Distinguishing immune activation and inflammatory signatures of multisystem inflammatory syndrome in children (MIS-C) versus hemophagocytic lymphohistiocytosis (HLH)

Deepak Kumar, Christina A Rostad, Preeti Jaggi, D Sofia Villacis Nunez, Chengyu Prince, Austin Lu, Laila Hussaini, Thinh H Nguyen, Sakshi Malik, Lori A Ponder, Sreekala P V Shenoy, Evan J Anderson, Michael Briones, Ignacio Sanz, Sampath Prahalad, Shanmuganathan Chandrakasan, Deepak Kumar, Christina A Rostad, Preeti Jaggi, D Sofia Villacis Nunez, Chengyu Prince, Austin Lu, Laila Hussaini, Thinh H Nguyen, Sakshi Malik, Lori A Ponder, Sreekala P V Shenoy, Evan J Anderson, Michael Briones, Ignacio Sanz, Sampath Prahalad, Shanmuganathan Chandrakasan

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.

Keywords: COVID-19; HLH; MIS-C; T-cell activation; cardiac dysfunction; hyperinflammation.

Copyright © 2022 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig 1
Fig 1
MIS-C and HLH display both unique and shared inflammatory signature. A, Heat map showing expression of cytokines and chemokines in HCs (n = 19), COVID-19 (n = 10), HLH (n = 8), MIS-C (n = 19), and MIS-C follow-up (n = 10) samples. B, Multidimensional cytokine/chemokine data were represented as 2-dimensional PCA space showing clusters for HCs, COVID-19, MIS-C, HLH, and MIS-C follow-up samples. Individuals are shown by small-size colored circles, whereas overall group is shown by large-size colored circles. PC, Principal component; PCA, principal-component analysis.
Fig 2
Fig 2
A-G, Cytokine families showing differences in patients with MIS-C and patients with HLH. Plasma levels of important cytokines belonging to multiple cytokine families were represented by dot plots in HCs (n = 19), COVID-19 (n = 10), HLH (n = 8), MIS-C (n = 19), and MIS-C follow-up (n = 10) samples. Conc., Concentration; ns, not significant. Kruskal-Wallis 1-way ANOVA followed by Dunn’s multiple comparison test for nonnormally distributed samples and ordinary 1-way ANOVA followed by Tukey’s multiple comparison test for normally distributed samples were used for statistical comparison. ∗P < .05, ∗∗P < .01, ∗∗∗P < .001; ∗∗∗∗P < .0001.
Fig 3
Fig 3
MIS-C and HLH are marked by increase in activation of CD8+ and CD4+ EM T cells. A and B, Representative FACS plots showing surface expression of HLA-DR+ CD38+ markers on the EM compartment of CD8+ and CD4+ T cells in HCs and COVID-19, HLH, MIS-C, and MIS-C follow-up patients. C-F, Percentage of HLA-DR+ CD38+ and HLA-DR+PD-1+ expression in HCs (n = 22) and COVID-19 (n = 24), HLH (n = 6), MIS-C (n = 69), and MIS-C follow-up (n = 31) patients in CD8+ and CD4+ EM compartments. FACS, Fluorescence-activated cell sorting; ns, not significant. Data represent median with interquartile range values for each group. Kruskal-Wallis 1-way ANOVA followed by Dunn’s multiple comparison test for nonnormally distributed samples and ordinary 1-way ANOVA followed by Tukey’s multiple comparison test for normally distributed samples were used for statistical comparison. ∗∗∗P < .001; ∗∗∗∗P < .0001.
Fig 4
Fig 4
Comparison of different laboratory parameters in patients with COVID-19, HLH, and MIS-C. Dot plots showing the plasma levels of ferritin (A), sIL-2R (B), and sCD163 (C) in different patient cohorts. Dotted lines represent ferritin cutoff levels of 500 ng/mL and sIL-2R cutoff levels of 2400 U/mL. D, Plots showing NLR in different patient cohorts. E and F, Plots showing ratio of ANC with CD8+ and CD4+ EM T-cell activation. ALC, Absolute lymphocyte count; ANC, absolute neutrophil count; Conc., concentration; NLR, neutrophil to lymphocyte ratio.
Fig 5
Fig 5
Cardiac dysfunction markers correlate with T-cell activation in patients with MIS-C and COVID-19. A-D, Scatter plots showing correlation of serum BNP and troponin levels with CD8+ and CD4+ EM T-cell activation. E-H, Scatter plots showing correlation of serum BNP and troponin levels with ferritin and CRP. Spearman correlation coefficient and P values are indicated.
Fig 6
Fig 6
Correlation of laboratory features and immune activation markers in MIS-C and COVID-19. A and B, Plots showing inverse correlation between platelets and T-cell activation. Spearman’s correlation coefficient and P values are shown. C, Correlation matrix showing positive and inverse correlations between different clinical parameters in patients with COVID-19 and patients with MIS-C. ALC, Absolute lymphocyte count; ALT, alanine transaminase; ANC, absolute neutrophil count; WBC, white blood cell. Positive correlation is shown as blue-colored circles, whereas inverse correlation is shown as red-colored circles. Size and intensity of colored circles show the strength of correlation. Only significant correlations with P less than .05 are shown as colored circles.
Fig E1
Fig E1
Distribution of follow-up blood sampling of patients with MIS-C. “0” represents the first blood sample drawn, and circles represent when follow-up samples were obtained since initial diagnosis.
