eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2022
vol. 47
 
Share:
Share:
Clinical immunology

Immune profile of children diagnosed with multisystem inflammatory syndrome associated with SARS-CoV-2 infection (MIS-C)

Ewelina Gowin
1
,
Grzegorz Dworacki
2
,
Bartosz Siewert
1
,
Jacek Wysocki
1
,
Danuta Januszkiewicz-Lewandowska
3

1.
Health Promotion Department, Poznan University of Medical Sciences, Poland
2.
Department of Clinical Pathomorphology, Poznan University of Medical Sciences, Poland
3.
Department of Oncology, Hematology, and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poland
Cent Eur J Immunol 2022; 47 (2): 151-159
Online publish date: 2022/05/30
Article file
- Immune profile.pdf  [0.16 MB]
Get citation
 
PlumX metrics:
 

Introduction

First cases of the pediatric multisystem inflammatory syndrome associated with SARS-CoV-2 infection (MIS-C) were reported in EU/EEA countries and the UK in spring 2020 [1-5]. In Poland, we did not observe them until autumn. According to the European Centre for Disease Prevention and Control (ECDC), in May 2020, Poland was still before the peak of the epidemic. From March 13, 2020, people were advised to self-isolate, and schools and kindergartens were closed. As pediatricians, we observed a marked decrease in infections in children [6]. The number of children consulted in an emergency room and hospitalized was much lower compared to previous months. Recent studies revealed that children are at a similar risk of COVID-19 infection to the general population. In other countries, MIS-C was reported relatively late during the waning tail of the first epidemic curve [7]. Countries with large outbreaks of SARS-CoV-2 (France, Italy, Spain, UK, US) have seen the occurrence of cases of pediatric inflammatory MIS-C in the late stages of the first wave of the COVID-19 pandemic [8-13]. Verdoni and colleagues, in an article published in The Lancet, described a recent outbreak of Kawasaki disease (KD) in Italy [1]. Clinicians in Europe and the United States have identified clusters of similar cases [14-17]. The observed epidemic of MIS-C disease in many countries is attributed to the SARS-CoV-2 pandemic. However, it has become evident that shock, gastrointestinal symptoms, and coagulopathy, which are rarely seen in classic KD, are prominent features of this unique syndrome. The pathophysiology of MIS-C and the T and B cell response to SARS-CoV-2 infection remains poorly understood. Peripheral natural killer (NK) cells are depleted and exhibit an exhausted phenotype in patients with severe COVID-19 [14]. Many analyses suggest that extravasation of T and NK lymphocytes and activation and chemotaxis of neutrophils and nonclassical monocytes likely contribute to the underlying disease pathogenesis [14]. In MIS-C, there is observed an elevated level of chemokines that recruit NK cells and T cells from the circulation (CCL19, CXCL10, and CDCP1) [15-17]. Alternatively, perturbations in hematopoiesis cannot be ruled out entirely.

This study aimed to define peripheral blood immune features and the clinical evolution of symptoms in patients with MIS-C.

Material and methods

Between April 1 and May 15, 2021, at the Infectious Diseases Department in St. Joseph’s Children’s Hospital in Poznan (Poland), eight children were hospitalized with an inflammatory disease similar to KD. We analyzed only patients with fever without an identified source and no response to antibiotic therapy who fulfilled diagnostic criteria for MIS-C [18]. On admission, laboratory tests including complete blood count, C-reactive protein (CRP), potassium, sodium, and transaminase levels, urinalysis, blood and urine cultures, chest X-ray, and abdominal ultrasound were performed. Children underwent cardiology consultation. KD was diagnosed in patients with fever lasting five days or longer accompanied by four or more of the following clinical criteria: bilateral nonexudative conjunctivitis, erythema of the lips and oral mucosa, changes in the extremities, rash, and cervical lymphadenopathy [6].

MIS-C case definition criteria proposed by the Royal College of Pediatrics and Child Health are described be- low [18]:

A child is presenting with persistent fever, inflammation (neutrophilia, elevated CRP, and lymphopenia), and evidence of single or multi-organ dysfunction (shock, cardiac, respiratory, renal, gastrointestinal, or neurological disorder) with other additional clinical, laboratory, or imaging and electrocardiogram (ECG) features.

  • Children fulfilling full or partial criteria for KD may be included.

  • Exclusion of any other microbial cause, including bacterial sepsis, staphylococcal or streptococcal shock syndromes, infections associated with myocarditis such as enterovirus.

  • SARS-CoV-2 PCR testing positive or negative.

