key: cord-0686930-rnn2p19c authors: Noval Rivas, Magali; Arditi, Moshe title: Kawasaki disease: pathophysiology and insights from mouse models date: 2020-05-26 journal: Nat Rev Rheumatol DOI: 10.1038/s41584-020-0426-0 sha: 68395f1560eedc0535d85d57d815e0922822277c doc_id: 686930 cord_uid: rnn2p19c Kawasaki disease is an acute febrile illness and systemic vasculitis of unknown aetiology that predominantly afflicts young children, causes coronary artery aneurysms and can result in long-term cardiovascular sequelae. Kawasaki disease is the leading cause of acquired heart disease among children in the USA. Coronary artery aneurysms develop in some untreated children with Kawasaki disease, leading to ischaemic heart disease and myocardial infarction. Although intravenous immunoglobulin (IVIG) treatment reduces the risk of development of coronary artery aneurysms, some children have IVIG-resistant Kawasaki disease and are at increased risk of developing coronary artery damage. In addition, the lack of specific diagnostic tests and biomarkers for Kawasaki disease make early diagnosis and treatment challenging. The use of experimental mouse models of Kawasaki disease vasculitis has considerably improved our understanding of the pathology of the disease and helped characterize the cellular and molecular immune mechanisms contributing to cardiovascular complications, in turn leading to the development of innovative therapeutic approaches. Here, we outline the pathophysiology of Kawasaki disease and summarize and discuss the progress gained from experimental mouse models and their potential therapeutic translation to human disease. mucosal surfaces in the lung 29 (Fig. 1 ). This hypothesis is supported by the seasonality of Kawasaki disease outbreaks, which is similar to that of other respiratory infections. In Japan, two seasonal peaks have been observed, one in winter and another in summer, whereas in the USA, the incidence peaks are observed during spring and winter 30 . Development of Kawasaki disease is age specific, with children from 6 months to 5 years of age at greatest risk 3, 30, 31 , which suggests a protective maternal passive immunity against the causative agent from birth to 6 months of age and the importance of immune system maturation in children ≥6 years of age 29 . The clinical features of Kawasaki disease, such as high fever, skin rash and peeling, conjunctivitis and intense release of pro-inflammatory cytokines, are reminiscent of other infectious diseases such as staphylococcal and streptococcal toxic shock syndromes 32 . Some studies have shown that, compared with healthy control individuals, patients with Kawasaki disease have a skewed Vβ T cell repertoire and increased frequencies of circulating Vβ2 + and Vβ8.1 + T cells, leading to the early suggestion that a superantigen toxin might have a role in triggering Kawasaki disease [33] [34] [35] . However, similar results were not reproduced in later studies 36, 37 , leading to the more generalized hypothesis that the development of Kawasaki disease might be triggered by multiple conventional antigens. Several early studies showed reduced prevalence of antibodies to the Epstein-Barr virus (EBV) capsid antigen in Japanese children with Kawasaki disease compared with age and sex-matched control patients [38] [39] [40] , suggesting the involvement of an abnormal immune response to EBV in disease development. However, this difference in EBV antibody seropositivity could not be reproduced in other studies [41] [42] [43] . A human coronavirus was detected more frequently in respiratory secretions of patients with Kawasaki disease than in control individuals 44 , although, again, other studies could not replicate this finding 45, 46 , indicating that the original association might have been coincidental. The possibility that a retrovirus is the triggering agent for Kawasaki disease has also been proposed, owing to detection of retrovirus-specific reverse transcriptase activity in the co-culture supernatant of peripheral blood mononuclear cells (PBMCs) from patients with Kawasaki disease but not controls 47, 48 . However, this result could not be replicated in later studies [49] [50] [51] . A peptide recognized by antibodies produced during the acute phase of Kawasaki disease has been identified in 2020 (reF. 52 ). Although the protein epitopes seem similar to hepaciviruses 53 , further studies are required to determine the specific gene sequence from which this peptide emerges. Altogether, the absence of consistent and reproducible studies pinpointing a specific aetiological agent suggests that Kawasaki disease is caused not by one but by multiple infectious agents. Acute Kawasaki disease is associated with infiltration of IgA + plasma cells in the respiratory tract, implying that the upper airways act as a portal of entry 25, 26 . One suggestion is that the triggering agent might be an environmental toxin or antigen transported by wind currents 54 ; however, this possibility cannot be rigorously assessed until precise identification of the aetiological agents is achieved 29 . Although Kawasaki disease has been observed around the world and in multiple ethnic groups, geographical differences exist in incidence. The highest incidence is in Asian countries such as Korea and Japan, where it has increased over the past decades and is now 10-20 times more prevalent than in North America and Europe 30 . This increased susceptibility in Asian children, as well as in children with Asian ancestry living in North America 31 , indicates that genetic components predispose to disease susceptibility. In Japan, siblings of children with Kawasaki disease are at increased risk of developing the disease 55 . Single nucleotide polymorphisms (SNPs) in multiple genes have been associated with increased susceptibility to Kawasaki disease (Fig. 1) ; however, mechanisms linking those SNPs with Kawasaki disease progression are not yet well understood and require more investigation. Calcium signalling pathway. Inositol 1,4,5-trisphosphate 3-kinase C (ITPKC), a kinase that phosphorylates inositol 1,4,5-triphosphate (IP 3 ), is involved in many signalling processes in a wide array of cells. In T cells, IP 3 is released after T cell receptor stimulation, thus increasing levels of intracellular Ca 2+ through IP 3 receptors expressed on the endoplasmic reticulum and leading to nuclear translocation of nuclear factor of activated T cells (NFAT), IL-2 production and T cell activation 56 . By blocking the interaction of IP 3 with its receptor, ITPKC negatively regulates T cell activation. A functional SNP in ITPKC has been associated with increased risk of coronary artery lesions in Taiwanese 57 , Japanese and American patients with Kawasaki disease 58 . Mechanistically, this ITPKC polymorphism might directly contribute to T cell hyperactivity, and more importantly, it might promote NLRP3 inflammasome activation and increase production of IL-1β and IL-18 (reF. 59 ). ORAI1 is a membrane-bound Ca 2+ channel protein encoded by ORAI1 that is involved in the Ca 2+ -calcineurin-NFAT signalling pathway. Although no significant association between ORAI1 polymorphisms and Kawasaki disease • Kawasaki disease is a childhood systemic vasculitis leading to the development of coronary artery aneurysms; it is the leading cause of acquired heart disease in children in developed countries. • The cause of Kawasaki disease is unknown, although it is suspected to be triggered by an unidentified infectious pathogen in genetically predisposed children. • Kawasaki disease might not be a normal immune response to an unusual environmental stimulus, but rather a genetically determined unusual and uncontrolled immune response to a common stimulus. • Although the aetiological agent in humans is unknown, mouse models of Kawasaki disease vasculitis demonstrate similar pathological features and have substantially accelerated discoveries in the field. • Genetic and transcriptomic analysis of blood samples from patients with Kawasaki disease and experimental evidence generated using mouse models have demonstrated the critical role of IL-1β in the pathogenesis of this disease and the therapeutic potential of targeting this pathway (currently under investigation in clinical trials). www.nature.com/nrrheum susceptibility or IVIG treatment response was initially reported in the Taiwanese population 60 , an SNP in exon 2 of ORAI1 is associated with Kawasaki disease susceptibility in the Japanese population 61 , and interestingly this SNP is 20 times more frequent in the general Japanese population than in the general European population 61 . Another SNP in SLC8A1, which encodes the Na + -Ca 2+ exchanger, is also associated with susceptibility to Kawasaki disease and aneurysm formation 62 , further highlighting the critical role of calcium signalling pathways in development of Kawasaki disease. Crucially, the Ca 2+ -NFAT signalling pathway is also key to intracellular Ca 2+ regulation and therefore to NLRP3 inflammasome activation and IL-1β production 63, 64 . CD40 ligand. CD40 ligand (CD40L) is a protein expressed by a large array of cells including activated T cells, B cells, monocytes and platelets. CD40L receptor, CD40, is expressed by antigen-presenting cells as well as endothelial cells 65 . CD40 engagement is associated with cell survival, activation, proliferation and cyto kine production 65 . Compared with control patients with other febrile illnesses, patients with Kawasaki disease have increased CD40L expression on CD4 + T cells and platelets, which correlates with increased development of coronary artery lesions and is reduced by IVIG treatment 66 . An SNP in CD40L has been reported in Japanese patients with Kawasaki disease and is more frequent in male patients with coronary artery lesions than in female patients 67 . This polymorphism was not observed in a cohort of Taiwanese patients 68 ; however, another SNP in the CD40 gene has been reported in an independent cohort of Taiwanese patients and is associated with increased susceptibility to Kawasaki disease and development of coronary artery lesions 69 . These results indicate a role of the CD40-CD40L pathway in the development and severity of Kawasaki disease and highlight this pathway as a potential therapeutic target. Mannose-binding lectin (MBL), a pattern recognition molecule of the innate immune system, binds the surface of pathogenic organisms and activates the complement pathway 70 . A polymorphism in MBL2 was found to be an age-related risk factor for development of coronary artery lesions in a Dutch cohort of patients 71, 72 require another round of IVIG treatment or the use of adjunctive therapies 15, 19, 20, 80 . The exact mechanisms by which IVIG mediates its therapeutic effect and how IVIG resistance develops remain unknown, and the potential involvement of this FcγRIIA polymorphism in IVIG resistance requires further investigation. The innate immune response. The immune response associated with Kawasaki disease is complex and involves the activation and infiltration of the coronary artery wall by both innate and adaptive immune cells (Fig. 2 ). On the basis of studies of post-mortem tissue from patients with Kawasaki disease, Kawasaki disease vascular pathology has been classified into three sequential linked pathological processes 81 . Necrotizing arteritis develops in the first 2 weeks of the disease and is associated with neutrophilic infiltrations, which gradually destroy the coronary artery intima, media and some portions of the adventitia. Alarmins from the S100 protein family, which are present in the cytoplasm of neutrophils, monocytes and macrophages 82 , also participate in this inflammatory process. Concentrations of circulating S100A8/A9 hetero dimers (calprotectin) and S100A12 are substantially higher in patients with Kawasaki disease during the acute phase than in control patients with other febrile illnesses and decline after IVIG treatment [83] [84] [85] . After the acute phase of Kawasaki disease, plasma concentrations of S100A8/A9 heterodimers only remain elevated in patients with giant CAAs 84 , highlighting its potential utility as a biomarker to monitor long-term persistence of inflammation. S100A12 also contributes to the acute inflammatory response by directly stimulating monocytes to produce IL-1β, which in turn activates coronary endothelial cells 85 . Necrotizing arteritis might result in the formation of CAAs and is followed by two other processes, subacute or chronic vasculitis and luminal myofibroblast proliferation (LMP), which occur simultaneously and might be observed for months to years after disease onset 81 . The inflammatory infiltrates are composed of CD8 + T cells, IgA + plasma cells, eosinophils and macrophages, which release pro-inflammatory cytokines contributing to cardiovascular pathology. Meanwhile, myofibroblasts, mainly derived from smooth muscle cells, and their matrix products progressively obstruct the coronary lumen 81 (Fig. 2) . Persistent subacute and chronic vasculitis and LMP can lead to stenosis and thrombosis after acute illness 6,9 . Matrix metalloproteinases. Matrix metalloproteinases (MMPs; zinc-dependent endopeptidases that degrade extracellular matrix components) are known to have an important role in both inflammation and tissue remodelling processes 86 . Increased expression and activity of a diverse set of MMPs has been demonstrated in acute Kawasaki disease [87] [88] [89] . The expression levels of MMP3 and MMP9, both known to mediate vascular smooth muscle cell migration and neointimal formation 90 , are increased in patients with Kawasaki disease 91 , and the circulating levels of these MMPs correlate with the development of CAAs in these patients 92 . MMP3 SNPs are also associated with the development of CAAs 88 , and The normal coronary artery is composed of three general layers: the tunica intima, tunica media and tunica adventitia. The intima is mainly composed of endothelial cells, the media of smooth muscle cells and the adventitia of loose connective tissue. In Kawasaki disease, necrotizing arteritis develops in the first 2 weeks of the disease and is associated with neutrophilic infiltration, which gradually destroys the intima, media and some portions of the adventitia of the coronary artery. CD8 + T cells, IgA + plasma cells, monocytes and macrophages compose the inflammatory infiltrate during subacute chronic arteritis. These cells release pro-inflammatory cytokines such as IL-1β and TNF, which contribute to luminal myofibroblast proliferation, in which myofibroblasts, mainly derived from smooth muscle cells, and their matrix products progressively obstruct the coronary lumen. www.nature.com/nrrheum this protease is considered to be a driving factor allowing IL-1-induced signalling to lead to migration of vascular smooth muscle cells and their transition to proliferating myofibroblasts [93] [94] [95] . Whereas MMP9 has been studied and implicated in elastin breakdown in the Lactobacillus casei cell wall extract (LCWE)-induced Kawasaki disease mouse model 96, 97 , information about the role of MMP3 in this mouse model is lacking. MicroRNAs. MicroRNAs (miRNAs; a class of small non-coding RNAs that regulate mRNA expression) are emerging as critical gene regulators in a host of cellular processes, including inflammation 98 . Of human coding genes, 60-70% are estimated to be regulated by miRNAs 99 . Several studies attempting to discover Kawasaki disease biomarkers have found that the miRNA profiles of serum exosome or coronary artery tissues are associated with acute Kawasaki disease 100-104 . These miRNAs include miR-23a 100-103 , miR-27b 100 , miR-223 (reFs 100-103 ) and miR-145 (reF. 103 ). These miRNAs might provide clues as to the molecular mechanisms involved in the development of the cardiovascular lesions associated with Kawasaki disease. For example, miR-145 is highly expressed in vascular smooth muscle cells and has been reported to promote their switching to neointimal proliferating cells 105, 106 and to regulate the transforming growth factor-β signalling pathway 103 . Increased levels of miR-23a contribute to cardiomyocyte apoptosis and may promote inflammatory responses by blocking macrophage autophagy activity 107, 108 . However, improved understanding and characterization of the molecular and cellular mechanisms underlying the different roles of miRs during Kawasaki disease require further studies with animal models. research and clinical practice has focused on the development of CAAs and long-term complications of coronary artery stenosis and ischaemia 109 . However, the subacute and chronic inflammation of Kawasaki disease is also associated with the development of myocarditis 3,6,110-112 . Myocarditis has been described as the 'hidden face of the moon' in Kawasaki disease 110 . Reports indicate that myocarditis occurs frequently during acute Kawasaki disease 111 , and serial myocardial biopsy studies have documented that histological myocarditis develops in the majority of patients with Kawasaki disease, even in the absence of coronary aneurysms 113, 114 . More recent data indicate that myocardial inflammation can be documented in 50-70% of patients using gallium citrate ( 67 Ga) scans and technetium-99 ( 99m Tc)labelled white blood cell scans 115 . Another study has shown that myocardial inflammatory changes and myocardial oedema in Kawasaki disease occur even before coronary artery abnormalities and without concurrent ischaemic damage 112 . Myocarditis in Kawasaki disease tends to develop early, and acute left ventricular dysfunction is generally transient and responds readily to anti-inflammatory treatment 116 . However, Kawasaki disease myocarditis might be associated with fatal arrhythmias in infants, and in certain cases might lead to long-term complications including myocardial fibrosis 81, 117 . Therefore, myocarditis during Kawasaki disease and its potential consequences deserve serious investigation, and long-term studies into late adulthood are needed. Kawasaki disease affects small and medium sized