SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation. / Jarlhelt, Ida; Nielsen, Sif Kaas; Jahn, Camilla Xenia Holtermann; Hansen, Cecilie Bo; Pérez-Alós, Laura; Rosbjerg, Anne; Bayarri-Olmos, Rafael; Skjoedt, Mikkel-Ole; Garred, Peter.

In: Frontiers in Immunology, Vol. 12, 767981, 2021.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Jarlhelt, I, Nielsen, SK, Jahn, CXH, Hansen, CB, Pérez-Alós, L, Rosbjerg, A, Bayarri-Olmos, R, Skjoedt, M-O & Garred, P 2021, 'SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation', Frontiers in Immunology, vol. 12, 767981. https://doi.org/10.3389/fimmu.2021.767981

APA

Jarlhelt, I., Nielsen, S. K., Jahn, C. X. H., Hansen, C. B., Pérez-Alós, L., Rosbjerg, A., Bayarri-Olmos, R., Skjoedt, M-O., & Garred, P. (2021). SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation. Frontiers in Immunology, 12, [767981]. https://doi.org/10.3389/fimmu.2021.767981

Vancouver

Jarlhelt I, Nielsen SK, Jahn CXH, Hansen CB, Pérez-Alós L, Rosbjerg A et al. SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation. Frontiers in Immunology. 2021;12. 767981. https://doi.org/10.3389/fimmu.2021.767981

Author

Jarlhelt, Ida ; Nielsen, Sif Kaas ; Jahn, Camilla Xenia Holtermann ; Hansen, Cecilie Bo ; Pérez-Alós, Laura ; Rosbjerg, Anne ; Bayarri-Olmos, Rafael ; Skjoedt, Mikkel-Ole ; Garred, Peter. / SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation. In: Frontiers in Immunology. 2021 ; Vol. 12.

Bibtex

@article{96c20bb13b2e4f3687cb48515f6c4ba6,
title = "SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation",
abstract = "The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to constitute a serious public health threat worldwide. Protective antibody-mediated viral neutralization in response to SARS-CoV-2 infection has been firmly characterized. Where the effects of the antibody response are generally considered to be beneficial, an important biological question regarding potential negative outcomes of a SARS-CoV-2 antibody response has yet to be answered. We determined the distribution of IgG subclasses and complement activation levels in plasma from convalescent individuals using in-house developed ELISAs. The IgG response towards SARS-CoV-2 receptor-binding domain (RBD) after natural infection appeared to be mainly driven by IgG1 and IgG3 subclasses, which are the main ligands for C1q mediated classical complement pathway activation. The deposition of the complement components C4b, C3bc, and TCC as a consequence of SARS-CoV-2 specific antibodies were depending primarily on the SARS-CoV-2 RBD and significantly correlated with both IgG levels and disease severity, indicating that individuals with high levels of IgG and/or severe disease, might have a more prominent complement activation during viral infection. Finally, freshly isolated monocytes and a monocyte cell line (THP-1) were used to address the cellular mediated inflammatory response as a consequence of Fc-gamma receptor engagement by SARS-CoV-2 specific antibodies. Monocytic Fc gamma receptor charging resulted in a significant rise in the secretion of the pro-inflammatory cytokine TNF-α. Our results indicate that SARS-CoV-2 antibodies might drive significant inflammatory responses through the classical complement pathway and via cellular immune-complex activation that could have negative consequences during COVID-19 disease. We found that increased classical complement activation was highly associated to COVID-19 disease severity. The combination of antibody-mediated complement activation and subsequent cellular priming could constitute a significant risk of exacerbating COVID-19 severity.",
keywords = "antibody response, complement, monocytes, receptor binding domain, SARS-CoV-2, spike",
author = "Ida Jarlhelt and Nielsen, {Sif Kaas} and Jahn, {Camilla Xenia Holtermann} and Hansen, {Cecilie Bo} and Laura P{\'e}rez-Al{\'o}s and Anne Rosbjerg and Rafael Bayarri-Olmos and Mikkel-Ole Skjoedt and Peter Garred",
note = "Publisher Copyright: {\textcopyright} Copyright {\textcopyright} 2021 Jarlhelt, Nielsen, Jahn, Hansen, P{\'e}rez-Al{\'o}s, Rosbjerg, Bayarri-Olmos, Skjoedt and Garred.",
year = "2021",
doi = "10.3389/fimmu.2021.767981",
language = "English",
volume = "12",
journal = "Frontiers in Immunology",
issn = "1664-3224",
publisher = "Frontiers Research Foundation",