Flow cytometry analysis was used for the detection of lymphocyte subsets. Samples of EDTA anticoagulated peripheral blood (2 ml) were obtained from all patients on admission. CD4+, CD8+ T-cell, and B-cell counts (cells/µl) were measured using multiple-color flow cytometry. Lymphocyte subpopulations were characterized by combining monoclonal antibodies for T, B, and NK cells:

– T cells: (CD45+, CD3+), (CD45+, CD3+, CD4+), (CD45+, CD3+, CD8+)

Naive Th CD4+RA+CD27+

Central memory Th CD4+RA–CD27+

Effector memory Th RA–CD27–

Terminally diff. memory Th CD4+RA+CD27–

Recent thymic emigrants Th CD4+CD31+ RA+

Treg CD4+CD25+CD127–

Naive Tc CD8+CD127+CD27+

Central memory CD8RA-CD197+CD27

Effector memory Tc CD8+RA–CD197–CD27–

Terminally diff. memory Tc CD8+RA+CD197–CD27–

– B cells: (CD45+, CD19+)

CD27+ Memory

CD27– Non-memory

CD27+sIgD+ Non-Switched-Mem

CD27–sIgD+ Naive

CD27+sIgD– Switched-Mem

CD38hi sIgMhi Transitional

CD38lo CD21lo Activated

CD21lo Immature

– NK cells: (CD45+, CD56+, CD3–).

The Bioethical Committee of Poznan University of Medical Sciences approved the study. Informed consent was obtained from all participants’ guardians. Blood was collected as a part of routine tests during hospitalization – no additional procedures were performed.

Results

Of the eight hospitalized children, seven agreed to take part in the study. There were three girls and four boys, aged from four to nine years. They all presented with fever not responding to antibiotic treatment, irritability, and abdominal pain. Inflammatory markers were elevated: neutrophilia, significantly increased CRP, ferritin levels, raised troponin and BNP levels. Echo and ECG were performed in all patients – in two coronary artery dilatation was found. One patient had markedly decreased heart contractility (ejection fraction 24%) and he was diagnosed with dilated cardiomyopathy.

Three patients were diagnosed with pneumonia. Typical findings on abdominal ultrasound were ascites and hepatosplenomegaly. Details are presented in Table 1. One patient underwent an appendectomy before being diagnosed with MIS-C.