vessels, particularly the coronary arteries; however, dilatations and aneurysms can occur systemically, including in the axillary, subclavian, brachial, renal and iliac arteries as well as the abdominal aorta 23, [118] [119] [120] . Post-mortem findings have revealed that 73% of patients with Kawasaki disease have renal artery involvement and acute kidney injury 121 involving glomerulonephritis with intracapillary changes and deposition of immune complex composed of IgA and complement component 3 (C3) 22, 122, 123 . These findings are comparable to those in two other human vasculitis diseases, IgA vasculitis (IgAV) and IgA nephropathy (IgAN), which are similarly characterized by IgA immune complexes with C3 deposition in kidney glomeruli (see below). Increased concentrations of circulating IgA and secretory IgA (sIgA) have been reported in the serum of children with Kawasaki disease during the acute phase 124 . IgA + plasma cells are present in the coronary artery wall and in non-vascular tissues, such as the kidney, trachea and pancreas of patients with Kawasaki disease 25, 26 . This IgA response is oligoclonal, seems to be antigen driven and might be caused by Kawasaki disease-triggering agents 125, 126 . The IL-1 signalling pathway. Evidence from mouse models of Kawasaki disease 11, 127, 128 , as well as transcriptome analysis performed on whole blood of patients with Kawasaki disease during the acute or convalescent phase 129, 130 , demonstrate the involvement of innate immune cells and inflammasome overactivation throughout the acute phase of the disease. In vitro cultured PBMCs isolated from patients with Kawasaki disease spontaneously release IL-1β into the supernatant, and this process is substantially reduced after IVIG treatment 28 . Serum concentrations of both IL-1β and IL-18 are also higher in children with acute Kawasaki disease than in control patients with other febrile illnesses, and markedly decrease during the convalescent phase 59 , supporting the concept of activation of the NLRP3 inflammasome complex. Similarly, IL-1 and NLRP3-related gene transcripts are upregulated in PBMCs from patients with acute Kawasaki disease and are decreased during the convalescent phase of the disease 59 , and an IL1B-gene-related signature is associated with acute phase disease and IVIG resistance 130 . Furthermore, a study has shown that differential expression of IL-1β and related signalling genes might have a role in mediating the sex-based differences seen in patients with Kawasaki disease 131 . In the LCWE mouse model of Kawasaki disease, the activation of caspase 1, IL-1α and IL-1β is key to the development of coronary arteritis, aneurysms, myocarditis and abdominal aorta aneurysms 127, 128, 132 . IL-1 has the capacity to expand and promote the differentiation of antigen-specific CD8 + T cells 133 , and indeed the frequencies of circulating CD4 + and CD8 + T cells are increased in patients with Kawasaki Nature reviews | Rheumatology disease 134 . Infiltrations of mature dendritic cells as well as activated cytotoxic CD8 + T cells have been reported in arterial layers of coronary aneurysms 24, 135 . Therefore, blocking the NLRP3-IL-1β pathway seems to be a valid therapeutic option in Kawasaki disease. Intestinal permeability. The intestinal barrier has a critical role in maintaining intestinal homeostasis and health by preventing harmful organisms and luminal antigens from entering the circulation. A dysfunctional intestinal barrier, characterized by increased intestinal permeability, is recognized as a pathogenic factor in many inflammatory diseases 136 . In Kawasaki disease, abdominal pain, diarrhoea and vomiting are often observed at the onset of acute illness, affecting up to 60% of diagnosed patients and indicating that the gastrointestinal tract is also affected 4,137-140 . A multicentre study of >300 patients revealed that gastrointestinal manifestations at onset of disease complicate diagnosis, delay adequate treatment and correlate with IVIG resistance and severity of CAAs 141 . Immunohistochemical studies have revealed higher numbers of activated CD4 + T cells and macrophages along with lower numbers of CD8 + T cells in the jejunum lamina propria in patients with Kawasaki disease than in control patients with diarrhoea from cows' milk protein intolerance 142 . However, these cellular abnormalities are specific to the acute phase of the disease and return to normal during the convalescent phase 142 . IgA + plasma cells have also been observed in a variety of different vascular and non-vascular tissues in patients with Kawasaki disease 26 , and patients with Kawasaki disease also have increased concentrations of sIgA, which is produced at the intestinal mucosal surface, in their serum 124 . These studies indicate that the gastrointestinal tract is affected during Kawasaki disease and that mucosal immune activation might compensate and protect from defective intestinal barriers. The role of gut-related immunity in the induction of inflammation in organ systems distant from the gut has been the subject of intensive investigation. We have observed increased intestinal permeability and a dysregulated intestinal immune response characterized by increased numbers of IgA + B cells in the Peyer's patches in the LCWE-induced mouse model of Kawasaki disease 143 (Fig. 3) 149, 150 and to improve intestinal barrier function by increasing the expression of intestinal tight junctions 151, 152 , enhancing the intestinal mucus layer 153 and modulating the intestinal microbiota composition 154 . Lactobacilli have also been shown to boost innate and immune functions against a variety of bacterial infections [155] [156] [157] , and their disappearance during acute Kawasaki disease might lead to the blooming of other bacterial pathogens, which might further promote intestinal barrier dysfunction and inflammation. Intriguingly, a retrospective study of 364 patients with Kawasaki disease showed that children who received microbiomealtering antibiotics in the week before Kawasaki disease diagnosis were substantially more likely to have IVIGresistant disease than those who did not receive antibiotics 158 . Antibiotics alter the abundance, taxonomic richness and diversity of the bacterial 159,160 as well as fungal 161 intestinal microbiome, and those alterations might persist from weeks to years after treatment discontinuation 159, 160, 162 . A longitudinal metagenomic study of faecal samples derived from patients with Kawasaki disease showed a marked increase of five Streptococcus spp. during the acute phase of Kawasaki disease 163 ; however, all patients in that study were treated with antibiotics in the early stage of disease, therefore this observation might be reflective of antibiotic-induced dysbiosis and not Kawasaki disease itself. Nonetheless, how this intestinal dysbiosis occurs and how its effect on intestinal permeability affects the development of cardiovascular lesions during Kawasaki disease vasculitis remains unknown and under-appreciated. Link with IgA vasculitis IgAV, or Henoch-Schönlein purpura, is an IgA-mediated necrotizing vasculitis resulting in fibrinoid destruction of the affected small vessels. Renal involvement, characterized by IgA deposition in the kidney glomeruli, is also observed in IgAV 164 . IgAV nephritis is closely related to another glomerular disease, IgAN, wherein accumulation and deposition of IgA and IgA immune complexes in the kidney glomerular mesangium drive glomerular inflammation 165 . As IgA is mainly found at mucosal surfaces, a 'gut-kidney axis' , influenced by a mix of genetic, microbial and dietary factors, has been suggested to be involved in the development of both IgAN 166 and IgAV in paediatric and adult patients 167 . We have demonstrated that the LCWE-induced mouse model of Kawasaki disease vasculitis is associated with the deposition of IgA and IgA-C3 immune complexes in vascular tissues, such as the inflamed coronary artery and abdominal aorta 143 . Deposited IgA and IgA-C3 immune complexes might result in overactivation of the immune cells present in the cardiovascular lesions and subsequent amplification of inflammation 143 . Substantial evidence indicates that immune complexes might promote vascular damage during human Kawasaki disease through the www.nature.com/nrrheum activation and aggregation of platelets, the release of vasoactive mediators, and the subsequent recruitment of neutrophils and leukocytes to the site of inflammation (reviewed elsewhere 168 ). Interestingly, we have also observed IgA and C3 deposition in the kidney glomeruli of LCWE-injected mice developing Kawasaki disease 143 , and immune complex-mediated nephropathy has also been observed in Kawasaki disease 123 . However, to date IgA deposition has not been reported in CAAs of patients with Kawasaki disease. Given that availability of human tissue samples is limited, and those that are available are usually collected at the end stage of the disease, they might not be representative of active Kawasaki disease pathological