Abstract
Background
Multisystem inflammatory syndrome in children (MIS-C) is a potentially life-threatening sequela of severe acute respiratory syndrome coronavirus 2 infection characterized by hyperinflammation and multiorgan dysfunction. Although hyperinflammation is a prominent manifestation of MIS-C, there is limited understanding of how the inflammatory state of MIS-C differs from that of well-characterized hyperinflammatory syndromes such as hemophagocytic lymphohistiocytosis (HLH).
Objectives
We sought to compare the qualitative and quantitative inflammatory profile differences between patients with MIS-C, coronavirus disease 2019, and HLH.
Methods
Clinical data abstraction from patient charts, T-cell immunophenotyping, and multiplex cytokine and chemokine profiling were performed for patients with MIS-C, patients with coronavirus disease 2019, and patients with HLH.
Results
We found that both patients with MIS-C and patients with HLH showed robust T-cell activation, markers of senescence, and exhaustion along with elevated TH1 and proinflammatory cytokines such as IFN-γ, C-X-C motif chemokine ligand 9, and C-X-C motif chemokine ligand 10. In comparison, the amplitude of T-cell activation and the levels of cytokines/chemokines were higher in patients with HLH when compared with patients with MIS-C. Distinguishing inflammatory features of MIS-C included elevation in TH2 inflammatory cytokines such as IL-4 and IL-13 and cytokine mediators of angiogenesis, vascular injury, and tissue repair such as vascular endothelial growth factor A and platelet-derived growth factor. Immune activation and hypercytokinemia in MIS-C resolved at follow-up. In addition, when these immune parameters were correlated with clinical parameters, CD8+ T-cell activation correlated with cardiac dysfunction parameters such as B-type natriuretic peptide and troponin and inversely correlated with platelet count.
Conclusions
Overall, this study characterizes unique and overlapping immunologic features that help to define the hyperinflammation associated with MIS-C versus HLH.
Key words: MIS-C, COVID-19, HLH, T-cell activation, hyperinflammation, cardiac dysfunction
Graphical abstract
Coronavirus disease 2019 (COVID-19) is less likely to result in hospitalization and death in children as compared with adults and older individuals1 , 2; however, children can develop a life-threating complication following exposure to severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) known as multisystem inflammatory syndrome in children (MIS-C).3 , 4 MIS-C is characterized by a prominent cardiac dysfunction with elevation of both B-type natriuretic peptide (BNP) and troponin. In addition to the heart, MIS-C affects multiple organs including the gastrointestinal tract, lungs, kidneys, brain, skin, or eyes.5 , 6 Because of prominent cardiac complications and hemodynamic instability, initial studies focused on differentiating MIS-C from Kawasaki disease (KD) or toxic shock syndrome. Systematic studies identified unique cardiac phenotypes in MIS-C characterized by more global cardiac dysfunction and myocarditis than in KD.7 , 8 Furthermore, KD is more commonly seen in children younger than 5 years,9 whereas MIS-C is observed more frequently after age 5 years.1 Further studies focused on immunobiology comparing the pathophysiology of MIS-C and KD revealed that MIS-C differs from KD. Lower frequency of T follicular helper cells was found in patients with MIS-C10 and more IL-17A–mediated inflammation in KD.11
Another pathognomonic manifestation of MIS-C is the systemic hyperinflammation.12 Some features such as hyperferritinemia, cytopenia, and elevated liver enzymes are reminiscent of hemophagocytic lymphohistiocytosis (HLH).12, 13, 14 In addition, management with high-dose steroids and biologics overlaps with the management of HLH.15 , 16 Thus far, limited data differentiating the immune signatures of MIS-C and HLH are available to provide insights into pathogenesis and ultimately patient management. Also, quantitative comparison of the T-cell activation state in MIS-C versus HLH has not been previously evaluated.
Using T-cell immunophenotyping and multiplex cytokine and chemokine profiling, we evaluated the immune signatures of children hospitalized for MIS-C and COVID-19 and compared them with those of children with HLH and healthy controls (HCs). We also evaluated how inflammatory markers correlated with cardiac dysfunction. Our findings could help understand the unique inflammatory milieu that contributes to the clinical manifestations observed in MIS-C.
Methods
Human subjects
Children with MIS-C and COVID-19 were enrolled between April 2020 and April 2021 at Children’s Healthcare of Atlanta. Patients with MIS-C were enrolled on the basis of case definition as described by the Centers for Disease Control and Prevention.17 Patients with HLH and age-matched HCs were included as a comparison cohort for MIS-C and COVID-19. Patients with HLH had mainly primary HLH, or EBV infection–associated HLH, and all of them fulfilled HLH-2004 diagnostic criteria.13 All patients were enrolled following informed consent, with approval by the Emory University Institutional Review Board.
Flow cytometry and analysis
PBMCs were used for immunophenotyping. Antibody information is provided in this article’s Online Repository at www.jacionline.org. Flow cytometry data were acquired on BD FACSymphony A5 (BD Biosciences, Franklin Lakes, NJ) and analyzed using FlowJo software v10 (BD, Ashland, Ore). T-cell activation was defined by coexpression of HLA-DR+ and CD38+ on effector memory (EM) CD4+ and CD8+ T cells.18, 19, 20 Coexpression of PD-1+ and Tim3+ on EM CD4+ and CD8+ T cells was defined as a marker for exhaustion.21 T-cell senescence was assessed by expression of CD57+ on the EM population of CD4+ and CD8+ T cells.22, 23, 24, 25 Definitions of T-cell populations analyzed in this study are presented in Table E1 in this article’s Online Repository at www.jacionline.org.
Plasma cytokine/chemokine profiling
Cytokine/chemokine profiling of plasma samples from patients with COVID-19 (n = 10), HLH (n = 8), MIS-C (n = 19), and MIS-C follow-up (n =10) and from HCs (n = 19) was performed on a luminex platform. The plasma samples for cytokine/chemokine profiling were chosen randomly from a cohort of 69 patients with MIS-C and 24 patients with COVID-19. A list of these analytes is provided in Table E2 in this article’s Online Repository at www.jacionline.org. Soluble IL-2 receptor (sIL-2R) and ferritin levels were done in clinical lab as standard of care. Plasma levels of sCD163 were measured using human CD163 ELISA kit (Abcam, ab274394; Boston, Mass) as per manufacturer’s instructions.
Statistics
Cytokine/chemokine data were represented as dot plots showing all the data points using Graphpad Prism version 9 (GraphPad Software, San Diego, Calif). One-way ANOVA with multiple comparison test was used for calculating significant differences between groups. Frequencies of immune parameters were represented with median and interquartile range. Fisher exact test was used to compute significance for categorical data. We applied a principal-component analysis on all the cytokines showing differences among patient groups based on principal-component 1 and principal-component 2 and created using Factoextra R package. Heatmap of all the reported cytokines was generated in pheatmap R package. Spearman correlation was used to find the correlation between clinical and flow-based parameters; correlation coefficients and P values were reported. Correlation matrix was made in “corrplot” package in R.
Results
Cohort description and clinical characteristics
We enrolled 69 patients with MIS-C (age, 2-19 years; median, 11 years), 24 patients hospitalized with COVID-19 but without MIS-C (age, 2-17 years; median, 11.5 years), 13 patients with HLH (age, 1 day-19.3 years; median, 1.2 years), and 22 HCs (age, 8-25 years; median, 17 years).
The demographic characteristics, clinical parameters, and standard of care laboratory tests for patients with MIS-C, COVID-19, and HLH are detailed in Table I and in Table E3 in this article’s Online Repository at www.jacionline.org. A follow-up immune evaluation was performed for 31 patients with MIS-C, median follow-up of 2 months postdiagnosis of MIS-C (range, 1-7 months) (see Fig E1 in this article’s Online Repository at www.jacionline.org).
Table I.
Parameters | n | MIS-C | n | COVID-19 | P value |
---|---|---|---|---|---|
Demographic data | |||||
Age (y), median (range) | 69 | 11 (2-19) | 24 | 11.5 (2-17) | .13 |
Sex (male:female) | 69 | 50:19 | 24 | 9:15 | .003 |
Clinical data | |||||
WBC∗ (cells/μL), median (range) | 69 | 14,340 (4,140-47,240) | 21 | 12140 (5,260-65,720) | .083 |
ANC∗ (cells/μL), median (range) | 69 | 12,040 (2,620-34,020) | 21 | 7,200 (2,410-22,450) | .009 |
ALC† (cells/μL), median (range) | 69 | 648 (140-2,706) | 20 | 996 (200-6,776) | .024 |
Platelet counts† (× 103 cells/μL), median (range) | 69 | 140 (18-726) | 22 | 226 (19-447) | <.0001 |
Creatinine∗ (mg/dL), median (range) | 69 | 0.66 (0.32-7.49) | 20 | 0.59 (0.25-1.11) | .1 |
ALT∗ (U/L), median (range) | 69 | 47 (12-2,109) | 19 | 47 (16-205) | .98 |
BNP∗ (pg/mL), median (range) | 69 | 690.1 (19.8-6,995) | 15 | 24.5 (10-852) | <.0001 |
Troponin∗ (ng/mL), median (range) | 69 | 0.15 (0.015-13.8) | 15 | 0.02 (0.015-1.92) | .002 |
Ferritin∗ (ng/mL), median (range) | 67 | 594.7 (89.73-4,744) | 16 | 137.66 (9.1-982.39) | <.0001 |
sIL-2R (U/mL), median (range) | 48 | 4792 (525.6-20,816) | 21 | 1091 (328.5-6,106) | <.0001 |
CRP∗ (mg/dL), median (range) | 69 | 15.7 (3.9-43.7) | 18 | 8.3 (0.3-23.9) | <.0001 |
ICU admission | 69 | 49 | 24 | 14 | .31 |
Low flow O2 | 69 | 31 | 24 | 12 | .81 |
High flow O2 | 69 | 24 | 24 | 8 | >.9999 |
Values in parentheses represent observed range of each parameter.