Table 1

Demographics, clinical findings, imaging findings, treatment, and outcome

Patients comorbiditiesClinical
presentation
Laboratory resultsMicrobiology
results
Imaging resultsPharmacological
treatment
Outcome
Patient 1
male
age < 5 years
Caucasian
no comorbidities
7 days > 39°C, abdominal pain, conjunctivitis,
rash,
lymphadenopathy,
vomiting
CRP 169 mg/l
PCT 11.8 ng/ml
NT-proBNP 13 496 pg/ml
troponin 30 ng/l
D-dimers 6.67 mg/l
ALT 97 U/l
AST 80 U/l
Albumin 27 g/l
Ferritin 1900 µg/l
Platelets 40 × 109/l
Lymphocytes 1.36 × 109/l
Neutrophils 6.36 × 109/l
Monocytes 0
Hemoglobin 11.9 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative
SARS-CoV-2 IgG positive
EBV IgG negative
EBV IgM negative
No abnormalities on chest X-ray, hepatomegalyCeftriaxone,
IVIG,
methylprednisolone, aspirin
Resolution
of symptoms,
no abnormalities on echo at 2 and 6 weeks
Patient 2
female
age > 5 years
Caucasian
no comorbidities
5 days > 39°C, abdominal pain, myalgia, headache, conjunctivitis, lymphadenopathy, pharyngitis, strawberry tongue, hemorrhagic rash, peripheral edemaCRP 204 mg/l
PCT 1.67 ng/ml
NT-proBNP 20 756 pg/ml
ALT 16U/l
AST 31U/l
Albumin 29 g/l
Ferritin 322 µg/l
D-dimers 10 mg/l
Troponin 45 ng/l
Platelets 217 × 109/l
Lymphocytes 1.14 × 109/l
Neutrophils 15.02 × 109/l
Monocytes 0.66
Hemoglobin 12.3 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative
SARS-CoV-2 IgG positive
EBV IgG positive
EBV IgM negative
Bilateral basal
lung consolidations, MI
Ceftriaxone,
IVIG,
methylprednisolone, aspirin
Resolution
of symptoms,
no abnormalities on echo
at 2 weeks
Patient 3
male
age < 5 years
Caucasian
no comorbidities
7 days > 39°C, abdominal pain, irritability, conjunctivitis, lymphadenopathy, cracked lips, pharyngitis, strawberry tongue, rash, peripheral edema, diarrheaCRP 190 mg/l
PCT 0.22 ng/ml
NT-proBNP 5805 pg/ml
ALT 18U/l
AST 20U/l
Albumin 41 g/l
Troponin 0.01 ng/l
D-dimers 2.93 mg/l
Ferritin 93 µg/l
Platelets 145 × 109/l
Lymphocytes 4.94 × 109/l
Neutrophils 8.94 × 109/l
Monocytes 0.48
Hemoglobin 10.5 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative SARS-CoV-2 IgG positive
EBV IgG negative
EBV IgM negative
No abnormalities on chest X-ray and abdominal ultrasoundCefuroxime,
IVIG,
methylprednisolone, aspirin
Good response,
no abnormalities on echo at 2 and 6 weeks
Patient 4 female
age < 5 years
Caucasian
no comorbidities
5 days > 39°C,
abdominal pain, hemorrhagic rash, lymphadenopathy, peripheral edema, diarrhea
CRP 113 mg/l
PCT 1.51 ng/ml
ALT 35 U/l
AST 43 U/l
Albumin 28 g/l
Ferritin 148 µg/l
Platelets 237 × 109/l
Lymphocytes 1.15 × 109/l
Neutrophils 9.0 × 109/l
Hemoglobin 9.7 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative
SARS-CoV-2 IgG positive
EBV IgG negative
EBV IgM negative
Ascites, MICeftriaxone,
IVIG,
methylprednisolone, aspirin
Good response
Patient 5
male
age > 5 years
Caucasian
no comorbidities
Fever 5 days > 39°C,
lymphadenopathy, abdominal pain
CRP 104 mg/l
PCT 0.19 ng/ml
ALT 9 U/l
AST 9 U/l
Albumin 44 g/l
Ferritin 320 µg/l
Platelets 257 × 109/l
Lymphocytes 0.8 × 109/l
Neutrophils 4.37 × 109/l
Monocytes 0.833 × 109/l
Hemoglobin 8.8 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative
SARS-CoV-2 IgG positive
EBV IgG negative
EBV IgM negative
Pleuropneumonia,
dilated cardiomyopathy, MI, AI
Ceftriaxone,
IVIG, methylprednisolone,
aspirin
Poor response, intensive cardiac treatment
Patient 6
male
age > 5 years
no comorbidities
7 days > 39°C, abdominal pain,
conjunctivitis, rash, lymphadenopathy, strawberry tongue
CRP 45 mg/l
PCT 0.85 ng/ml
ALT 25 U/l
AST 38 U/l
Albumin 33 g/l
Ferritin 358 µg/l
Platelets 261 × 109/l
Lymphocytes 0.931 × 109/l
Neutrophils 6.69 × 109/l
Monocytes 0.29 × 109/l
Hemoglobin 10.3 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative,
SARS-CoV-2 IgG positive,
EBV IgG positive
EBV IgM negative
Ascites, pleural
effusion, dilated RCA
ceftriaxone,
IVIG,
methylprednisolone, aspirin
Good response
Patient 7
female
age > 5 years
no comorbidities
7 days > 39°C, abdominal pain,
conjunctivitis, lymphadenopathy
CRP 58.7 mg/l
PCT 3.57 ng/ml
ALT 31 U/l
AST 23 U/l
Albumin 40 g/l
Ferritin 807 µg/l
Platelets 92 × 109/l
Lymphocytes 0.522 × 109/l
Neutrophils 4.61 × 109/l
Monocytes 0.18 × 109/l
Hemoglobin 11.8 g/l
Blood culture sterile
Influenza PCR negative
SARS-CoV-2 PCR negative
SARS-CoV-2 IgG positive
EBV IgG positive
EBV IgM negative
MICeftriaxone,
IVIG, methylprednisolone,
aspirin
Good response

[i] ALT – alanine aminotransferase, AST – aspartate aminotransferase, CRP – C-reactive protein, EBV – Epstein-Barr virus, IVIG – human intravenous immunoglobulin, MI – mitral insufficiency,

[ii] NT-proBNP – N-terminal pro-B-type natriuretic peptide, PCR – polymerase chain reaction, PCT – procalcitonin, RCA – right coronary artery

There was no COVID-19 history in one patient, one patient had a family history of COVID-19 four weeks earlier, and one five months earlier. Three patients had asymptomatic or mildly symptomatic COVID-19 three/four weeks earlier. None of the tested children was found to be positive for SARS-CoV-2 by PCR. All patients were positive for IgG SARS-CoV-2 antibodies. The median level of antibodies was 198.27 BAU/ml (positive ≥ 7.1 BAU/ml). The lowest level was 14 BAU/ml (2 ULN), detected in a patient who had exposure to SARS-CoV-2 five months earlier.

