Skip to main content
Clinical Medicine Insights. Pediatrics logoLink to Clinical Medicine Insights. Pediatrics
. 2022 Feb 16;16:11795565221075319. doi: 10.1177/11795565221075319

A Review of Coronaviruses Associated With Kawasaki Disease: Possible Implications for Pathogenesis of the Multisystem Inflammatory Syndrome Associated With COVID-19

Fatima Farrukh Shahbaz 1, Russell Seth Martins 1, Abdullah Umair 1, Ronika Devi Ukrani 1, Kausar Jabeen 2, M Rizwan Sohail 3, Erum Khan 4,
PMCID: PMC8859668  PMID: 35197719

Abstract

Multisystem Inflammatory Syndrome in Children (MIS-C), representing a new entity in the spectrum of manifestations of COVID-19, bears symptomatic resemblance with Kawasaki Disease (KD). This review explores the possible associations between KD and the human coronaviruses and discusses the pathophysiological similarities between KD and MIS-C and proposes implications for the pathogenesis of MIS-C in COVID-19. Since 2005, when a case-control study demonstrated the association of a strain of human coronavirus with KD, several studies have provided evidence regarding the association of different strains of the human coronaviruses with KD. Thus, the emergence of the KD-like disease MIS-C in COVID-19 may not be an unprecedented phenomenon. KD and MIS-C share a range of similarities in pathophysiology and possibly even genetics. Both share features of a cytokine storm, leading to a systemic inflammatory response and oxidative stress that may cause vasculitis and precipitate multi-organ failure. Moreover, antibody-dependent enhancement, a phenomenon demonstrated in previous coronaviruses, and the possible superantigenic behavior of SARS-CoV-2, possibly may also contribute toward the pathogenesis of MIS-C. Lastly, there is some evidence of complement-mediated microvascular injury in COVID-19, as well as of endotheliitis. Genetics may also represent a possible link between MIS-C and KD, with variations in FcγRII and IL-6 genes potentially increasing susceptibility to both conditions. Early detection and treatment are essential for the management of MIS-C in COVID-19. By highlighting the potential pathophysiological mechanisms that contribute to MIS-C, our review holds important implications for diagnostics, management, and further research of this rare manifestation of COVID-19.

Keywords: MIS-C, Kawasaki-like disease, PIMS-TS, SARS-CoV-2, human coronaviruses

Background

On May 14, 2020, the CDC issued an advisory regarding a disease with similar symptoms to Kawasaki Disease (KD) in pediatric patients exposed to SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2). However, despite being life-threatening, the disease is considered a rare complication of SARS-CoV-2 infection. 1 The WHO classified this Kawasaki-like disease as “multisystem inflammatory syndrome in children and adolescents temporally related to COVID-19 (MIS-C)” (also known as Pediatric Multisystem Inflammatory Syndrome temporally associated with SARS-CoV-2; PIMS-TS). 2 With the COVID-19 (coronavirus disease 2019) pandemic looming large, the emergence of MIS-C has brought renewed attention and interest to KD. 1

While the specifics are yet unknown, the pathogenesis of KD is commonly thought to involve a post-infectious etiology. 3 It has been repeatedly hypothesized that there exists a connection between respiratory viruses and Kawasaki Disease, 4 with positive viral polymerase chain reaction (PCR) results frequently seen in KD patients. 5 Moreover, some studies propose the existence of a hitherto unidentified respiratory virus (the “KD agent”).4,6 The viral theory of KD pathogenesis is supported by the epidemiological, clinical, histopathological, and laboratory features of KD. 3 One group of respiratory viruses mentioned in association with Kawasaki-like symptoms are the human coronaviruses (HCoVs). 7 Moreover, though KD and MIS-C are separate entities that differ when it comes to age and geographic areas, similarities exist in how they present clinically, as is summarized in Table 1. This review summarizes the literature exploring the association and commonalities between KD and the human coronaviruses and discusses the implications for the pathogenesis of MIS-C in COVID-19.

Table 1.

Similarities and differences in MIS-C and KD.

MIS-C KD
Age 0-19 years 8 (median age 8-99) <5 years 3 (median age 39)
Geographic Area Europe, North America, South America 9 Asia (Japan, South Korea, Taiwan) 3
Clinical similarities8,10 Fever ⩾ 3 days Fever ⩾ 5 days
Rash Polymorphous rash
Bilateral non-purulent conjunctivitis Bilateral non-purulent conjunctivitis
Mucocutaneous inflammation signs (oral, hands, feet) Oral mucous membrane changes (cracked and erythematous lips, strawberry tongue)
Cardiovascular: Features of myocardial dysfunction, pericarditis, valvulitis, or coronary abnormalities Changes in extremities (erythema of hands and feet, desquamation)
Gastrointestinal: Diarrhea, vomiting, abdominal pain Cardiovascular: Coronary artery abnormalities, decreased LV function, mitral regurgitation, pericardial effusion
Gastrointestinal: Diarrhea, nausea, vomiting, abdominal pain

Abbreviations: KD, Kawasaki disease; LV, left ventricle; MIS-C, multisystem inflammatory syndrome related to COVID-19.

The Human Coronaviruses and KD

In 2005, Esper et al 7 reported a possible association of a strain of HCoV-NL63 (termed New Haven Coronavirus) with KD in a case-control study of 11 KD patients and 22 controls (72.7% of KD patients NL63+ vs 4.5% of controls NL63+; P = .015).Subsequently, though several case-control studies have explored the association of coronaviruses with KD, there has been a great deal of conflicting evidence4,5,7,11-16 (Table 2). The differences in results may be due to differences in geographical location, study population, study design, sampling (timing and method of collection, handling, and testing of samples), criteria for selection of cases (inclusion of exclusion of patients with incomplete/atypical KD) and control patients, and coronavirus species tested for.4,5,7,11-22 Furthermore, case series by Shimizu et al, 17 Baker et al 21 (both USA and The Netherlands), Chang et al 18 (Taiwan) and Cho et al 20 (South Korea) have reported HCoV positivity rates ranging from 0% to 2% in KD patients. A case report by Bohlok et al 19 (Lebanon) in 2020 also described atypical KD following HCoV-OC43 infection in 13.5-year-old male, while one by Giray et al 22 (Turkey) in 2016 described HCoV-OC43/HKU1 infection in a 17-month-old female. Interestingly, Rowley et al 23 identified a “corona” of “spikes” surrounding viral particles in bronchial epithelium visualized using transmission electron microscopy, a description that is reminiscent of the crown-like projection of spike proteins that gives the coronaviruses their name. 23 However, despite more than a decade of research exploring the association of the HCoVs with KD, no conclusive evidence has been discovered.

