Next Article in Journal
Induction of Sustained Remissions Associated with Immune Activation by Idelalisib in Patients with Follicular Lymphoma
Previous Article in Journal
A Review of KSHV/HHV8-Associated Neoplasms and Related Lymphoproliferative Lesions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Landscape of Kawasaki Disease: An Update

Paediatric Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh 160012, India
*
Authors to whom correspondence should be addressed.
Lymphatics 2025, 3(3), 21; https://doi.org/10.3390/lymphatics3030021
Submission received: 22 April 2025 / Revised: 16 June 2025 / Accepted: 12 July 2025 / Published: 20 July 2025

Abstract

Kawasaki disease (KD), first identified in 1967 by Dr. Tomisaku Kawasaki, is an acute, self-limited vasculitis and remains the leading cause of acquired heart disease in children worldwide, particularly affecting those under the age of five. Clinically, it presents with persistent fever, mucocutaneous inflammation, skin rashes, and lymphadenopathy, with a marked tendency to involve the coronary arteries, potentially leading to serious complications such as coronary artery aneurysms. Despite extensive research spanning more than five decades, the precise etiology of KD remains unclear. However, accumulating evidence supports the significant role of genetic predisposition, highlighting the contribution of inherited factors in modulating immune responses and influencing disease susceptibility and severity. Emerging evidence highlights genetic susceptibility as pivotal, with genome-wide studies identifying polymorphisms in immune-related genes, such as ITPKC, CASP3, BLK, CD40, and ORAI1, which modulate disease risk and coronary complications. Epigenetic mechanisms, including DNA methylation and non-coding RNAs, bridge the gap between genetic and environmental factors, regulating immune responses and endothelial activation. Furthermore, emerging insights into autophagy-related processes provide a deeper understanding of the molecular mechanisms underlying the disease. This review aims to explore the current knowledge on the genetic landscape of KD, examine how these findings contribute to our understanding of its pathophysiology, and investigate the potential for genetically targeted therapeutic strategies in the future.

1. Introduction

Kawasaki disease (KD) is an acute, self-limiting vasculitis that primarily affects children, especially those of East Asian descent. It is now considered to be the most common cause of acquired heart disease in children worldwide [1]. Due to the absence of a single causative agent or pathognomonic biomarker, diagnosis relies on a constellation of clinical features, including prolonged fever, bilateral conjunctival injection, mucosal changes (e.g., strawberry tongue), polymorphous rash, extremity changes, and unilateral cervical lymphadenopathy. Several children with KD may not fulfil the diagnostic criteria; they are said to have incomplete/atypical KD. The proportion of children with incomplete/atypical KD may be as high as 30–50% in large cohorts [2].
A hallmark of KD is the inflammation of medium-sized arteries, especially those involving the coronary arteries, which can lead to thrombosis, aneurysms, and long-term cardiac complications. Early detection of KD is crucial, as most children respond to treatment. However, delays in diagnosis or the initiation of appropriate therapy can result in the development of coronary artery abnormalities (CAAs) in 15–25% of cases [3].
Despite advances, the pathophysiology of KD remains unclear. Current research suggests an abnormal immune response triggered by environmental or infectious factors in genetically predisposed individuals [4,5,6]. The seasonal nature of KD and its occurrence primarily in infants and young children suggests an infectious trigger, though no specific pathogen has yet been identified. Several lines of evidence suggest that genetics plays a crucial role in the susceptibility to KD. Family studies indicate that KD may cluster in certain families, with siblings of affected children at a significantly increased risk [7,8,9]. In addition, studies of twin populations have suggested a higher concordance for the disease in monozygotic twins compared to dizygotic twins, further supporting the genetic basis of KD [10]. To uncover disease-associated genes in KD, two main approaches have been employed: candidate gene studies and genome-wide association studies (GWASs). Both approaches involve genotyping genetic differences, primarily single-nucleotide polymorphisms (SNPs), in cases and controls [11]. Over the past two decades, numerous genetic association studies have identified potential risk loci for KD. The most significant findings have emerged from GWASs, which have provided a deeper understanding of the genetic underpinnings of the disease. This article focuses on SNPs in genes associated with KD susceptibility and CAAs, aiming to summarize current findings and guide future genetic research in KD.

2. Genetic Susceptibility and the Role of Host Genomic Variability

2.1. Immune System-Related Genes

The pathogenesis of KD is also known to involve a hyperactive immune response to an environmental or infectious trigger in genetically susceptible individuals [4,6]. The activation of both innate and adaptive immune responses leads to widespread inflammation, particularly in medium-sized arteries, with a predilection for the coronary arteries.

2.1.1. MHC Class I and II Genes

Glycoproteins on the cell surface, known as major histocompatibility complex (MHC) molecules, are essential to the immune system as they present peptides to the CD4+ T cells’ antigen receptor. Antigen presentation is crucial for maintaining self-tolerance and controlling immune responses. In fact, during CD4+ T cell maturation in the thymus, MHC expression guides both positive and negative selection mechanisms that influence the specificity of the T cell receptor repertoire. Studies have reported the significant role played by the MHC region in the susceptibility to autoimmunity, transplant acceptance, and infection resistance [12]. Despite this, the association between MHC and KD has not been well studied.
However, the HLA (human leukocyte antigen) region, particularly HLA-DRB1, has been known to be associated with the risk of developing KD, suggesting that T cell-mediated immune responses may play a key role in disease development. Several studies have examined the role of the HLA region in KD, with varying results. For instance, in a GWAS conducted on the Japanese population, HLA-DQB2-DOB at the rs2857151 locus was found to be significantly associated with the risk of KD [13]. It has been observed that specific HLA genotypes are related to certain populations in the context of susceptibility to KD. HLA types B5, B44, Bw51, DR2, and DRB3*0301 are reportedly associated with KD in Caucasians [14,15]; B54, Bw15, and Bw35 are associated with KD in Japanese individuals [16]; and Bw51 is associated with KD in Israeli individuals [17].
In contrast to the Japanese, the Korean population was found to have a higher prevalence of HLA-B35, B75, and Cw09 alleles, making them more susceptible to KD [18]. Nonetheless, the correlation with specific HLA-B alleles, which have been documented in some populations, has not been reliably validated in others [19]. Huang et al. reported the link between the MHC class I chain-related gene A (MICA) gene in a Southern Chinese population and suggested that the allele A4 has a protective role against developing CAA in children with KD [20].
Despite numerous research studies, the precise HLA allele or haplotype that confers vulnerability to KD has yet to be consistently identified. This inconsistency and variability across the population make it imperative for future researchers to study and better understand the role of HLA and MHC regions in KD.

2.1.2. FCGR2A

The term “Fc receptor” (abbreviated as “FcR”) refers to a transmembrane glycoprotein that binds to the Fc segment of IgG and is a member of the immunoprotein superfamily. The FCGR2A gene encodes the Fc gamma receptor IIa (FcγRIIa), which is expressed on the surface of various immune cells, including macrophages, neutrophils, and platelets, that bind to the Fc region of antibodies. When the Fc region of an antibody binds to FcR on an immune cell, it can activate a variety of cellular effector molecules and functions. Various GWASs have identified SNPs that result in amino acid substitutions and may increase susceptibility to KD across diverse ethnicities; however, the effect is more pronounced in Europeans and Asians. A plethora of studies have reported that a substitution of arginine for histidine at position 131 (H131R) in the FCGR2A gene results in an altered binding affinity of FcγRIIa, which in turn leads to an altered immune response. In a study, it was observed that individuals with the AA genotype demonstrated a higher efficacy in IgG2-mediated phagocytosis than those with the GG genotype [21].
According to Khor and colleagues, the A allele of the polymorphism rs1801274 is associated with an increased risk of KD. Onouchi and colleagues reached a similar conclusion [7]. Furthermore, a comparison of genotypes revealed that individuals with the AG + GG genotypes exhibited a trend toward a lower odds ratio for KD compared to those with the AA genotype. However, this difference did not reach statistical significance [16]. In contrast, Kwon and colleagues [21] found no substantial link between polymorphism and susceptibility to KD in females; other studies have similarly reported no apparent effect of the polymorphism rs1801274 on the overall occurrence of KD [22,23,24].
A recent meta-analysis revealed that the CD32a polymorphism rs1801274 allele A and the AA genotype could significantly alter susceptibility to KD, especially in Asians. In particular, the polymorphism increased the disease incidence in every subgroup under the corresponding genetic models following stratification analyses by ethnicity and control source, respectively, in addition to the total analysis under all five comparisons of AA vs. GG, AA + GA vs. GG, AA vs. GG + GA, A vs. G, and GA vs. GG [25,26]. Overall, the role of FCGR2A in KD remains unclear, and further studies are necessary to understand the relationship between this gene and the disease fully.

2.1.3. CD40/CD40L Pathway

CD40 is a co-stimulatory receptor expressed on antigen-presenting cells (APCs) like dendritic cells, macrophages, and B cells, while CD40L (also known as CD154) is primarily expressed on activated T helper cells. Owing to its involvement in B cell activation, T cell differentiation, and antibody production, it has a significant role in the pathogenesis of various autoimmune and inflammatory diseases. The excessive activation of immune cells, leading to inflammation and vascular injury, particularly in the coronary arteries in KD patients, has also been found to be an effect of the CD40/CD40L pathway.
Studies indicate that a higher expression of CD40L on CD4+ T cells and platelets is significantly associated with the occurrence of CAL in KD patients [27]. A decreased expression of CD40L after treatment with intravenous immunoglobulin (IVIg) suggests the modulation of this pathway and may mitigate the vascular damage associated with KD. Onouchi et al. reported that the presence of specific genetic polymorphisms (IVS4+121 A>G) in the CD40L gene has also been linked to an increased risk of CAL, particularly in male patients [28]. In contrast, Huang et al. found that a similar variant had no impact on Taiwanese children [29]. Nevertheless, an independent cohort of Taiwanese patients with rs1535045 was found to have a higher risk of developing CAA [30]. More recently, a study from Chandigarh, North India, reported no significant variation in the expression of CD40 and CD40 SNPs (rs4810485 and rs153045) between KD patients and healthy controls [31].

2.1.4. IL-1β (Interleukin-1 Beta)

IL-1β plays a crucial role in the inflammatory process by promoting the recruitment of immune cells, the production of other cytokines, and the activation of endothelial cells, which contributes to the initiation and resolution of inflammation. IL-1β not only promotes the inflammatory cascade but also affects endothelial cells and vascular smooth muscle cells, potentially leading to vascular injury and the development of coronary artery aneurysms, which are some of the most severe complications of KD.
Several single-nucleotide polymorphisms (SNPs) have been identified in the IL-1β gene that may influence its expression and the subsequent inflammatory response [32]. One of the most studied polymorphisms is the IL-1β-511 C/T polymorphism, located in the promoter region of the gene. This variant has been associated with increased IL-1β expression, which may lead to a more vigorous inflammatory response in susceptible individuals. Studies have shown that the T allele of the IL-1β-511 polymorphism is linked to higher levels of IL-1β production, which may contribute to the exaggerated inflammatory response observed in KD. Similar to the -511 C/T polymorphism, the T allele of the IL-1β -31 polymorphism has been linked to increased IL-1β expression and an enhanced inflammatory response [33]. Weng et al. demonstrated that the IL-1β (511 TT and IL-1β-31 CC) genotype polymorphism is associated with the failure of early intravenous immunoglobulin treatment in Taiwanese children with KD [34]. Furthermore, IL-1β has been implicated in the development of CAAs due to its ability to promote endothelial cell activation, vascular smooth muscle cell proliferation, and the recruitment of inflammatory cells to the vascular wall. For instance, the rs16944 GG and rs1143627 AA genotypes of the IL-1β gene may substantially increase the risk of CAL development in children under 12 months [35].