Coinfection with other pathogens has been investigated, and no pathogens have been detected: blood cultures were negative, and there were no serological markers of EBV infection.

All patients received empiric antibiotic therapy – two patients received only one dose of ceftriaxone, while the rest received a full 7-day course. All children fulfilled the MIS-C case definition criteria proposed by the Royal College of Pediatrics and Child Health. All the children received the following treatment: intravenous immunoglobulin (IVIG) 1-2 g/kg given over 12 h, methylprednisolone 2 mg/kg/day for five days, and a low dose of aspirin (3-5 mg/kg max. 75 mg).

All patients survived, and their health status improved with time. A rapid response was observed after introduction of the treatment (IVIG and methylprednisolone), fever and abdominal pain subsided, and inflammatory markers and lymphocyte level return to normal. One child had severe cardiac dysfunction that required vasopressor treatment. At the follow-up visit after six weeks all patients presented with good health. One patient experienced permanent heart dysfunction and was unable to return to daily activities.

Lymphocyte subsets in peripheral blood of patients

Lymphopenia was detected in all patients. A marked decrease in both CD4+ and CD8+ T cells was observed, with levels much below age-matched reference ranges (Table 2). In CD4+ T cells, majority subsets were: naive cells, CD45RA+ cells, recent thymic emigrants. The percentages of T regulatory cells (Tregs) ranged from 2.9% to 8.2%. Details are presented in Figure 1.

Table 2

Lymphocyte subsets in peripheral blood of patients with MIS-C

Patient 1Patient 2Patient 3Patient 4Patient 5Patient 6Patient 7
CD45+93%87%95%86%84%83%81%
CD19152/10%426/32%977/44%253/19%269/28%240/21%6/1%
CD31086/73%484/37%949/43%783/59%404/42%514/46%380/59%
CD3 (+) HLA-DR (+)22%3%3%2%2%10%12%
CD3 (–) HLA DR (+)13%37%50%25%29%30%21%
CD4761/51%375/28%674/31%546/41%233/24%243/22%262/41%
CD8280/19%77/6%189/9%217/16%140/155207/18%87/14%
CD4/CD82.724.903.562.521.671.173.00
NK50/3%158/12%132/6%48/4%238/25%200/18%109/17%
CD4
Naive CD4503/66.2%345/92.1%494/73.4%382/69.9%176/75.5%172/70.7%160/61%
Central mem CD4250/32.9%22/5.8%172/25.5%156/28.5%43/18.4%62/25.3%75/28.4%
Effector mem CD46/0.8%2/0.5%7/1.0%1/0.3%10/4.1%6/2.3%18/6.7%
Terminally Diff CD43/1.2%6/1.6%1/0.2%7/1.3%5/2%4/1.7%10/3.9%
CD4 CD45RO+173/22.8%98/26.2%191/28.4%143/26.2%84/36.2%86/35.3%100/38.1%
CD45RA+587/77.2%277/73.8%482/71.6%403/73.8%149/63.85157/64.7%162/61.9%
Recent Thymic Emigrants Th521/68.4%271/72.2%362/53.7%334/61%167/71.4%159/65.6%134/51.1%
Treg54/7%17/4.6%39/5.7%45/8.2%12/5.4%13/5.3%8/2.9%
CD8
Naive CD8227/81.3%46/60.7%143/75.7%198/78.3%116/83%108/52.3%22/25.4%
Central mem CD820/7.3%012/6.3%14/6.3%08/3.62/2.4%
Effector mem CD82/0.8%3/3.6%1/0.5%05/3.8%6/3.1%21/24.6%
Terminally Diff CD83/1.2%11/14.3%2/1.1%1/0.6%8/5.7%2/1%7/7.9%
Lymphocytes B
CD27+ Mem29/19.1%68.16%89/9.15%53/21%25/9.4%56/23.2%1/15%
CD27– Non mem123/80.9%358/84%888/90.9%200/79%243/90.6%184/76.8%5/85%
CD27+sIgD+ Non-Sw-Mem13/8.5%26/6%50/5.1%19/7.7%17/6.3%44/18.4%0
CD27–sIgD+ Naive109/71.35354/83%863/88.3%198/78.3%239/89%182/75.8%5/76.6%
CD27+sIgD– Sw-Mem16/10.6%43/10%39/4.0%34/13.3%8/3.1%11/4.7%0
CD27–sigD–15/9.6%4/1.0%26/2.7%2/0.7%4/1.6%3/1.1%1/8,4%
CD38hi sIgMhi Transitional16/10.6%14/3.2%203/20.8%31/12.3%10/3.7%21/8.8%1/10.6%
CD38loCD21lo Activated8/5.2%40/9.5%25/2.6%4/1.5%8/2.8%21/8.8%0
CD21lo Immature10/6.5%47/11.1%43/4.4%14/5.4%15/5.5%35/14.7%0
PLASMABLASTS00010/3.8%000