Table 2.

Case control studies exploring the association of Human Coronaviruses with KD.

Author (Year) Country Duration (months) HCoV Species Sample size Mean/Median Age Sample used Method Findings
Case (n) Control (n) Case Control
Esper et al 7 USA (New Haven) 30 NL63 24 Total: 11 Total: 22 24.4 M 23.7 M Respiratory Secretions RT-PCR Cases:
72.7% NL63 +
Controls:
4.5% NL63 +
MH matched OR [95% CI]: 16.0 [3.4-74.4]
P = 0.015
Lehmann et al 11 Germany (South East) 28 NL63
229E
OC43
HKU1
SARS-CoV-1
Total: 21
Incomplete KD: 9
Total: 33 4.5 Y 5.2 Y Serum Serology Cases:
NL63: 48% IgG +, 10% IgM +, 29% IgA +
229E: 19% IgG +, 10% IgM +, 10% IgA +
OC43: 52% IgG +, 5% IgM +, 19% IgA +
HKU1: 29% IgG +, 5% IgM +, 24% IgA +
SARS-CoV-1: 0% IgG +, 0% IgM +, 0% IgA+
Controls:
NL63: 67% IgG +, 3% IgM +, 21% IgA +
229E: 21% IgG +, 3% IgM +, 6% IgA +
OC43: 30% IgG +, 3% IgM +, 12% IgA +
HKU1: 12% IgG +, 0 % IgM +, 6% IgA
SARS-CoV-1: 0% IgG +, 0% IgM +, 0% IgA+
Chi-Squared Test: all p > 0.05
Belay et al 12 USA (San Diego) 1 NL63
229E
OC45
Total: 10 Total: 6 3.6 Y 3.3 Y OP/NP Swab RT-PCR All samples for Cases and Control tested negative for all strains of HCoV
Shirato et al 13 Japan (Tokyo) 24 NL63
229E
Total: 15 Total: 42
Control 1:
23
(NT test with VSV pseudotyped
viruses)
Control 2:
29
(NT Test with HCoV-229E)
2.3 Y 2.2-2.4 Y Serum Serology Cases:
NL63: 47% + in Acute & Recovery Phases
229E: 33% + in Acute % 40% + in Recovery Phase
Control 1:
NL63: 39% +
229E: 9% +
Control 2:
NL63: 51% +
229E: 10% +
Chi-Squared P < 0.05:
229E in Recovery Phase vs. Control 1 and 2
Ebihara et al 14 Japan (Sapporo) 7 NL63 24 Total: 19 Total: 208 22.6 M 21.6 M NP Swab RT-PCR Cases:
0% NL63 +
Controls:
2.4% NL63 +
Dominguez et al 15 USA (Denver) 7 NL63 24 Total: 26
Incomplete KD: 6
Total: 52 46.2 M 40.1 M NP wash RT-PCR Cases:
7.7% NL63 +
Controls:
7.7% NL63 +
Chang et al 4 Taiwan (Taipei and Tao-Yuan County) 73 Pancoronaviruses Total: 226 Total: 226 2.07 Y 2.01 Y OP and NP Swab RT-PCR
SN-PCR
Cases:
7.1% + Pan-HCoV
Controls:
0.9% + Pan-HCoV
Turnier et al 5 USA (Aurora) 52 NL63
229E
OC43
HKU1
Total: 222 Total: 16415 2.92-3.52 Y - NP Wash RT-PCR Cases:
NL63: 1.6%
229E: 1%
OC43: 1%
HKU1: 0%
Controls:
NL63: 0.8%
229E: 0.5%
OC43: 1.4%
HKU1: 0.5%
Kim et al. 16 South Korea (Seoul) 8 NL63
229E
OC43
Total: 55 Total: 78 2.77 Y 3.95 Y NP Swab RT-PCR Cases:
OC43/229E: 3.6%
NL63: 0%
Controls:
OC43/229E: 1.3%
NL63: 1.3%
P = .028

Abbreviations: MH, Mantel-Haenszel; NP, nasopharyngeal; NT, neutralizing test; OP, oropharyngeal; RT-PCR, reverse transcription polymerase chain reaction; SN-PCR, semi-nested polymerase chain reaction; USA, United States of America.

COVID-19’s Cytokine Storm

Cytokines is a broad term to describe a category of proteins that play a key role in cell signaling and the immune system. Homeostasis is maintained by the body by a balance between pro- and anti-inflammatory cytokines. In KD, it is proposed that abnormal activation of the immune system results in the release of pro-inflammatory cytokines. 25 Similarly, COVID-19 has been shown to cause a massive release of pro-inflammatory cytokines (“cytokine storm”) and immune dysregulation, increasing disease severity24,26 and possibly precipitating multi-organ failure. 27 Pro-inflammatory cytokines activate more immune cells and promote leukocyte extravasation, causing local tissue damage and vasculitis. 25 Amongst these cytokines is believed to be TNF-α, which mediates elastin breakdown and eventual aneurysm formation. 28 IL-6 is thought to play a major role, as is supported by its increased levels in MIS-C 29 and the effectiveness of tocilizumab (IL-6R antagonist) in treating it. 30 IL-6 expression is increased by TNF- α, 30 and its effects include promoting CD8+, Th17, and self-reactive CD4+ cells, while inhibiting Treg cells. 31 NF-κB in COVID-19’s cytokine storm induces the IL-6 amplifier (IL-6 Amp), which results in a positive feedback loop of further pro-inflammatory cytokine release, 32 and also induces TNF- α. 32 Moreover, a study demonstrated increased proinflammatory Th17 cell activity with concomitantly decreased anti-inflammatory Treg cell function in KD. 33 This change in Th17 and Treg activity is akin to what is seen in KD, 33 and murine models have shown that inhibition of Th17 cytokines via JAK2 inhibitor Fedratinib may prevent the adverse outcomes of Th17-associated cytokine storm in COVID-19. 34 Thus, understandably, most MIS-C cases have demonstrated recovery after treatment with immunomodulatory agents, including anti-TNF and IL-6 inhibitors. 35

Superantigenic Behavior of SARS-CoV-2

There has been long-standing speculation that the pathogenesis of KD might involve superantigens that cause immune dysregulation, and ultimately, a cytokine storm. 36 Though SAGs are classically bacterial, known and potential viral SAGs include in mouse mammary tumor virus (MMTV), CMV, and EBV. 36 Cheng et al 37 used structural-based computational models to demonstrate the possible superantigenic activity of SARS-CoV-2,and noted that a rare mutation due to a single amino acid substitution from a European strain of the virus resulted in stabilization and stronger binding of SARS-CoV-2’s spike protein with TCR Vβ. 37 Considering this, it is possible that the superantigenic activity of SARS-CoV-2 could contribute to the cytokine storm that characterizes the MIS-C. 38 Table 3 summarizes the cytokine and superantigenic associations in KD and COVID-19.