ALC, Absolute lymphocyte count; ALT, alanine transaminase; ANC, absolute neutrophil count; ICU, intensive care unit; WBC, white blood cell count.
Maximum values recorded.
Minimum values recorded.
Seventy-one percent of patients with MIS-C required intensive care unit care, whereas 58.3% of patients with COVID-19 were admitted to intensive care unit. Most patients with MIS-C were treated with steroids and intravenous immunoglobulin (see Fig E2 in this article’s Online Repository at www.jacionline.org). In our cohort, children with MIS-C had higher ferritin, C-reactive protein (CRP), neutrophil count, and thrombocytopenia compared with those with COVID-19 (Table I), consistent with the previously published reports.6 , 11 , 26 , 27
Inflammatory milieu reveals differences and similarities between MIS-C and HLH
To investigate how qualitatively the inflammatory response of patients with MIS-C differs from that of patients with HLH in plasma, we performed multiplex cytokine/chemokine profiling of 66 analytes in MIS-C (n = 19), MIS-C follow-up (n = 10), COVID-19 (n = 10), HLH (n = 8), and HCs (n = 19) (Fig 1 , A). Principal-component analysis of all these cytokines showed that both patients with HLH and patients with MIS-C formed distinct clusters in comparison with patients with COVID-19, HCs, and follow-up MIS-C patients (Fig 1, B).
We observed a cluster of 17 cytokines, that is, IFN-γ, C-X-C motif chemokine ligand 9 (CXCL9), C-X-C motif chemokine ligand 10 (CXCL10), TNF, IL-6, IL-8, IL-10, IL-15, IL-18, IL-27, IL-1α, IL-1RA, CXCL13, FLT-3L, M-CSF, MIP-1β, and MCP-2, that were significantly elevated in both patients with MIS-C and patients with HLH. A decreased level of macrophage-derived chemokine or CCL22 was observed in patients with HLH and patients with MIS-C. Of these cytokines, concentrations of CXCL9, IL-6, IL-8, IL-18, and M-CSF were higher in patients with HLH as compared with patients with MIS-C.
Next, we also looked at the unique cytokine signature in patients with MIS-C. Importantly, significant elevation in vascular endothelial growth factor A (VEGF-A), sCD40L, IFN-α2, IL-4, IL-13, platelet-derived growth factor (PDGF)-AA, and TARC was found only in patients with MIS-C but not in patients with HLH. However, eotaxin, GRO-α, MCP-1, RANTES, I309, cutaneous T-cell–attracting chemokine, MIP-1δ, and IL-12p40 were significantly increased in patients with HLH but not in patients with MIS-C. Cytokines such as MIP-1α, TGF-α, lymphotoxin-alpha, eotaxin-2, eotraxin-3, and IL-16 were significantly elevated in patients with MIS-C as compared with controls, but we did not find a significant difference between patients with HLH and patients with MIS-C. Similarly, fractalkine, G-CSF, and TPO were significantly elevated in patients with HLH as compared with HCs but did not show differences with respect to patients with MIS-C. In children with COVID-19, a limited number of cytokines such as IL-1RA, IL-6, PDGF-AA, MIP-1β, and MCP2 were significantly elevated.
Next, we sought to investigate the cytokine families that were differentially regulated in these cohorts. In general, patients with MIS-C showed prominent elevations in different cytokine families (Fig 2 , A-F). Cytokines and chemokines related to T-cell activation such as IFN-γ, IL-6, TNF, CXCL9, and CXCL10 were significantly elevated in both MIS-C and HLH groups. However, the amplitude of these cytokines was higher in patients with HLH when compared with the amplitude in patients with MIS-C (Fig 2, A). Interestingly, cytokine mediators responsible for angiogenesis, vascular injury, and tissue repair such as VEGF-A (P < .0001), PDGF-AA (P < .05), PDGF-AA/AB (P = .07), and FGF-2 (P = .06) were elevated in patients with MIS-C when compared with HCs but not in patients with HLH (Fig 2, E). Increased levels of these cytokines were also observed in some patients with COVID-19, although overall we did not find significance between COVID-19 and HC cohorts except for PDGF-AA (P = .04). TH2 inflammatory cytokines (ie, IL-4, IL-13) were significantly elevated in patients with MIS-C but not in patients with HLH. All the remaining cytokines are shown in Fig E3 in this article’s Online Repository at www.jacionline.org. As previously reported, we also evaluated whether the values of TNF and IL-10 in combination can differentiate between MIS-C and COVID-19 in our cohorts.28 We found similar observations where the sum of the plasma levels of TNF and IL-10 was significantly elevated in patients with MIS-C versus patients with COVID-19 (Fig E3 extended).
Overall, these data demonstrate that patients with MIS-C and patients with HLH have some overlap in the inflammatory milieu; however, the amplitude of the inflammation is much higher in patients with HLH. In addition, differential expression of certain inflammatory cytokines and chemokines in patients with MIS-C suggests unique inflammatory pathways that are active in patients with MIS-C but not in patients with HLH.
T-cell activation is higher in MIS-C and overlaps with HLH
Increased T-cell activation has been reported in both pediatric COVID-19 and MIS-C.29, 30, 31 A profound elevation of IFN-γ and its induced chemokines, that is, CXCL9 and CXCL1032, 33, 34 and other inflammatory markers such as IL-6 and TNF, in patients with HLH when compared with patients with MIS-C suggests higher T-cell activation in HLH in comparison to MIS-C. To further strengthen these observations and to investigate how T-cell activation in MIS-C and COVID-19 compares with that of HLH, we assessed the expression of activation markers on CD4+ and CD8+ T-cell subsets. The gating strategy for these analyses is shown in Fig E4 in this article’s Online Repository at www.jacionline.org. We first evaluated HLA-DR+ CD38+ expression in the EM compartment of CD4+ and CD8+ T cells. CD8+ EM T-cell activation was noted in both patients with COVID-19 and patients with MIS-C. In COVID-19, the T-cell activation was modest (5-fold; P = .0022), whereas MIS-C showed higher activation (13-fold; P < .0001) when compared with the median for HCs. Quantitatively, the amplitude of CD8+ EM T-cell activation in patients with HLH was 2-fold greater than that in patients with MIS-C. Although CD8+ EM T-cell activation was significantly higher in patients with HLH than in patients with MIS-C, we observed that a subset of patients with MIS-C had similar CD8+ EM T-cell activation as of patients with HLH (Fig 3 , A and C). Similarly, although the CD4+ EM T compartment was activated in both patients with MIS-C and patients with HLH (Fig 3, B and D), CD4+ EM T-cell activation was 6 times higher in patients with HLH than in patients with MIS-C. Using receiver-operating characteristic (ROC) statistics, we calculated the optimal threshold value for CD8+ EM T-cell activation that can differentiate patients with MIS-C from HCs, patients with COVID-19, and patients with HLH with high sensitivity and specificity (see Fig E5, A-C, in this article’s Online Repository at www.jacionline.org). We observed that CD8+ EM T-cell activation more than 79.9% can differentiate patients with HLH from patients with MIS-C with high area under the ROC curve (0.99) along with high sensitivity (100%) and specificity (98.6%). Similarly, cutoff of CD8+ EM T-cell activation more than 15.9% (area under the ROC curve, 0.98; sensitivity, 89.9%; specificity, 95.5%) and more than 27.4% (area under the ROC curve, 0.79; sensitivity, 72.5%; specificity, 75%) could help differentiate MIS-C from HC and COVID-19, respectively. To provide a more global picture of T-cell activation, we also evaluated HLA-DR+ CD38+ expression on central memory and TEMRA populations of CD4+ and CD8+ T cells and also on total CD4+ and CD8+ cells. A similar trend of activation was observed as seen previously in the EM compartment (see Fig E6 in this article’s Online Repository at www.jacionline.org).