[i] CD4+ normal range

[ii] 1–5 years 1000 cells/mm3; 6–12 years 500 cells/mm3

[iii] CD8+ normal values

[iv] 1–5 years 300-1800 cells/mm3; 6–12 years 300–1600 cells/mm3

[v] CD3+ normal values

[vi] 1–5 years 900–4500 cells/mm3; 6–12 years 700–4200 cells/mm3

Fig. 1

Lymphocyte T CD4 subsets (terminally differentiated, effector memory, central memory, naive) in peripheral blood of patients with MIS-C

/f/fulltexts/CEJOI/47059/CEJI-47-47059-g001_min.jpg

The majority (6/7) of the analyzed patients had a higher CD4+/CD8+ T cell ratio than average values. Apart from low CD4+ levels, also a marked reduction in CD8+ T cells was detected. The majority of the T cells were naive cells (Figs. 1, 2). B lymphocytes were within the normal range – the majority were non-memory cells. Plasmablasts (PBs) were detected only in one patient. The number of NK cells was low in all patients.

Fig. 2

Lymphocyte T CD8 subsets (terminally differentiated, effector memory, central memory, naive) in peripheral blood of patients with MIS-C

/f/fulltexts/CEJOI/47059/CEJI-47-47059-g002_min.jpg

Discussion

The clinical symptoms and laboratory results of the observed children in MIS-C resemble those of children with KD or adult patients with acute COVID-19. The laboratory results such as high inflammatory markers, lymphopenia, neutrophilia, anemia, low platelets, altered coagulation, and high cardiac markers present in our patients are typical for adult patients with active severe COVID-19 [19, 20].

Upon recovery from COVID-19, results return to normal. Complications of COVID-19 and so-called “long COVID” seen in adults weeks after primary infection are not accompanied by high inflammatory marker levels [21].

During acute viral infections or after vaccination, PB responses are transiently detectable in the blood and correlate with CD4+ T cell responses [22, 23].

In the six out of seven observed patients, PBs were not detected. So anti-spike antibodies cannot be related to the mechanism of MIS-C development. This finding is significant regarding the vaccination which is now offered to adolescents. The risk of developing MIS-C cannot be established based on the IgG level. None of the children had severe COVID-19 in the past – three of them had no symptoms or mild symptoms – and did not need medical help with this. It appears that MIS-C is an inflammatory immune-mediated SARS-CoV-2 postinfectious process triggered by the virus rather than the effect of the prolonged viral infection.

Lymphopenia was observed in all our patients. Transient lymphopenia is quite common at the beginning of viral infections, but it resolves over time [23, 24]. Low lymphocyte levels, including reduced CD4+ and CD8+ T cells, are also seen in patients with acute COVID-19 – both children and adults [25, 26]. By contrast, COVID-19 patients had high polymorphonuclear leukocyte (PMN) counts; monocyte, eosinophil, and basophil counts were normal [25-27]. This is also commonly seen in both COVID-19 and MIS-C patients [28, 29]. A higher total T cell count, including both CD4+ and CD8+, is a predictor of less severe disease and more favorable clinical outcome. Robust CD4+ T cell activation, lack of cTFH cells with proliferating effector or exhausted CD8+ T cells, and PB involvement were associated with more severe disease [25, 26, 30]. Low levels of CD4+ in COVID-19 patients can be explained by extravasation and emigration to the site of infection. Effusions composed of T cells were detected in the lungs of patients with severe COVID-19 [31]. But this mechanism cannot explain lymphopenia in patients with mild COVID-19 or children. There are several causes of the observed lymphopenia in severe COVID-19 patients. SARS-CoV-2 can induce pyroptosis in lymphocytes via induction of the NLRP3 inflammasome [32]. Massive lymphocyte death can also be attributed to high levels of interleukin 6 (IL-6) as well as Fas-FasL interactions [33]. COVID-19 infection can result in exhaustion of T cells. Markers of T cell exhaustion – programmed cell death protein 1 (PD-1), T cell immunoglobulin and mucin domain 3 (Tim-3) – were found on the surface of CD4+ and CD8+ T cells from COVID-19 patients [34]. High levels of CXCL10 and CCL2 can suppress the development of hematopoietic precursor cells. T cell count returns to normal in recovered COVID patients [25].