Table 3.

Cytokines and superantigens in COVID-19 and Kawasaki disease.

Cytokines/
Superantigens
COVID-19 Kawasaki disease
Proinflammatory ILs ● IL-1B, IL-RA, IL-7, IL-8, and IL-9 are increased in both ICU and non-ICU patients with COVID-1926
● IL-6/ IFN-γ ratio is higher in patients with COVID-1931
● IL-2R and IL-6 are significantly elevated in patients with COVID-19, and expression increases with disease severity 39
● IL-6 is significantly increased in COVID-19 patients, and its levels in ICU patients are significantly higher than in non-ICU patients 24
● 97% of patients with MIS-C who were tested had IL-6 ⩾ 5.0 pg/ml, with a median of 116.3 pg/ml overall 29
● IL-1, IL-2, IL-6, and IL-8 are upregulated in acute stage KD 40
● IL-6 is significantly elevated in KD, and it decreases post-IVIG therapy 41
● Patients with IL-6 >66.7 pg/ml are at higher risk for KD shock syndrome (KDSS) 42
● IL-17, IL-6, and IL-23 (TH17 cytokines) are significantly upregulated 33
● IL-1 has been found to facilitate antibody-mediated cytotoxicity in patients with KD 43
Proinflammatory Non-IL cytokines ● Basic FGF, G-CSF, GM-CSF, IFN-γ, IP-10, MCP-1, MIP-1A, MIP-1B, PDGF, TNF-α, and VEGF are increased in both ICU and non-ICU patients with COVID-19, and ICU patients have higher concentrations of G-CSF, IP-10, MCP-1, MIP-1A, and TNF-α than non-ICU patients 26
● TNF-α is significantly increased in COVID-19 patients, and its levels in ICU patients are significantly higher than in non-ICU patients 24
● TNF-α, IFN-γ, and MCP-1 are upregulated in acute stage KD 40
● IFN-γ and TNF-α are significantly elevated in KD, and they decrease post-IVIG therapy. TNF-α is significantly decreased post-IVIG in patients without coronary artery lesions (CALs) and in IVIG responders, but is slightly increased in post-IVIG patients with CALs and in non-IVIG responders. 41
● IFN- γ and TNF have been found to facilitate antibody-mediated cytotoxicity in patients with KD 44
● Patients with IFN-γ >8.35 pg/ml are at higher risk for KD shock syndrome (KDSS) 42
Anti-inflammatory cytokines (IL and Non-IL) IL-10 is increased in both ICU and non-ICU patients with COVID-19, and ICU patients have higher concentrations of IL-10 than non-ICU patients24,26 ● IL-4, and IL-10 are upregulated in acute stage KD 40
● IL-10 is significantly increased in KD, and it decreases post-IVIG 41
● Patients with IL-10 >20.85 pg/ml are at a higher risk for KD shock syndrome (KDSS) 44
● TGF-β (Treg cytokine) is low in patients with KD 33
Superantigens The Spike (S) glycoprotein of SARS-CoV-2 has a high affinity TCR Vβ-binding epitope (P681RRA684) that is similar in sequence and structure to bacterial SAGs, such as Staphylococcal enterotoxin B (SEB), and there is greater stabilization and binding of S with TCR Vβ in a European strain (D839Y/N/E) 37 ● KD-specific serum molecules are similar to molecule associated molecular patterns (MAMPs) from Bacillus cereus, Bacillus subtilis, Yersinia pseudotuberculosis, and Staphylococcus aureus 45
● TSST-1 producing strains of S. aureus have been isolated in patients with KD 45
● Hemolytic Streptococci SAGs have been implicated in KD 46
● Murine models of KD have demonstrated the induction of coronary arteritis using a SAG found within Lactobacillus cillus casei cell wall extract (LCWE) 47

Abbreviations: G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; ICU, intensive care unit; IFN, interferon; IP-10, interferon-γ-inducible protein 10; MCP-1, monocyte chemoattractant protein-1; MIP, macrophage inflammatory protein; PDGF, platelet derived growth factor; TGF, transforming growth factor; TNF, tumor necrosis factor; TSST-1, toxic shock syndrome toxin 1.

Genetics: Possible Connections Between SARS-CoV-2 and KD

Genetics are widely considered to play a role in the susceptibility to KD. 48 Genome-wide association studies have demonstrated several gene variants associated with KD. These include FCGR2A (FcγRII), ITPKC (inositol 1,4,5 triphosphate kinase C), CD40, CASP3 (Caspase 3), multiple alleles of HLA I and II (Human Leukocyte Antigens I and II), CD209 (CD209/DC-SIGN), and BLK (B lymphoid tyrosine kinase). 48

Due to the nature of the novel coronavirus, genetic studies regarding SARS-CoV-2 are still nascent. However, a few potential developments have been made. HLA associations have been demonstrated, with HLA-B*46:01 having the lowest number of predicted binding peptides for SARS-CoV-2. 49 This indicates that individuals with the allele may be particularly vulnerable to COVID-19, as was the case previously with SARS-CoV-1. 49 Moreover, HLA-B*15:03 demonstrated the greatest ability for presenting highly conserved SARS-CoV-2 peptides shared among the HCoVs, suggesting a possibility that it may provide cross-protective T-cell-based immunity. 49 Although neither HLA-B*46:01 nor HLA-B*15:03 have been associated with KD, HLA-B*54:01 has been found to increase susceptibility to the disease. 50 A small proportion of COVID-19 patients develop features of a cytokine release syndrome (CRS) 1-2 weeks after onset of infection. Recent evidence suggests that IL-6 may be a key mediatory in the CRS associated severe SARS-CoV-2 infection, 51 and it is hypothesized that IL-6 polymorphisms may predispose to severe COVID-19. 52 Increased IL-6 levels also play a role in the acute hyperinflammatory response in KD, showing a potential commonality between SARS-CoV-2 infection and KD. 51