features, and further studies are warranted. Like Kawasaki disease, IgAV develops mostly in children, affects males more than females, is more predominant in Asian countries such as Japan and Korea, and is also associated with abdominal pain, diarrhoea, skin rash and IgA deposition in the affected small vessels 169 . IgAN also shares pathological features with Kawasaki disease, such as increased intestinal permeability, low to moderate intestinal inflammation associated with activation of inflammatory cells in the small intestinal mucosa and colocalization of sIgA-complement in the glomerular mesangium 165, 170 . Moreover, a polymorphism in the promoter of the lipopolysaccharide (LPS) receptor CD14 (CD14/159) is associated with coronary artery abnormalities in patients with Kawasaki disease 171 and has been linked to progression of IgAN to more severe renal disease 172 . IL-1β has a key pathogenic role during Kawasaki disease and also seems to be implicated in renal complications related to IgAV 173 and IgAN 174 180 demonstrated that a single intraperitoneal injection of LCWE induces a dose-dependent and chronic polyarthritis in rats. However, when injected into mice, LCWE induces instead a focal coronary arteritis 176 . How and which element of LCWE triggers Kawasaki disease vasculitis is unknown. LCWE is mainly composed of peptidoglycans, contains high levels of rhamnose and is resistant to lysozyme degradation 176 . The cardiovascular lesions induced in mice by LCWE are histologically similar to those observed in human disease. LCWE-induced Kawasaki disease vasculitis is characterized by infiltration of inflammatory cells in the aortic root, development of necrotizing arteritis in the coronary artery followed by luminal obstruction due to LMP that can lead to complete coronary artery stenosis 181 , recapitulating the three pathological processes of human Kawasaki disease described above (Fig. 4a-d) . In children with Kawasaki disease, thrombotic occlusion of the inflamed coronary artery leads to ischaemic heart disease 23, 120 , and similarly, occluding organizing thrombus in the coronary artery can be observed in LCWE-injected mice (Fig. 4e) . Acute myocarditis and chronic scarring of the coronary arteries with the formation of stenotic fragments are also observed in LCWE-induced Kawasaki disease vasculitis (Fig. 4f) , even long after the acute phase 182 , which is similar to the fibrotic lesions that might lead children with Kawasaki disease to develop long-term cardiovascular sequelae in adulthood 8, 9 . MRI and echocardiography in LCWE-injected mice demonstrate the presence of electrocardiographic changes (as observed in human Kawasaki disease) and myocardial dysfunction, which are responsive to anakinra therapy 183, 184 . The LCWE-induced Kawasaki disease vasculitis in mice is dependent on intact TLR2 and MyD88 signalling and the subsequent release of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF 10 . Genetic depletion of the TNF receptor or pharmacological blockade of the TNF signalling pathway (with infliximab (monoclonal antibodies to TNF) or etanercept (soluble TNF receptors)) protects mice from LCWEinduced Kawasaki disease vasculitis 132, 185 . This model is also T cell dependent, as Rag1 -/mice develop fewer cardiovascular lesions 11 . CD8 + T cells are specifically required for LCWE-induced Kawasaki disease vasculitis as treatment of LCWE-injected mice with an anti-CD8-depleting antibody prevents the development of vasculitis 181 . This finding correlates with human disease, in which infiltrations of CD3 + T cells 135 , and particularly CD8 + T cells, are detected in the CAAs 24 . The LCWE model has also confirmed the importance 59 . Interestingly, the relatively mild development of coronary arteritis in LCWE-injected CBA/N mice -which are characterized by a defective B cell maturation process and poor humoral immune responses -suggests that the humoral immune response might participate in amplification of the disease 186 . IgA + plasma cells infiltrate vascular and non-vascular tissues during the acute phase of Kawasaki disease 25, 26 , resulting in the development of an oligoclonal IgA response in the coronary artery 125, 126 . Interestingly, we have observed increased numbers of IgA + plasmablasts in the spleen, Peyer's patches and abdominal aorta draining lymph nodes of LCWE-injected mice, as well as increased concentrations of circulating IgA and IgA deposition in heart tissues, abdominal aorta and kidney glomureli 143 . Mouse models also provide a useful opportunity to evaluate the efficacy of therapeutic regimens on the development and healing of cardiovascular lesions. When given up to 5 days after LCWE injection, IVIG substantially decreases the severity of cardiovascular lesions in mice 187 , mirroring the effects of IVIG treatment in humans. As described above, IL-1β signalling is higher in patients with Kawasaki disease than in age-matched control patients with other febrile illnesses 91, 188 , and studies using the LCWE model helped lead to the discovery of the importance of this pathway in the pathogenesis of the disease and the therapeutic potential of IL-1 blockade. Depletion of macrophages or blocking the IL-1 pathway either genetically using IL1R −/− , IL1α −/− or IL1β −/− mice or with antibodies targeting IL-1α or IL-1β, or anakinra (IL1Ra), strongly reduces cardiovascular lesion development as well as myocardial dysfunction in LCWE-injected mice 128, 132, 184 . The CAWS mouse model. C. albicans is a harmless commensal fungus normally present in the human gastrointestinal tract that can transition into a pathogen capable of inducing inflammation in immune-impaired hosts. In 1979, Murata demonstrated that an alkaline extract made from C. albicans isolated from faeces from a patient with Kawasaki disease induced coronary arteritis in mice 177 . CAWS is composed of polysaccharides, mainly β-glucans and α-mannan proteins of the yeast cell wall 189 , and needs to be injected intraperitoneally for five consecutive days in the first week of the disease to induce vasculitis in the aortic valves and the coronary arteries 189, 190 . In this model, recognition of α-mannan proteins by the dectin-2 receptor seems to be essential, as CAWS-injected Dectin-2 −/− mice do not develop vasculitis 191 . The CAWS model shares some histological similarities with human Kawasaki disease pathology in that inflammation affects both the aortic root and the proximal region of the coronary arteries 190 . Inflammation can also affect non-coronary artery sites in 25% of CAWS-injected mice and can be observed in the lymph nodes, the kidneys and the liver 190, 192 . CAWS-induced coronary artery lesions resemble those of human Kawasaki disease and are typically proliferative, granulomatous and characterized by intimal thickening with destruction of the elastic lamina and media 190 . Wild-type mice underwent intraperitoneal injection with Lactobacillus casei cell wall extract (LCWE), and heart tissues were harvested 2 weeks later. Haematoxylin and eosin (H&E) and trichrome staining were performed on heart sections. a | Inflammatory cell infiltration in the aortic route (H&E staining; ×40). b | Arteritis development in epicardial muscular coronary artery (H&E staining; ×20). c | Luminal myofibroblast proliferation (LMP) and non-specific neointimal proliferation injury to the arterial wall (trichrome staining; ×200). d | Complete occlusion of the coronary artery by LMP (trichrome staining; ×20). e | Organized thrombus in the coronary artery (H&E staining; ×200). f | Myocarditis (H&E staining; ×200). Ao; aorta, CA; coronary artery. Echocardiography in CAWS-injected mice indicates a marked decrease of cardiac function, which can be restored by IL-10 supplementation 193 . IL-10 is a potent anti-inflammatory cytokine that might improve the outcome of CAWS-induced vasculitis by inhibiting the release of pro-inflammatory mediators, such as TNF and IL-1β, from tissue-infiltrating innate immune cells 194 . Interestingly, CAWS-induced Kawasaki disease vasculitis is also strain dependent, as CAWS injections lead to a high incidence of vasculitis in CD-1, C3H/HeN, DBA/2 and C57BL/6N mice, but the CBA/JN strain is resistant to coronary arteritis 190, 195 . The DBA/2 strain is the most sensitive, with the highest mortality rate resulting from a more intense coronary arteritis 195 . The sensitivity of DBA/2 mice is associated with increased production of the pro-inflammatory cytokines TNF, IL-6 and IFNγ 195, 196 , whereas resistance of CBA/JN mice is explained by increased levels of IL-10 production in that strain 197 . Despite the presence of T cell and B cell infiltration in the inflamed coronary artery, mice lacking T cells still develop moderate to typical cardiac inflammation, indicating that T cells might not be required in the development of Kawasaki disease vasculitis in this particular model 198, 199 . Absence of both T cells and B cells in Rag1 −/− mice leads to lower incidence of CAWS-induced Kawasaki disease vasculitis; reconstitution of Rag1 −/− mice with wild-type, but not CCR2 −/− , T cells and B cells restores cardiovascular lesions, suggesting roles for both T cells and B cells and the modulation of disease development by CCR2 expression 200 . The innate immune response also participates in vasculitis development; resident macrophages recognize the CAWS antigens through the dectin-2 receptor, leading to their activation, release of CCL2, and recruitment of neutrophils and inflammatory monocytes producing IL-1β in the aortic root 201 . CAWS-induced vasculitis is also associated with the rapid production of granulocyte-monocyte colonystimulating factor in the heart, which subsequently drives inflammatory myocarditis by activating tissue macrophages and promoting recruitment of neutrophils and monocytes 199 . TNF is also produced during the acute phase of CAWS-induced Kawasaki disease vasculitis and is essential for the development of acute myocarditis, as TNF receptor-deficient mice are protected from the development of CAWS vasculitis 202 . IVIG administration substantially reduces CAWS-induced heart vessel inflammation 203 . Like the LCWE model, the CAWS model is also dependent on the IL-1 pathway, as IL1R −/− , IL1β −/− , Asc −/− and Nlrp3 −/− mice are protected from induction of vasculitis, and treatment with anti-IL-1β agents substantially attenuates CAWS vasculitis 202, 204, 205 . The Nod1 ligand mouse model. Endothelial cells are equipped to sense microbial components through Toll-like receptors and nucleotide-binding oligomerization domain-containing protein like receptors. Subcutaneous injection or oral delivery of FK565, a specific synthetic Nod1 ligand, in mice primed with LPS results in a diffuse cellular inflammation of the aortic root and transmural infiltration of inflammatory cells in the coronary artery wall 178, 206 . Other arteries, such as the iliac and renal arteries, also show signs of inflammation associated with a thickening of the intima 206 . The mechanisms by which FK565 induces coronary arteritis in mice remain unknown. When administered orally, FK565 does not induce intestinal mucosa inflammation, but specifically activates vascular cells to produce a diverse array of pro-inflammatory cytokines, including IL-1β 206 , and chemokines such as CCL2, resulting in the recruitment of inflammatory cells in the tissues 178 . This model seems to be independent of T cells, B cells and natural killer T cells, as LPS-primed Rag-1 −/− mice still develop aortitis and coronary arteritis after FK565 injection 207 . The inflammatory infiltrates observed around the inflamed aortic root and coronary arteries mainly comprise neutrophils and CD11c + cardiac macrophages; their specific depletion considerably reduces the development of FK565-induced Kawasaki disease vasculitis 178, 207 . The concentration of circulating IL-1β is substantially increased in the serum of FK565-injected mice compared with control or CAWS-injected animals, and higher IL-1β levels correlate with a larger inflammation area 206 . However, specific studies further investigating the role of IL-1β in this model are needed. Traditional and novel therapies in humans. The current standard of care for Kawasaki disease is the use of high-dose IVIG together with aspirin. If given during the first 10 days of the disease, IVIG reduces the risk of development of coronary arteritis and aneurysms from about 30% to 5-7% 14, 15 . The mechanisms by which IVIG treatment reduces the inflammatory responses are still unknown; however, IVIG is suspected to have a wide spectrum of action targeting multiple arms of the immune response 18 . IVIG has been shown to inhibit IL-1β production from in vitro stimulated macrophages and to stimulate the production of IL-1Ra 208, 209 . During Kawasaki disease, IVIG reduces production of inflammatory cytokines and chemokines, and decreases the activation and number of circulating neutrophils, monocytes, macrophages and activated T cells by saturating Fc receptors 18 . The majority of patients with Kawasaki disease who are treated with IVIG improve and do not develop coronary artery damage; however, up to 20% of children with Kawasaki disease do not respond to treatment or have fever recurrence after initial IVIG treatment, and these patients are at the highest risk of developing coronary artery lesions 3, 20, 210 . The involvement of pro-inflammatory cytokines in the acute phase of Kawasaki disease suggests that combinational therapy, composed of IVIG associated with TNF inhibitors, steroids, calcineurin inhibitors or anakinra, might be useful to treat patients with IVIG-resistant disease. The use of TNF inhibitors in combination with IVIG has had mixed results thus far. Infliximab was associated with decreased fever duration and reduced markers of inflammation (C-reactive protein and neutrophil counts), suggesting a possible improvement of coronary artery outcomes 211 ; however, etanercept treatment www.nature.com/nrrheum resulted in a substantial reduction in IVIG resistance only in patients >1 year old 212 . An important area of research is the use of biomarkers to predict IVIG resistance in Kawasaki disease. The Kobayashi scoring system, based on a combination of laboratory test results (for example, C-reactive protein levels, neutrophil percentages, platelets counts and levels of aspartate and alanine aminotransferase) and demographic variables (sex, age and number of days of illness before the start of the treatment) has been successfully used to predict IVIG-resistance in Japanese patients 213 , but not in North American children with Kawasaki disease 214 . The combination of prednisolone and IVIG to treat Japanese patients with Kawasaki disease predicted to have IVIG-resistant disease according to the Kobayashi score (RAISE study) resulted in more rapid fever resolution, reduced development of CAAs and lower incidence of additional rescue treatment 215 compared with IVIG alone. As discussed above, Kawasaki disease susceptibility and increased coronary artery lesion risk are associated with an SNP in ITPKC 58 that results in a lack of NFAT regulation and activation of the T cell compartment owing to increased IL-2 production 216 . CD8 + cytotoxic T cells are present in the inflamed arterial wall during Kawasaki disease 24, 135 ; therefore, targeting T cell expansion might be an efficient approach to preventing CAAs during Kawasaki disease. A combination treatment of IVIG and ciclosporin, a calcineurin inhibitor that suppresses IL-2 production and T cell activation, was tested in a clinical trial in Japanese patients with Kawasaki disease predicted to have IVIG-resistant disease based on the Kobayashi score (KAICA trial) 217 . In this trial, the combination treatment was shown to be safe and associated with a lower incidence of CAAs; however, treatment was linked with increased risk of relapse 217 . Furthermore, the scoring system used to identify IVIG-non-responders is poorly predictive in European children with Kawasaki disease, limiting the conclusions of this study. The important role of the IL-1β-IL-1 receptor pathway in Kawasaki disease development has been demonstrated in both human patients 27, 28, 129, 130 and mouse models 127, 132, 202, 204 . Therefore, clinical trials investigating IL-1 pathway inhibition by using anakinra, which blocks both IL-1α and IL-1β, have been initiated in North America (ANAKID; ClinicalTrials.gov identifier NCT02179853) 218 and Europe (Kawakinra; European Clinical Trials number 2014-002715-4) 219 . Already, multiple case reports exist of the successful use of anakinra to treat patients with IVIG-resistant Kawasaki disease 220-224 , indicating the promise of this second-line therapy. Therapeutic insights from mouse models. Although no animal model can fully mimic human disease, the LCWE-induced Kawasaki disease mouse model has been accepted by many in the research community as a reliable experimental model providing novel insights that can be tested in patients. For example, IVIG efficiently prevents coronary arteritis development in LCWE-injected mice 187 as well as in the CAWS mouse model of Kawasaki disease 203 . The effects of the calcineurin inhibitors ciclosporin and tacrolimus have been investigated in the Nod1 ligand-induced mouse model of Kawasaki disease vasculitis 225 . This approach was rational given the established role of T cells and calcium signalling in Kawasaki disease. However, contrary to the expected outcome, these inhibitors exacerbated the coronary arteritis 225 . Notably, however, this result was probably related to the choice of mouse model, as the Nod1 ligand-mediated mouse model of Kawasaki disease vasculitis has previously been shown to be T cellindependent 207 . Indeed, in an independent study using the CAWS mouse model, which is T cell dependent, ciclosporin suppressed CAWS-induced vasculitis 226 , emphasizing the importance of model selection in preclinical studies. Most importantly, results in human studies bear out the therapeutic potential of calcineurin inhibition, as the Japanese phase III trial (KAICA