Fig E2
Fig E2
Treatment and blood sampling timeline of patients with MIS-C. Timeline for patients with MIS-C indicating blood sampling with respect to start of steroid treatment (vertical dotted line). IVIG, Intravenous immunoglobulin. Each row represents an individual patient with MIS-C. “0” represents blood sampling within first 24 hours of initiation of steroid treatment. “1” represents blood sampling within 24 to 48 hours of steroid initiation and so on. ∗ represents patients with MIS-C who did not receive any steroids during hospital stay. Φ represents patients in whom blood sampling was done before IVIG. Δ represents patients who did not receive IVIG. Rest all patients received IVIG before blood sampling.
Fig E3
Fig E3
Comparison of serum levels of selected cytokines in different patient groups. Dot plots showing serum concentrations of selected cytokines/chemokines in HCs and patients with COVID-19, patients with HLH, and patients with MIS-C. Conc., Concentration; ns, not significant. Kruskal-Wallis 1-way ANOVA followed by Dunn’s multiple comparison test for nonnormally distributed samples and ordinary 1-way ANOVA followed by Tukey’s multiple comparison test for normally distributed samples were used for statistical comparison. ∗P < .05, ∗∗P < .01, ∗∗∗P < .001; ∗∗∗∗P < .0001.
Fig E3
Fig E3
Comparison of serum levels of selected cytokines in different patient groups. Dot plots showing serum concentrations of selected cytokines/chemokines in HCs and patients with COVID-19, patients with HLH, and patients with MIS-C. Conc., Concentration; ns, not significant. Kruskal-Wallis 1-way ANOVA followed by Dunn’s multiple comparison test for nonnormally distributed samples and ordinary 1-way ANOVA followed by Tukey’s multiple comparison test for normally distributed samples were used for statistical comparison. ∗P < .05, ∗∗P < .01, ∗∗∗P < .001; ∗∗∗∗P < .0001.
Fig E4
Fig E4
Gating strategy used to define CD4+ and CD8+ T-cell activation in different patient cohorts. FSC-A, Forward scatter-area; FSC-H, forward scatter-height; SSC-A, side scatter-area.
Fig E5
Fig E5
Evaluation of differences in CD8+ EM T-cell activation in patient cohorts. ROC curves showing optimal threshold value with corresponding percentage sensitivity and specificity for frequency of HLA-DR+CD38+ on EM CD8+ T cells between HCs vs MIS-C (A), MIS-C vs COVID-19 (B), and HLH vs MIS-C (C). AUC, Area under the ROC curve.
Fig E6
Fig E6
Comparison of T-cell activation in different subsets of CD8+ and CD4+ T-cell populations. Dot plots showing HLA-DR+ CD38+ coexpression in CM (A and B) and TEMRA (C and D) subsets of CD8+ and CD4+ T cells and also on total CD8+ and CD4+ T-cell populations (E and F). CM, Central memory; ns, nonsignificant.
Fig E7
Fig E7
Quantitation of T-cell perturbations among different patient cohorts. (A) Plots showing ratio of CD8+ EM vs naive compartment and (B) CD4+ vs CD8+ ratio in different patient cohorts. Dot plots showing frequencies of CD8+ and CD4+ TEMRA populations (C and D). Plots showing percentage coexpression of PD-1+ and Tim3+ (E and F) and expression of CD57+ in the EM compartment of CD8+ and CD4+ T cells (G and H) in HCs (n = 22) and COVID-19 (n = 24), HLH (n = 6), MIS-C (n = 69), and MIS-C follow-up (n = 31) patients. ns, Nonsignificant.
Fig E8
Fig E8
Follow-up analysis of patients with MIS-C displays decrease in activation, exhaustion, and senescence markers on T cells along with improvement in clinical markers of inflammation. A-E, Dot plots showing paired analysis of different states of T cells and its subsets in patients with MIS-C at onset and follow-up (n = 18). F, Paired analysis of patients with MIS-C showing levels of CRP and ferritin at patient admission and 7 days postadmission. ns, Nonsignificant.
FIG E9
FIG E9
Quantitation of BNP and troponin levels in MIS-C and COVID-19. Plots showing serum levels of BNP (A) and troponin (B) in patients with COVID-19 (n = 15) and patients with MIS-C (n = 69). Based on % optimal threshold value of CD8+ T activation, patients with MIS-C and patients with COVID-19 were categorized into 2 groups having low (<15.9%) and high CD8+ (>15.9%) EM T-cell activation. Dot plots showing differences between BNP (C) and troponin (D) levels in groups having low and high CD8+ T-cell activation. Act., Activation.
Fig E10
Fig E10
Correlation of laboratory features and immune markers in MIS-C and COVID-19. Correlation matrix showing positive and inverse correlations between different laboratory and immune parameters in patients with COVID-19 (n = 13) and patients with MIS-C (n = 40). Positive correlation is shown as blue-colored circles, whereas inverse correlation is shown in red-colored circles. Size and intensity of colored circles show the strength of correlation. Only significant correlations with P less than .05 are shown as colored circles. ALC, Absolute lymphocyte count; ALT, alanine transaminase; ANC, absolute neutrophil count; WBC, white blood cell.