The average CD4+/CD8+ ratio in healthy patients is not well defined. Ratios between 1.5 and 2.5 are typically regarded as normal [25]. The CD4+/CD8+ ratio was higher than usual in the observed group – a profound decrease in CD8+ cells caused it.

This was also found in other studies on adults with COVID-19 [34]. In MIS-C patients, the frequency of CD8+ T cells was lower compared to adults with COVID-19. In some studies in severe COVID-19 elevated levels of CD8+ T cells persist over time [22]. Low CD8 levels were observed in COVID-19 patients with a bad prognosis [21]. In HIV infected patients loss of CD4+ T cells is accompanied with an increase in CD8+ T cells, which compensates for the loss of CD4+ T cells [23].

Analyzing T cells subsets, we detected that the frequency of naive CD4+ and CD8+ T cells was higher than the reference range – this is typical for pediatric patients. For both CD4+ and CD8+ T cells, the frequency of central memory (CM, CD45RA–CD27+CCR7+) and effector memory 1 (EM1, CD45RA–CD27+CCR7–) was lower than the reference range. Similar findings were described by Vella et al. [28]. Terminally differentiated effector memory T cells (EMRA, CD45RA+ CD27- CCR7-) were present at a lower frequency in our pediatric cohorts. The naive, effector and central memory subset distributions were not different between the pediatric COVID-19 and MIS-C.

The period for peak CD8+ or CD4+ T cell responses during many acute viral infections and the window for PB detection in peripheral blood are usually short. A prolonged period of peak immune responses (CD8+, CD4+ T cell activation, and PB presence) observed during COVID-19 suggests a failure to appropriately down-regulate reactions in some patients. In the analyzed group B, cells were usually within the normal range. This was also noted in other studies [27-29]. Plasmablasts were not detected in our patients. A notable feature of some patients was a decrease in memory cells, effector memory cells, and terminally differentiated memory cells. In a study by Vella et al., they observed prolonged plasmablast responses and a profile of B-cell subsets similar to that in MIS-C and COVID-19 patients [28]. In this American study on 14 patients, 86% were PCR positive up to a cycle threshold of 45. They suggested that MIS-C can be caused by continued activation of the adaptive immune response, driven by persisting antigen in the later stage of a poorly controlled primary infection. Persistent antigen stimulation cannot explain all cases of MIS-C. Their data suggested that PB responses were altered compared to pediatric COVID-19. Prolonged PB presence was not detected in adults weeks after COVID-19. Elevated PB frequencies were surprising given that MIS-C occurs weeks after SARS-CoV-2 infection. In our group a boy with the lowest level of antibodies had a history of positive SARS-CoV-2 PCR six months earlier. But unfortunately, he was also the most affected patient. He developed severe cardiac complications.

Anemia is a prevalent finding in patients with severe COVID-19. It was previously reported that COVID-19-related anemia showed several RBC shape abnormalities [23]. Erythrocyte deformability may be involved in COVID-19 severity, causing tissue hypoxia. In three patients, we detected abnormalities in blood smear – the formation of erythrocyte “rouleaux”. These morphological changes may contribute to the microvascular thrombosis typical of COVID-19. In all patients with MIS-C, D-dimers were elevated.

Vascular inflammation, particularly endotheliitis, can explain many of the organ changes observed in patients with MIS-C and in KD patients. Similar to observational studies, our analysis shows that MIS-C is of higher preponderance in the age group of 5-14 years. This age distribution is different from that of classical KD, which is common in children of younger age. Lymphopenia and low platelets are not commonly seen in KD [35]. The immune profile of KD patients is very diverse. They do not have one immune trait. In a study published by Burns et al., CD4+ T cells were in the normal and low range; some patients showed significant CD8+ lymphocytosis [36].