2.1.5. BLK

The BLK gene (B-lymphoid tyrosine kinase) phosphorylates the ITAM (immunoreceptor tyrosine-based activation motif) residues of Igα and Igβ molecules, playing a critical role in B cell receptor signaling and influencing B cell development, proliferation, differentiation, and immune tolerance. Reduced BLK expression in peripheral B cells during the acute phase of KD may impair antibody production and promote abnormal immune complex formation, which is implicated in the vascular inflammation characteristic of KD. Recently, Chen et al. reported a significant association between rs2254546, located in the intergenic region between the FAM167A and BLK genes, and susceptibility to KD [36]. Similar findings were reported earlier [37], thereby suggesting that a genetically determined differential in the rate or type of antibody generated in response to an infection is implicated in KD [38]. Furthermore, BLK variants underscore the significance of B cell-mediated immunity in KD pathogenesis, suggesting a genetic basis for the disease that involves both humoral and cellular immune mechanisms.

2.1.6. TNF (Tumour Necrosis Factor)

Polymorphisms in the tumor necrosis factor (TNF) gene, particularly the −308 G/A variant, have been extensively studied for their association with KD susceptibility. Studies have demonstrated that individuals with the A allele are more susceptible and may have an increased predisposition to KD and contribute to vascular complications such as arterial stiffness and intima–media thickening [39,40,41,42]. The association between the TNF-α-308 polymorphism and KD risk vary by ethnicity, with some studies and meta-analyses indicating no significant link [43]. These disparities are most likely caused by genetic variation, variances in sample size, and demographic diversity.

2.2. Calcium Signaling Pathway

2.2.1. ITPKC

Inositol-1,4,5-triphosphate (IP3) is an essential second messenger that causes the sarcoplasmic and endoplasmic reticulum to release calcium (Ca). Inositol-1,4,5-trisphosphate kinase C (ITPKC), in conjunction with other kinases, phosphorylates IP3, leading to the production of inositol-1,3,4,5-tetrakisphosphate (IP4), which inhibits Ca release through IP3. Signaling cascades that activate T cells and immune cells are triggered by the Ca influx, which in turn initiates the Ca2+/NFAT pathway and may result in autoimmune reactions or other immunological diseases. Furthermore, any mutation in the ITPKC gene would result in an imbalance in the T cell activation pathway. Among others, the ITPKC gene has been studied extensively for its role in the pathogenesis of KD. In a landmark study, Onouchi et al. reported that a functional polymorphism (rs28493229) in the ITPKC gene is substantially linked to an elevated incidence of CAAs and a vulnerability to KD in children from both Japan and the United States [44]. Following this discovery, several studies have reported a significant association between this variant and KD in the Indian, Chinese, and Japanese populations [45,46]. The mechanistic link is attributed to an alteration in the splicing efficiency of intron 1, resulting in reduced transcription of ITPKC mRNA. The significance of the CG + GG genotype of rs2290692 in the ITPKC gene was highlighted by a meta-analysis in Indian children, which also supported this connection [47]. Further evidence of the impact of this genetic polymorphism on the development and prognosis of KD comes from studies conducted in Korean children [48]. Similar findings were strongly reinforced by a recent meta-analysis comprising 2771 cases and 5357 controls from 10 different studies across various genetic models [49]. The findings from all the studies suggest that, compared to people without the allele, those who carry the risk allele for rs28493229 have a 52% higher chance of developing KD. While multiple studies [44,45,46,47,48,50] have reported an association between rs28493229 and KD susceptibility, inconsistencies in methodology, limited statistical power, and lack of replication across diverse cohorts undermine the robustness of these results. Natividad and colleagues, on the other hand, reported that a superantigen could play a role in KD, while the HLA-DRB1 exon 2, the TNF-α-308 region, and the ITPKC SNP rs28493229 may not be linked to the disease [51]. Therefore, comprehensive subgroup analyses and replication studies are required to substantiate this genetic association.
In contrast to the abovementioned variant, the rs7251246 polymorphism of the ITPKC gene did not show any significant association with KD susceptibility. However, there was an ambiguity in the data obtained from other studies. For instance, the first study examining the association between the severity of KD and the SNP rs7251246 in the ITPKC gene revealed a strong link between the development of CAL and the SNP. A haplotype analysis further strengthened these findings [52]. Recently, Liu et al. reported that despite not being substantially associated with KD susceptibility, the ITPKC rs7251246 T > C polymorphism was strongly associated with CAA risk in children with KD. Additionally, males with the CT/TT genotype were less susceptible to thrombosis [53].

2.2.2. ORAI1

ORAI1 (calcium release-activated calcium modulator 1) is a membrane-bound Ca2+ channel protein encoded by the ORAI1 gene that is involved in the Ca2+–calcineurin–NFAT signaling pathway. The role of ORAI1 gene polymorphisms in KD was first investigated in 2011 by Lee et al., who examined five tagging SNPs in Taiwanese populations but found no significant associations with KD susceptibility, CAL, or IVIG resistance [54]. While Lee et al. pioneered the investigation of the ORAI1 gene in KD, Onouchi et al. provided the first evidence of genetic associations, highlighting ORAI1’s potential role in KD pathogenesis through calcium channel regulation [55]. They reported that the risk allele frequency of rs3741596 was 20 times higher in Japanese populations compared to Europeans. Moreover, the risk allele of rs3741596 (G), as documented in the 1000 Genomes Project, is regarded as an ancestral allele of this SNP; nonetheless, it is nearly nonexistent in gene pools in groups other than East Asians and people of African heritage. Yet, given the conservation of the LD pattern between rs3741596 and other variants, it is quite likely that the rs3741596 G allele descended from a founder haplotype.
Recently, a whole-genome sequencing analysis on a patient with a presumptive diagnosis of KD revealed two single-nucleotide variants (SNVs) in disease susceptibility genes in Japanese KD patients: ORAI1 (rs3741596) and BLK (rs2254546). The presence of these variants was found to be linked to the refractory nature of the case [38]. The results were corroborated by another study that found a gain-of-function mutation in the ORAI1 gene and a missense mutation with a minor allele frequency of <0.001 that was strongly linked to KD in a collapsing method analysis. Additional evidence suggested that these differences could lead to an upregulation of the Ca/NFAT pathway, thereby increasing susceptibility to KD [56].

2.2.3. CASP3

The role of CASP3 gene polymorphisms in KD was first investigated by Onouchi et al. in 2010 via a genome-wide association study. They identified the rs72689236 variant in the 5′ untranslated region of the CASP3 gene as a key susceptibility factor in both Japanese and US populations of European ancestry [57]. This G-to-A substitution disrupts the binding of the nuclear factor of activated T cells (NFAT), thereby impairing the regulation of T cell activation and increasing the susceptibility to KD. Furthermore, the A allele of rs72689236 abolishes NFAT binding, resulting in reduced CASP3 expression in immune cells. This alters apoptosis regulation and promotes immune hyperactivation, the cornerstone of KD pathogenesis [58]. These findings were then confirmed by meta-analyses, which reported that the A allele increased KD susceptibility (OR = 1.34–1.44) and the risk of CAL (OR = 1.51–1.59) [59]. In Taiwanese cohorts, the A allele showed a borderline association with KD susceptibility (p = 0.053–0.057) but a significant association with aneurysm formation (p = 0.009) [60]. However, no significant association was found between IVIG resistance and the other variables. In a recent study from India, individuals with the CT genotype of rs113420705 were found to be more susceptible to KD. They also ascertained that the overexpression of the C allele was associated with a greater risk of developing CAAs [61]. While ITPKC SNPs (e.g., rs28493229) influence KD susceptibility and CAA development, CASP3 variants specifically affect disease onset but not treatment outcomes [58]. A later study [57] suggested that CASP3 and calcium-signaling genes, such as ORAI1, may interact to amplify immune dysregulation in KD.

2.2.4. STIM1

STIM1 and ORAI1 form the core components of the store-operated calcium (SOC) influx pathway, which is essential for T cell activation and immune responses. A specific polymorphism in the STIM1 gene (rs2304891) was associated with coronary artery dilation in KD patients, suggesting that variations in the STIM1 gene may influence the vascular complications of the disease. However, this polymorphism was not linked to resistance to IVIG treatment [62]. A few studies have also investigated the effect of rs1561876 on KD susceptibility. Additionally, this variant is predicted to influence transcription factor binding site activity and miRNA binding site interactions, offering potential directions for future investigation [63,64]. The involvement of STIM1 gene polymorphisms in KD patients further suggests that calcium signaling and immune cell activation pathways, including the Ca2+/NFAT pathway, play critical roles in KD pathophysiology.

2.2.5. SLC8A1

SLC8A1 (solute carrier family 8 member A1), also known as NCX1 (sodium/calcium exchanger 1), encodes a sodium/calcium exchanger involved in maintaining intracellular calcium homeostasis. Dysregulated calcium signaling through SLC8A1 can affect immune cell activation, vascular tone, and cardiac function, processes that are implicated in the pathogenesis of KD. Multiple single-nucleotide polymorphisms (SNPs) in the SLC8A1 gene, particularly rs13017968, have been consistently associated with an increased susceptibility to KD and a higher risk of coronary artery abnormalities in diverse populations, including European and Japanese cohorts. Patients homozygous for the A (risk) allele of rs13017968 showed significantly higher rates of coronary artery aneurysms and dilation (p = 0.029) [65]. A recent study evaluated the polymorphism rs13017968 in the SLC8A1 gene and hematological parameters as predictors of KD and observed no significant difference between the KD cases and controls, unlike other studies, which could be attributed to population-specific variations. However, a trend emerged suggesting that patients with the TT genotype of rs13017968 may have a higher tendency to develop CAA compared to those with the GG genotype (Table 1).

2.3. TGF-β Pathway

Transforming growth factor-beta (TGF-β) is a multifunctional cytokine that is essential for orchestrating the balance between cell growth, differentiation, immune responses, and tissue remodeling, making it fundamental to both normal physiology and disease [69]. Genetic polymorphisms in key genes of this pathway, including TGFB2, TGFBR2, and SMAD3, have been consistently associated with increased KD susceptibility, coronary artery aneurysm formation, and aortic root dilatation in multiple populations of mainly European descent [69]. Notably, an SMAD3 haplotype and the intronic SNP rs4776338 showed strong associations with KD susceptibility [70]. Transcript and protein analyses revealed dynamic changes in TGF-β pathway gene expression and TGF-β2 plasma levels during the disease course, underscoring its involvement in immune activation and cardiovascular remodeling in KD. While some studies in Asian populations confirmed SMAD3 polymorphisms’ link to KD susceptibility, associations with coronary artery lesions and IVIG resistance varied. Overall, these findings highlighted the TGF-β pathway as a key genetic and molecular contributor to KD pathogenesis and a potential target for therapeutic intervention. Furthermore, a two-locus model, wherein the combined effect of SNPs in ADAM17 and other TGF-β signaling pathway genes, including TGFβ2 and SMAD3, was studied, suggesting that they had a greater impact on KD phenotypes than single SNPs [71].