T-cell activation measured by the expression of HLA-DR+ PD-1+ on the EM compartment of CD4+ and CD8+ T cells also showed similar trends noted with HLA-DR+ and CD38+ expression. The expression of HLA-DR+ PD-1+ was significantly elevated in both CD4+ and CD8+ T cells in patients with MIS-C as well as patients with HLH; however, the expression of HLA-DR+ PD-1+ was much higher in patients with HLH as compared with patients with MIS-C (Fig 3, E and F). Because we observed increased activation in CD8+ T EM compartment in MIS-C cohorts, we aimed to further evaluate whether increased activation in CD8 EM compartment resulted in relative expansion of CD8+ EM compartment. Therefore, we calculated the ratio of the frequencies of EM and naive compartments. Interestingly, we found a relative expansion of the EM compartment in patients with HLH but not in patients with MIS-C (see Fig E7, A, in this article’s Online Repository at www.jacionline.org). The CD4+/CD8+ ratio was similar between patients with MIS-C and patients with HLH. When compared with patients with MIS-C, patients with COVID-19 had overall higher CD4+/CD8+ ratio, although nonsignificant (Fig E7, B). Next, we also evaluated CD4+ and CD8+ TEMRA populations. Although TEMRA populations were similar among different patient cohorts, we found an increase of this subset in MIS-C follow-up patients when compared with onset MIS-C patients (Fig E7, C and D).
Patients with MIS-C are marked by high surface markers of T-cell exhaustion and senescence
Patients with MIS-C have been reported to have prolonged presence of severe acute respiratory syndrome coronavirus 2 in the gastrointestinal tract35; thus, we hypothesized that chronic antigenic exposure in MIS-C and associated T-cell activation could potentially lead to a postactivation exhaustion state of T cells and show features of proliferation-induced senescence. To test these hypotheses, we evaluated the expression of T-cell exhaustion and senescence surface markers in CD4+ and CD8+ EM T cells for these patient cohorts. T-cell exhaustion was evaluated by coexpression of PD-1+ and Tim3+ on EM CD4+ and CD8+ T cells. Significant increase in T-cell exhaustion markers was observed in both patients with MIS-C and patients with HLH in both CD4+ and CD8+ EM T cells; however, the frequency of cells expressing exhaustion markers was much higher in patients with HLH when compared with patients with MIS-C (Fig E7, E and F). We also observed a modest increase in exhaustion markers in patients with COVID-19 when compared with HCs in both CD4+ and CD8+ T EM compartments. Also, patients with MIS-C displayed significantly higher CD57+ expression on CD8+ EM cells (Fig E7, G). Interestingly, we did not find any difference in CD57+ expression for COVID-19 and HLH cohorts when compared with HCs. Similarly, we found a significant increase in the expression of CD57+ on CD4+ EM T cells in patients with MIS-C (Fig E7, H). Although patients with COVID-19 did not show any difference, patients with HLH showed a marked increase in CD57+ expression when compared with HCs. These phenotypic markers of exhaustion and senescence on T cells reassuringly return to HCs range at follow-up in patients with MIS-C.
Clinical parameters of MIS-C and COVID-19 overlap with HLH
To understand where patients with MIS-C and patients with COVID-19 fall into the clinical spectrum of HLH, we evaluated patients with MIS-C and patients with COVID-19 based on clinical laboratory parameters defined in the 2004 HLH criteria.13 We compared some important clinical and laboratory features such as cytopenias, ferritin, fibrinogen, and sIL-2R in patients with MIS-C and patients with COVID-19. About 83.3% and 33.3% of patients with MIS-C and patients with COVID-19, respectively, had sIL-2R levels greater than or equal to 2400 U/mL; 61.2% and 18.8% of patients with MIS-C and patients with COVID-19, respectively, were noted to have ferritin levels greater than or equal to 500 ng/mL. In patients with MIS-C, 16.7% had platelets less than 100 × 109/L, 3.4% had absolute neutrophil count less than 1.0 × 109/L, 3.3% had hemoglobin less than 90 g/L, and 1.8% had fibrinogen less than or equal to 1.5 g/L. However, none of the patients with COVID-19 had low platelets, absolute neutrophil count, hemoglobin, and fibrinogen as per 2004 HLH criteria (see Table E4 in this article’s Online Repository at www.jacionline.org). Recent publications have evaluated overlap of MIS-C and COVID-19 with HLH, and our cohort validates some of these findings.36 Although we compared important parameters of HLH 2004 criteria, a formal application of these criteria was not possible because all the clinical and lab parameters needed for evaluation were not obtained in all patients. However, individual laboratory parameters that were consistently available for these patients were assessed. A direct comparison of standard parameters of HLH such as ferritin, sIL-2R, and sCD163 revealed that patients with HLH had significantly higher plasma levels of ferritin, sIL-2R, and sCD163 when compared with patients with MIS-C, whereas patients with COVID-19 had lower levels of ferritin, sIL-2R, and sCD163 when compared with patients with MIS-C (Fig 4 , A-C). These findings validate the T-cell activation and inflammatory signature as seen in our previous observations.
Neutrophil to lymphocyte ratio has been proposed as a distinguishing parameter in MIS-C and COVID-19 illness.26 , 37 As reported previously, neutrophil to lymphocyte ratio was higher in patients with MIS-C when compared with patients with COVID-19, with a trend toward significance (median, 8.2 vs 3.8; P = .06). However, this ratio was significantly lower in patients with HLH compared with patients with COVID-19 (median, 0.34 vs 3.8; P = .005) and patients with MIS-C (median, 0.34 vs 8.2; P < .0001) (Fig 4, D). In addition, to determine whether the neutrophil counts differ with respect to T-cell activation in these patient cohorts, we assessed the ratio of ANC with CD8+ and CD4+ EM T-cell activation. We observed that these ratios could easily distinguish these clinical entities because these were higher in patients with COVID-19 when compared with patients with MIS-C (P = .08) and patients with HLH (P < .001), whereas patients with HLH had significantly lower ratios when compared with patients with MIS-C and patients with COVID-19 (Fig 4, E and F).
Inflammatory markers resolve at follow-up in patients with MIS-C
Although previous studies describing immunophenotypic differences in MIS-C have only focused on the acute disease state at onset, follow-up studies are limited.26 In our study, we assessed T-cell–based immune markers as well as cytokine/chemokine profile during follow-up. Immune evaluation of patients with MIS-C at follow-up revealed a significant decrease in both CD4+ and CD8+ EM T-cell activation with return to HCs (Fig 3, C-F; see Fig E8 in this article’s Online Repository at www.jacionline.org). In addition, we observed a decrease in T-cell exhaustion as well as senescence markers at follow-up in both CD4 and CD8 EM subsets (Fig E7, E-H, and Fig E8). In a subset of patients with MIS-C, we also assessed the levels of cytokines and chemokines at follow-up. Almost all of the elevated cytokines had resolved to normal levels in patients with MIS-C (Fig 2, G). We also assessed some clinical inflammatory markers such as ferritin and CRP at hospital admission and after 7 days of follow-up in some patients with MIS-C for whom longitudinal data were available and found a significant decrease in their levels on follow-up, suggesting rapid response of systemic inflammation with treatment (Fig E8, F).
T-cell activation correlates with cardiac inflammatory markers in patients with MIS-C
MIS-C and severe COVID-19 have been found associated with rise in acute myocardial markers such as BNP and troponin.26 , 38, 39, 40 To investigate whether T-cell activation correlates with the cardiac inflammatory markers and disease severity in COVID-19 and MIS-C, we first compared the BNP and troponin levels in patients with COVID-19 and patients with MIS-C. In our cohort, we found that both BNP and troponin are significantly higher in patients with MIS-C as compared with patients with COVID-19, validating the high occurrence of cardiac dysfunction and cardiac injury in patients with MIS-C (see Fig E9, A and B, in this article’s Online Repository at www.jacionline.org). We next checked whether there is an association of cardiac dysfunction markers with T-cell activation in patients with MIS-C and patients with COVID-19. We found a correlation of troponin levels with CD8+ and CD4+ EM T-cell activation (Fig 5 , A and B). Similarly, BNP levels correlated with CD8+ EM T-cell activation as well as CD4+ EM T-cell activation (Fig 5, C and D). We found that patients with T-cell activation greater than the threshold value of more than 15.9% CD8+ EM activation (threshold determined on the basis of ROC between MIS-C and HCs) have significantly high BNP and troponin levels as compared with patients with T-cell activation lower than the cutoffs values (Fig E9, C and D). Next, we chose cutoff values for BNP (200 pg/mL; 2 times the upper limit of normal) and troponin (0.09 ng/mL; 2 times the upper limit of normal) for calculating the odds ratio of finding high BNP or troponin in patients with high CD8+ EM T-cell activation. Our data showed that patients having higher CD8+ EM T-cell activation (>15.9%) were 9.1 times (95% CI, 2.7-30.1) more likely to have elevated BNP levels (>200 pg/mL) than patients with low T-cell activation. Similarly, patients with high CD8+ EM T-cell activation were 6.2 (95% CI, 1.8-21.3) times more likely to have high troponin (>0.09 ng/mL) levels than the patients who had lower T-cell activation. This suggests that T-cell activation might directly or indirectly contribute to cardiac pathology with elevation of BNP and troponin levels in these children with MIS-C.