Since SARS-CoV-2 bears considerable genetic similarity to SARS-CoV-1, it is in the realm of thought that genetic associations with SARS-CoV-1 may hold true to some extent for SARS-CoV-2 as well. Assuming the association holds true, we can postulate genetic links between SARS-CoV-2 and KD. To begin with, there is an association between the FcγRIIA-R/R131 genotype and a severe course of SARS-CoV-1, 53 and the FCGR2A (FcγRII) has been found to be associated with increased susceptibility to KD. 48 Similarly, a single nucleotide polymorphism in the CD209 promoter is associated with severe SARS-CoV-1 infection, 54 while the CD209 gene has been shown to be associated with increased susceptibility to KD. 48 The +874 A/T polymorphism in the IFN-γ (interferon gamma) is associated with a higher risk of developing SARS-CoV-1 infection, 55 and the IFN-γ gene has been identified in a mouse model as a candidate gene for association with KD through its regulatory cytokine-suppressing inflammatory response. 28

Currently, there is insufficient genetic research pertaining to SARS-CoV-2 to support any associations with KD, and the links remain highly speculative. However, based on previous experience with SARS-CoV-1 and MERS, MBL (Mannose-Binding Lectin), CD147, CCL2, IL-12, and HLA genes have been highlighted for consideration in playing a role in susceptibility to SARS-CoV-2 and severe COVID-19. 56 With the initiation of the COVID-19 Host Genetics Initiative, 57 the interplay of genetics with SARS-CoV-2 infection and COVID-19 will be increasingly unraveled. This will serve to shed more light upon the genetic associations between KD and SARS-CoV-2.

Antibody-Dependent Enhancement

Antibody-dependent enhancement (ADE), the phenomenon whereby dysfunctional antibodies amplify viral entry and infection instead of diminishing it, has been demonstrated in the past with viruses, such as HIV 58 and the coronaviruses, including SARS-CoV-1 and MERS-CoV.59,60 The most common mechanism of ADE is Fc receptor (FcR)-dependent, where binding of FcR to the virus-antibody complex augments the attachment between the cell and the virus, thus resulting in enhanced infectivity. 58 ADE may also occur via binding of virus-antibody complexes to complement receptors or by induction of a conformational change in the viral glycoproteins, resulting in enhanced fusion. 58

SARS-CoV-1 has demonstrated ADE in several in vitro experiments. Antibodies generated in response to a SARS-CoV-1 in a hamster model enhanced FcγRII (CD32)-dependent viral entry into B cells. 59 Another study using anti-Spike immune serum reinforced that ADE in SARS-CoV-1 is predominantly dependent on FcγRII (CD32). 61 This study also showed that certain immune cells are only infected by SARS-CoV-1 in the presence of anti-Spike immune serum. 61 A different study used an HL-CZ human pneumocyte cell line isolated from a leukemia patient to demonstrate ADE. 62 The HL-CZ cells expressed both ACE2 as well as higher levels of FcγRII (CD32), and when they were treated with diluted anti-sera collected from SARS-CoV-1 patients, there was increased viral infectivity, as opposed to when they were treated with a more concentrated anti-sera. 62 Furthermore, a study found that anti-Spike antibodies increased infection in macrophages, and this depended on an intact FcγRII extracellular and intracellular signaling domain. 63 FcγRII with mutated intracellular and normal extracellular domains were unable to trigger ADE, despite being able to bind to the ligand. 63

Similarly, MERS-CoV has also shown ADE in vitro. Wan et al. 60 demonstrated a novel mechanism for ADE in MERS-CoV using Mersmab 1, a monoclonal antibody that binds to the receptor-binding domain of MERS-CoV’s Spike-ectodomain (S-e). Mersmab 1 was shown to bind MERS-CoV S-e with the Fc receptors on human HEK293T (human kidney) cells. Controlled experiments demonstrated that Mersmab1 mediated viral entry into HEK293T cells with exogenously expressed Fc-receptors as well as into macrophages with endogenously expressed Fc-receptors, but blocked viral entry into cells expressing MERS-CoV’s receptor dipeptidyl peptidase-4 (DPP-4). The ADE varied with the dosage of Mersmab 1, and with the expression of viral receptors and Fc-receptors. Cells with both Fc-receptors and DPP-4 had the highest viral entry at the intermediate dose. Moreover, the study demonstrated that neutralizing antibodies mimic the function of viral receptors, binding to the spike protein and triggering a conformational change in it, which results in sequential proteolytic cleavage, thus mediating viral entry into cells expressing FcR.

Various studies in the past have found that there is significant cross-reactivity between antibodies generated in response to different coronavirus strains, suggesting the possibility of non-specific ADE. One such study found that 7/28 (25%) of SARS-CoV-1 patients had cross-reactive neutralizing anti-MERS antibodies, 64 while another found appreciable cross-reactivity between MERS-CoV with 3 out of 4 of the seasonal human coronaviruses (HKU1, 229E, OC43, and NL63 CoV). 65 Moreover, serum samples from COVID-19 patients cross-react with nucleocapsid antigens of SARS-CoV-1, 66 while antibodies against the Spike protein and receptor-binding domain (RBD) cross react with plasma from SARS-CoV-1 patients. 67 The cross-reactivity is mostly with the non-RBD regions, resulting in non-neutralizing antibodies. 67

As MIS-C occurs late after infection with SARS-CoV-2, once antibodies have developed, it may be possible that ADE may contribute to the underlying mechanism of a pro-inflammatory cytokine storm, yet this remains to be explored.35,68 This speculation is supported by the geographical distribution of MIS-C, as prior outbreaks in areas with different coronavirus strains may prime for ADE due to antigenic epitope heterogeneity. 68 In the in vitro model that demonstrated ADE of SARS-CoV-1 in HL-CZ cells, the cells were found to have increased pro-inflammatory cytokine secretion. 62 A study using SARS-CoV-1/macaque models 69 demonstrated that anti-spike IgG in infected lungs prior to viral clearance resulted in recruitment of proinflammatory macrophages and a 5–10 fold increase in IL-6, MCP1, and IL-8. 69 Lastly, since by adulthood most individuals develop natural immunity against coronaviruses, the resultant antibodies may be passed to the infant transplacentally and via breastfeeding, and may cross-react with SARS-CoV-2. With age and with weaning, as the titers of these passive antibodies fall, ADE may occur in the presence of SARS-CoV-2 infection. Studies have highlighted this dose-dependent correlation between antibodies resulting in ADE.60,62 Such a phenomenon has been observed with other viruses such as the Dengue virus, in which infants <1 year of age can develop dengue hemorrhagic fever with a primary dengue virus infection due to the maternal dengue-specific antibodies. 70