The weak aspect of this study is the small group of patients. It was difficult to enroll more patients over a short period of time. This can be explained by the fact that MIS-C is a rare clinical entity; blood samples had to be collected and immediately tested before introduction of the treatment. It is not uncommon for MIS-C studies to be performed on such a small group of patients. These preliminary results will be further developed when new MIS-C patients are treated.

The immune profile of the studied patients differs from that of children with KD, but it is similar to that of adults with severe COVID-19. Immune exhaustion can be a possible mechanism of MIS-C. The proposed explanation is profound lymphopenia caused by SARS-CoV-2 infection – which persists for weeks, leading to uncontrolled inflammation and immune dysregulation. Our data suggest that MIS-C is an inflammatory disease similar to severe COVID-19 in adults. This study helps to understand the immune status of MIS-C patients before initiation of treatment and gives a hint of possible triggering mechanisms.

There is a question of whether lymphopenia observed in MIS-C is a consequence of lymphopenia caused by SARS-CoV-2. According to our data, the activation-induced cell death (AICD) mechanism may explain both lymphopenia as well as depletion of activated T cells and suppression of the B cell response.

In COVID-19 patients the T cell level returns to normal after the second week of the disease. Our data suggest that in children prolonged lymphopenia after COVID-19 can be a practical marker for possible MIS-C alert. If there is a continuum from lymphopenia to MIS-C, there is a room for screening and prevention. Further studies are needed to determine whether steroid treatment introduced in a child with prolonged lymphopenia could stop the inflammatory process.

Conclusions

  1. Lymphopenia and functional aberrations in T-cell subpopulations were detected in all patients with MIS-C.

  2. Patients with MIS-C displayed reduced CD4+ and CD8+ T cell levels.

  3. The CD4+/CD8+ratio was increased in all patients.

  4. Predominance of naive CD4+ and CD8+ cells and suppression of terminally differentiated B and T cells were observed. The observed low activation of memory T cells usually occurs in viral infections.

  5. Possible mechanism of MIS-C – prolonged lymphopenia caused by viral infection leads to an inflammatory response.

Notes

[9] Conflicts of interest The authors declare no conflict of interest.

References

1 

Verdoni L, Mazza A, Gervasoni A, et al. (2020): An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet 2020; 395: 1771-1778.

2 

European Centre for Disease Prevention and Control (ECDC). Rapid risk assessment: Paediatric inflammatory multisystem syndrome and SARS-CoV-2 infection in children. Available online: https://www.ecdc.europa.eu/en/publications-data/paediatric-inflammatory-multisystem-syndrome-and-sars-cov-2-rapid-risk-assessment.

3 

Whittaker E, Bamford A, Kenny J, et al. (2020): Clinical characteristics of 58 children with a pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2. JAMA 2020; 324: 259-269.

4 

World Health Organization (2020): Multisystem inflammatory syndrome in children and adolescents with COVID-19. Published May 15, 2020. www.who.int/publications-detail/multisystem-inflammatory-syndrome-in-children-and-adolescents-with-covid-19 (accessed: June 5, 2021).

5 

American Academy of Pediatrics. CDC details COVID-19-related inflammatory syndrome in children. www.aappublications.org/news/2020/05/14/covid19inflammatory051420 (accessed: June 5, 2021).

6 

Walker DM, Tolentino VR (2020): COVID-19: The impact on pediatric emergency care. Pediatr Emerg Med Pract 17 (Suppl 6-1): 1-27.

7 

Riphagen S, Gomez X, Gonzalez-Martinez C, et al. (2020): Hyperinflammatory shock in children during COVID-19 pandemic. Lancet 395: 1607-1608.

8 

Guimarães D, Pissarra R, Reis-Melo A, Guimarães H (2021): Multisystem inflammatory syndrome in children (MISC): a systematic review. Int J Clin Pract 75: e14450.

9 

Cattalini M, Della Paolera S, Zunica F, et al.; Rheumatology Study Group of the Italian Pediatric Society (2021): Defining Kawasaki disease and pediatric inflammatory multisystem syndrome-temporally associated to SARS-CoV-2 infection during SARS-CoV-2 epidemic in Italy: results from a national, multicenter survey. Pediatr Rheumatol Online J 19: 29.

10 

Belhadjer Z, Méot M, Bajolle F, et al. (2020): Acute heart failure in multisystem inflammatory syndrome in children (MIS-C) in the context of global SARS-CoV-2 pandemic. Circulation (published online May 17).

11 

Belot A, Antona D, Renolleau S, et al. (2020): SARS-CoV-2-related paediatric inflammatory multisystem syndrome, an epidemiological study, France, March 1 to May 17 2020. Euro Surveill 25: 2001010.