trial) showed that adding ciclosporin to IVIG in patients with Kawasaki disease who were at high risk of IVIG resistance was beneficial in diminishing overall incidence of CAAs 217 . The role of TNF has been investigated in both the LCWE and the CAWS mouse models of Kawasaki disease vasculitis 185 . Initially, etanercept treatment or genetic deletion of TNF receptor 1 was shown to protect mice from LCWE-induced coronary arteritis 185, 202 . Infliximab treatment also prevented the development of both LCWE-induced coronary arteritis and myocarditis 132 . Similar results were obtained in the CAWS mouse model of Kawasaki disease vasculitis, in which etanercept 226, 227 suppressed the incidence and decreased the severity of vasculitis. Mechanistically, TNF has been proposed to be produced by myeloid cells in the acute phase and to promote myocarditis and recruitment of immune cells by acting on cardiac stromal cells 202 . However, infliximab and etanercept might not directly target the TNF signalling pathway, and their observed effects might be indirect. Indeed, infliximab is not able to bind mouse TNF 227, 228 ; therefore, the anti-inflammatory effect of infliximab might be attributable to the binding of Fc receptors at the surface of activated cells 229, 230 . The overwhelming evidence for the critical role of IL-1β in promoting LCWE-induced Kawasaki disease vasculitis in mice 127, 128, 132 led to the initiation of clinical trials testing the effect of anakinra for blocking IL-1β as a second therapy option to treat children with IVIG-resistant Kawasaki disease. Multiple case reports now outline the successful use of anakinra to treat patients with IVIG-resistant Kawasaki disease [221] [222] [223] [224] . Alternatively, direct inhibition of the NLRP3 inflammasome might be a more targeted therapeutic strategy to treat Kawasaki disease, as it would affect several pathways beyond IL-1β, including IL-1α and IL-18. Several NLRP3 inhibitors have been identified 231 and tested in mouse models of inflammatory diseases, such as experimental autoimmune encephalomyelitis and cryopyrin-associated periodic syndrome 232 . It would be interesting to determine if such drugs could be used to prevent and reduce the cardiovascular complications in mouse models of Kawasaki disease vasculitis. Over the past 40 years, research has improved our understanding of Kawasaki disease pathology and the development of coronary vasculitis. However, some questions still remain unanswered, such as the identification of the aetiological agents, how the disease is triggered, and the specific immune pathways associated with coronary vasculitis development and IVIG resistance. Owing to the rarity of human tissues from patients with Kawasaki disease, the use of animal models reproducing human Kawasaki disease features is invaluable. Many advances have been made over the decades by combining biological observations in human samples with mechanistic insights from experimental animal models. This 'bench to bedside' approach successfully led to the identification of the critical role of IL-1β in Kawasaki disease and resulted in the development of clinical trials in which anakinra is being used to treat children with IVIG-resistant Kawasaki disease. LCWE-injected mice exhibit a dysfunctional intestinal barrier, and the increased IgA response and elevated sIgA levels in both LCWE-injected mice and children with Kawasaki disease reveal the existence of a 'gutvascular' axis 143 . In evaluating this model system and the role of IgA, it should not be forgotten that injection of identically prepared LCWE induces chronic polyarthritis in selected inbred rat strains 180, 233 . This observation implies that a common immunogenetic pathway might underlie a variety of autoimmune illnesses, with disease expression moderated not by the inducing agent, but rather by host genetics. The fact that cell wall fragments of common gut bacteria can produce varying disease manifestations in the face of inflammation-induced increased gut permeability suggests that some autoimmune diseases might not in fact be induced by the normal response to an unusual agent, but rather an unusual response to a common agent. Similarly, we hypothesize that vasculitic diseases, including Kawasaki disease, are not a usual response to an unusual environmental stimulus, but rather an unusual response (genetically determined) to a common environmental stimulus. This hypothesis has major implications for understanding the aetiology and pathogenesis of not only Kawasaki disease but also IgA-mediated diseases and perhaps others. In addition, it strongly suggests that inhibition of IL-1β might be effective for the many chronic inflammatory diseases in which IgA deposition is a key finding. Published online xx xx xxxx A new infantile acute febrile mucocutaneous lymph node syndrome (MLNS) prevailing in Japan Diagnosis, treatment, and long-term management of Kawasaki disease: a scientific statement for health professionals from the American Heart Association Diagnosis, treatment, and long-term management of Kawasaki disease: a statement for health professionals from the Committee on Rheumatic Fever, Endocarditis and Kawasaki Disease, Council on Cardiovascular Disease in the Young The epidemiology of Kawasaki disease: a global update When children with Kawasaki disease grow up: myocardial and vascular complications in adulthood Prevalence of Kawasaki disease in young adults with suspected myocardial ischemia Acute myocardial ischemia in adults secondary to missed Kawasaki disease in childhood The spectrum of cardiovascular lesions requiring intervention in adults after Kawasaki disease TLR2 and MyD88 contribute to Lactobacillus casei extract-induced focal coronary arteritis in a mouse model of Kawasaki disease Involvement of innate and adaptive immunity in a murine model of coronary arteritis mimicking Kawasaki disease Searching for the cause of Kawasaki disease-cytoplasmic inclusion bodies provide new insight Ultrastructural, immunofluorescence, and RNA evidence support the hypothesis of a new virus associated with Kawasaki disease Intravenous gammaglobulin treatment and retreatment in Kawasaki disease. US/Canadian Kawasaki Syndrome Study Group Resistance to intravenous immunoglobulin in children with Kawasaki disease Long-term consequences of Kawasaki disease. A 10-to 21-year follow-up study of 594 patients Coronary artery aneurysms in Kawasaki disease: risk factors for progressive disease and adverse cardiac events in the US population The immunomodulatory effects of intravenous immunoglobulin therapy in Kawasaki disease Epidemiology of immunoglobulin resistant Kawasaki disease: results from a large, national database Kawasaki disease outcomes and response to therapy in a multiethnic community: a 10-year experience Nationwide epidemiologic survey of Kawasaki disease in Japan Kawasaki disease as a systemic vasculitis in childhood Histopathological aspects of cardiovascular lesions in Kawasaki disease CD8 T lymphocytes and macrophages infiltrate coronary artery aneurysms in acute Kawasaki disease IgA plasma cells in vascular tissue of patients with Kawasaki syndrome IgA plasma cell infiltration of proximal respiratory tract, pancreas, kidney, and coronary artery in acute Kawasaki disease Two monokines, interleukin 1 and tumor necrosis factor, render cultured vascular endothelial cells susceptible to lysis by antibodies circulating during Kawasaki syndrome Endothelial cell activation and high interleukin-1 secretion in the pathogenesis of acute Kawasaki disease Is Kawasaki disease an infectious disorder? Epidemiology of Kawasaki disease in Asia, Europe, and the United States Hospitalizations for Kawasaki syndrome among children in the United States The mechanism of superantigenmediated toxic shock: not a simple Th1 cytokine storm Selective expansion of T cells expressing T-cell receptor variable regions V beta 2 and V beta 8 in Kawasaki disease Characterization of T cell repertoire changes in acute Kawasaki disease Evidence for a superantigen mediated process in Kawasaki disease Characterization of the T-cell receptor V-beta repertoire in Kawasaki disease TCR V beta family repertoire and T cell activation markers in Kawasaki disease Kawasaki disease and Epstein-Barr virus Recurrence of Kawasaki disease and Epstein-Barr virus infection Epstein-Barr virus genome-positive T lymphocytes in a boy with chronic active EBV infection associated with Kawasaki-like disease Epstein-Barr virus and other herpesvirus infections in Kawasaki syndrome Children with Kawasaki disease are not infected with Epstein-Barr virus Epstein-Barr virus antibodies in Kawasaki disease Association between a novel human coronavirus and Kawasaki disease Lack of association between New Haven coronavirus and Kawasaki disease Kawasaki disease and human coronavirus Does Kawasaki disease have a retroviral aetiology Polymerase activity in lymphocyte culture supernatants from patients with Kawasaki disease Retrovirus serology and Kawasaki syndrome Lack of evidence of retroviral