. Circulation. 118:2763–2772. 2008.PubMed/NCBI View Article : Google Scholar | |
Newburger JW, Takahashi M and Burns JC: Kawasaki disease. J Am Coll Cardiol. 67:1738–1749. 2016.PubMed/NCBI View Article : Google Scholar | |
McCrindle BW, Rowley AH, Newburger JW, Burns JC, Bolger AF, Gewitz M, Baker AL, Jackson MA, Takahashi M, Shah PB, et al: Diagnosis, treatment, and long-term management of kawasaki disease: A scientific statement for health professionals from the American heart association. Circulation. 135:e927–e999. 2017.PubMed/NCBI View Article : Google Scholar | |
Duan C, Du ZD, Wang Y and Jia LQ: Effect of pravastatin on endothelial dysfunction in children with medium to giant coronary aneurysms due to Kawasaki disease. World J Pediatr. 10:232–237. 2014.PubMed/NCBI View Article : Google Scholar | |
Singh S, Vignesh P and Burgner D: The epidemiology of Kawasaki disease: A global update. Arch Dis Child. 100:1084–1088. 2015.PubMed/NCBI View Article : Google Scholar | |
Kainth R and Shah P: Kawasaki disease: Origins and evolution. Arch Dis Child. 106:413–414. 2021.PubMed/NCBI View Article : Google Scholar | |
Dietz SM, van Stijn D, Burgner D, Levin M, Kuipers IM, Hutten BA and Kuijpers TW: Dissecting Kawasaki disease: A state-of-the-art review. Eur J Pediatr. 176:995–1009. 2017.PubMed/NCBI View Article : Google Scholar | |
Nakamura A, Ikeda K and Hamaoka K: Aetiological significance of infectious stimuli in Kawasaki disease. Front Pediatr. 7(244)2019.PubMed/NCBI View Article : Google Scholar | |
Rypdal M, Rypdal V, Burney JA, Cayan D, Bainto E, Skochko S, Tremoulet AH, Creamean J, Shimizu C, Kim J and Burns JC: Clustering and climate associations of Kawasaki disease in San Diego County suggest environmental triggers. Sci Rep. 8(16140)2018.PubMed/NCBI View Article : Google Scholar | |
Singh S, Jindal AK and Pilania RK: Diagnosis of Kawasaki disease. Int J Rheum Dis. 21:36–44. 2018.PubMed/NCBI View Article : Google Scholar | |
Ng YM, Sung RYT, So LY, Fong NC, Ho MH, Cheng YW, Lee SH, Mak WC, Wong DM, Yam MC, et al: Kawasaki disease in Hong Kong, 1994 to 2000. Hong Kong Med J. 11:331–335. 2005.PubMed/NCBI | |
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021.PubMed/NCBI View Article : Google Scholar | |
Angeli JPF, Shah R, Pratt DA and Conrad M: Ferroptosis inhibition: Mechanisms and opportunities. Trends Pharmacol Sci. 38:489–498. 2017.PubMed/NCBI View Article : Google Scholar | |
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017.PubMed/NCBI View Article : Google Scholar | |
Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW and Kopf M: T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 212:555–568. 2015.PubMed/NCBI View Article : Google Scholar | |
Wu X, Qin K, Iroegbu CD, Xiang K, Peng J, Guo J, Yang J and Fan C: Genetic analysis of potential biomarkers and therapeutic targets in ferroptosis from coronary artery disease. J Cell Mol Med. 26:2177–2190. 2022.PubMed/NCBI View Article : Google Scholar | |
Wen H, Hun M, Zhao M, Han P and He Q: Serum ferritin as a crucial biomarker in the diagnosis and prognosis of intravenous immunoglobulin resistance and coronary artery lesions in Kawasaki disease: A systematic review and meta-analysis. Front Med (Lausanne). 9(941739)2022.PubMed/NCBI View Article : Google Scholar | |
Wright VJ, Herberg JA, Kaforou M, Shimizu C, Eleftherohorinou H, Shailes H, Barendregt AM, Menikou S, Gormley S, Berk M, et al: Diagnosis of Kawasaki disease using a minimal whole-blood gene expression signature. JAMA Pediatr. 172(e182293)2018.PubMed/NCBI View Article : Google Scholar | |
Jaggi P, Mejias A, Xu Z, Yin H, Moore-Clingenpeel M, Smith B, Burns JC, Tremoulet AH, Jordan-Villegas A, Chaussabel D, et al: Whole blood transcriptional profiles as a prognostic tool in complete and incomplete Kawasaki disease. PLoS One. 13(e0197858)2018.PubMed/NCBI View Article : Google Scholar | |
Fury W, Tremoulet AH, Watson VE, Best BM, Shimizu C, Hamilton J, Kanegaye JT, Wei Y, Kao C, Mellis S, et al: Transcript abundance patterns in Kawasaki disease patients with intravenous immunoglobulin resistance. Hum Immunol. 71:865–873. 2010.PubMed/NCBI View Article : Google Scholar | |
Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015.PubMed/NCBI View Article : Google Scholar | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