2.4. Autophagy Pathway

The dysregulation of autophagy has been implicated in aberrant immune responses and endothelial dysfunction, both of which are central to the pathophysiology of KD. Comprehensive bioinformatics analyses identified a set of 20 autophagy-related genes (ARGs) with altered expression in KD, highlighting the central role of autophagy and macroautophagy pathways in the disease’s pathogenesis. Key genes such as WIPI1 and GBA were significantly upregulated in KD patients, suggesting their potential roles as biomarkers and therapeutic targets [72]. These genes are involved in crucial autophagic processes, including autophagosome formation and lysosomal degradation, which are essential for maintaining immune homeostasis and regulating inflammation. Studies have demonstrated that impaired autophagy and mitophagy exacerbate vascular inflammation and damage in KD, contributing to the development of coronary artery lesions [73]. In KD, altered expression or polymorphisms in these genes may disrupt normal autophagic flux, leading to excessive or dysregulated inflammation that contributes to vascular endothelial injury and the coronary artery lesions characteristic of the disease. Moreover, the expression levels of autophagy genes, such as ATG16L1, were altered in KD patients, particularly those with coronary artery lesions, indicating a protective role of autophagy in mitigating vascular injury [74]. Additionally, the interplay between ARG polymorphisms and immune cell infiltration, especially involving CD8+ T cells and neutrophils, further underscores the immunomodulatory effect of autophagy pathways in KD. Furthermore, the expression levels of autophagy markers such as ATG16L1, BECN1, and LC3II were significantly lower in IVIG-resistant KD patients, and these markers served as independent risk factors for predicting resistance to intravenous immunoglobulin therapy [75].

2.5. Miscellaneous Genes

Additionally, rare variants in genes associated with inflammatory regulation and vascular biology, such as PLCG2, RIG-I-like receptors, and TGFB2, suggest that diverse pathways influence disease pathology beyond classical immune activation. A groundbreaking family-based whole-genome sequencing study revealed a Toll-like receptor gene variant (TLR6) associated with fungal antigen recognition, suggesting that pathogen–host interactions contribute to susceptibility. Distinct genetic loci have also been linked to CAAs, including variants near MMP12 and FGFR2, which affect vascular remodeling and integrity. These findings highlight a dual genetic architecture: one set of variants increases general KD susceptibility through exaggerated immune responses to infections, while another causes aneurysm formation via vascular wall destabilization (Table 2). Epigenetic modifications, including DNA methylation changes in regulatory regions of immune-related genes, are also being explored for their role in modulating disease onset and progression [76]. These emerging insights highlight the polygenic and multifactorial nature of KD pathogenesis, underscoring the need for further studies that integrate multi-omics approaches—combining genomics, transcriptomics, and epigenomics—to unravel the intricate genetic networks involved. Furthermore, the overlap between KD-associated genes and those involved in multisystem inflammatory syndrome in children (MIS-C) post-COVID-19 further emphasizes shared pathways in immune dysregulation [73]. Understanding these lesser-known genetic contributors not only enhances the biological understanding of KD but also opens the door for the development of more precise diagnostic biomarkers and targeted therapies, ultimately improving patient outcomes and paving the way for personalized medical strategies in pediatric vasculitis. As the genetic landscape continues to unfold, the potential for earlier diagnoses and more effective interventions in KD increases.

2.6. Host Defense and Microbial Interactions

KD has been hypothesized to result from an abnormal immune response to an infectious agent in genetically predisposed individuals. Although no specific pathogen has been definitively linked to KD, the seasonal patterns and clustering of cases support an infectious etiology, likely involving common viral or bacterial agents that initiate an exaggerated immune response. The innate immune system, particularly components that recognize pathogen-associated molecular patterns (PAMPs), is central to this response. Toll-like receptors (TLRs), particularly TLR2, TLR4, and TLR9, have been implicated in KD, as they mediate the recognition of bacterial and viral components, leading to the production of pro-inflammatory cytokines. Polymorphisms in these TLR genes may modulate host susceptibility by altering signaling pathways that regulate inflammation [96]. Another key player is the NLRP3 inflammasome, which regulates the maturation of IL-1β, a cytokine strongly linked to vascular inflammation in KD. Variants in NLRP3 and other inflammasome-related genes can enhance this response, potentially contributing to coronary artery complications [97]. A study has investigated the role of the NLRP3 inflammasome in a murine model of KD-like vasculitis induced by the water-soluble fraction of Candida albicans (CAWS) and concluded that NLRP3 inflammasome-driven IL-1 production is necessary for vasculitis development. The same study also discovered that both the priming and activation of the NLRP3 inflammasome were mediated by the Dectin-2/Syk/JNK/NF-B pathway and the Dectin-2/Syk/JNK/mitochondrial reactive oxygen species (mtROS) pathway, respectively [98]. In a global gene expression study, the whole-blood transcription profiles of KD patients in the acute (pre-treatment) and convalescent phases were examined, revealing a significant upregulation in the mRNA levels of TIFA, NLRP3, CASP1, CASP4, CASP5, and IL1B. The findings from this study again highlight the significance of the IL-1 signaling pathway, as well as a prominent signature of innate immunity and cell migration, in the acute phase of the illness [99]. These findings thus shed light on the role and mechanism of the NLRP3 inflammasome in the pathophysiology of KD, suggesting that the NLRP3 inflammasome may be a therapeutic target. Additionally, genes such as IFIH1 (which encodes MDA5, a viral RNA sensor), RIG-I, and OAS1 are part of the host’s antiviral defense. They may affect the severity of KD depending on the individual’s response to viral infections [2]. The interplay between these genetic factors and microbial agents may trigger an uncontrolled inflammatory cascade, ultimately resulting in the vascular injury characteristic of KD.

2.7. Impact of Genetic Variants on KD Susceptibility, CAAs, IVIg Resistance, and BCG Site Reactivation

A wide array of genes has been associated with the pathogenesis and clinical manifestations of KD, including susceptibility, CAAs, IVIg resistance, and BCG site reactivation (Table 1). Key immune-related genes such as ITPKC, CASP3, FCGR2A, CD40, and IL-1β have been consistently linked to KD susceptibility. For instance, polymorphisms in ITPKC (rs28493229) and CASP3 (rs113420705) predispose individuals to KD and correlate with IVIg resistance and CAAs. The FCGR2A (rs1801274) variant modulates phagocytosis and inflammatory response, influencing KD risk and CAA development. IL-1β polymorphisms are associated with increased inflammatory cytokine production and resistance to IVIg. Genes such as CD40 and CD40L further influence endothelial inflammation and immune activation, thereby affecting coronary outcomes. Additionally, BCG reactivation—a classical yet underrecognized clinical sign in KD—has been genetically linked to ITPKC and SLC11A1 polymorphisms. This phenomenon, most common in infants and young children, indicates hyperresponsiveness of innate immunity and is considered a surrogate marker of immune dysregulation in genetically susceptible individuals.

2.8. Gene–Environment Interactions

Although genetic susceptibility plays a significant role in KD, environmental triggers, such as viral or bacterial infections, are believed to be crucial in initiating the disease in genetically susceptible individuals. The identification of gene–environment interactions is a growing area of research. For example, it has been proposed that an infectious agent may trigger a dysregulated immune response in individuals with specific genetic predispositions, leading to the development of KD [4,5,6]. Moreover, geographic variations in KD incidence suggest that environmental factors, such as exposure to specific pathogens, may modulate the expression of genetic risk factors.

2.9. Advances in Genetic Testing and Personalized Medicine

The identification of genetic variants associated with KD has significant clinical implications. Understanding an individual’s genetic predisposition could help predict disease severity, make treatment decisions, and potentially identify those at higher risk for CAAs. For instance, genetic screening may allow for the identification of patients at higher risk for developing aneurysms, enabling early interventions with more aggressive treatment strategies, such as IVIG and corticosteroid therapy. Standard therapy (IVIG + aspirin) is effective in most patients, but ~10–20% are IVIG-resistant and have a higher risk of complications. Given the heterogeneity of immune responses in KD, personalized immune-modulating therapy offers an opportunity to optimize treatment efficacy and safety. Advances in biomarker discovery and immunophenotyping may enable the stratification of patients to receive targeted immunosuppressive or immunomodulatory interventions, such as corticosteroids, TNF-α inhibitors, IL-1 blockers, or novel biologics.

3. Future Directions

A. Functional Genomics: Understanding how identified genetic variants contribute to KD pathogenesis is essential for developing targeted therapies. Functional studies that investigate the molecular mechanisms through which these genes influence immune activation, inflammation, and cardiovascular outcomes are critical to advancing our understanding of the disease.
B. Longitudinal Cohorts and Genomic Databases: Large-scale, longitudinal cohort studies that integrate genomic data with clinical outcomes are necessary to better elucidate the role of genetics in KD. These studies could help identify biomarkers predicting disease course and treatment response.
C. Therapeutic Implications: As the genetic landscape of KD becomes clearer, targeted therapies that modulate the immune response or address specific genetic defects may offer new avenues to better treatment. Personalized medicine, guided by genetic profiles, may enhance the effectiveness of interventions and reduce the risk of long-term cardiovascular complications.

4. Conclusions

KD remains a major challenge in pediatric healthcare, with its complex pathogenesis involving genetic, immune, and environmental factors. Advances in genetic research have provided significant insights into the disease’s molecular basis, particularly regarding immune system regulation and host defense mechanisms. While much progress has been made, further studies are necessary to understand the intricate genetic networks involved in KD fully. As our knowledge expands, there is hope that genetic-based approaches will not only improve diagnosis and risk stratification but also pave the way for more effective, personalized therapies for KD patients.

Author Contributions

Design, conception of the study: T.G., S.S. (Saniya Sharma), R.K.P. and S.S. (Surjit Singh); Data collection and review: K.J., S.C., M.S., A.K., M.R., U.A., and V.T.; Draft preparation: T.G., S.S. (Saniya Sharma), R.K.P. and K.J.; Critical review of the manuscript for important intellectual content: M.D., V.P., A.R., and R.K.P.; Final approval of the version to be published: S.S. (Saniya Sharma), S.S. (Surjit Singh) and A.R. All authors have read and agreed to the published version of the manuscript.

Funding

Rakesh Kumar Pilania received a research grant from the Indian Council of Medical Research, New Delhi, India, vide Grant ID: IIRP-2023-0409. However, the funding agency had no role in the preparation of the manuscript or its final approval.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no competing interests. The authors have no relevant affiliations or financial involvement with any organizations or entities with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimonies, grants or patents received or pending, and royalties.