We also correlated other inflammatory markers with cardiac dysfunction markers in patients with MIS-C and COVID-19. Ferritin and CRP levels correlated with both BNP and troponin levels (Fig 5, E-H). Ferritin and CRP levels also correlated with CD8+ EM T-cell activation (Fig 6 , C). Thrombocytopenia was frequent in MIS-C. We observed in patients with MIS-C and COVID-19 platelet counts were inversely correlated with CD4+ and CD8+ EM T-cell activation (Fig 6, A and B). In addition to T-cell activation, other inflammatory markers such as ferritin levels correlated with elevation of liver enzyme alanine transaminase and creatinine, but inversely correlated with platelet count in these patients (Fig 6, C). We also compared these parameters separately in patients with MIS-C and patients with COVID-19. Because steroid treatment might affect some of activation readout and laboratory features over time, we evaluated patients with MIS-C in whom sampling was done either presteroids or within the first 48 hours of steroid initiation. We found similar correlations in patients with MIS-C as observed in patients with MIS-C and COVID-19. However, because of the limited number of patients with COVID-19, we observed much lesser correlations between these variables (Fig E10).
Discussion
MIS-C is an immune dysregulation state characterized by hyperinflammation, with multisystem manifestations including myocarditis, cardiac dysfunction, respiratory failure, acute kidney injury, or gastrointestinal, dermatologic, or neurological involvement.1 , 17 , 26 , 41 Initially, MIS-C was identified in children, but later a similar presentation was also reported in adults.42, 43, 44 Although several groups have demonstrated an increase in inflammatory markers and T-cell activation in MIS-C,11 , 28 , 31 , 45 , 46 the exact nature and amplitude of hyperinflammation is still poorly defined. Hence, a comparison with an established hyperinflammatory state such as HLH offers additional insight into the immunopathogenesis of MIS-C. A number of similarities exist, but there are also qualitative and quantitative differences in clinical presentation and management of patients with MIS-C and patients with HLH. For example, hyperinflammation in MIS-C has been treated with steroids, and anakinra.16 , 47 Similar cytokine blockade and steroids are used for the management of patients with secondary forms of HLH.48 Although there are similarities between MIS-C and HLH, they differ in some clinical manifestations such as presence of myocarditis leading to cardiac dysfunction, and gastrointestinal manifestation such as acute abdomen or inflammatory bowel disease–like presentations, which are common in MIS-C but usually not a part of the disease process in HLH.14 , 48 In addition, pancytopenia and liver function test abnormities are more commonly seen in HLH but are infrequent in MIS-C. We hypothesized that despite certain clinical similarities, the amplitude and nature of hyperinflammation might be different in MIS-C when compared with HLH.
We found several similarities as well as striking differences between MIS-C and HLH. High T-cell activation was found in both MIS-C and HLH; however, the degree of T-cell activation was lower in MIS-C when compared with HLH. Despite the higher T-cell activation in HLH, we found some patients with MIS-C having CD8+ EM T-cell activation comparable to that seen in patients with HLH. Although CD4 versus CD8 ratio was similar in MIS-C and HLH, high CD8+ EM/naive ratio was observed in HLH, but not in MIS-C, suggesting more dramatic EM T-cell expansion in HLH. Children with COVID-19 had a modest increase in T-cell activation, this finding was not universal in all hospitalized children with COVID-19. In fact, most of these children had only modest T-cell activation when compared with controls, suggesting that unlike in MIS-C, T-cell activation may not be an important driver of disease morbidity in COVID-19 in children. Though we have shown upregulation of surface markers of exhaustion and senescence in T cells, the fact that they resolve during follow-up could suggest that a postactivation state induced transient upregulation of exhaustion markers rather than true exhaustion of T cells.49 Similarly, transient proliferative stress in the EM compartment of T cells could be the cause of upregulation of senescence marker.
Despite increased T-cell activation in MIS-C, the qualitative nature of T-cell activation is still poorly understood. Hence, we performed additional evaluation of plasma cytokines and chemokines in all patient cohorts. We found elevated plasma IFN-γ and associated chemokines (ie, CXCL9, CXCL10) in both MIS-C and HLH. Interestingly, differences in the degree of T-cell activation were also seen at the cytokine levels where median values for IFN-γ, CXCL9, and CXCL10 were high in HLH when compared with MIS-C. In addition to these cytokines, elevated IL-6 and TNF might be responsible for amplifying the cytokine storm in patients with MIS-C. Innate inflammation can also be a driver of T-cell activation.50 , 51 Elevated innate inflammatory cytokines such as IL-18, IL-15, IL-1α, and IL-1RA in MIS-C suggest that innate inflammatory pathways upregulation could play a role in modulating the T-cell activation noted in MIS-C.
Steroids and intravenous immunoglobulin remain the mainstay of therapy for MIS-C.15 , 16 , 52 However, additional biologics were needed for a subset of severely ill patients. Anakinra, tocilizumab, and infliximab are among the common biologics used in the steroid-refractory settings.27 , 53, 54, 55 Increase in plasma IL-1RA56 and IL-1857 , 58 in patients with MIS-C suggest the significant autoinflammatory component of this disease and reinforces the use of IL-1–blockade therapies such as anakinra in MIS-C.59 Increase in innate inflammatory signature and elevation of IL-6 and TNF in children with MIS-C noted in our study and previous reports could support the use of IL-6 and TNF blockade.28 , 45 , 46 , 55 , 60 Because these biologics are broadly used even in adults with severe COVID-19,61, 62, 63 medication shortages have become a concern. In this context, expanding our armamentarium of drugs available to manage MIS-C hyperinflammation would be advantageous. Agents such as Janus kinase and signal transducer and activator of transcription inhibitors have been used in both HLH and severe COVID-19.64, 65, 66, 67, 68 Given the overlapping hyperinflammation with HLH, similar strategies might be beneficial in patients with MIS-C and further studies are warranted. Similarly, IFN-γ–neutralizing antibody emapalumab has demonstrated efficacy in the management of HLH,69 and it is currently being evaluated for the management of severe COVID-19 (NCT04324021). Based on our data and previous findings of elevated INF-γ and its chemokines,28 , 55 emapalumab might also be another potential agent of interest to evaluate in the management of severe or refractory MIS-C.
We found some unique disease-defining markers in both MIS-C and HLH. For example, cytokines responsible for elevated TH2 inflammation, IL-4 and IL-13, were uniquely elevated in the MIS-C cohort but not in patients with HLH. Similarly, VEGF-A, PDGF-AA, PDGF-AA/AB, and FGF-2, which mainly contribute to angiogenesis, vascular injury, tissue repair, and healing, were found elevated in MIS-C and not in HLH. Elevated VEGF-A levels and angiogenesis have been shown to be involved in cardiovascular diseases.70 , 71 Thrombotic microangiopathy (TMA) has been reported as an important part of pathophysiology in patients with COVID-19 and MIS-C,72, 73, 74, 75, 76 and its etiology is likely multifactorial. Complement dysregulation characterized by IFN-γ signature, elevated soluble C5b9, and TMA is noted in both HLH and MIS-C.75, 76, 77 In addition to complement dysregulation, elevated VEGF levels might contribute to TMA-like phenotype in MIS-C. Previous literature from other systemic inflammatory diseases showed that increase in VEGF might contribute to TMA.78 , 79 Elevated VEGF might be related to high angiotensin II (AngII) from dysregulation of physiological balance of angiotensin-converting enzyme 2-angiotensin II pathway caused by SARS-COV-2 infection. Increased AngII is known to increase VEGF expression and levels80 , 81; together, they could contribute to endothelial activation and dysfunction. It is possible that angiotensin-converting enzyme 2-angiotensin II dysregulation persists in MIS-C after initial infection of SARS-COV-2, hence contributing to microangiopathy and organ injury. In addition to complement dysregulation and VEGF contributing to TMA, platelet activation as evident by increase in plasma PDGF levels82 may lead to platelet aggregation and consumption and contribute to thrombocytopenia and microangiopathy.83
Despite higher T-cell activation in HLH, significant cardiac dysfunction is uncommon. Elevation of troponin as well as VEGF-A in MIS-C but not in HLH suggests increased angiogenetic inflammation. Also, cardiac dysfunction along with TH2 phenotype might drive an antibody-driven vasculitis-like process in patients with MIS-C. The observation of concomitant TH2 activation and increase in angiogenic inflammatory markers might explain why most patients with HLH do not develop this degree of myocarditis and gastrointestinal vasculopathy when compared with patients with MIS-C. This suggests that although the nature of T-cell activation in HLH is overwhelmingly TH1 biased, in MIS-C, it is both TH1 and TH2. Moreover, the tropism of activated T cells might be different. In HLH, the tropism might be directed more to the bone marrow, liver, and other lymphoid organs, leading to more pancytopenia, liver dysfunction, and organomegaly, whereas in MIS-C, it might be directed more toward the heart, the gastrointestinal tract, and vascular inflammation.
Our correlation data revealed that global markers of inflammation such as ferritin and CRP are predictive markers of cardiac morbidity. In addition, more specific T-cell activation was shown to significantly correlate with cardiac inflammatory markers, suggesting a role of T-cell activation in cardiac and systemic manifestations in MIS-C. Such global markers of inflammation can also be used to track the disease course in these patients. Furthermore, most patients with MIS-C even at 2-month follow-up were found to have T-cell activation and cytokine and chemokine milieu comparable to that in HCs, suggesting that hyperinflammation in MIS-C caused by cytokine storm is transient, amenable to be controlled by therapies, and almost universally resolves with time.