involvement in Kawasaki disease A protein epitope targeted by the antibody response to Kawasaki disease Monoclonal antibodies from children with Kawasaki disease (KD) recognize hepacivirus peptides Tropospheric winds from northeastern China carry the etiologic agent of Kawasaki disease from its source to Japan Kawasaki disease in families The inositol 1,4,5-trisphosphate receptor is essential for T-cell receptor signaling ITPKC single nucleotide polymorphism associated with the Kawasaki disease in a Taiwanese population ITPKC functional polymorphism associated with Kawasaki disease susceptibility and formation of coronary artery aneurysms Inositol-triphosphate 3-kinase C mediates inflammasome activation and treatment response in Kawasaki disease Lack of association between ORAI1/CRACM1 gene polymorphisms and Kawasaki disease in the Taiwanese children Variations in ORAI1 gene associated with Kawasaki disease Genetic variation in the SLC8A1 calcium signaling pathway is associated with susceptibility to Kawasaki disease and coronary artery abnormalities Critical role for calcium mobilization in activation of the NLRP3 inflammasome Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors Molecular mechanism and function of CD40/CD40L engagement in the immune system Expression of CD40 ligand on CD4+ T-cells and platelets correlated to the coronary artery lesion and disease progress in Kawasaki disease CD40 ligand gene and Kawasaki disease Genetic polymorphisms in the CD40 ligand gene and Kawasaki disease CD40 gene polymorphisms associated with susceptibility and coronary artery lesions of Kawasaki disease in the Taiwanese population The role of mannose-binding lectin in health and disease Polymorphisms in the mannose-binding lectin gene as determinants of age-defined risk of coronary artery lesions in Kawasaki disease Association of mannose-binding lectin genotype with cardiovascular abnormalities in Kawasaki disease Association of mannose-binding lectin gene polymorphisms with Kawasaki disease in the Japanese Involvement of mannose-binding lectin in the pathogenesis of Kawasaki disease-like murine vasculitis Fcgamma receptor IIa polymorphism in Caucasian patients with systemic lupus erythematosus: association with clinical symptoms Immunoglobulin G fc-receptor (FcgammaR) IIA, IIIA, and IIIB polymorphisms related to disease severity in rheumatoid arthritis Fc gamma receptor IIa (CD32) polymorphism in fulminant meningococcal septic shock in children The involvement of Fc gamma receptor gene polymorphisms in Kawasaki disease Genome-wide association study identifies FCGR2A as a susceptibility locus for Kawasaki disease Adjunctive therapies in Kawasaki disease Three linked vasculopathic processes characterize Kawasaki disease: a light and transmission electron microscopic study S100 proteins in rheumatic diseases Gene expression profiling of the effect of high-dose intravenous Ig in patients with Kawasaki disease Circulating markers of inflammation persist in children and adults with giant aneurysms after Kawasaki disease Monocyte-derived interleukin-1β as the driver of S100A12-induced sterile inflammatory activation of human coronary artery endothelial cells: implications for the pathogenesis of Kawasaki disease Matrix metalloproteinases in vascular physiology and disease The pathophysiology of coronary artery aneurysms in Kawasaki disease: role of matrix metalloproteinases Matrix metalloproteinase haplotypes associated with coronary artery aneurysm formation in patients with Kawasaki disease Cell distribution differences of matrix metalloproteinase-9 and tissue inhibitor of matrix metalloproteinase-1 in patients with Kawasaki disease Matrix metalloproteinase (MMP)-3 activates MMP-9 mediated vascular smooth muscle cell migration and neointima formation in mice Gene-expression patterns reveal underlying biological processes in Kawasaki disease Tissue inhibitor of metalloproteinases-1 and matrix metalloproteinase-3 in Japanese healthy children and in Kawasaki disease and their clinical usefulness in juvenile rheumatoid arthritis Interleukin-1 promotes proliferation of vascular smooth muscle cells in coordination with PDGF or a monocyte derived growth factor Essential roles of IκB kinases α and β in serum-and IL-1-induced human VSMC proliferation Genetic inactivation of IL-1 signaling enhances atherosclerotic plaque instability and reduces outward vessel remodeling in advanced atherosclerosis in mice Matrix metalloproteinase 9 activity leads to elastin breakdown in an animal model of Kawasaki disease Inhibition of matrix metalloproteinase-9 activity improves coronary outcome in an animal model of Kawasaki disease Anti-inflammatory microRNAs and their potential for inflammatory diseases treatment The art of microRNA research Bone marrow-derived microRNA-223 works as an endocrine genetic signal in vascular endothelial cells and participates in vascular injury from Kawasaki disease Next-generation sequencing identifies micro-RNA-based biomarker panel for Kawasaki disease A study of cardiovascular miRNA biomarkers for Kawasaki disease Differential expression of miR-145 in children with Kawasaki disease miR-483 targeting of CTGF suppresses endothelial-to-mesenchymal transition: therapeutic implications in Kawasaki disease miR-143 and miR-145: molecular keys to switch the phenotype of vascular smooth muscle cells MicroRNA-modulated targeting of vascular smooth muscle cells miR-23a downregulation modulates the inflammatory response by targeting ATG12-mediated autophagy miR-23a regulates cardiomyocyte apoptosis by targeting manganese superoxide dismutase Active remodeling of the coronary arterial lesions in the late phase of Kawasaki disease: immunohistochemical study Not just coronary arteritis, Kawasaki disease is a myocarditis, too Myocarditis and Kawasaki disease Histopathological characteristics of myocarditis in acute-phase Kawasaki disease Cardiac biopsy of Kawasaki disease Myocarditis in Kawasaki syndrome. A minor villain? The detection of ventricular dysfunction and carditis in children with Kawasaki disease using equilibrium multigated blood pooling ventriculography and 99Tcm-HMPAO-labelled WBC heart scans Noncoronary cardiac abnormalities are associated with coronary artery dilation and with laboratory inflammatory markers in acute Kawasaki disease Kawasaki disease update Transient dilatation of the abdominal aorta in an infant with Kawasaki disease associated with thrombocytopenia Atypical Kawasaki disease with aortic aneurysm On the morphological alterations corresponding to the clinical manifestations Kawasaki disease as a systemic vasculitis in childhood Kidney and urinary tract involvement in Kawasaki disease Clinical features of acute kidney injury in patients with Kawasaki disease High levels of IgA-containing circulating immune complex and secretory IgA in Kawasaki disease Cloning the arterial IgA antibody response during acute Kawasaki disease Oligoclonal IgA response in the vascular wall in acute Kawasaki disease IL-1 signaling is critically required in stromal cells in Kawasaki disease vasculitis mouse model: role of both IL-1α and IL-1β Role of interleukin-1 signaling in a mouse model of Kawasaki disease-associated abdominal aortic aneurysm Global gene expression profiling identifies new therapeutic targets in acute Kawasaki disease Transcript abundance patterns in Kawasaki disease patients with intravenous immunoglobulin resistance Iinterleukin-1-mediated sex differences in Kawasaki disease vasculitis development and response to treatment Interleukin-1β is crucial for the induction of coronary artery inflammation in a mouse model of Kawasaki disease IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells Circulating CD4+CD8+ T lymphocytes in patients with Kawasaki disease Activated myeloid dendritic cells accumulate and co-localize with CD3+ T cells in coronary artery lesions in patients with Kawasaki disease Intestinal permeability-a new target for disease prevention and therapy Small bowel pseudo-obstruction in Kawasaki disease The surgical manifestations of the intestinal tract in Kawasaki disease Associated symptoms in the ten days before diagnosis of Kawasaki disease Intestinal involvement in Kawasaki disease Gastrointestinal presentation of Kawasaki disease: a red flag for severe disease? Immunohistochemical studies on small intestinal mucosa in Kawasaki disease Intestinal permeability and IgA provoke immune vasculitis linked to cardiovascular inflammation The contributory role of gut microbiota in cardiovascular disease The gut microbiome and its role in cardiovascular diseases Characteristic profile of intestinal microflora in Kawasaki disease Microbiologic studies on the small intestine in Kawasaki disease Heat shock proteins and superantigenic properties of bacteria from the gastrointestinal tract of patients with Kawasaki disease Probiotics for the prevention of pediatric antibiotic-associated diarrhea Probiotics for treating acute infectious diarrhoea Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier Lactobacillus