References

  1. Pilania, R.K.; Tremoulet, A.H.; Prinja, S.; Dahdah, N.; Singh, S. Kawasaki disease: The most common cause of acquired heart disease among children globally. Cardiol. Young 2025, 35, 441–443. [Google Scholar] [CrossRef] [PubMed]
  2. Jone, P.-N.; Tremoulet, A.; Choueiter, N.; Dominguez, S.R.; Harahsheh, A.S.; Mitani, Y.; Zimmerman, M.; Lin, M.-T.; Friedman, K.G. Update on Diagnosis and Management of Kawasaki Disease: A Scientific Statement From the American Heart Association. Circulation 2024, 150, e481–e500. [Google Scholar] [CrossRef] [PubMed]
  3. Kuo, H.-C. Diagnosis, Progress, and Treatment Update of Kawasaki Disease. Int. J. Mol. Sci. 2023, 24, 13948. [Google Scholar] [CrossRef]
  4. Rowley, A.H.; Shulman, S.T. The Epidemiology and Pathogenesis of Kawasaki Disease. Front. Pediatr. 2018, 6, 374. [Google Scholar] [CrossRef]
  5. Rodó, X.; Curcoll, R.; Robinson, M.; Ballester, J.; Burns, J.C.; Cayan, D.R.; Lipkin, W.I.; Williams, B.L.; Couto-Rodriguez, M.; Nakamura, Y.; et al. Tropospheric winds from northeastern China carry the etiologic agent of Kawasaki disease from its source to Japan. Proc. Natl. Acad. Sci. USA 2014, 111, 7952–7957. [Google Scholar] [CrossRef]
  6. Noval Rivas, M.; Arditi, M. Kawasaki disease: Pathophysiology and insights from mouse models. Nat. Rev. Rheumatol. 2020, 16, 391–405. [Google Scholar] [CrossRef]
  7. Onouchi, Y. The genetics of Kawasaki disease. Int. J. Rheum. Dis. 2018, 21, 26–30. [Google Scholar] [CrossRef]
  8. Fujita, Y.; Nakamura, Y.; Sakata, K.; Hara, N.; Kobayashi, M.; Nagai, M.; Yanagawa, H.; Kawasaki, T. Kawasaki disease in families. Pediatrics 1989, 84, 666–669. [Google Scholar] [CrossRef]
  9. Xie, X.; Shi, X.; Liu, M. The Roles of Genetic Factors in Kawasaki Disease: A Systematic Review and Meta-analysis of Genetic Association Studies. Pediatr. Cardiol. 2018, 39, 207–225. [Google Scholar] [CrossRef] [PubMed]
  10. Hayashida, K.; Ae, R.; Masuda, H.; Kosami, K.; Matsumura, M.; Makino, N.; Matsubara, Y.; Sasahara, T.; Nakamura, Y.M. Clinical Characteristics of Patients With Kawasaki Disease Whose Siblings Had the Same Disease. Pediatr. Infect. Dis. J. 2021, 40, 531–536. [Google Scholar] [CrossRef]
  11. Amos, W.; Driscoll, E.; Hoffman, J.I. Candidate genes versus genome-wide associations: Which are better for detecting genetic susceptibility to infectious disease? Proc. R. Soc. B Biol. Sci. 2011, 278, 1183–1188. [Google Scholar] [CrossRef] [PubMed]
  12. González-Gay, M.Á.; Heras-Recuero, E.; Blázquez-Sánchez, T.; Caraballo-Salazar, C.; Rengifo-García, F.; Castaneda, S.; Martín, J.; Martín, J. Genetics of vasculitis. Best Pract. Res. Clin. Rheumatol. 2024, 38, 101969. [Google Scholar] [CrossRef] [PubMed]
  13. Onouchi, Y.; Ozaki, K.; Burns, J.C.; Shimizu, C.; Terai, M.; Hamada, H.; Honda, T.; Suzuki, H.; Suenaga, T.; Takeuchi, T.; et al. A genome-wide association study identifies three new risk loci for Kawasaki disease. Nat. Genet. 2012, 44, 517–521. [Google Scholar] [CrossRef] [PubMed]
  14. Krensky, A.M.; Grady, S.; Shanley, K.M.; Berenberg, W.; Yunis, E.J. Epedimic and endemic HLA-B and DR associations in mucocutaneous lymph node syndrome. Hum. Immunol. 1983, 6, 75–77. [Google Scholar] [CrossRef]
  15. Kaslow, R.A.; Bailowitz, A.; Lin, F.Y.; Koslowe, P.; Simonis, T.; Israel, E. Association of epidemic kawasaki syndrome with the HLA-A2, B44, Cw5 antigen combination. Arthritis Rheum. 1985, 28, 938–940. [Google Scholar] [CrossRef]
  16. Kato, S.; Kimura, M.; Tsuji, K.; Kusakawa, S.; Asai, T.; Juji, T.; Kawasaki, T. HLA antigens in Kawasaki disease. Pediatrics 1978, 61, 252–255. [Google Scholar] [CrossRef]
  17. Keren, G.; Danon, Y.L.; Orcad, S.; Kalt, R.; Gazit, E. HLA Bw51 is increased in mucocutaneous lymph node syndrome in Israeli patients. Tissue Antigens 1982, 20, 144–146. [Google Scholar] [CrossRef]
  18. Oh, J.H.; Han, J.W.; Lee, S.J.; Lee, K.Y.; Suh, B.K.; Koh, D.K.; Lee, J.S.; Oh, C.K.; Kim, T.G.; Choi, H.B. Polymorphisms of Human Leukocyte Antigen Genes in Korean Children with Kawasaki Disease. Pediatr. Cardiol. 2008, 29, 402–408. [Google Scholar] [CrossRef]
  19. Huang, F.Y.; Chang, T.Y.; Chen, M.R.; Hsu, C.H.; Lee, H.C.; Lin, S.P.; Kao, H.-A.; Chiu, N.-C.; Chi, H.; Liu, T.Y.-C.; et al. Genetic Variations of HLA-DRB1 and Susceptibility to Kawasaki Disease in Taiwanese Children. Hum. Immunol. 2007, 68, 69–74. [Google Scholar] [CrossRef]
  20. Huang, F.-Y.; Lee, Y.-J.; Chen, M.-R.; Hsu, C.-H.; Lin, S.-P.; Sung, T.-C.; Chang, S.-C.; Chang, J.-G. Polymorphism of Transmembrane Region of MICA Gene and Kawasaki Disease. Exp. Clin. Immunogenet. 2000, 17, 130–137. [Google Scholar] [CrossRef]
  21. Kwon, Y.-C.; Kim, J.-J.; Yun, S.W.; Yu, J.J.; Yoon, K.L.; Lee, K.-Y.; Kil, H.-R.; Kim, G.B.; Han, M.-K.; Song, M.S.; et al. Male-specific association of the FCGR2A His167Arg polymorphism with Kawasaki disease. PLoS ONE 2017, 12, e0184248. [Google Scholar] [CrossRef]
  22. Lou, J.; Zhong, R.; Shen, N.; Lu, X.-Z.; Ke, J.-T.; Duan, J.-Y.; Qi, Y.-Q.; Wang, Y.-J.; Zhang, Q.; Wang, W.; et al. Systematic Confirmation Study of GWAS-Identified Genetic Variants for Kawasaki Disease in A Chinese Population. Sci. Rep. 2015, 5, 8194. [Google Scholar] [CrossRef] [PubMed]
  23. Chatzikyriakidou, A.; Aidinidou, L.; Giannopoulos, A.; Papadopoulou-Legbelou, K.; Kalinderi, K.; Fidani, L. Absence of association of FCGR2A gene polymorphism rs1801274 with Kawasaki disease in Greek patients. Cardiol. Young 2015, 25, 681–683. [Google Scholar] [CrossRef] [PubMed]
  24. Taniuchi, S.; Masuda, M.; Teraguchi, M.; Ikemoto, Y.; Komiyama, Y.; Takahashi, H.; Kino, M.; Kobayashi, Y. Polymorphism of Fcγ RIIa May Affect the Efficacy of γ-Globulin Therapy in Kawasaki Disease. J. Clin. Immunol. 2005, 25, 309–313. [Google Scholar] [CrossRef] [PubMed]
  25. Biezeveld, M.; Geissler, J.; Merkus, M.; Kuipers, I.M.; Ottenkamp, J.; Kuijpers, T. The involvement of Fc gamma receptor gene polymorphisms in Kawasaki disease. Clin. Exp. Immunol. 2006, 147, 106–111. [Google Scholar] [CrossRef]
  26. Wang, Z.; Geng, P.-L. CD32a polymorphism rs1801274 affects the risk of Kawasaki disease. Artif. Cells Nanomed. Biotechnol. 2020, 48, 620–626. [Google Scholar] [CrossRef]
  27. Wang, C.-L.; Wu, Y.-T.; Liu, C.-A.; Lin, M.-W.; Lee, C.-J.; Huang, L.-T.; Yang, K.D. Expression of CD40 Ligand on CD4+ T-Cells and Platelets Correlated to the Coronary Artery Lesion and Disease Progress in Kawasaki Disease. Pediatrics 2003, 111, e140–e147. [Google Scholar] [CrossRef]
  28. Onouchi, Y.; Onoue, S.; Tamari, M.; Wakui, K.; Fukushima, Y.; Yashiro, M.; Nakamura, Y.; Yanagawa, H.; Kishi, F.; Ouchi, K.; et al. CD40 ligand gene and Kawasaki disease. Eur. J. Hum. Genet. 2004, 12, 1062–1068. [Google Scholar] [CrossRef]
  29. Huang, F.-Y.; Chang, T.-Y.; Chen, M.-R.; Chiu, N.-C.; Chi, H.; Lee, H.-C.; Lin, S.-P.; Chen, C.-K.; Chan, H.-W.; Chen, W.-F.; et al. Genetic Polymorphisms in the CD40 Ligand Gene and Kawasaki Disease. J. Clin. Immunol. 2008, 28, 405–410. [Google Scholar] [CrossRef]
  30. Kuo, H.-C.; Chao, M.-C.; Hsu, Y.-W.; Lin, Y.-C.; Huang, Y.-H.; Yu, H.-R.; Hou, M.-F.; Liang, C.-D.; Yang, K.D.; Chang, W.-C.; et al. CD40 Gene Polymorphisms Associated with Susceptibility and Coronary Artery Lesions of Kawasaki Disease in the Taiwanese Population. Sci. World J. 2012, 2012, 520865. [Google Scholar] [CrossRef]
  31. Patra, P.K.; Jindal, A.K.; Rikhi, R.; Kaur, A.; Srivastava, P.; Suri, D.; Rawat, A.; Pilania, R.; Singh, S. CD40 gene polymorphism and its expression in children with Kawasaki disease from North India: A preliminary case–control study and meta-analysis. Front. Pediatr. 2023, 11, 1252024. [Google Scholar] [CrossRef]
  32. Porritt, R.A.; Markman, J.L.; Maruyama, D.; Kocaturk, B.; Chen, S.; Lehman, T.J.A.; Lee, Y.; Fishbein, M.C.; Rivas, M.N.; Arditi, M. Interleukin-1 Beta-Mediated Sex Differences in Kawasaki Disease Vasculitis Development and Response to Treatment. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 802–818. [Google Scholar] [CrossRef] [PubMed]
  33. Zou, D.; Ahmed Mohamed, E.; Jiang, J.; Tian, L.; Chen, J.; Li, Z.; Yang, Z. Correlation between -31 T/C polymorphisms of interleukin-1β gene and Kawasaki disease. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2017, 42, 8–12. [Google Scholar] [PubMed]
  34. Weng, K.-P.; Hsieh, K.-S.; Ho, T.-Y.; Lai, C.-R.; Chiu, Y.-T.; Huang, S.-C.; Lin, C.-C.; Hwang, Y.-T.; Ger, L.-P. IL-1B Polymorphism in Association With Initial Intravenous Immunoglobulin Treatment Failure in Taiwanese Children With Kawasaki Disease. Circ. J. 2010, 74, 544–551. [Google Scholar] [CrossRef] [PubMed]
  35. Fu, L.Y.; Qiu, X.; Deng, Q.L.; Huang, P.; Pi, L.; Xu, Y.; Che, D.; Zhou, H.; Lu, Z.; Tan, Y.; et al. The IL-1B Gene Polymorphisms rs16944 and rs1143627 Contribute to an Increased Risk of Coronary Artery Lesions in Southern Chinese Children with Kawasaki Disease. J. Immunol. Res. 2019, 2019, 4730507. [Google Scholar] [CrossRef]
  36. Chen, M.-R.; Chang, T.-Y.; Chiu, N.-C.; Chi, H.; Yang, K.D.; Chang, L.; Huang, D.T.-N.; Huang, F.-Y.; Lien, Y.-P.; Lin, W.-S.; et al. Validation of genome-wide associated variants for Kawasaki disease in a Taiwanese case–control sample. Sci. Rep. 2020, 10, 11756. [Google Scholar] [CrossRef]
  37. Lee, Y.-C.; Kuo, H.-C.; Chang, J.-S.; Chang, L.-Y.; Huang, L.-M.; Chen, M.-R.; Liang, C.-D.; Chi, H.; Huang, F.-Y.; Lee, M.-L.; et al. Two new susceptibility loci for Kawasaki disease identified through genome-wide association analysis. Nat. Genet. 2012, 44, 522–525. [Google Scholar] [CrossRef]