12 

Morris SB, Schwartz NG, Patel P, et al. (2020): Case series of multisystem inflammatory syndrome in adults associated with SARS-CoV-2 infection–United Kingdom and United States, March-August 2020. MMWR Morb Mortal Wkly Rep 69: 1450-1456.

13 

Ozsurekci Y, Gurlevik S, Kesici S, et al. (2021): Multisystem inflammatory syndrome in children during the COVID-19 pandemic in Turkey: first report from the Eastern Mediterranean. Clin Rheumatol 40: 3227-3237.

14 

Maucourant C, Filipovic I, Ponzetta A, et al.; Karolinska COVID-19 Study Group (2020): Natural killer cell immunotypes related to COVID-19 disease severity. Sci Immunol 5: eabd6832.

15 

Mathew D, Giles JR, Baxter AE, et al. (2020): Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369: eabc8511.

16 

Laing AG, Lorenc A, Del Molino Del Barrio I, et al. (2020): A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat Med 26: 1623-1635.

17 

Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. (2020): Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181: 1036-1045.e9.

18 

Royal College of Paediatrics and Child Health Guidance – Paediatric multisystem inflammatory syndrome temporally associated with COVID-19. https://www.rcpch.ac.uk/resources/guidance-paediatric-multisystem-inflammatory-syndrome-temporally-associated-covid-19 (May 5, 2020).

19 

Liu J, Li S, Liang B, et al. (2020): Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMed 55: 102763.

20 

Feng X, Li S, Sun Q (2020): Immune-inflammatory parameters in COVID-19 cases: a systematic review and meta-analysis. Front Med (Lausanne) 7: 301.

21 

Yong SJ (2021): Long COVID or post-COVID-19 syndrome: putative pathophysiology, risk factors, and treatments. Infect Dis (Lond) 53: 737-754.

22 

Ni M, Tian FB, Xiang DD, Yu B (2020): Characteristics of inflammatory factors and lymphocyte subsets in patients with severe COVID-19. J Med Virol 92: 2600-2606.

23 

Yuan X, Huang W, Ye B, et al. (2020): Changes of hematological and immunological parameters in COVID-19 patients. Int J Hematol 112: 553-559.

24 

Wang F, Nie J, Wang H, et al. (2020): Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J Infect Dis 221: 1762.e1769.

25 

Liu Z, Long W, Tu M, et al. (2020): Lymphocyte subset (CD4+, CD8+) counts reflect the severity of infection and predict the clinical outcomes in patients with COVID-19. J Infect 81: 318-356.

26 

Diao B, Wang C, Tan Y, et al. (2020): Reduction and functional exhaustion of t cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol 11: 827.

27 

Wilk AJ, Rustagi A, Zhao NQ, et al. (2020): A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med 26: 1070-1076.

28 

Vella L, Giles JR, Baxter AE, et al. (2020): Deep immune profiling of MIS-C demonstrates marked but transient immune activation compared to adult and pediatric COVID-19. medRxiv 2020.09.25.20201863.

29 

Beckmann ND, Comella PH, Cheng E, et al. (2020): Cytotoxic lymphocytes are dysregulated in multisystem inflammatory syndrome in children. medRxiv 2020.08.29.20182899.

30 

Hou H, Zhang B, Huang H, et al. (2020): Using IL-2R/lymphocyte for predicting the clinical progression of patients with COVID-19. Clin Exp Immunol 201: 76-84.

31 

Xiong Y, Liu Y, Cao L, et al. (2020): Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg Microbes Infect 9: 761-770.

32 

Ouyang Y, Yin J, Wang W, et al. (2020): Down-regulated gene expression spectrum and immune responses changed during the disease progression in COVID-19 patients. Clin Infect Dis 71: 2052-2060.

33 

Mazzoni A, Salvati L, Maggi L, et al. (2020): Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest 130: 4694-4703.

34 

Consiglio CR, Cotugno N, Sardh F, et al. (2020): The immunology of multisystem inflammatory syndrome in children with COVID-19. Cell 183: 968-981.e7.

35 

Brown TJ, Crawford SE, Cornwall ML, et al. (2001): CD8 T lymphocytes and macrophages infiltrate coronary artery aneurysms in acute Kawasaki disease. J Infect Dis 184: 940-943.

36 

Burns JC, Hsieh LE, Kumar J, et al. (2020): Characterization of circulating immune cells in acute Kawasaki disease suggests exposure to different antigens. Clin Exp Immunol 202: 263-272.

Copyright: © 2022 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.