Our study had a few limitations, and future studies are needed to further strengthen these observations. Because of its rarity, we could include only a limited number of patients with HLH. In future studies, these observations can be compared with a larger cohort of patients with HLH. Many patients with MIS-C had received some immunosuppressive intervention such as steroids before study enrollment. However, the high acuity of clinical presentation of patients with MIS-C and need to start immunomodulatory interventions immediately precludes access to biological samples of truly treatment-naive patients. These practical limitations were noted in other MIS-C and HLH studies describing immune activation and dysregulation.20 , 31 To minimize the effect of these interventions, most patients in our study were enrolled either pretherapy or within the first couple of days of steroid initiation, thus minimizing the effect of therapy on the research observations. Nonetheless, future studies focused on treatment-naive cohorts with MIS-C would be more desirable.
Conclusions
In this study, we characterized the similarities and differences in hyperinflammatory states of MIS-C and HLH. We found high T-cell activation and TH1-type inflammation in both MIS-C and HLH; however, the amplitude of T-cell activation and TH1 cytokines was higher in HLH versus MIS-C. In addition to TH1, elevation of TH2-type and angiogenic cytokines and chemokines was unique to MIS-C. We also found that T-cell activation as well as other clinical parameters such as ferritin and CRP correlated with cardiac dysfunction markers. Importantly, the hyperinflammation in MIS-C was transient and resolved at follow-up. Overall, our data revealed a comprehensive comparison elucidating both shared features and differences in immune signatures that might help guide treatment strategies for these distinct clinical entities.
Clinical implications.
Although the hyperinflammatory profiles of MIS-C and HLH overlap, they differ in amplitude and qualitative nature of immune activation. Global markers of inflammation and CD8+ EM T-cell activation markers correlate with cardiac morbidity in MIS-C. The inflammatory milieu in MIS-C resolves during follow-up.
Acknowledgments
Emory + Children’s (Pediatric) Flow Cytometry Core facility was used for flow cytometry studies.
Footnotes
This work was supported by the National Heart, Lung, and Blood Institute (grant no. 1K08HL141635-01A1), Atlanta Pediatric Scholars Program K12 Scholar (grant nos. K12HD072245 and U54AI082973 to S.C.), and the National Institute of Allergy and Infectious Diseases 3U19AI110483-07S1 to I.S. (PI) and S.C. (Co-I) and S.P. (Co-I). S.P. is supported in part by the Marcus Foundation, Inc, Atlanta.
Disclosure of potential conflict of interest: S. Chandrakasan serves on the advisory committee for SOBI. S. Prahalad serves on a macrophage activation syndrome adjudication committee for Novartis Pharmaceuticals and is an Op-Med fellow for Doximity for 2021-2022. E. J. Anderson has consulted for Pfizer, Sanofi-Pasteur, Janssen, and Medscape, and his institution receives funds to conduct clinical research unrelated to this manuscript from MedImmune, Regeneron, PaxVax, Pfizer, GlaxoSmithKline (GSK), Merck, Sanofi-Pasteur, Janssen, and Micron. He also serves on a safety monitoring board for Kentucky BioProcessing, Inc, and Sanofi-Pasteur. His institution has also received funding from the National Institutes of Health to conduct clinical trials of Moderna and Janssen coronavirus disease 2019 vaccines. C. A. Rostad’s institution has received funds to conduct clinical research unrelated to this manuscript from BioFire, Inc, GSK, MedImmune, Micron, Janssen, Merck, Moderna, Novavax, PaxVax, Pfizer, Regeneron, and Sanofi-Pasteur. The rest of the authors declare that they have no relevant conflicts of interest.
Antibodies used in this study
CD3-PerCP/Cy5.5 (Biolegend, Cat. #300430; San Diego, Calif), CD4-PE-Cy7 (BD Biosciences, Cat. #560649; Franklin Lakes, NJ), CD8-BUV395 (BD Biosciences, Cat. #563795), CD45RA-APC (Biolegend, Cat. #304112), CCR7-PE (Biolegend, Cat. #353204), HLA-DR-BV711 (BD Biosciences, Cat. #563696), CD38-BUV496 (BD Biosciences, Cat. #612947), PD-1-BV421 (BD Biosciences, Cat. #562516), CD57-BB515 (BD Biosciences, Cat. #565285), and Tim-3-BV650 (Biolegend, Cat. #345028) antibodies were used for staining cells. Live/dead fixable aqua dead cell stain (Thermofisher, Cat. # L34957; Waltham, Mass) was used to exclude dead cells in the analysis.
Table E1.
Population | Markers | References |
---|---|---|
CM CD4+ T | CD4+ CCR7+ CD45RA− | E1 |
CM CD8+ T | CD8+ CCR7+ CD45RA− | E1 |
EM CD4+ T | CD4+ CCR7− CD45RA− | E1 |
EM CD8+ T | CD8+ CCR7− CD45RA− | E1 |
TEMRA CD8+T | CD8+ CCR7− CD45RA+ | E1 |
TEMRA CD4+T | CD4+ CCR7− CD45RA+ | E1 |
Naive CD4+ T | CD4+ CCR7+ CD45RA+ | E1 |
Naive CD8+ T | CD8+ CCR7+ CD45RA+ | E1 |
Activated CD4+ EM | CD4+CD45RA−CCR7− HLA-DR+CD38+ | E2, E3, E4 |
Activated CD8+ EM | CD8+CD45RA−CCR7− HLA-DR+CD38+ | E2, E3, E4 |
Senescent CD4+ T | CD4+ CD45RA−CCR7− CD57+ | E5, E6, E7, E8 |
Senescent CD8+ T | CD8+ CD45RA−CCR7− CD57+ | E5, E6, E7, E8 |
Exhausted CD4+ | CD4+ CD45RA−CCR7− PD-1+ Tim3+ | E9 |
Exhausted CD8+ | CD8+ CD45RA−CCR7− PD-1+ Tim3+ | E9 |
CM, Central memory.
Table E2.
MCP-1 | EGF |
MCP-3 | Eotaxin |
M-CSF | FGF-2 |
CCL22 | FLT-3L |
MIG/CXCL9 | Fractalkine |
MIP-1α | G-CSF |
MIP-1β | GROα |
PDGF-AA | IFN-α2 |
PDGF-AB/BB | IFN-γ |
RANTES | IL-1α |
TGF-α | IL-1β |
TNF | IL-1RA |
LT-α | IL-4 |
VEGF-A | IL-5 |
Eotaxin-2 | IL-6 |
MCP-2 | IL-8 |
CXCL13 | IL-9 |
MCP-4 | IL-10 |
I-309 | IL-12p40 |
IL-16 | IL-12p70 |
TARC | IL-13 |
6CKine | IL-15 |
Eotaxin-3 | IL-17A |
LIF | IL-17E/IL-25 |
TPO | IL-17F |
TSLP | IL-18 |
IL-33 | IL-22 |
IL-20 | IL-27 |
IL-21 | IP-10/CXCL10 |
IL-23 | MIP-1δ |
TRAIL | IL-28A |
CTACK | ENA-78 |
SDF-1α+β | |
sCD40L |
CTACK, Cutaneous T-cell–attracting chemokine; LT-α, lymphotoxin-alpha.
Table E3.
Patient ID | Age | Sex | Causal factor (gene defect) | WBC (cells/μL) | ALC (cells/μL) | ANC (cells/μL) | Hemoglobin (g/L) | Platelets (cells × 103/μL) | Ferritin maximum (ng/mL) | sIL-2R (U/ mL) |
---|---|---|---|---|---|---|---|---|---|---|
1 | 5 mo | M | p-HLH (STXBP2) | 4,140 | 2,649 | 890 | 79 | 81 | 40,000 | 37,874 |
2 | 3 mo | M | p-HLH (UNC13D) | 6,370 | 4,650 | 1,210 | 82 | 40 | 13,672 | 47,474 |
3 | 1 d | F | p-HLH (STXBP2) | 10,370 | 6,222 | 2,177 | 117 | 63 | 12,488 | 11,165 |
4 | 3 mo | M | p-HLH (RAB27a) | 3,520 | 2,675 | 180 | 72 | <10 | 14,855 | 44,256 |
5 | 2 mo | M | p-HLH (RAB27a) | 2,600 | 1,508 | 390 | 72 | 23 | 5,473 | 22,268 |
6 | 2 y 8 mo | M | Suspected p-HLH∗ | 4,130 | 950 | 2,740 | 81 | 70 | 5,753 | 7,125 |
7 | 1 y 4 mo | M | Suspected p-HLH∗ | 890 | 480 | 400 | 112 | 80 | 5,067 | 13,625 |
8 | 1 y | M | EBV-HLH | 200 | 160 | 10 | 47 | <10 | 150,000 | 46,723 |
9 | 1 y | M | EBV-HLH | 1,930 | 1,022 | 870 | 85 | 82 | 170,000 | 10,426 |
10 | 19 y 4 mo | F | EBV-HLH | 2,390 | 1,888 | 300 | 84 | 47 | 29,094 | 11,849 |
11 | 7 y 4 mo | F | EBV-HLH | 1,260 | 441 | 720 | 79 | 10 | 40,000 | 15,132 |
12 | 3 y 2 mo | M | EBV-HLH | 5,720 | 4,347 | 1,020 | 83 | 85 | 28,410 | 5,422 |
13 | 12 y 6 mo | F | Infection-HLH† | 100 | 70 | 30 | 88 | 34 | 38,408 | 6,917 |
F, Female; M, male; p-HLH, primary HLH; RSV, respiratory syncytial virus.