  38. Kanda, S.; Fujii, Y.; Hori, S.-I.; Ohmachi, T.; Yoshimura, K.; Higasa, K.; Kaneko, K. Combined Single Nucleotide Variants of ORAI1 and BLK in a Child with Refractory Kawasaki Disease. Children 2021, 8, 433. [Google Scholar] [CrossRef]
  39. Cheung, Y.F.; Huang, G.Y.; Chen, S.B.; Liu, X.Q.; Xi, L.I.; Liang, X.C.; Huang, M.-R.; Chen, S.; Huang, L.-S.; Liu, X.-Q. Inflammatory Gene Polymorphisms and Susceptibility to Kawasaki Disease and Its Arterial Sequelae. Pediatrics 2008, 122, e608–e614. [Google Scholar] [CrossRef]
  40. Chien, Y.-H.; Chang, K.-W.; Yang, Y.-H.; Lu, M.-Y.; Lin, Y.-T.; Chiang, B.-L. Association between levels of TNF-alpha and TNF-alpha promoter -308 A/A polymorphism in children with Kawasaki disease. J. Formos. Med. Assoc. 2003, 102, 147–150. [Google Scholar]
  41. Yang, J.; Li, C.-R.; Li, Y.-B.; Li, R.-X.; Sun, L.-B.; Huang, H.-J.; Wang, G.-B. The correlation between Kawasaki disease and polymorphisms of Tumor necrosis factor alpha and interleukin-10 gene promoter. Zhonghua Er Ke Za Zhi 2003, 41, 598–602. [Google Scholar]
  42. Arj-Ong, S.; Thakkinstian, A.; McEvoy, M.; Attia, J. A systematic review and meta-analysis of tumor necrosis factor α-308 polymorphism and Kawasaki disease. Pediatr. Int. 2010, 52, 527–532. [Google Scholar] [CrossRef]
  43. Yuan, Y.; Piao, J.; Lu, N. Tumor necrosis factor-α-308 polymorphism is not associated with Kawasaki disease: A meta-analysis of case–control studies. Medicine 2019, 98, e15963. [Google Scholar] [CrossRef]
  44. Onouchi, Y.; Gunji, T.; Burns, J.C.; Shimizu, C.; Newburger, J.W.; Yashiro, M.; Nakamura, Y.; Yanagawa, H.; Wakui, K.; Fukushima, Y.; et al. ITPKC functional polymorphism associated with Kawasaki disease susceptibility and formation of coronary artery aneurysms. Nat. Genet. 2008, 40, 35–42. [Google Scholar] [CrossRef]
  45. Lin, M.-T.; Wang, J.-K.; Yeh, J.-I.; Sun, L.-C.; Chen, P.-L.; Wu, J.-F.; Chang, C.-C.; Lee, W.-L.; Shen, C.-T.; Wang, N.-K. Clinical Implication of the C Allele of the ITPKC Gene SNP rs28493229 in Kawasaki Disease: Association With Disease Susceptibility and BCG Scar Reactivation. Pediatr. Infect. Dis. J. 2011, 30, 148–152. [Google Scholar] [CrossRef]
  46. Chi, H.; Huang, F.-Y.; Chen, M.-R.; Chiu, N.-C.; Lee, H.-C.; Lin, S.-P.; Chen, W.-F.; Lin, C.-L.; Chan, H.-W.; Liu, H.-F.; et al. ITPKC gene SNP rs28493229 and Kawasaki disease in Taiwanese children. Hum. Mol. Genet. 2010, 19, 1147–1151. [Google Scholar] [CrossRef] [PubMed]
  47. Bhattarai, D.; Kumrah, R.; Kaur, A.; Kaur, A.; Srivastava, P.; Rawat, A.; Singh, S. Association of ITPKC gene polymorphisms rs28493229 and rs2290692 in North Indian children with Kawasaki disease. Pediatr. Res. 2022, 92, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
  48. Kim, K.Y.; Bae, Y.-S.; Ji, W.; Shin, D.; Kim, H.-S.; Kim, D.S. ITPKC and SLC11A1 Gene Polymorphisms and Gene-Gene Interactions in Korean Patients with Kawasaki Disease. Yonsei Med. J. 2018, 59, 119–127. [Google Scholar] [CrossRef] [PubMed]
  49. Habibi, A.; Talebi, H.; Bahrami, R.; Golshan-Tafti, M.; Shahbazi, A.; Dastgheib, S.A.; Tahooni, A.; Vafapour, M.; Rashnavadi, H.; Pourkazemi, M.; et al. A comprehensive integration of data on the association of ITPKC polymorphisms with susceptibility to Kawasaki disease: A meta-analysis. BMC Med. Genom. 2025, 18, 56. [Google Scholar] [CrossRef]
  50. Lou, J.; Xu, S.; Zou, L.; Zhong, R.; Zhang, T.; Sun, Y.; Lu, X.; Liu, L.; Li, C.; Wang, L.; et al. A functional polymorphism, rs28493229, in ITPKC and risk of Kawasaki disease: An integrated meta-analysis. Mol. Biol. Rep. 2012, 39, 11137–11144. [Google Scholar] [CrossRef]
  51. Natividad, M.F.; Torres-Villanueva, C.A.T.; Saloma, C.P. Superantigen involvement and susceptibility factors in Kawasaki disease: Profiles of TCR Vβ2+ T cells and HLA-DRB1, TNF-α and ITPKC genes among Filipino patients. Int. J. Mol. Epidemiol. Genet. 2013, 4, 70–76. [Google Scholar]
  52. Kuo, H.-C.; Hsu, Y.-W.; Lo, M.-H.; Huang, Y.-H.; Chien, S.-C.; Chang, W.-C.; Gaetano, C. Single-Nucleotide Polymorphism rs7251246 in ITPKC Is Associated with Susceptibility and Coronary Artery Lesions in Kawasaki Disease. PLoS ONE 2014, 9, e91118. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, J.; Yuan, P.; Pang, Y.; Su, D. ITPKC polymorphism (rs7251246 T > C), coronary artery aneurysms, and thrombosis in patients with Kawasaki disease in a Southern Han Chinese population. Front. Immunol. 2023, 14, 1184162. [Google Scholar] [CrossRef] [PubMed]
  54. Kuo, H.C.; Lin, Y.J.; Juo, S.H.H.; Hsu, Y.W.; Chen, W.C.; Yang, K.D.; Liang, C.-D.; Yang, S.; Chao, M.-C.; Yu, H.-R.; et al. Lack of association between ORAI1/CRACM1 gene polymorphisms and Kawasaki disease in the Taiwanese children. J. Clin. Immunol. 2011, 31, 650–655. [Google Scholar] [CrossRef] [PubMed]
  55. Onouchi, Y.; Fukazawa, R.; Yamamura, K.; Suzuki, H.; Kakimoto, N.; Suenaga, T.; Takeuchi, T.; Hamada, H.; Honda, T.; Yasukawa, K.; et al. Variations in ORAI1 Gene Associated with Kawasaki Disease. PLoS ONE 2016, 11, e0145486. [Google Scholar] [CrossRef]
  56. Japan Kawasaki Disease Genome Consortium; Thiha, K.; Mashimo, Y.; Suzuki, H.; Hamada, H.; Hata, A.; Hara, T.; Tanaka, T.; Ito, K.; Onouchi, Y. Investigation of novel variations of ORAI1 gene and their association with Kawasaki disease. J. Hum. Genet. 2019, 64, 511–519. [Google Scholar] [CrossRef]
  57. Onouchi, Y.; Ozaki, K.; Buns, J.C.; Shimizu, C.; Hamada, H.; Honda, T.; Terai, M.; Honda, A.; Takeuchi, T.; Shibuta, S.; et al. Common variants in CASP3 confer susceptibility to Kawasaki disease. Hum. Mol. Genet. 2010, 19, 2898–2906. [Google Scholar] [CrossRef]
  58. Yoon, K.L. Update of genetic susceptibility in patients with Kawasaki disease. Korean J. Pediatr. 2015, 58, 84. [Google Scholar] [CrossRef]
  59. Peng, Q.; Chen, C.-H.; Wu, Q.; Yang, Y. Association of new functional SNP rs72689236 of CASP3 with Kawasaki disease: A meta-analysis. Zhongguo Dang Dai Er Ke Za Zhi 2013, 15, 477–483. [Google Scholar]
  60. Kuo, H.-C.; Yu, H.-R.; Juo, S.-H.H.; Yang, K.D.; Wang, Y.-S.; Liang, C.-D.; Huang, C.-F.; Lee, C.-P.; Lin, L.-Y.; Liu, Y.-C.; et al. CASP3 gene single-nucleotide polymorphism (rs72689236) and Kawasaki disease in Taiwanese children. J. Hum. Genet. 2011, 56, 161–165. [Google Scholar] [CrossRef]
  61. Das, K.G.; Bhattarai, D.; Kaur, A.; Kaur, A.; Kumrah, R.; Srivastava, P.; Rawat, A.; Singh, S. Association of single nucleotide polymorphism rs113420705 of CASP3 in children with Kawasaki disease from North India. J. Fam. Med. Prim. Care 2022, 11, 5404–5409. [Google Scholar] [CrossRef]
  62. Hsu, Y.-W.; Chien, S.-C.; Liang, C.-C.; Yang, K.D.; Lee, J.-A.; Kuo, H.-C.; Chang, W.-C. Stromal Interaction Molecule 1 Polymorphisms are Associated with Coronary Artery Dilation but not with Aneurysm Formation in Patients with Kawasaki Disease. J. Exp. Clin. Med. 2013, 5, 73–76. [Google Scholar] [CrossRef]
  63. Frischauf, I.; Fahrner, M.; Jardín, I.; Romanin, C. The STIM1: Orai Interaction. Adv. Exp. Med. Biol. 2016, 898, 25–46. [Google Scholar] [PubMed]
  64. Oh-Hora, M.; Yamashita, M.; Hogan, P.G.; Sharma, S.; Lamperti, E.; Chung, W.; Prakriya, M.; Feske, S.; Rao, A. Dual functions for the endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat. Immunol. 2008, 9, 432–443. [Google Scholar] [CrossRef] [PubMed]
  65. Shimizu, C.; Eleftherohorinou, H.; Wright, V.J.; Kim, J.; Alphonse, M.P.; Perry, J.C.; Cimaz, R.; Burgner, D.; Dahdah, N.; Hoang, L.T.; et al. Genetic Variation in the SLC8A1 Calcium Signaling Pathway Is Associated With Susceptibility to Kawasaki Disease and Coronary Artery Abnormalities. Circ. Cardiovasc. Genet. 2016, 9, 559–568. [Google Scholar] [CrossRef]
  66. Duan, J.; Lou, J.; Zhang, Q.; Ke, J.; Qi, Y.; Shen, N.; Zhu, B.; Zhong, R.; Wang, Z.; Liu, L.; et al. A Genetic Variant rs1801274 in FCGR2A as a Potential Risk Marker for Kawasaki Disease: A Case-Control Study and Meta-Analysis. PLoS ONE 2014, 9, e103329. [Google Scholar] [CrossRef]
  67. Kuo, H.-C.; Yang, K.D.; Juo, S.-H.H.; Liang, C.-D.; Chen, W.-C.; Wang, Y.-S.; Lee, C.-H.; Hsi, E.; Yu, H.-R.; Woon, P.-Y.; et al. ITPKC single nucleotide polymorphism associated with the Kawasaki disease in a Taiwanese population. PLoS ONE 2011, 6, e17370. [Google Scholar] [CrossRef]
  68. Onouchi, Y.; Suzuki, Y.; Suzuki, H.; Terai, M.; Yasukawa, K.; Hamada, H.; Suenaga, T.; Honda, T.; Honda, A.; Kobayashi, H.; et al. ITPKC and CASP3 polymorphisms and risks for IVIG unresponsiveness and coronary artery lesion formation in Kawasaki disease. Pharmacogenomics J. 2013, 13, 52–59. [Google Scholar] [CrossRef]
  69. Shimizu, C.; Jain, S.; Davila, S.; Hibberd, M.L.; Lin, K.O.; Molkara, D.; Frazer, J.R.; Sun, S.; Baker, A.L.; Newburger, J.W.; et al. Transforming growth factor-beta signaling pathway in patients with Kawasaki disease. Circ. Cardiovasc. Genet. 2011, 4, 16–25. [Google Scholar] [CrossRef]
  70. Cho, J.H.; Han, M.Y.; Cha, S.H.; Jung, J.H.; Yoon, K.L. Genetic polymorphism of SMAD5 is associated with Kawasaki disease. Pediatr. Cardiol. 2014, 35, 601–607. [Google Scholar] [CrossRef]
  71. Peng, Q.; Deng, Y.; Yang, X.; Leng, X.; Yang, Y.; Liu, H. Genetic variants of ADAM17 are implicated in the pathological process of Kawasaki disease and secondary coronary artery lesions via the TGF-β/SMAD3 signaling pathway. Eur. J. Pediatr. 2016, 175, 705–713. [Google Scholar] [CrossRef]
  72. Zhu, H.; Xu, B.; Hu, C.; Li, A.; Liao, Q. Identification and validation of autophagy-related genes in Kawasaki disease. Hereditas 2023, 160, 17. [Google Scholar] [CrossRef]