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.
    1. Brodin P. Why is COVID-19 so mild in children? Acta Paediatr. 2020;109:1082–1083.
    1. 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
    1. Riphagen S., Gomez X., Gonzalez-Martinez C., Wilkinson N., Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–1608.
    1. 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.
    1. 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.
    1. 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
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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
    1. 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.
    1. Chandrakasan S., Filipovich A.H. Hemophagocytic lymphohistiocytosis: advances in pathophysiology, diagnosis, and treatment. J Pediatr. 2013;163:1253–1259.
    1. 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.
    1. 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.
    1. Centers for Disease Control and Prevention 2020 Multisystem inflammatory syndrome in children (MIS-C) associated with coronavirus disease 2019 (COVID-19) Available at:
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Focosi D., Bestagno M., Burrone O., Petrini M. CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol. 2010;87:107–116.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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
    1. 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
    1. 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
    1. 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.
    1. 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.
    1. 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.
    1. 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
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Canna S.W., Marsh R.A. Pediatric hemophagocytic lymphohistiocytosis. Blood. 2020;135:1332–1343.
    1. Wherry E.J., Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–499.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Dinarello C.A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281:8–27.
    1. 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.
    1. 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.
    1. 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
    1. 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
    1. 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.
    1. 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.
    1. 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
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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 2021.02.22.432177.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Maecker H.T., McCoy J.P., Nussenblatt R. Standardizing immunophenotyping for the Human Immunology Project. Nat Rev Immunol. 2012;12:191–200.
    1. 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.
    1. 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.
    1. 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.
    1. Focosi D., Bestagno M., Burrone O., Petrini M. CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol. 2010;87:107–116.
    1. 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.
    1. 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.
    1. 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.
    1. 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.

Source: PubMed

3
Abonnieren