Li Y, Li Y, Bai Z, Pan J, Wang J and Fang F: Identification of potential transcriptomic markers in developing pediatric sepsis: A weighted gene co-expression network analysis and a case-control validation study. J Transl Med. 15(254)2017.PubMed/NCBI View Article : Google Scholar | |
Hara T, Nakashima Y, Sakai Y, Nishio H, Motomura Y and Yamasaki S: Kawasaki disease: A matter of innate immunity. Clin Exp Immunol. 186:134–143. 2016.PubMed/NCBI View Article : Google Scholar | |
Jackson H, Menikou S, Hamilton S, McArdle A, Shimizu C, Galassini R, Huang H, Kim J, Tremoulet A, Thorne A, et al: Kawasaki disease patient stratification and pathway analysis based on host transcriptomic and proteomic profiles. Int J Mol Sci. 22(5655)2021.PubMed/NCBI View Article : Google Scholar | |
Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, Drummer C IV, Johnson C, Xu K, Jiang X, et al: Vascular endothelial cells and innate immunity. Arterioscler Thromb Vasc Biol. 40:e138–e152. 2020.PubMed/NCBI View Article : Google Scholar | |
Qian G, Xu L, Qin J, Huang H, Zhu L, Tang Y, Li X, Ma J, Ma Y, Ding Y and Lv H: Leukocyte proteomics coupled with serum metabolomics identifies novel biomarkers and abnormal amino acid metabolism in Kawasaki disease. J Proteomics. 239(104183)2021.PubMed/NCBI View Article : Google Scholar | |
Kuijpers TW, Wiegman A, van Lier RA, Roos MT, Wertheim-van Dillen PM, Pinedo S and Ottenkamp J: Kawasaki disease: A maturational defect in immune responsiveness. J Infect Dis. 180:1869–1877. 1999.PubMed/NCBI View Article : Google Scholar | |
Ikeda K, Yamaguchi K, Tanaka T, Mizuno Y, Hijikata A, Ohara O, Takada H, Kusuhara K and Hara T: Unique activation status of peripheral blood mononuclear cells at acute phase of Kawasaki disease. Clin Exp Immunol. 160:246–255. 2010.PubMed/NCBI View Article : Google Scholar | |
Hoang LT, Shimizu C, Ling L, Naim AN, Khor CC, Tremoulet AH, Wright V, Levin M, Hibberd ML and Burns JC: Global gene expression profiling identifies new therapeutic targets in acute Kawasaki disease. Genome Med. 6(541)2014.PubMed/NCBI View Article : Google Scholar | |
Hara T, Yamamura K and Sakai Y: The up-to-date pathophysiology of Kawasaki disease. Clin Transl Immunology. 10(e1284)2021.PubMed/NCBI View Article : Google Scholar | |
Sakurai Y: Autoimmune aspects of Kawasaki disease. J Investig Allergol Clin Immunol. 29:251–261. 2019.PubMed/NCBI View Article : Google Scholar | |
Popper SJ, Shimizu C, Shike H, Kanegaye JT, Newburger JW, Sundel RP, Brown PO, Burns JC and Relman DA: Gene-expression patterns reveal underlying biological processes in Kawasaki disease. Genome Biol. 8(R261)2007.PubMed/NCBI View Article : Google Scholar | |
Xie Z, Huang Y, Li X, Lun Y, Li X, He Y, Wu S, Wang S, Sun J and Zhang J: Atlas of circulating immune cells in Kawasaki disease. Int Immunopharmacol. 102(108396)2022.PubMed/NCBI View Article : Google Scholar | |
Cargnello M and Roux PP: Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 75:50–83. 2011.PubMed/NCBI View Article : Google Scholar | |
Zheng T, Zhang B, Chen C, Ma J, Meng D, Huang J, Hu R, Liu X, Otsu K, Liu AC, et al: Protein kinase p38α signaling in dendritic cells regulates colon inflammation and tumorigenesis. Proc Natl Acad Sci USA. 115:E12313–E12322. 2018.PubMed/NCBI View Article : Google Scholar | |
Yan R and Zhou T: Identification of key biomarkers in neonatal sepsis by integrated bioinformatics analysis and clinical validation. Heliyon. 8(e11634)2022.PubMed/NCBI View Article : Google Scholar | |
Reddy ABM, Srivastava SK and Ramana KV: Aldose reductase inhibition prevents lipopolysaccharide-induced glucose uptake and glucose transporter 3 expression in RAW264.7 macrophages. Int J Biochem Cell Biol. 42:1039–1045. 2010.PubMed/NCBI View Article : Google Scholar | |
Fu Y, Maianu L, Melbert BR and Garvey WT: Facilitative glucose transporter gene expression in human lymphocytes, monocytes, and macrophages: A role for GLUT isoforms 1, 3, and 5 in the immune response and foam cell formation. Blood Cells Mol Dis. 32:182–190. 2004.PubMed/NCBI View Article : Google Scholar | |
Khan GB, Qasim M, Rasul A, Ashfaq UA and Alnuqaydan AM: Identification of Lignan compounds as new 6-phosphogluconate dehydrogenase inhibitors for lung cancer. Metabolites. 13(34)2022.PubMed/NCBI View Article : Google Scholar | |
Liu T, Qi J, Wu H, Wang L, Zhu L, Qin C, Zhang J and Zhu Q: Phosphogluconate dehydrogenase is a predictive biomarker for immunotherapy in hepatocellular carcinoma. Front Oncol. 12(993503)2022.PubMed/NCBI View Article : Google Scholar |