Suspected p-HLH, but negative workup for classical cytotoxic pathway gene defects.
Presumed triggered by RSV.
Table E4.
Laboratory parameters in HLH 2004 criteria | MIS-C∗ | Percentage† | COVID-19∗ | Percentage† |
---|---|---|---|---|
Fibrinogen ≤1.5 g/L | 1 (57) | 1.8 | 0 (15) | 0 |
Platelets <100 × 109/L | 10 (60) | 16.7 | 0 (18) | 0 |
ANC <1.0 × 109/L | 2 (59) | 3.4 | 0 (19) | 0 |
Hemoglobin <90 g/L | 2 (60) | 3.3 | 0 (18) | 0 |
Ferritin ≥500 ng/mL | 41 (67) | 61.2 | 3 (16) | 18.8 |
sIL-2R ≥2400 U/mL | 40 (48) | 83.3 | 7 (21) | 33.3 |
The number of subjects who had abnormal lab parameters, and the values in parentheses represent the total number of subjects for whom the data were available.
Percentage of subjects who had abnormal lab parameters as defined in HLH 2004 criteria.
References
- 1.Feldstein L.R., Rose E.B., Horwitz S.M., Collins J.P., Newhams M.M., Son M.B.F., et al. Multisystem inflammatory syndrome in U.S. children and adolescents. N Engl J Med. 2020;383:334–346. doi: 10.1056/NEJMoa2021680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Brodin P. Why is COVID-19 so mild in children? Acta Paediatr. 2020;109:1082–1083. doi: 10.1111/apa.15271. [DOI] [PubMed] [Google Scholar]
- 3.Payne A.B., Gilani Z., Godfred-Cato S., Belay E.D., Feldstein L.R., Patel M.M., et al. Incidence of multisystem inflammatory syndrome in children among US persons infected with SARS-CoV-2. JAMA Netw Open. 2021;4 doi: 10.1001/jamanetworkopen.2021.16420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Riphagen S., Gomez X., Gonzalez-Martinez C., Wilkinson N., Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–1608. doi: 10.1016/S0140-6736(20)31094-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jiang L., Tang K., Levin M., Irfan O., Morris S.K., Wilson K., et al. COVID-19 and multisystem inflammatory syndrome in children and adolescents. Lancet Infect Dis. 2020;20:e276–e288. doi: 10.1016/S1473-3099(20)30651-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Dufort E.M., Koumans E.H., Chow E.J., Rosenthal E.M., Muse A., Rowlands J., et al. Multisystem inflammatory syndrome in children in New York State. N Engl J Med. 2020;383:347–358. doi: 10.1056/NEJMoa2021756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lee M.S., Liu Y.C., Tsai C.C., Hsu J.H., Wu J.R. Similarities and differences between COVID-19-related multisystem inflammatory syndrome in children and Kawasaki disease. Front Pediatr. 2021;9 doi: 10.3389/fped.2021.640118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sperotto F., Friedman K.G., Son M.B.F., VanderPluym C.J., Newburger J.W., Dionne A. Cardiac manifestations in SARS-CoV-2-associated multisystem inflammatory syndrome in children: a comprehensive review and proposed clinical approach. Eur J Pediatr. 2021;180:307–322. doi: 10.1007/s00431-020-03766-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tacke C.E., Breunis W.B., Pereira R.R., Breur J.M., Kuipers I.M., Kuijpers T.W. Five years of Kawasaki disease in the Netherlands: a national surveillance study. Pediatr Infect Dis J. 2014;33:793–797. doi: 10.1097/INF.0000000000000271. [DOI] [PubMed] [Google Scholar]
- 10.Nakra N.A., Blumberg D.A., Herrera-Guerra A., Lakshminrusimha S. Multi-system inflammatory syndrome in children (MIS-C) following SARS-CoV-2 infection: review of clinical presentation, hypothetical pathogenesis, and proposed management. Children (Basel) 2020;7:69. doi: 10.3390/children7070069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Consiglio C.R., Cotugno N., Sardh F., Pou C., Amodio D., Rodriguez L., et al. The immunology of multisystem inflammatory syndrome in children with COVID-19. Cell. 2020;183:968–981.e7. doi: 10.1016/j.cell.2020.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hennon T.R., Yu K.O.A., Penque M.D., Abdul-Aziz R., Chang A.C., McGreevy M.B., et al. COVID-19 associated multisystem inflammatory syndrome in children (MIS-C) guidelines; a Western New York approach as the pandemic evolves. Prog Pediatr Cardiol. 2021;62 doi: 10.1016/j.ppedcard.2021.101407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Henter J.I., Horne A., Aricó M., Egeler R.M., Filipovich A.H., Imashuku S., et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2007;48:124–131. doi: 10.1002/pbc.21039. [DOI] [PubMed] [Google Scholar]
- 14.Chandrakasan S., Filipovich A.H. Hemophagocytic lymphohistiocytosis: advances in pathophysiology, diagnosis, and treatment. J Pediatr. 2013;163:1253–1259. doi: 10.1016/j.jpeds.2013.06.053. [DOI] [PubMed] [Google Scholar]
- 15.Son M.B.F., Murray N., Friedman K., Young C.C., Newhams M.M., Feldstein L.R., et al. Multisystem inflammatory syndrome in children—initial therapy and outcomes. N Engl J Med. 2021;385:23–34. doi: 10.1056/NEJMoa2102605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ouldali N., Toubiana J., Antona D., Javouhey E., Madhi F., Lorrot M., et al. Association of intravenous immunoglobulins plus methylprednisolone vs immunoglobulins alone with course of fever in multisystem inflammatory syndrome in children. JAMA. 2021;325:855–864. doi: 10.1001/jama.2021.0694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Centers for Disease Control and Prevention 2020 Multisystem inflammatory syndrome in children (MIS-C) associated with coronavirus disease 2019 (COVID-19) https://emergency.cdc.gov/han/2020/han00432.asp Available at:
- 18.Khandelwal P., Chaturvedi V., Owsley E., Lane A., Heyenbruch D., Lutzko C.M., et al. CD38(bright)CD8(+) T cells associated with the development of acute GVHD are activated, proliferating, and cytotoxic trafficking cells. Biol Blood Marrow Transplant. 2020;26:1–6. doi: 10.1016/j.bbmt.2019.08.008. [DOI] [PubMed] [Google Scholar]
- 19.Khandelwal P., Lane A., Chaturvedi V., Owsley E., Davies S.M., Marmer D., et al. Peripheral blood CD38 bright CD8+ effector memory T cells predict acute graft-versus-host disease. Biol Blood Marrow Transplant. 2015;21:1215–1222. doi: 10.1016/j.bbmt.2015.04.010. [DOI] [PubMed] [Google Scholar]
- 20.Chaturvedi V., Marsh R.A., Lorenz A.Z., Owsley E., Chaturvedi V., Nguyen T., et al. T cell activation profiles distinguish hemophagocytic lymphohistiocytosis and early sepsis. Blood. 2021;137:2337–2346. doi: 10.1182/blood.2020009499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Anderson A.C., Joller N., Kuchroo V.K. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44:989–1004. doi: 10.1016/j.immuni.2016.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cura Daball P., Ventura Ferreira M.S., Ammann S., Klemann C., Lorenz M.R., Warthorst U., et al. CD57 identifies T cells with functional senescence before terminal differentiation and relative telomere shortening in patients with activated PI3 kinase delta syndrome. Immunol Cell Biol. 2018;96:1060–1071. doi: 10.1111/imcb.12169. [DOI] [PubMed] [Google Scholar]
- 23.Lopez-Vergès S., Milush J.M., Pandey S., York V.A., Arakawa-Hoyt J., Pircher H., et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010;116:3865–3874. doi: 10.1182/blood-2010-04-282301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Focosi D., Bestagno M., Burrone O., Petrini M. CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol. 2010;87:107–116. doi: 10.1189/jlb.0809566. [DOI] [PubMed] [Google Scholar]
- 25.Brenchley J.M., Karandikar N.J., Betts M.R., Ambrozak D.R., Hill B.J., Crotty L.E., et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood. 2003;101:2711–2720. doi: 10.1182/blood-2002-07-2103. [DOI] [PubMed] [Google Scholar]
- 26.Feldstein L.R., Tenforde M.W., Friedman K.G., Newhams M., Rose E.B., Dapul H., et al. Characteristics and outcomes of US children and adolescents with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19. JAMA. 2021;325:1074–1087. doi: 10.1001/jama.2021.2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Whittaker E., Bamford A., Kenny J., Kaforou M., Jones C.E., Shah P., et al. Clinical characteristics of 58 children with a pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2. JAMA. 2020;324:259–269. doi: 10.1001/jama.2020.10369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Diorio C., Henrickson S.E., Vella L.A., McNerney K.O., Chase J., Burudpakdee C., et al. Multisystem inflammatory syndrome in children and COVID-19 are distinct presentations of SARS-CoV-2. J Clin Invest. 2020;130:5967–5975. doi: 10.1172/JCI140970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kuri-Cervantes L., Pampena M.B., Meng W., Rosenfeld A.M., Ittner C.A.G., Weisman A.R., et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci Immunol. 2020;5 doi: 10.1126/sciimmunol.abd7114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mathew D., Giles J.R., Baxter A.E., Oldridge D.A., Greenplate A.R., Wu J.E., et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369 doi: 10.1126/science.abc8511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Vella L.A., Giles J.R., Baxter A.E., Oldridge D.A., Diorio C., Kuri-Cervantes L., et al. Deep immune profiling of MIS-C demonstrates marked but transient immune activation compared to adult and pediatric COVID-19. Sci Immunol. 2021;6 doi: 10.1126/sciimmunol.abf7570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lin H., Scull B.P., Goldberg B.R., Abhyankar H.A., Eckstein O.E., Zinn D.J., et al. IFN-γ signature in the plasma proteome distinguishes pediatric hemophagocytic lymphohistiocytosis from sepsis and SIRS. Blood Advances. 2021;5:3457–3467. doi: 10.1182/bloodadvances.2021004287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Qian L., Yu S., Yin C., Zhu B., Chen Z., Meng Z., et al. Plasma IFN-γ-inducible chemokines CXCL9 and CXCL10 correlate with survival and chemotherapeutic efficacy in advanced pancreatic ductal adenocarcinoma. Pancreatology. 2019;19:340–345. doi: 10.1016/j.pan.2019.01.015. [DOI] [PubMed] [Google Scholar]