Complement activation

Due to its ability to cause inflammation, the complement pathway may be involved in the pathogenesis of MIS-C in COVID-19. A study found that the nucleocapsid (N) proteins of SARS-CoV-1, MERS-CoV, and SARS-CoV-2 bind to MASP-2, the serine protease in the lectin pathway of the complement cascade, causing inflammatory lung injury. 71 The study also found complement hyper-activation in COVID-19 patients, and observed a promising suppressive effect when the patients were treated with anti-C5a monoclonal antibody. 71 Moreover, in the advanced stages of COVID-19, C3 inhibition has the potential to broadly control systemic inflammation affecting the microvascular beds of vital organs of the body. 72 A study exploring complement-associated microvascular injury in cases of severe COVID-19 found striking septal capillary injury, in addition to extensive deposits of terminal complement complexes (C5b-9) in the lungs. 73 As the defining feature of KD is vasculitis and general systemic inflammation, further investigation into the role of complement in its pathogenesis is warranted.

Endotheliitis

Being a multisystem inflammatory disease that affects mainly the medium-sized arteries, KD is known to cause endotheliitis. 74 Angiotensin Converting Enzyme 2 (ACE2) receptors are present in arterial cells of several organs such as the lungs, heart, kidneys, and intestines.75-77 As ACE2 receptors promote SARS-CoV-2′s entry into cells and its replication, 75 SARS-CoV-2 may cause endotheliitis via endothelial ACE2 receptors. 76 Viral inclusion bodies in capillary endothelial cells, along with cellular infiltrates and apoptosis of endothelial cells, confirm endotheliitis in SARS-CoV-2 infection. 76 The endotheliitis and resultant endothelial dysfunction may lead to tissue edema, a procoagulant state, and potentially, multi-organ dysfunction. 77 Thus, SARS-CoV-2 infection and inflammation in COVID-19 may function as “priming triggers” that accelerate KD development, and highlight an important possible connection between COVID-19 and KD. 74 Given this, it can be hypothesized that endotheliitis may contribute to the pathophysiological mechanism of association between KD and MIS-C.

Reactive oxygen species

Given the multisystemic inflammatory nature of KD, it has been suggested that enhanced oxidative stress may contribute to the pathogenesis of the disease. 78 The overproduction of ROS upregulates cytokine/chemokine gene expression, raising serum levels of TNF-α, IL-1, and IL-6. 79 In addition, increased ROS may lead to activation of kinase pathways and redox-sensitive transcription factors, such as NF-κB, which further upregulate cytokines. 79 These amplify the inflammatory process and cause tissue migration of neutrophils and macrophages. 79 In KD, the increased oxidative stress and resultant cytokine release may promote the vasculitic changes and characteristic coronary artery abnormalities. 79

Increased ROS production may play a role in the development of MIS-C. Viral infections of the respiratory system are often associated with pathophysiological processes culminating in oxidative stress through dysregulation of antioxidant mechanisms. 80 In SARS-CoV-2 infection, inflammatory changes may lead to a dysregulated anti-oxidant system, causing overproduction of reactive oxygen species (ROS), which could contribute to the pathogenesis of COVID-19 and its progression into severe disease.80,81 In addition, inflammation associated with SARS-CoV-2 may also cause macrophages and neutrophils to increase their production of ROS. 82 The levels of angiotensin II (ATII) have been shown to increase linearly in SARS-CoV-2 infection, 83 and this increase mediates an NADH/NADPH oxidase-induced production of ROS. 81 Regardless of the source, increased levels of ROS may ultimately cause significant damage to the vascular endothelium in COVID-19. 81

Conclusion

MIS-C represents a new entity in the spectrum of manifestations of COVID-19, with cases being reported from all over the world. It is important to understand the pathophysiological mechanisms behind this new condition, as this will guide progress toward diagnostics and treatments. Building on the clinical similarities between KD and MIS-C, our review aimed to compare pathophysiological mechanisms between the2 conditions in order to shed light on this rare manifestation of COVID-19.

Given the history of association of KD with coronaviruses and other viruses,4,5,7,11-22 this review demonstrates how the emergence of MIS-C may not be a wholly unique and unprecedented phenomenon. KD and MIS-C share a range of commonalities, including in their clinical presentation and proposed pathophysiology. Though there is no definitive genetic link between the two, the possibility of one cannot be excluded. Both conditions share features of a hyper-inflammatory syndrome or cytokine storm,25,27 leading to a downstream inflammatory response, and to systemic oxidative stress, that may cause vasculitis and precipitate multi-organ failure.77,80,81 It is possible that antibody-dependent enhancement, a phenomenon seen in previous coronaviruses, may play a role in advancing the cytokine storm in COVID-19.59-63 Additionally, our review also highlights the possible role of the superantigenic behavior of SARS-CoV-2, and of the complement cascade in contributing toward the multi-system inflammatory features of MIS-C. Lastly, given the rarity of MIS-C, this review also speculates, based on evidence, that geographic and genetic factors may predispose to this uncommon manifestation of COVID-19 (Figure 1). The potential immunological and molecular mechanisms for the pathophysiology of MIS-C, as discussed in this review, are summarized in Figure 2. The clinical similarities between MIS-C and KD, as well as the potential underlying pathophysiology in both conditions has important implications when it comes to treatment options, which are contrasted in Table 4.

Figure 1.

Figure 1.

Proposed model for pathogenesis of MIS-C in COVID-19.

Abbreviations: HLA, human leukocyte antigen; IL, interleukin; ROS, reactive oxygen species; TNF, tumor necrosis factor.

Figure 2.

Figure 2.

Immune responses and viral mechanisms in the pathogenesis of MIS-C.