  73. Wessels, P.A.; Bingler, M.A. A comparison of Kawasaki Disease and multisystem inflammatory syndrome in children. Prog. Pediatr. Cardiol. 2022, 65, 101516. [Google Scholar] [CrossRef]
  74. Huang, F.-C.; Huang, Y.-H.; Kuo, H.-C.; Li, S.-C. Identifying Downregulation of Autophagy Markers in Kawasaki Disease. Children 2020, 7, 166. [Google Scholar] [CrossRef] [PubMed]
  75. Zhou, Y.; Wu, Y.; Yuan, C.; Yin, W.; Wang, B.; Ding, Y. The expression of autophagy markers in IVIG-resistant Kawasaki disease and the establishment of prediction model. BMC Pediatr. 2023, 23, 642. [Google Scholar] [CrossRef] [PubMed]
  76. Sharma, K.; Vignesh, P.; Srivastava, P.; Sharma, J.; Chaudhary, H.; Mondal, S.; Kaur, A.; Kaur, H.; Singh, S. Epigenetics in Kawasaki Disease. Front. Pediatr. 2021, 9, 673294. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, X.; Sun, Y.; Meng, L.; Ye, C.; Han, H.; Zhang, T.; Feng, Y.; Li, J.; Duan, L.; Chen, Y. Whole-exome sequencing analysis identifies novel variants associated with Kawasaki disease susceptibility. Pediatr. Rheumatol. 2023, 21, 78. [Google Scholar] [CrossRef]
  78. Wang, Y.; Lin, K.; Zhang, L.; Lin, Y.; Yu, H.; Xu, Y.; Fu, L.; Pi, L.; Li, J.; Mai, H.; et al. The rs7404339 AA Genotype in CDH5 Contributes to Increased Risks of Kawasaki Disease and Coronary Artery Lesions in a Southern Chinese Child Population. Front. Cardiovasc. Med. 2022, 9, 760982. [Google Scholar] [CrossRef]
  79. Yu, H.; Liu, F.; Chen, K.; Xu, Y.; Wang, Y.; Fu, L.; Zhou, H.; Pi, L.; Che, D.; Li, H. The EIF2AK4/rs4594236 AG/GG Genotype Is a Hazard Factor of Immunoglobulin Therapy Resistance in Southern Chinese Kawasaki Disease Patients. Front. Genet. 2022, 13, 868159. [Google Scholar] [CrossRef]
  80. Lu, Z.; Zheng, Z.; Xu, Y.; Wang, C.; Lin, Y.; Lin, K.; Fu, L.; Zhou, H.; Pi, L.; Che, D.; et al. The Associated of the Risk of IVIG Resistance in Kawasaki Disease with ZNF112 Gene and ZNF180 Gene in a Southern Chinese Population. J. Inflamm. Res. 2022, 15, 5053–5062. [Google Scholar] [CrossRef]
  81. Buda, P.; Chyb, M.; Smorczewska-Kiljan, A.; Wieteska-Klimczak, A.; Paczesna, A.; Kowalczyk-Domagała, M.; Okarska-Napierała, M.; Sobalska-Kwapis, M.; Grochowalski, Ł.; Słomka, M.; et al. Association Between rs12037447, rs146732504, rs151078858, rs55723436, and rs6094136 Polymorphisms and Kawasaki Disease in the Population of Polish Children. Front. Pediatr. 2021, 9, 624798. [Google Scholar] [CrossRef]
  82. Li, W.; Pi, L.; Yuan, J.; Gu, X.; Wang, Z.; Liu, Y.; Deng, Q.; Wang, Y.; Huang, P.; Zhang, L.; et al. Impact of Platelet Glycoprotein Ia/IIa C807T Gene Polymorphisms on Coronary Artery Aneurysms of KD Patients. Cardiol. Res. Pract. 2021, 2021, 4895793. [Google Scholar] [CrossRef] [PubMed]
  83. Hoggart, C.; Shimizu, C.; Galassini, R.; Wright, V.J.; Shailes, H.; Bellos, E.; Herberg, J.A.; Pollard, A.J.; O’cOnnor, D.; Choi, S.W.; et al. Identification of novel locus associated with coronary artery aneurysms and validation of loci for susceptibility to Kawasaki disease. Eur. J. Hum. Genet. 2021, 29, 1734–1744. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.; Xu, Y.; Huang, P.; Che, D.; Wang, Z.; Huang, X.; Xie, X.; Li, W.; Zhang, L.; Gu, X. Homozygous of MRP4 Gene rs1751034 C Allele Is Related to Increased Risk of Intravenous Immunoglobulin Resistance in Kawasaki Disease. Front. Genet. 2021, 12, 510350. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, K.; Zhang, L.; Wang, Y.; Li, J.; Xu, Y.; Che, D.; Mai, H.; Yu, H.; Fu, L.; Wei, B.; et al. FNDC1 Polymorphism (rs3003174 C > T) Increased the Incidence of Coronary Artery Aneurysm in Patients with Kawasaki Disease in a Southern Chinese Population. J. Inflamm. Res. 2021, 14, 2633–2640. [Google Scholar] [CrossRef]
  86. Kuo, K.-C.; Yang, Y.-L.; Lo, M.-H.; Cai, X.-Y.; Kuo, H.-C.; Huang, Y.-H. The Expression of Glycoprotein Genes in the Inflammatory Process of Kawasaki Disease. Front. Pediatr. 2020, 8, 592122. [Google Scholar] [CrossRef]
  87. Shi, R.; Luo, Y.; Li, S.; Kong, M.; Liu, X.; Yu, M.; Wu, J.; Huang, L.; Yang, Z. Single-nucleotide Polymorphism rs17860041 A/C in the Promoter of the PPIA Gene is Associated with Susceptibility to Kawasaki Disease in Chinese Children. Immunol. Investig. 2021, 50, 230–242. [Google Scholar] [CrossRef]
  88. Nie, H.; Wang, S.; Wu, Q.; Xue, D.; Zhou, W. Five immune-gene-signatures participate in the development and pathogenesis of Kawasaki disease. Immun. Inflamm. Dis. 2021, 9, 157–166. [Google Scholar] [CrossRef]
  89. Wu, J.; Yu, M.; Huang, L.; Qian, Y.; Kong, M.; Kang, Z.; Yang, Z. Association of MnSOD gene polymorphism with susceptibility to Kawasaki disease in Chinese children. Cardiol. Young 2021, 31, 179–185. [Google Scholar] [CrossRef]
  90. Wang, Z.; Xu, Y.; Zhou, H.; Wang, Y.; Li, W.; Lu, Z.; Jiang, Z.; Gu, X.; Zheng, H.; Zeng, L.; et al. Association between P2RY12 Gene Polymorphisms and IVIG Resistance in Kawasaki Patients. Cardiovasc. Ther. 2020, 2020, 3568608. [Google Scholar] [CrossRef]
  91. Amano, Y.; Akazawa, Y.; Yasuda, J.; Yoshino, K.; Kojima, K.; Kobayashi, N.; Matsuzaki, S.; Nagasaki, M.; Kawai, Y.; Minegishi, N.; et al. A low-frequency IL4R locus variant in Japanese patients with intravenous immunoglobulin therapy-unresponsive Kawasaki disease. Pediatr. Rheumatol. 2019, 17, 34. [Google Scholar] [CrossRef]
  92. Korean Kawasaki Disease Genetics Consortium; Kwon, Y.-C.; Kim, J.-J.; Yu, J.J.; Yun, S.W.; Yoon, K.L.; Lee, K.-Y.; Kil, H.-R.; Kim, G.B.; Han, M.-K.; et al. Identification of the TIFAB Gene as a Susceptibility Locus for Coronary Artery Aneurysm in Patients with Kawasaki Disease. Pediatr. Cardiol. 2019, 40, 483–488. [Google Scholar] [CrossRef]
  93. Ahn, J.G.; Bae, Y.; Shin, D.; Nam, J.; Kim, K.Y.; Kim, D.S. HMGB1 gene polymorphism is associated with coronary artery lesions and intravenous immunoglobulin resistance in Kawasaki disease. Rheumatology 2019, 58, 770–775. [Google Scholar] [CrossRef]
  94. Zha, L.; Li, S.; Liu, X.; Li, Z.; Jiang, J.; Huang, L.; Yang, Z. Association of miR-146a Gene Polymorphism at loci rs2910164 G/C, rs57095329 A/G, and rs6864584 T/C with Susceptibility to Kawasaki Disease in Chinese Children. Pediatr. Cardiol. 2019, 40, 504–512. [Google Scholar] [CrossRef] [PubMed]
  95. Kim, J.-J.; Yun, S.W.; Yu, J.J.; Yoon, K.L.; Lee, K.-Y.; Kil, H.-R.; Kim, G.B.; Han, M.-K.; Song, M.S.; Lee, H.D.; et al. Identification of SAMD9L as a susceptibility locus for intravenous immunoglobulin resistance in Kawasaki disease by genome-wide association analysis. Pharmacogenomics J. 2020, 20, 80–86. [Google Scholar] [CrossRef] [PubMed]
  96. Rowley, A.H.; Wylie, K.M.; Kim, K.-Y.A.; Pink, A.J.; Yang, A.; Reindel, R.; Baker, S.C.; Shulman, S.T.; Orenstein, J.M.; Lingen, M.W.; et al. The transcriptional profile of coronary arteritis in Kawasaki disease. BMC Genom. 2015, 16, 1076. [Google Scholar] [CrossRef] [PubMed]
  97. Jia, C.; Zhang, J.; Chen, H.; Zhuge, Y.; Chen, H.; Qian, F.; Zhou, K.; Niu, C.; Wang, F.; Qiu, H.; et al. Endothelial cell pyroptosis plays an important role in Kawasaki disease via HMGB1/RAGE/cathespin B signaling pathway and NLRP3 inflammasome activation. Cell Death Dis. 2019, 10, 778. [Google Scholar] [CrossRef]
  98. Anzai, F.; Watanabe, S.; Kimura, H.; Kamata, R.; Karasawa, T.; Komada, T.; Nakamura, J.; Nagi-Miura, N.; Ohno, N.; Takeishi, Y.; et al. Crucial role of NLRP3 inflammasome in a murine model of Kawasaki disease. J. Mol. Cell. Cardiol. 2020, 138, 185–196. [Google Scholar] [CrossRef]
  99. Hoang, L.T.; Shimizu, C.; Ling, L.; Naim, A.N.M.; Khor, C.C.; Tremoulet, A.H.; Victoria, W.; Levin, M.; Hibberd, M.L.; Burns, J.C. Global gene expression profiling identifies new therapeutic targets in acute Kawasaki disease. Genome Med. 2014, 6, 541. [Google Scholar] [CrossRef]
Table 1. Studies highlighting genetic association of Kawasaki disease.
Table 1. Studies highlighting genetic association of Kawasaki disease.
Author/YearGene/Gene LocusStudy PopulationAssociation with KDFunction
Association with the occurrence of KD
Onouchi et al., 2012
[13]
Intergenic region between HLA-DQB2 and HLA-DOB
(rs2857151),
FAM167A-BLK (rs2254546), and CD40 (rs4813003)
Japanese
KD patients (n = 428); healthy Controls (n = 3379)
Strong association with KD HLA-DQB2/DOB: MHC class I alleles mediate antigen presentation to CD4+ T cells.
BLK: Tyrosine kinase involved in B cell receptor signaling and affects B cell-mediated immune response
Duan et al. 2014
[66]
FCGR2A (rs1801274),
HLA-DQB2 and HLA-DOB (rs2857151), and BLK (rs2254546)
Han Chinese
KD patients (n = 358); healthy controls (n = 815)
Significant association with KD FCGR2A: The FCGR2A gene encodes FcγRIIA, a low-affinity receptor for the Fc portion of immunoglobulin G (IgG). It helps in the phagocytosis of immune complexes and activation of pro-inflammatory signaling pathways (via ITAM motifs)
OH et al. 2008
[18]
HLA-B35, HLA-B75, and HLA-Cw09 allelesKorean
KD patients (n= 74);
healthy controls (n = 159)
Increased susceptibility to KDHLA-B35/B75: MHC class I alleles mediate antigen presentation to CD8+ T cells.
Kwon et al. 2017
[21]
FCGR2A (rs1801274),
SEMA6A (rs12516652),
IL17REL (rs5771303)
Korean
KD patients (n = 249);
healthy controls (n = 1000)
Replication study in 671 Japanese cases and 3553 controls
FCGR2A, SEMA6A, and IL17REL variants were significantly associated with KD in males but not in females
Functional polymorphism of FCGR2A (rs1801274; p. His167Arg) of FCGR2A was significantly associated with KD in males
SEMA6A: Regulates immune cell guidance and endothelial function
IL17REL: Involved in modulating immune and inflammatory responses, potentially as part of the IL-17 cytokine signaling pathway.