- 34.Han J.H., Suh C.-H., Jung J.-Y., Ahn M.-H., Han M.H., Kwon J.E., et al. Elevated circulating levels of the interferon-γ–induced chemokines are associated with disease activity and cutaneous manifestations in adult-onset Still’s disease. Sci Rep. 2017;7:46652. doi: 10.1038/srep46652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yonker L.M., Gilboa T., Ogata A.F., Senussi Y., Lazarovits R., Boribong B.P., et al. Multisystem inflammatory syndrome in children is driven by zonulin-dependent loss of gut mucosal barrier. J Clin Invest. 2021;131 doi: 10.1172/JCI149633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Reiff D.D., Cron R.Q. Performance of cytokine storm syndrome scoring systems in pediatric COVID-19 and multisystem inflammatory syndrome in children. ACR Open Rheumatol. 2021;3:820–826. doi: 10.1002/acr2.11331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Song C.Y., Xu J., He J.Q., Lu Y.Q. Immune dysfunction following COVID-19, especially in severe patients. Sci Rep. 2020;10:15838. doi: 10.1038/s41598-020-72718-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Godfred-Cato S., Bryant B., Leung J., Oster M.E., Conklin L., Abrams J., et al. COVID-19-associated multisystem inflammatory syndrome in children—United States, March-July 2020. MMWR Morb Mortal Wkly Rep. 2020;69:1074–1080. doi: 10.15585/mmwr.mm6932e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Valverde I., Singh Y., Sanchez-de-Toledo J., Theocharis P., Chikermane A., Di Filippo S., et al. Acute cardiovascular manifestations in 286 children with multisystem inflammatory syndrome associated with COVID-19 infection in Europe. Circulation. 2021;143:21–32. doi: 10.1161/CIRCULATIONAHA.120.050065. [DOI] [PubMed] [Google Scholar]
- 40.Zhao Y., Patel J., Huang Y., Yin L., Tang L. Cardiac markers of multisystem inflammatory syndrome in children (MIS-C) in COVID-19 patients: a meta-analysis. Am J Emerg Med. 2021;49:62–70. doi: 10.1016/j.ajem.2021.05.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Miller J., Cantor A., Zachariah P., Ahn D., Martinez M., Margolis K.G. Gastrointestinal symptoms as a major presentation component of a novel multisystem inflammatory syndrome in children that is related to coronavirus disease 2019: a single center experience of 44 Cases. Gastroenterology. 2020;159:1571–1574.e2. doi: 10.1053/j.gastro.2020.05.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Morris S.B., Schwartz N.G., Patel P., Abbo L., Beauchamps L., Balan S., et al. 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. 2020;69:1450–1456. doi: 10.15585/mmwr.mm6940e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Weatherhead J.E., Clark E., Vogel T.P., Atmar R.L., Kulkarni P.A. Inflammatory syndromes associated with SARS-CoV-2 infection: dysregulation of the immune response across the age spectrum. J Clin Invest. 2020;130:6194–6197. doi: 10.1172/JCI145301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shaigany S., Gnirke M., Guttmann A., Chong H., Meehan S., Raabe V., et al. An adult with Kawasaki-like multisystem inflammatory syndrome associated with COVID-19. Lancet. 2020;396:e8–e10. doi: 10.1016/S0140-6736(20)31526-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gruber C.N., Patel R.S., Trachtman R., Lepow L., Amanat F., Krammer F., et al. Mapping systemic inflammation and antibody responses in multisystem inflammatory syndrome in children (MIS-C) Cell. 2020;183:982–995.e14. doi: 10.1016/j.cell.2020.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lee P.Y., Day-Lewis M., Henderson L.A., Friedman K.G., Lo J., Roberts J.E., et al. Distinct clinical and immunological features of SARS-CoV-2-induced multisystem inflammatory syndrome in children. J Clin Invest. 2020;130:5942–5950. doi: 10.1172/JCI141113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dove M.L., Jaggi P., Kelleman M., Abuali M., Ang J.Y., Ballan W., et al. Multisystem inflammatory syndrome in children: survey of protocols for early hospital evaluation and management. J Pediatr. 2021;229:33–40. doi: 10.1016/j.jpeds.2020.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Canna S.W., Marsh R.A. Pediatric hemophagocytic lymphohistiocytosis. Blood. 2020;135:1332–1343. doi: 10.1182/blood.2019000936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wherry E.J., Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–499. doi: 10.1038/nri3862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lee H.-G., Cho M.-Z., Choi J.-M. Bystander CD4+ T cells: crossroads between innate and adaptive immunity. Exp Mol Med. 2020;52:1255–1263. doi: 10.1038/s12276-020-00486-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kim T.-S., Shin E.-C. The activation of bystander CD8+ T cells and their roles in viral infection. Exp Mol Med. 2019;51:1–9. doi: 10.1038/s12276-019-0316-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kaushik S., Aydin S.I., Derespina K.R., Bansal P.B., Kowalsky S., Trachtman R., et al. Multisystem inflammatory syndrome in children associated with severe acute respiratory syndrome coronavirus 2 infection (MIS-C): a multi-institutional study from New York City. J Pediatr. 2020;224:24–29. doi: 10.1016/j.jpeds.2020.06.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Abdel-Haq N., Asmar B.I., Deza Leon M.P., McGrath E.J., Arora H.S., Cashen K., et al. SARS-CoV-2-associated multisystem inflammatory syndrome in children: clinical manifestations and the role of infliximab treatment. Eur J Pediatr. 2021;180:1581–1591. doi: 10.1007/s00431-021-03935-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dolinger M.T., Person H., Smith R., Jarchin L., Pittman N., Dubinsky M.C., et al. Pediatric Crohn disease and multisystem inflammatory syndrome in children (MIS-C) and COVID-19 treated with infliximab. J Pediatr Gastroenterol Nutr. 2020;71:153–155. doi: 10.1097/MPG.0000000000002809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Carter M.J., Fish M., Jennings A., Doores K.J., Wellman P., Seow J., et al. Peripheral immunophenotypes in children with multisystem inflammatory syndrome associated with SARS-CoV-2 infection. Nat Med. 2020;26:1701–1707. doi: 10.1038/s41591-020-1054-6. [DOI] [PubMed] [Google Scholar]
- 56.Dinarello C.A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281:8–27. doi: 10.1111/imr.12621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Weiss E.S., Girard-Guyonvarc’h C., Holzinger D., de Jesus A.A., Tariq Z., Picarsic J., et al. Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood. 2018;131:1442–1455. doi: 10.1182/blood-2017-12-820852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.de Jesus A.A., Hou Y., Brooks S., Malle L., Biancotto A., Huang Y., et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J Clin Invest. 2020;130:1669–1682. doi: 10.1172/JCI129301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Della Paolera S., Valencic E., Piscianz E., Moressa V., Tommasini A., Sagredini R., et al. Case report: use of anakinra in multisystem inflammatory syndrome during COVID-19 pandemic. Front Pediatr. 2021;8 doi: 10.3389/fped.2020.624248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Esteve-Sole A., Anton J., Pino-Ramirez R.M., Sanchez-Manubens J., Fumadó V., Fortuny C., et al. Similarities and differences between the immunopathogenesis of COVID-19-related pediatric multisystem inflammatory syndrome and Kawasaki disease. J Clin Invest. 2021;131 doi: 10.1172/JCI144554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Robinson P.C., Richards D., Tanner H.L., Feldmann M. Accumulating evidence suggests anti-TNF therapy needs to be given trial priority in COVID-19 treatment. Lancet Rheumatol. 