Abbreviations: Ab, antibody; ACE2, angiotensin-converting enzyme 2; APC, antigen presenting cell; CD4+, T helper cell; CTL, cytotoxic T cell; FcR, Fc receptor; MASP-2, mannan-binding lectin serine protease 2; MHC, major histocompatibility complex; N protein, nucleocapsid protein; SAG, Superantigen; TCR, T cell receptor.

Table 4.

Summary of established and potential management options for MIS-C and KD.

MIS-C1,84 KD10,85,86
Anti-inflammatory IVIG IVIG
Steroids Aspirin (high dose)
Aspirin Steroids (can consider in IVIG resistance)
Biologic IL-1Ra inhibitors (Anakinra) If IVIG resistant, can consider:
IL-6 inhibitors (tocilizumab, siltuximab) IL-1 inhibitors
IL-6 inhibitors
Anti-TNF (infliximab)
Ancillary Broad-spectrum antibiotics Antiplatelet (low-dose aspirin, clopidogrel)
Thromboprophylaxis (LMWH) Thromboprophylaxis (LMWH, warfarin) if needed

Abbreviations: IVIG, intravenous immunoglobulins; IL, interleukin; KD, Kawasaki disease; LMWH, low molecular weight heparin; MIS-C, multisystem inflammatory syndrome related to COVID-19; TNF, tumor necrosis factor.

Early detection and treatment are essential for the management of MIS-C in COVID-19. The WHO has published a preliminary case framework for diagnosis, and has suggested physicians suspect this condition when presented with a patient who displays some or all features of KD or of toxic shock syndrome. 2 Treatment options include immunomodulatory agents, such as IV immunoglobulins (IVIG) and corticosteroids, which have shown positive results. 35 As the pandemic continues to challenge healthcare systems with unprecedented disease manifestations, it is vital that the medical community be prepared to meet this challenge with evidence-based management responses. By highlighting the potential pathophysiological mechanisms that contribute to the condition, our review holds important implications for the diagnosis and management of the MIS-C.

Acknowledgments

We also acknowledge the Research and Development Wing of The Society for Promoting Innovation in Education (SPIE) for providing mentorship to author Ronika Devi Ukrani (medical student) on this project. SPIE is actively involved with innovation, education and research in the academic and public health sector.

Footnotes

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Authors’ Note: M Rizwan Sohail is now affiliated to Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA.

Author Contributions: FFS and RSM conceived the idea for this review. AU, RDU, FFS and RSM performed the literature search and data extraction. The article was written by FFS, RSM, AU and RDU, and critically reviewed by KJ, MRS and EK. FFS and RSM produced the figures in this article. The final manuscript was approved by all authors.

Ethical Standards: This review was exempt from ethical approval by the institutional review board at the Aga Khan University Hospital, Pakistan.

ORCID iDs: Fatima Farrukh Shahbaz Inline graphic https://orcid.org/0000-0002-5889-0048