rhamnosus GG prevents epithelial barrier dysfunction induced by interferongamma and fecal supernatants from irritable bowel syndrome patients in human intestinal enteroids and colonoids Probiotics inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression Regulation of intestinal homeostasis and immunity with probiotic lactobacilli Lactobacillus paracasei feeding improves immune control of influenza infection in mice Lactobacillus paracasei feeding improves the control of secondary experimental meningococcal infection in flu-infected mice Lactobacillus gasseri suppresses the production of proinflammatory cytokines in Helicobacter pylori-infected macrophages by inhibiting the expression of ADAM17 Variability in response to intravenous immunoglobulin in the treatment of Kawasaki disease Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation Antibiotics and the gut microbiota Immunological consequences of intestinal fungal dysbiosis Long-term ecological impacts of antibiotic administration on the human intestinal microbiota Characterization of the gut microbiota of Kawasaki disease patients by metagenomic analysis IgA vasculitis (Henoch-Shönlein purpura) in adults: diagnostic and therapeutic aspects The mucosa-kidney axis in IgA nephropathy The gut-kidney axis in IgA nephropathy: role of microbiota and diet on genetic predisposition Is there IgA from gut mucosal origin in the serum of children with Henoch-Schönlein purpura? Kawasaki disease: the role of immune complexes revisited Childhood IgA vasculitis (Henoch Schonlein Purpura)-advances and knowledge gaps A pathogenic role for secretory IgA in IgA nephropathy A polymorphism in the promoter of the CD14 gene (CD14/-159) is associated with the development of coronary artery lesions in patients with Kawasaki disease Association of the CD14 gene -159C polymorphism with progression of IgA nephropathy Partial response to anakinra in life-threatening Henoch-Schönlein purpura: case report NLRP3 localizes to the tubular epithelium in human kidney and correlates with outcome in IgA nephropathy Interleukin-1 targeting drugs in familial Mediterranean fever: a case series and a review of the literature Coronary arteritis in mice following the systemic injection of group B Lactobacillus casei cell walls in aqueous suspension Experimental candida-induced arteritis in mice. Relation to arteritis in the mucocutaneous lymph node syndrome Nod1 ligands induce site-specific vascular inflammation The Lactobacillus casei group: history and health related applications Polyarthritis in rats following the systemic injection of Lactobacillus casei cell walls in aqueous suspension CD8+ T cells contribute to the development of coronary arteritis in the Lactobacillus casei cell wall extract-induced murine model of Kawasaki disease Myocardial fibrosis after adrenergic stimulation as a long-term sequela in a mouse model of Kawasaki disease vasculitis IL-1-dependent electrophysiological changes and cardiac neural remodeling in a mouse model of Kawasaki disease vasculitis IL-1 receptor antagonist, anakinra, prevents myocardial dysfunction in a mouse model of Kawasaki disease vasculitis and myocarditis TNF-α is necessary for induction of coronary artery inflammation and aneurysm formation in an animal model of Kawasaki disease Variable expression of Lactobacillus casei cell wall-induced coronary arteritis: an animal model of Kawasaki's disease in selected inbred mouse strains Proceedings of the 5th International Kawasaki Disease Symposium Circulating interleukin-1β in patients with Kawasaki disease Chemistry and biology of angiitis inducer, Candida albicans water-soluble mannoprotein-β-glucan complex (CAWS) Histopathological features of murine systemic vasculitis caused by Candida albicans extract-an animal model of Kawasaki disease Recognition of alpha-mannan by dectin 2 is essential for onset of Kawasaki disease-like murine vasculitis induced by Candida albicans cell-wall polysaccharide JNK is critical for the development of Candida albicans-induced vascular lesions in a mouse model of Kawasaki disease Adeno-associated virus vector-mediated interleukin-10 induction prevents vascular inflammation in a murine model of Kawasaki disease Biology of interleukin-10 Murine model of Kawasaki disease induced by mannoprotein-beta-glucan complex, CAWS, obtained from Candida albicans IL-10 is a negative regulatory factor of CAWS-vasculitis in CBA/J mice as assessed by comparison with Bruton's tyrosine kinase-deficient CBA/N mice A murine model of vasculitis induced by fungal polysaccharide Coronary arteritis induced by CAWS (Candida albicans water-soluble fraction) in various strains of mice GM-CSF primes cardiac inflammation in a mouse model of Kawasaki disease Important role of CCR2 in a murine model of coronary vasculitis Dectin-2-induced CCL2 production in tissue-resident macrophages ignites cardiac arteritis TNF and IL-1 play essential but temporally distinct roles in driving cardiac inflammation in a murine model of Kawasaki disease Administration of human immunoglobulin suppresses development of murine systemic vasculitis induced with Candida albicans water-soluble fraction: an animal model of Kawasaki disease Interleukin-1beta inhibition attenuates vasculitis in a mouse model of Kawasaki disease Crucial role of NLRP3 inflammasome in a murine model of Kawasaki disease Characterization of a murine model with arteritis induced by Nod1 ligand, FK565: a comparative study with a CAWS-induced model Identification of pathogenic cardiac CD11c+ macrophages in Nod1-mediated acute coronary arteritis Antipyretic activity of a human immunoglobulin preparation for intravenous use in an experimental model of fever in rabbits Selective induction of interleukin-1 receptor antagonist and interleukin-8 in human monocytes by normal polyspecific IgG (intravenous immunoglobulin) Analysis of potential risk factors associated with nonresponse to initial intravenous immunoglobulin treatment among Kawasaki disease patients in Japan Infliximab for intensification of primary therapy for Kawasaki disease: a phase 3 randomised, double-blind, placebo-controlled trial Etanercept with IVIg for acute Kawasaki disease: a randomized controlled trial Prediction of intravenous immunoglobulin unresponsiveness in patients with Kawasaki disease Evaluation of Kawasaki disease risk-scoring systems for intravenous immunoglobulin resistance Efficacy of immunoglobulin plus prednisolone for prevention of coronary artery abnormalities in severe Kawasaki disease (RAISE study): a randomised, open-label, blinded-endpoints trial Revisiting the concept of targeting NFAT to control T cell immunity and autoimmune diseases Efficacy of primary treatment with immunoglobulin plus ciclosporin for prevention of coronary artery abnormalities in patients with Kawasaki disease predicted to be at increased risk of non-response to intravenous immunoglobulin (KAICA): a randomised controlled, open-label, blindedendpoints, phase 3 trial Rationale and study design for a phase I/IIa trial of anakinra in children with Kawasaki disease and early coronary artery abnormalities (the ANAKID trial) KAWAKINRA: a phase IIA multicenter trial to assess the efficacy, and safety of anakinra in patients with intravenous immunoglobulin-resistant Kawasaki disease A child with severe relapsing Kawasaki disease rescued by IL-1 receptor blockade and extracorporeal membrane oxygenation High dose anakinra for treatment of severe neonatal Kawasaki disease: a case report Usefulness and safety of anakinra in refractory Kawasaki disease complicated by coronary artery aneurysm A child with resistant Kawasaki disease successfully treated with anakinra: a case report Severe late-onset Kawasaki disease successfully treated with anakinra Calcineurin inhibitors exacerbate coronary arteritis via the MyD88 signalling pathway in a murine model of Kawasaki disease Etanercept suppresses arteritis in a murine model of Kawasaki disease: a comparative study involving different biological agents The role of TNF-α in a murine model of Kawasaki disease arteritis induced with a Candida albicans cell wall polysaccharide Anti-inflammatory effects of infliximab in mice are independent of tumour necrosis factor α neutralization Differences in binding and effector functions between classes of TNF antagonists Anti-tumor necrosis factor-α antibodies induce regulatory macrophages in an Fc regiondependent manner Targeting the NLRP3 inflammasome in inflammatory diseases A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases Lactobacillus casei cell wall-induced arthritis in rats: cell wall fragment distribution and persistence in chronic arthritis-susceptible LEW/N and -resistant F344/N rats The work of M.A. is supported by the NIH Grant R01 AI072726 and M.N.R. is supported by the NIH grant R01 HL139766. Both authors contributed equally to all aspects of the manuscript. The authors declare no competing interests. Nature Reviews Rheumatology thanks Ho-Chang Kuo, Isabelle Kone-Paut and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.