}

RIS

TY - JOUR

T1 - SARS-CoV-2 Antibodies Mediate Complement and Cellular Driven Inflammation

AU - Jarlhelt, Ida

AU - Nielsen, Sif Kaas

AU - Jahn, Camilla Xenia Holtermann

AU - Hansen, Cecilie Bo

AU - Pérez-Alós, Laura

AU - Rosbjerg, Anne

AU - Bayarri-Olmos, Rafael

AU - Skjoedt, Mikkel-Ole

AU - Garred, Peter

N1 - Publisher Copyright: © Copyright © 2021 Jarlhelt, Nielsen, Jahn, Hansen, Pérez-Alós, Rosbjerg, Bayarri-Olmos, Skjoedt and Garred.

PY - 2021

Y1 - 2021

N2 - The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to constitute a serious public health threat worldwide. Protective antibody-mediated viral neutralization in response to SARS-CoV-2 infection has been firmly characterized. Where the effects of the antibody response are generally considered to be beneficial, an important biological question regarding potential negative outcomes of a SARS-CoV-2 antibody response has yet to be answered. We determined the distribution of IgG subclasses and complement activation levels in plasma from convalescent individuals using in-house developed ELISAs. The IgG response towards SARS-CoV-2 receptor-binding domain (RBD) after natural infection appeared to be mainly driven by IgG1 and IgG3 subclasses, which are the main ligands for C1q mediated classical complement pathway activation. The deposition of the complement components C4b, C3bc, and TCC as a consequence of SARS-CoV-2 specific antibodies were depending primarily on the SARS-CoV-2 RBD and significantly correlated with both IgG levels and disease severity, indicating that individuals with high levels of IgG and/or severe disease, might have a more prominent complement activation during viral infection. Finally, freshly isolated monocytes and a monocyte cell line (THP-1) were used to address the cellular mediated inflammatory response as a consequence of Fc-gamma receptor engagement by SARS-CoV-2 specific antibodies. Monocytic Fc gamma receptor charging resulted in a significant rise in the secretion of the pro-inflammatory cytokine TNF-α. Our results indicate that SARS-CoV-2 antibodies might drive significant inflammatory responses through the classical complement pathway and via cellular immune-complex activation that could have negative consequences during COVID-19 disease. We found that increased classical complement activation was highly associated to COVID-19 disease severity. The combination of antibody-mediated complement activation and subsequent cellular priming could constitute a significant risk of exacerbating COVID-19 severity.

AB - The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to constitute a serious public health threat worldwide. Protective antibody-mediated viral neutralization in response to SARS-CoV-2 infection has been firmly characterized. Where the effects of the antibody response are generally considered to be beneficial, an important biological question regarding potential negative outcomes of a SARS-CoV-2 antibody response has yet to be answered. We determined the distribution of IgG subclasses and complement activation levels in plasma from convalescent individuals using in-house developed ELISAs. The IgG response towards SARS-CoV-2 receptor-binding domain (RBD) after natural infection appeared to be mainly driven by IgG1 and IgG3 subclasses, which are the main ligands for C1q mediated classical complement pathway activation. The deposition of the complement components C4b, C3bc, and TCC as a consequence of SARS-CoV-2 specific antibodies were depending primarily on the SARS-CoV-2 RBD and significantly correlated with both IgG levels and disease severity, indicating that individuals with high levels of IgG and/or severe disease, might have a more prominent complement activation during viral infection. Finally, freshly isolated monocytes and a monocyte cell line (THP-1) were used to address the cellular mediated inflammatory response as a consequence of Fc-gamma receptor engagement by SARS-CoV-2 specific antibodies. Monocytic Fc gamma receptor charging resulted in a significant rise in the secretion of the pro-inflammatory cytokine TNF-α. Our results indicate that SARS-CoV-2 antibodies might drive significant inflammatory responses through the classical complement pathway and via cellular immune-complex activation that could have negative consequences during COVID-19 disease. We found that increased classical complement activation was highly associated to COVID-19 disease severity. The combination of antibody-mediated complement activation and subsequent cellular priming could constitute a significant risk of exacerbating COVID-19 severity.

KW - antibody response

KW - complement

KW - monocytes

KW - receptor binding domain

KW - SARS-CoV-2

KW - spike

U2 - 10.3389/fimmu.2021.767981

DO - 10.3389/fimmu.2021.767981

M3 - Journal article

C2 - 34804055

AN - SCOPUS:85119276867

VL - 12

JO - Frontiers in Immunology

JF - Frontiers in Immunology

SN - 1664-3224

M1 - 767981

ER -

ID: 302066574