Ronika Devi Ukrani Inline graphic https://orcid.org/0000-0001-9129-9041

References

  • 1. Feldstein LR, Rose EB, Horwitz SM, et al. Multisystem inflammatory syndrome in U.S. Children and adolescents. N Engl J Med. 2020;383:334-346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Stephen Freedman SF, Richard G. Multisystem Inflammatory Syndrome in Children and Adolescents Temporally Related to COVID-19. WHO; 2020. [Google Scholar]
  • 3. Rowley AH, Shulman ST. The epidemiology and pathogenesis of Kawasaki disease. Front Pediatr. 2018;6:374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Chang LY, Lu CY, Shao PL, et al. Viral infections associated with Kawasaki disease. J Formosan Medical Association = Taiwan yi zhi. 2014;113:148-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Turnier JL, Anderson MS, Heizer HR, Jone P-N, Glodé MP, Dominguez SR. Concurrent respiratory viruses and Kawasaki disease. Pediatrics. 2015;136:e609-e14. [DOI] [PubMed] [Google Scholar]
  • 6. Rowley AH, Baker SC, Shulman ST, et al. RNA-containing cytoplasmic inclusion bodies in ciliated bronchial epithelium months to years after acute Kawasaki disease. PLoS One. 2008;3:e1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Esper F, Shapiro ED, Weibel C, Ferguson D, Landry ML, Kahn JS. Association between a novel human coronavirus and Kawasaki disease. J Infect Dis. 2005;191:499-502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Organization WH. Multisystem Inflammatory Syndrome in Children and Adolescents With COVID-19: Scientific Brief. World Health Organization; 2020:2020. [Google Scholar]
  • 9. Sancho-Shimizu V, Brodin P, Cobat A, et al. SARS-CoV-2–related MIS-C: A key to the viral and genetic causes of Kawasaki disease? J Exp Med. 2021;218:e20210446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Freeman AF, Shulman ST. Kawasaki disease: summary of the American Heart Association guidelines. Am Fam Physician. 2006;74:1141-1148. [PubMed] [Google Scholar]
  • 11. Lehmann C, Klar R, Lindner J, Lindner P, Wolf H, Gerling S. Kawasaki disease lacks association with human coronavirus NL63 and human bocavirus. Pediatr Infect Dis J. 2009;28(6):553-554. [DOI] [PubMed] [Google Scholar]
  • 12. Belay ED, Erdman DD, Anderson LJ, et al. Kawasaki disease and human Coronavirus. J Infect Dis. 2005;192:352-NaN3; author rely 353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Shirato K, Imada Y, Kawase M, Nakagaki K, Matsuyama S, Taguchi F. Possible involvement of infection with human coronavirus 229E, but not NL63, in Kawasaki disease. J Med Virol. 2014;86:2146-2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ebihara T, Endo R, Ma X, Ishiguro N, Kikuta H. Lack of association between New Haven coronavirus and Kawasaki disease. J Infect Dis 2005;192(2):351-352; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Dominguez SR, Anderson MS, Glodé MP, Robinson CC, Holmes KV. Blinded case-control study of the relationship between human coronavirus NL63 and Kawasaki syndrome. J Infect Dis. 2006;194:1697-1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Kim JH, Yu JJ, Lee J, et al. Detection rate and clinical impact of respiratory viruses in children with Kawasaki disease. Korean J Pediatr. 2012;55:470-473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Shimizu C, Shike H, Baker S, et al. Human coronavirus NL63 is not detected in the respiratory tracts of children with acute Kawasaki disease. J Infect Dis. 2005;192:1767-1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chang LY, Chiang BL, Kao CL, et al. Lack of association between infection with a novel human coronavirus (HCoV), HCoV-NH, and Kawasaki disease in Taiwan. J Infect Dis. 2006;193:283-286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Bohlok F, Abou Hamdan Z, Rachini N, Rabah S, Slim Z, Mortada L. Atypical Kawasaki disease in adolescent male with liver involvement following human Coronavirus OC43 infection. Acta Sci Paediatr. 2020;3:01-03. [Google Scholar]
  • 20. Cho EY, Eun BW, Kim NH, et al. Association between Kawasaki disease and acute respiratory viral infections. Korean J Pediatr. 2009;52:1241. [Google Scholar]
  • 21. Baker SC, Shimizu C, Shike H, et al. Human coronavirus-NL63 infection is not associated with acute Kawasaki disease. Adv Exp Med Biol. 2006;581:523-526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Giray T, Biçer S, Küçük Ö, et al. Four cases with Kawasaki disease and viral infection: aetiology or association. Le infezioni in medicina. 2016;24:340-344. [PubMed] [Google Scholar]
  • 23. Rowley AH, Baker SC, Shulman ST, et al. Ultrastructural, immunofluorescence, and RNA evidence support the hypothesis of a “new” virus associated with Kawasaki disease. J Infect Dis. 2011;203:1021-1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Diao B, Wang C, Tan Y, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol. 2020;11:827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Takahashi K, Oharaseki T, Yokouchi Y. Pathogenesis of Kawasaki disease. Clin Exp Immunol. 2011;164 Suppl 1:20-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497-506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Jose RJ, Manuel A. COVID-19 cytokine storm: the interplay between inflammation and coagulation. Lancet Respir Med. 2020;8:e46-e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yeung RS. Kawasaki disease: update on pathogenesis. Curr Opin Rheumatol. 2010;22:551-560. [DOI] [PubMed] [Google Scholar]
  • 29. Dufort EM, Koumans EH, Chow EJ, et al. Multisystem inflammatory syndrome in children in New York state. New Engl J Med. 2020;383(4):347-358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Zhang C, Wu Z, Li JW, Zhao H, Wang G-Q. Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist Tocilizumab may be the key to reduce mortality. Int J Antimicrob Agents. 2020;55:105954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Lagunas-Rangel FA, Chávez-Valencia V. High IL-6/IFN-γ ratio could be associated with severe disease in COVID-19 patients. J Med Virol. 2020;92:1789-1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Hirano T, Murakami M. COVID-19: a new virus, but a familiar receptor and cytokine release syndrome. Immunity. 2020;52:731-733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jia S, Li C, Wang G, Yang J, Zu Y. The T helper type 17/regulatory T cell imbalance in patients with acute Kawasaki disease. Clin Exp Immunol. 2010;162:131-137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor Fedratinib. J Microbiol Immunol Infect. 2020;53:368-370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Levin M. Childhood multisystem inflammatory syndrome—a new challenge in the pandemic. New Engl J Med. 2020;383(4):393-395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Fraser J, Arcus V, Kong P, Baker E, Proft T. Superantigens – powerful modifiers of the immune system. Mol Med Today. 2000;6:125-132. [DOI] [PubMed] [Google Scholar]
  • 37. Cheng MH, Zhang S, Porritt RA, Arditi M, Bahar I. An insertion unique to SARS-CoV-2 exhibits superantigenic character strengthened by recent mutations. bioRxiv 2020 [Google Scholar]
  • 38. Garazzino S, Montagnani C, Donà D, et al. Multicentre Italian study of SARS-CoV-2 infection in children and adolescents, preliminary data as at 10 april 2020. Euro Surveill. 2020;25:2000600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Chen L, Liu H, Liu W, et al. Analysis of clinical features of 29 patients with 2019 novel coronavirus pneumonia. Zhonghua jie he he hu xi za zhi= Zhonghua jiehe he huxi zazhi= Chinese journal of tuberculosis and respiratory diseases. 2020;43:E005-E05. [DOI] [PubMed] [Google Scholar]
  • 40. Takahashi K, Oharaseki T, Yokouchi Y. Update on etio and immunopathogenesis of Kawasaki disease. Curr Opin Rheumatol. 2014;26:31-36. [DOI] [PubMed] [Google Scholar]
  • 41. Wang Y, Wang W, Gong F, et al. Evaluation of intravenous immunoglobulin resistance and coronary artery lesions in relation to Th1/Th2 cytokine profiles in patients with Kawasaki disease. Arthritis Rheum. 2013;65:805-814. [DOI] [PubMed] [Google Scholar]
  • 42. Li Y, Zheng Q, Zou L, et al. Kawasaki disease shock syndrome: clinical characteristics and possible use of IL-6, IL-10 and IFN-γ as biomarkers for early recognition. Pediatr Rheumatol Online J. 2019;17:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Nonoyama S. Immunological abnormalities and endothelial cell injury in Kawasaki disease. Pediatr Int. 1991;33:752-755. [DOI] [PubMed] [Google Scholar]