Wang et al.
2020
[26]
FCGR2A (CD32a) gene polymorphism
(rs1801274)
Asian
(meta-analysis)
Genotype AA and allele A of CD32a polymorphism (rs1801274) may increase the KD susceptibility
In Asian population
-
Onouchi et al.
2004
[28]
IL-1β gene (–31T/C polymorphism)Chinese
100 KD patients and 100 healthy controls
-31T/C polymorphism of IL-1β gene is associated with genetic susceptibility to KD.
TT genotype in is associated with low risk of KD.
IL-1β gene: Encodes IL-1β, a key inflammatory cytokine and is involved in stimulating the production of other cytokines (e.g., IL-6 and TNF-α) and enhancing endothelial activation and coagulation pathways
Thiha et al.
2019
[56]
ORAI1 gene novel variants3812 Japanese patients with KD and 2644 healthy controls6 novel, rare, and deleterious missense variants were detected only in KD patients, including 3 cases of refractory KDORAI1: Regulates calcium influx in immune cells (store-operated calcium entry).
Onouchi et al.
2010
[57]
CASP3 gene variants920 Japanese KD patients
1409 Japanese healthy controls and 249 controls of European descent
Multiple variants in CASP3 gene confer susceptibility to KDCASP3: It encodes caspase-3, a critical executioner enzyme in the apoptosis (programmed cell death) pathway. Regulates immune cell death and tissue remodeling.
Association with the occurrence of CAAs in KD
Duan et al. 2014
[66]
FCGR2A (rs1801274), HLA-DQB2, and DOB intergenic region (rs2857151) and CASP3 (rs11340705)Han Chinese
KD patients (n = 358); healthy controls (n = 815)
High-risk genotypes (rs1801274, rs2857151, and rs11340705) associated with significantly higher risk of CAAsFCGR2A: The FCGR2A gene encodes FcγRIIA, a low-affinity receptor for the Fc portion of immunoglobulin G (IgG). It helps in the phagocytosis of immune complexes and activation of pro-inflammatory signaling pathways (via ITAM motifs).
OH et al. 2008
[18]
HLA-DRB1 allelesKorean
KD patients (n = 74);
healthy controls (n = 159)
Significantly increased frequency of the HLA-DRB1*11 allele in KD patients with CAAs compared with healthy controls
Huang et al.
2000
[20]
MHC-class-I-chain-related gene A (MICA)70 Chinese children with KD and 154 healthy controlsAllele A4 was significantly less frequent and allele A5 was significantly more frequent in patients with CAAs.
Allele A4 protects against KD CAAs
MICA: It encodes a stress-induced ligand that binds to NKG2D, an activating receptor present on natural killer (NK) cells, CD8+ T cells, and γδ T cells
Taniuchi et al. 2005
[24]
Polymorphism of FCGR2A, FCGR3B, and FCGR3A56 Japanese
KD patients who received IVIg therapy
60% of patients with the HR and RR alleles of FCGR2A polymorphism developed CAAs compared to 23% with HH allele.
HR and RR alleles
may predict the progression of CAAs in KD before the
initiation of IVIg therapy
FCGR3A/FCGR3 B: IgG receptors on NK cells and neutrophils. Variants may alter immune complex clearance and inflammation.
Wang et al.
2020
[26]
CD40L polymorphism427 Japanese
KD patients and 476 healthy controls
SNP in intron 4 (IVS4+121 A>G) is marginally increased in KD patients compared to controls, especially in males.
CD40L may play a role in CAA development
CD40L: Ligand for CD40, expressed on activated T cells and modulates immune activation.
Onouchi et al.
2004
[28]
CD40 polymorphisms
(rs4810485 and rs1535045)
381 Taiwanese
KD patients and 569 healthy controls
CD40 polymorphism (rs1535045) was significantly associated with KD
CD40 polymorphism (rs4810485) showed a significant association with CAAs in KD patients
CD40: Encodes a co-stimulatory receptor expressed primarily on B cells, dendritic cells, monocytes, and endothelial cells. The CD40–CD40L interaction is essential for B cell activation, proliferation, and antibody class switching
Kuo et al., 2012
[30]
IL-1B Polymorphisms (rs16944 and rs1143627)719 Southern Chinese
KD patients and 1401 healthy children
rs16944 GG and rs1143627 AA genotypes may significantly increase the risk of CAAs in children below 12 monthsIL-1B: Encodes IL-1β, a key inflammatory cytokine.
Weng et al. 2010
[34]
SNPs within introns of NUMBL, ADCK4, ITPKC, and FLJ41131 genes637 Japanese
KD patients and 1034 healthy controls
ITPKC intron 1 functional SNP is significantly associated with KD and CAAs
C-allele may be associated with immune hyper-reactivity in KD
NUMBL: Notch signaling modulator that plays a role in vascular development and inflammation.
ITPKC: Inhibits T cell activation via Ca2+ signaling pathways.
ADCK4: Mitochondrial function and coenzyme Q biosynthesis and involvement in immune cell energetics.
Liu et al.,
2023
[53]
ITPKC polymorphism
(rs7251246 T > C)
221 Han Chinese
children with KD;
262 children as healthy controls
CC/CT genotype was significantly associated with the risk of CAA in children with KD.
Those with the CC genotype had a significantly higher risk of thrombosis.
ITPKC mRNA levels were lower in children with CAA that was complicated by thrombosis
ITPKC: Inhibits T cell activation via Ca2+ signaling pathways
Kuo et al.,
2011
[67]
ITPKC SNP
rs28493229
341 Taiwanese
KD patients and 1190 controls
C-allele is associated with the susceptibility to KD and coronary artery aneurysm formation in KD ITPKC: Inhibits T cell activation via Ca2+ signaling pathways
Kanda et al., 2021
[38]
ORAI1 (rs3741596) and BLK (rs2254546) SNPs8-month-old Japanese boyORAI1 (rs3741596) and BLK (rs2254546) SNPs were associated with refractory KD in the child along with CAAsORAI1: Regulates calcium influx in immune cells (store-operated calcium entry).
CASP3 SNP (rs113420705) 45 cases of KD and 50 healthy controlsC allele was significantly higher in frequency in patients with KD with CAAs CASP3: Executioner caspase in apoptosis. Regulates immune cell death; variants associated with KD and IVIg resistance
Association with the occurrence of IVIg resistance in KD
Kuo et al.,
2012
[30]
IL-1B polymorphism156 Taiwanese
KD children treated with high-dose IVIG (136 with IVIg response and 20 without IVIG response)
IL-1B (−511 TT) and IL-1B (−31 CC) genotypes may be associated with IVIG failure at initial therapyIL-1B: Encodes IL-1β, a key inflammatory cytokine.
Onouchi et al.,
2013
[68]
ITPKC (rs28493229) and CASP3 (rs113420705) polymorphisms539 Japanese
patients who received IVIg
In IVIg non-responders, the susceptibility allele of both SNPs was overrepresented.
Higher risk of IVIg unresponsiveness was found in patients with at least 1 susceptible allele at both loci
ITPKC: Inhibits T cell activation via Ca2+ signaling pathways
Kuo et al., 2010
[60]
CASP3 gene SNP (rs72689236)341 KD patients and 751 controlsThe A allele of rs72689236 was found in a higher frequency in KD patients and in patients and IVIg resistance and CAAsCASP3: Executioner caspase in apoptosis. Regulates immune cell death; variants associated with KD and IVIg resistance
Association with the occurrence of BCG site reactivation in KD
Chi et al.,
2010
[46]
ITPKC SNP (rs28493229)280 Taiwanese
children with KD and 492 healthy controls
The frequency of the C-allele (GC and CC genotypes) was higher in KD patients than in controls.
GC or CC genotypes had a higher frequency of Bacille Calmette–Guérin (BCG) inoculation site reactivation in the acute phase than GG genotypes, especially in patients younger than 20 months old.
This suggests the activation of a hyperimmune response due to this SNP.
ITPKC: Inhibits T cell activation via Ca2+ signaling pathways
Kim et al.,
2017
[48]
ITPKC and SLC11A1 gene polymorphisms299 Korean
KD patients and 210 healthy controls
ITPKC SNP
(rs28493229) was associated with KD and CAAs.
SLC11A1 SNP (rs77624405) was associated with KD and BCG site erythema.
Gene–gene interactions were also associated with
BCG site erythema.
SLC11A1: Involved in macrophage activation and response to pathogens.
KD: Kawasaki Disease; IVIg: Intravenous Immunoglobulin; CAAs: Coronary Artery Aneurysm; BCG: Bacillus Calmette–Guérin; SNP: Single-Nucleotide Polymorphism; Ca2+: Calcium; NK cells: Natural Killer Cells; BLK: B-Lymphoid Tyrosine Kinase; SLC11A1: Solute Carrier Family 11; ITPKC: Inositol-Trisphosphate 3-Kinase C; CASP3:Caspase-3; IL-1B: Interleukin 1 Beta; ORAI1: ORAI Calcium Release-Activated Calcium Modulator 1; NUMBL: NUMB Like Endocytic Adaptor Protein; MICA: MHC Class I Polypeptide-Related Sequence A; SEMA6A: Semaphorin 6A; FCGR2A: Fc Gamma Receptor IIa.
Table 2. Recent studies highlighting novel variants associated with Kawasaki disease.
Table 2. Recent studies highlighting novel variants associated with Kawasaki disease.
SNo.AuthorYearEthnicitySample SizeTechnique UsedResultGene Function
1.Zhang et al.
[77]
2023ChinaKD patients (n = 93);
non-KD control cases (n = 91)
WES and Sanger sequencingRare variants in MYH14 and RBP3MYH14: Encodes a myosin heavy chain involved in cytoskeletal structure and intracellular transport. Mutations may affect vascular integrity and inflammation.
RBP3: Encodes interphotoreceptor retinoid-binding protein; mainly retinal, but mutations may suggest broader immune or inflammatory roles.
2Zhu et al.
[72]
2023ChinaKD patients (n = 55); non-KD control cases (n = 37)GeneCards Database and Gene Expression Omnibus (GEO) databaseWIPI1 and GBA, which are autophagy-related genes, could serve as biomarkers and potential therapeutic targets.WIPI1: Autophagy-related gene involved in autophagosome formation; dysregulation may contribute to immune response anomalies in KD.
GBA: Encodes glucocerebrosidase and is important in lysosomal function. Mutations can affect immune regulation via lysosomal pathways.