2020;2:e653–e655. doi: 10.1016/S2665-9913(20)30309-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Feldmann M., Maini R.N., Woody J.N., Holgate S.T., Winter G., Rowland M., et al. Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed. Lancet. 2020;395:1407–1409. doi: 10.1016/S0140-6736(20)30858-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Castelnovo L., Tamburello A., Lurati A., Zaccara E., Marrazza M.G., Olivetti M., et al. Anti-IL6 treatment of serious COVID-19 disease: a monocentric retrospective experience. Medicine (Baltimore) 2021;100 doi: 10.1097/MD.0000000000023582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bronte V., Ugel S., Tinazzi E., Vella A., De Sanctis F., Canè S., et al. Baricitinib restrains the immune dysregulation in patients with severe COVID-19. J Clin Invest. 2020;130:6409–6416. doi: 10.1172/JCI141772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kalil A.C., Patterson T.F., Mehta A.K., Tomashek K.M., Wolfe C.R., Ghazaryan V., et al. Baricitinib plus remdesivir for hospitalized adults with Covid-19. N Engl J Med. 2021;384:795–807. doi: 10.1056/NEJMoa2031994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Patoulias D., Doumas M., Papadopoulos C., Karagiannis A. Janus kinase inhibitors and major COVID-19 outcomes: time to forget the two faces of Janus! A meta-analysis of randomized controlled trials. Clin Rheumatol. 2021;40:4671–4674. doi: 10.1007/s10067-021-05884-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Vannucchi A.M., Sordi B., Morettini A., Nozzoli C., Poggesi L., Pieralli F., et al. Compassionate use of JAK1/2 inhibitor ruxolitinib for severe COVID-19: a prospective observational study. Leukemia. 2021;35:1121–1133. doi: 10.1038/s41375-020-01018-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ahmed A., Merrill S.A., Alsawah F., Bockenstedt P., Campagnaro E., Devata S., et al. Ruxolitinib in adult patients with secondary haemophagocytic lymphohistiocytosis: an open-label, single-centre, pilot trial. Lancet Haematol. 2019;6:e630–e637. doi: 10.1016/S2352-3026(19)30156-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Locatelli F., Jordan M.B., Allen C., Cesaro S., Rizzari C., Rao A., et al. Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 2020;382:1811–1822. doi: 10.1056/NEJMoa1911326. [DOI] [PubMed] [Google Scholar]
- 70.Cho M.L., Jung Y.O., Moon Y.M., Min S.Y., Yoon C.H., Lee S.H., et al. Interleukin-18 induces the production of vascular endothelial growth factor (VEGF) in rheumatoid arthritis synovial fibroblasts via AP-1-dependent pathways. Immunol Lett. 2006;103:159–166. doi: 10.1016/j.imlet.2005.10.020. [DOI] [PubMed] [Google Scholar]
- 71.Kucukardali Y., Aydogdu S., Ozmen N., Yonem A., Solmazgul E., Ozyurt M., et al. The relationship between severity of coronary artery disease and plasma level of vascular endothelial growth factor. Cardiovasc Revasc Med. 2008;9:66–70. doi: 10.1016/j.carrev.2007.11.005. [DOI] [PubMed] [Google Scholar]
- 72.Ma L., Sahu S.K., Cano M., Kuppuswamy V., Bajwa J., McPhatter J., et al. Increased complement activation is a distinctive feature of severe SARS-CoV-2 infection. bioRxiv. 2021 doi: 10.1126/sciimmunol.abh2259. 2021.02.22.432177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Magro C., Mulvey J.J., Berlin D., Nuovo G., Salvatore S., Harp J., 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: 10.1016/j.trsl.2020.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ackermann M., Verleden S.E., Kuehnel M., Haverich A., Welte T., Laenger F., et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med. 2020;383:120–128. doi: 10.1056/NEJMoa2015432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Diorio C., McNerney K.O., Lambert M., Paessler M., Anderson E.M., Henrickson S.E., et al. Evidence of thrombotic microangiopathy in children with SARS-CoV-2 across the spectrum of clinical presentations. Blood Adv. 2020;4:6051–6063. doi: 10.1182/bloodadvances.2020003471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Diorio C., Shraim R., Vella L.A., Giles J.R., Baxter A.E., Oldridge D.A., et al. Proteomic profiling of MIS-C patients indicates heterogeneity relating to interferon gamma dysregulation and vascular endothelial dysfunction. Nat Commun. 2021;12:7222. doi: 10.1038/s41467-021-27544-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gloude N.J., Dandoy C.E., Davies S.M., Myers K.C., Jordan M.B., Marsh R.A., et al. Thinking beyond HLH: clinical features of patients with concurrent presentation of hemophagocytic lymphohistiocytosis and thrombotic microangiopathy. J Clin Immunol. 2020;40:699–707. doi: 10.1007/s10875-020-00789-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lee H., Lee Y., Lo A., Powell L., Shah B. Primarily VEGF-driven etiopathogenesis of Tafro syndrome and fibroblastic reticular cells as a probable Castleman cell—qualitative metasynthesis. Blood. 2020;136:27–28. [Google Scholar]
- 79.Sun P.P., Yu X.J., Wang S.X., Zhou X.J., Qu L., Zhang F., et al. Association of vascular endothelial growth factor and renal thrombotic microangiopathy-like lesions in patients with Castleman’s disease. Nephrology (Carlton) 2020;25:125–134. doi: 10.1111/nep.13630. [DOI] [PubMed] [Google Scholar]
- 80.Otani A., Takagi H., Oh H., Suzuma K., Matsumura M., Ikeda E., et al. Angiotensin II-stimulated vascular endothelial growth factor expression in bovine retinal pericytes. Invest Ophthalmol Vis Sci. 2000;41:1192–1199. [PubMed] [Google Scholar]
- 81.Funatsu H., Yamashita H., Nakanishi Y., Hori S. Angiotensin II and vascular endothelial growth factor in the vitreous fluid of patients with proliferative diabetic retinopathy. Br J Ophthalmol. 2002;86:311–315. doi: 10.1136/bjo.86.3.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Selheim F., Fukami M.H., Holmsen H., Vassbotn F.S. Platelet-derived-growth-factor-induced signalling in human platelets: phosphoinositide-3-kinase-dependent inhibition of platelet activation. Biochem J. 2000;350:469–475. [PMC free article] [PubMed] [Google Scholar]
- 83.Canzano P., Brambilla M., Porro B., Cosentino N., Tortorici E., Vicini S., et al. Platelet and endothelial activation as potential mechanisms behind the thrombotic complications of COVID-19 patients. JACC Basic Transl Sci. 2021;6:202–218. doi: 10.1016/j.jacbts.2020.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
References
- Maecker H.T., McCoy J.P., Nussenblatt R. Standardizing immunophenotyping for the Human Immunology Project. Nat Rev Immunol. 2012;12:191–200. doi: 10.1038/nri3158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khandelwal P., Chaturvedi V., Owsley E., Lane A., Heyenbruch D., Lutzko C.M., et al. CD38(bright)CD8(+) T cells associated with the development of acute GVHD are activated, proliferating, and cytotoxic trafficking cells. Biol Blood Marrow Transplant. 2020;26:1–6. doi: 10.1016/j.bbmt.2019.08.008. [DOI] [PubMed] [Google Scholar]
- Khandelwal P., Lane A., Chaturvedi V., Owsley E., Davies S.M., Marmer D., et al. Peripheral blood CD38 bright CD8+ effector memory T cells predict acute graft-versus-host disease. Biol Blood Marrow Transplant. 2015;21:1215–1222. doi: 10.1016/j.bbmt.2015.04.010. [DOI] [PubMed] [Google Scholar]
- Chaturvedi V., Marsh R.A., Lorenz A.Z., Owsley E., Chaturvedi V., Nguyen T., et al. T cell activation profiles distinguish hemophagocytic lymphohistiocytosis and early sepsis. Blood. 2021;137:2337–2346. doi: 10.1182/blood.2020009499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Focosi D., Bestagno M., Burrone O., Petrini M. CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol. 2010;87:107–116. doi: 10.1189/jlb.0809566. [DOI] [PubMed] [Google Scholar]
- Cura Daball P., Ventura Ferreira M.S., Ammann S., Klemann C., Lorenz M.R., Warthorst U., et al. CD57 identifies T cells with functional senescence before terminal differentiation and relative telomere shortening in patients with activated PI3 kinase delta syndrome. Immunol Cell Biol. 2018;96:1060–1071. doi: 10.1111/imcb.12169. [DOI] [PubMed] [Google Scholar]
- Lopez-Vergès S., Milush J.M., Pandey S., York V.A., Arakawa-Hoyt J., Pircher H., et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010;116:3865–3874. doi: 10.1182/blood-2010-04-282301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenchley J.M., Karandikar N.J., Betts M.R., Ambrozak D.R., Hill B.J., Crotty L.E., et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood. 2003;101:2711–2720. doi: 10.1182/blood-2002-07-2103. [DOI] [PubMed] [Google Scholar]
- Anderson A.C., Joller N., Kuchroo V.K. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44:989–1004. doi: 10.1016/j.immuni.2016.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]