  • 44. Lindquist ME, Hicar MD. B cells and antibodies in Kawasaki disease. Int J Mol Sci. 2019;20(8). doi: 10.3390/ijms20081834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Huang SM, Huang SH, Weng KP, Chien KJ, Lin CC, Huang YF. Update on association between Kawasaki disease and infection. J Chin Med Assoc. 2019;82:172-174. [DOI] [PubMed] [Google Scholar]
  • 46. Nagata S. Causes of Kawasaki disease-from past to present. Front Pediatr. 2019;7:18. doi: 10.3389/fped.2019.00018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Noval Rivas M, Lee Y, Wakita D, et al. CD8+ T cells contribute to the development of coronary arteritis in the Lactobacillus casei cell wall extract-induced murine model of Kawasaki disease. Arthritis Rheumatol. 2017;69:410-421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Lo MS. A framework for understanding Kawasaki disease pathogenesis. Clin Immunol. 2020;214:108385. [DOI] [PubMed] [Google Scholar]
  • 49. Nguyen A, David JK, Maden SK, et al. Human leukocyte antigen susceptibility map for severe acute respiratory syndrome Coronavirus 2. J Virol. 2020;94(13):e00510-e00520. doi: 10.1128/JVI.00510-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Kwon Y-C, Sim BK, Yu JJ, et al. HLA-B*54:01 Is associated with susceptibility to Kawasaki disease. Circ Genom Precis Med. 2019;12:e002365. [DOI] [PubMed] [Google Scholar]
  • 51. Woo Y-L, Kamarulzaman A, Augustin Y, Staines H, Altice F, Krishna S. A genetic predisposition for Cytokine Storm in life-threatening COVID-19 infection. 2020 [Google Scholar]
  • 52. Kirtipal N, Bharadwaj S. Interleukin 6 polymorphisms as an indicator of COVID-19 severity in humans. J Biomol Struct Dyn. 2021;39:4563-4565. [DOI] [PubMed] [Google Scholar]
  • 53. Yuan FF, Tanner J, Chan PK, et al. Influence of FcgammaRIIA and MBL polymorphisms on severe acute respiratory syndrome. Tissue Antigens. 2005;66:291-296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Chan KY, Xu MS, Ching JC, et al. Association of a single nucleotide polymorphism in the CD209 (DC-SIGN) promoter with SARS severity. Hong Kong medical journal = Xianggang yi xue za zhi. 2010;16:37-42. [PubMed] [Google Scholar]
  • 55. Chong WP, Ip WK, Tso GH, et al. The interferon gamma gene polymorphism +874 A/T is associated with severe acute respiratory syndrome. BMC Infect Dis. 2006;6:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Darbeheshti F, Rezaei N. Genetic predisposition models to COVID-19 infection. Med Hypotheses. 2020;142:109818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. COVID-19 Host Genetics Initiative. The COVID-19 Host Genetics initiative, a global initiative to elucidate the role of host genetic factors in susceptibility and severity of the SARS-CoV-2 virus pandemic. Eur J Hum Genet. 2020;28:715-718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Takada A, Kawaoka Y. Antibody-dependent enhancement of viral infection: molecular mechanisms and in vivo implications. Rev Med Virol. 2003;13:387-398. [DOI] [PubMed] [Google Scholar]
  • 59. Kam YW, Kien F, Roberts A, et al. Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV challenge despite their capacity to mediate FcgammaRII-dependent entry into B cells in vitro. Vaccine. 2007;25:729-740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wan Y, Shang J, Sun S, et al. Molecular mechanism for antibody-dependent enhancement of coronavirus entry. J Virol. 2020;94(5):e02015-e02019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Jaume M, Yip MS, Cheung CY, et al. Anti-Severe acute respiratory syndrome Coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent Fc R pathway. J Virol. 2011;85:10582-10597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Wang S-F, Tseng S-P, Yen C-H, et al. Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins. Biochem Biophys Res Commun. 2014;451:208-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Yip MS, Leung NH, Cheung CY, et al. Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus. Virol J. 2014;11:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Chan K-H, Chan JF-W, Tse H, et al. Cross-reactive antibodies in convalescent SARS patients’ sera against the emerging novel human coronavirus EMC (2012) by both immunofluorescent and neutralizing antibody tests. Infect J. 2013;67:130-140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Al Kahlout RA, Nasrallah GK, Farag EA, et al. Comparative serological study for the prevalence of anti-MERS coronavirus antibodies in high- and low-risk groups in Qatar. J Immunol Res. 2019;2019:1386740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Long Q-X, Liu B-Z, Deng H-J, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26(6):845-848. [DOI] [PubMed] [Google Scholar]
  • 67. Lv H, Wu NC, Tsang OT, et al. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep. 2020;31:107725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tetro JA. Is COVID-19 receiving ADE from other coronaviruses? Microbes Infect. 2020;22:72-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Liu L, Wei Q, Lin Q, et al. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight. 2019;4(4):e123158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Adimy M, Mancera PFA, Rodrigues DS, Santos FLP, Ferreira CP. Maternal passive immunity and dengue hemorrhagic fever in infants. Bull Math Biol. 2020;82:24. [DOI] [PubMed] [Google Scholar]
  • 71. Gao T, Hu M, Zhang X, et al. Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. MedRxiv. 2020. [Google Scholar]
  • 72. Risitano AM, Mastellos DC, Huber-Lang M, et al. Complement as a target in COVID-19? Nat Rev Immunol. 2020;20:343-344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Magro C, Mulvey JJ, Berlin D, et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res. 2020;220:1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Xu S, Chen M, Weng J. COVID-19 and Kawasaki disease in children. Pharmacol Res. 2020;159:104951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631-637. Online. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417-1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Bourgonje AR, Abdulle AE, Timens W, et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J Pathol. 2020;251:228-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Cheung YF, O K, Woo CW, et al. Oxidative stress in children late after Kawasaki disease: relationship with carotid atherosclerosis and stiffness. BMC Pediatr. 2008;8:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Sekine K, Mochizuki H, Inoue Y, et al. Regulation of oxidative stress in patients with Kawasaki disease. Inflammation. 2012;35:952-958. [DOI] [PubMed] [Google Scholar]
  • 80. Delgado-Roche L, Mesta F. Oxidative stress as key player in severe acute respiratory syndrome Coronavirus (SARS-CoV) infection. Arch Med Res. 2020;51:384-387. https://www.sciencedirect.com/science/article/pii/S0188440920305403?via%3Dihub [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Baqi HR, M. Farag HA, El Bilbeisi AHH, Askandar RH, El Afifi AM. Oxidative stress and its association with COVID-19: a narrative review. Kurdistan J Appl Res. 2020;2020:97-105. [Google Scholar]
  • 82. Wang J-Z, Zhang R-Y, Bai J. An anti-oxidative therapy for ameliorating cardiac injuries of critically ill COVID-19-infected patients. Int J Cardiol. 2020;312:137-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Liu Y, Yang Y, Zhang C, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci. 2020;63:364-374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Cattalini M, Taddio A, Bracaglia C, et al. Childhood multisystem inflammatory syndrome associated with COVID-19 (MIS-C): a diagnostic and treatment guidance from the rheumatology study group of the Italian Society of Pediatrics. Ital J Pediatr. 2021;47:1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Sosa T, Brower L, Divanovic A. Diagnosis and management of Kawasaki disease. JAMA Pediatr. 2019;173:278-279. [DOI] [PubMed] [Google Scholar]
  • 86. Chen M-R, Kuo H-C, Lee Y-J, et al. Corrigendum: phenotype, susceptibility, autoimmunity, and immunotherapy between Kawasaki disease and Coronavirus disease-19 associated multisystem inflammatory syndrome in children. Front Immunol. 2021;12:722582. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Medicine Insights. Pediatrics are provided here courtesy of SAGE Publications

RESOURCES