3Wang et al.
[78]
2022ChinaKD patients (n = 1335); healthy
controls (n = 1669)
Multiplex PCR for genotype of rs7404339 polymorphism in CDH5Increased susceptibility in individuals with the AA genotype of rs7404339 in CDH5CDH5 (VE-cadherin): Critical in endothelial cell adhesion and vascular integrity. Variants may lead to increased endothelial permeability and inflammation.
4Yu et al.
[79]
2022ChinaIVIg-responsive KD
patients (n = 795); IVIg-resistant KD patients (n = 234)
RT-PCRThe EIF2AK4/rs4594236 AG/GG genotype has a higher risk of IVIg resistance compared to the AA genotype.EIF2AK4 (GCN2): Regulates translation under stress. Implicated in immune modulation and possibly IVIg resistance in KD.
5Lu et al.
[80]
2022ChinaKD patients (n = 996)ZNF112/rs8113807 and ZNF180/rs2571051 genotyping by RT-PCR C and T carriers of ZNF112/rs8113807 and ZNF180/rs2571051, respectively, had a higher risk of IVIg resistance in KD.ZNF112/ZNF180: Zinc finger proteins likely involved in transcription regulation. Altered expression may impact immune cell gene networks and inflammation.
6Buda et al.
[81]
2021PolandKD patients (n = 119);
controls (n = 6071)
GWAS Increased susceptibility to KD is linked to rs12037447 in a non-coding sequence, rs146732504 in KIF25, rs151078858 in PTPRJ, rs55723436 in SPECC1L, and rs6094136 in RPN2. KIF25: Motor protein involved in mitotic spindle organization. May indirectly influence immune cell proliferation.
PTPRJ: Protein tyrosine phosphatase involved in immune signaling and cell growth regulation. Implicated in vascular and immune response modulation.
SPECC1L: Plays a role in cytoskeleton organization. Could affect immune cell migration or vascular structure.
RPN2: Part of the oligosaccharyltransferase complex. Involved in protein glycosylation, possibly influencing immune function.
7Li et al.
[82]
2021China KD patients (n = 818); healthy controls (n = 1401)Multiplex polymerase chain reactionsThe T allele carriers of the platelet glycoprotein Ia/IIa may have a reduced risk of CAAs in KD patients, particularly in females and children under 60 months. ITGA2 (Glycoprotein Ia/IIa): Platelet receptor involved in thrombosis. Variants may modulate the risk of coronary artery abnormalities (CAA).
8Hoggart et al.
[83]
2021UKKD patients (n = 200); controls (n = 276)GWASAn intergenic region on Chr. 20 is significantly associated with the formation of CAAs.-
9Wang et al.
[84]
2021China KD patients (n = 760)6 polymorphisms of the MRP4 gene using
TaqMan methods
The c allele of the MRP4 rs1751034 is associated with increased risk of IVIg resistance in KD patients MRP4: Encodes an ATP-binding cassette (ABC) transporter involved in the efflux of various endogenous and exogenous substances, including inflammatory mediators and drugs. Altered MRP4 function may impact immune regulation and drug responsiveness, thereby contributing to IVIg resistance
10Lin et al.
[85]
2021 China KD patients (n = 1459);
controls (n = 1611)
FNDC1 rs3003174 polymorphismThe C>T polymorphism may contribute to the development of CAAs in KD patients. FNDC1: Encodes a protein that contains fibronectin type III domains, which are often involved in cell adhesion, growth, and signaling. It has been implicated in vascular remodeling and the regulation of the immune system.
11Kuo et al.
[86]
2020TaiwanKD patients (n = 49); healthy controls (n = 24); disease controls (n = 24) Transcription levels of HP, CLEC4D, and GPR84 by RT-PCR Increased expression of CLEC4D, GPR84, and HP genes in peripheral leukocytes may indicate acute-phase KD when compared to control patients. HP, CLEC4D, GPR84: Acute-phase and immune signaling proteins. Upregulation is associated with involvement in innate immunity during KD.
12Shi et al.
[87]
2020 China KD patients (n = 101);
healthy controls (n = 105)
Three PPIA SNPs were genotyped by PCRIndividuals with promoter SNP (rs17860041 A/C) are more susceptible to KD in Chinese childrenPPIA (Cyclophilin A): Facilitates protein folding and is involved in immune cell signaling. Promoter variants may enhance susceptibility to inflammation.
13Nie et al.
[88]
2020ChinaKD patients (n = 173); healthy controls (n = 101)Gene Expression Omnibus databaseCXCL8, CCL5, CCR7, CXCR3, and CCR1 may have a significant role in the pathogenesis of KD CXCL8, CCL5, CCR7, CXCR3, CCR1: Chemokines and receptors involved in leukocyte migration. Their dysregulation contributes to vascular inflammation in KD.
14Wu et al
[89]
2020ChinaKD patients (n = 100); healthy controls (n = 102)RT-PCR for studying the polymorphism in the MnSOD gene Allele A in the MnSOD gene rs5746136 may be a risk factor for developing KDMnSOD (SOD2): Antioxidant enzyme protecting cells from oxidative stress. Polymorphisms may influence inflammatory response severity.
15Wang et al.
[90]
2020 China IVIg-resistant KD patients (n = 148); IVIg-responsive KD patients (n = 611) 5 polymorphisms of
P2RY12: rs9859538, rs1491974, rs7637803, rs6809699, and rs2046934 by PCR
The rs6809699 polymorphism in P2RY12 could predict IVIg resistance in
KD patients
P2RY12: Platelet receptor for ADP. Involved in clot formation and inflammation; variants may predict IVIg resistance.
16Amano et al.
[91]
2019JapanComplete KD patients (n = 75);
incomplete KD patients (n = 7); and allergic subjects (n = 99)
Pooled genome sequencing SNV rs563535954, located in the IL4R locus, could serve as a predictive indicator of IVIg unresponsivenessThe IL4R gene encodes the alpha chain of the interleukin-4 receptor, which binds both IL-4 and IL-13—key cytokines involved in Th2 immune responses, B cell proliferation, and immunoglobulin class switching. IL4R is critical in regulating allergic inflammation, antibody production, and immune homeostasis.
17 Kwon et al.
[92]
2018China KD patients (n = 713)GWASThe TIFAB gene SNP (rs899162) is significantly associated with CAA development (diameter ≥ 5 mm) in KD patients. TIFAB: Encodes a regulatory protein involved in modulating innate immune signaling, particularly through interaction with TRAF6, a key adaptor in the Toll-like receptor (TLR) and IL-1 receptor pathways. TIFAB negatively regulates TRAF6-mediated NF-κB activation, a major pathway in inflammation.
18Ahn et al.
[93]
2018Republic of KoreaKD patients (n = 265); controls (n = 203)Whole-genome sequencingrs 412125 in HMGB1 may contribute to CAA and IVIg resistance in KD patients.HMGB1: A nuclear protein that acts as a damage-associated molecular pattern (DAMP), triggering inflammatory responses
19Zha et al.
[94]
2018China KD patient (n = 120); healthy subjects (n = 126)RT-PCRThe rs2910164 of miR-146a G/C genotype and rs57095329 of miR-155 A/G allele were found to be risk factors for CAL.miR-146a: A microRNA regulating NF-κB pathway. Polymorphisms may lead to exaggerated inflammation.
miR-155: Regulates immune response; alterations linked to autoimmune and inflammatory diseases.
20Kim et al.
[95]
2018Republic of KoreaIVIg-resistant KD patients (n = 148); IVIg-responsive KD patients (n = 845) GWASrs28662 in the SAMD9L is significantly associated with IVIg resistance in KDSAMD9L: Associated with cell growth suppression and immune regulation. A variant may predispose individuals to IVIG resistance.
KD: Kawasaki Disease; IVIg: Intravenous Immunoglobulin; CAAs: Coronary Artery Aneurysm; GWAS: Genome-Wide Association Study; RT-PCR: Real-Time PCR; SAMD9L: Sterile Alpha Motif Domain Containing 9 Like; HMGB1: High Mobility Group Box 1 Protein; TIFAB: TRAF-Interacting Protein With Forkhead-Associated Domain, Family Member B; IL4R: Interleukin 4 Receptor; P2RY12: Purinergic Receptor P2Y12; MnSOD: Manganese Superoxide Dismutase; PPIA: Peptidylprolyl Isomerase A; HP: Haptoglobin; CLEC4D: C-Type Lectin Domain Family 4 Member D; GPR84: G-protein Coupled Receptor 84; FNDC1: Fibronectin Type III Domain Containing 1; MRP4: Multidrug Resistance-Associated Protein 4; ITGA2: Integrin Subunit Alpha 2; KIF25: Kinesin Family Member 25; PTPRJ: Protein Tyrosine Phosphatase Receptor Type J; SPECC1L: Sperm Antigen with Calponin Homology and Coiled-Coil Domains 1 Like; RPN2: Ribophorin II; ZNF112: Zinc Finger Protein 112; EIF2AK4: Eukaryotic Translation Initiation Factor 2 Alpha Kinase 4; CDH5: Cadherin 5; GBA: Glucosylceramidase Beta; WIPI1: WD Repeat Domain, Phosphoinositide Interacting 1; MYH14: Myosin Heavy Chain 14; RBP3: Retinol Binding Protein 3.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Goyal, T.; Sharma, S.; Pilania, R.K.; Jawallia, K.; Chawla, S.; Sharma, M.; Rawat, M.; Thakur, V.; Arya, U.; Kumar, A.; et al. Genetic Landscape of Kawasaki Disease: An Update. Lymphatics 2025, 3, 21. https://doi.org/10.3390/lymphatics3030021

AMA Style

Goyal T, Sharma S, Pilania RK, Jawallia K, Chawla S, Sharma M, Rawat M, Thakur V, Arya U, Kumar A, et al. Genetic Landscape of Kawasaki Disease: An Update. Lymphatics. 2025; 3(3):21. https://doi.org/10.3390/lymphatics3030021

Chicago/Turabian Style

Goyal, Taru, Saniya Sharma, Rakesh Kumar Pilania, Kajol Jawallia, Sanchi Chawla, Madhubala Sharma, Monica Rawat, Vaishali Thakur, Urvi Arya, Anoop Kumar, and et al. 2025. "Genetic Landscape of Kawasaki Disease: An Update" Lymphatics 3, no. 3: 21. https://doi.org/10.3390/lymphatics3030021

APA Style

Goyal, T., Sharma, S., Pilania, R. K., Jawallia, K., Chawla, S., Sharma, M., Rawat, M., Thakur, V., Arya, U., Kumar, A., Dhaliwal, M., Pandiarajan, V., Rawat, A., & Singh, S. (2025). Genetic Landscape of Kawasaki Disease: An Update. Lymphatics, 3(3), 21. https://doi.org/10.3390/lymphatics3030021

Article Metrics

Back to TopTop