Skip to main content
Top
Published in: Virology Journal 1/2023

Open Access 01-12-2023 | SARS-CoV-2 | Research

The detectable anti-interferon-γ autoantibodies in COVID-19 patients may be associated with disease severity

Authors: Po-Ku Chen, Kai-Jieh Yeo, Shih-Hsin Chang, Tsai-Ling Liao, Chia-Hui Chou, Joung-Liang Lan, Ching-Kun Chang, Der-Yuan Chen

Published in: Virology Journal | Issue 1/2023

Login to get access

Abstract

Background

Neutralizing anti-interferon (IFN)-γ autoantibodies are linked to adult-onset immunodeficiency and opportunistic infections.

Methods

To explore whether anti-IFN-γ autoantibodies are associated with disease severity of coronavirus disease 2019 (COVID-19), we examined the titers and functional neutralization of anti-IFN-γ autoantibodies in COVID-19 patients. In 127 COVID-19 patients and 22 healthy controls, serum titers of anti-IFN-γ autoantibodies were quantified using enzyme-linked immunosorbent assay, and the presence of autoantibodies was verified with immunoblotting assay. The neutralizing capacity against IFN-γ was evaluated with flow cytometry analysis and immunoblotting, and serum cytokines levels were determined using the MULTIPLEX platform.

Results

A higher proportion of severe/critical COVID-19 patients had positivity for anti-IFN-γ autoantibodies (18.0%) compared with non-severe patients (3.4%, p < 0.01) or healthy control (HC) (0.0%, p < 0.05). Severe/critical COVID-19 patients also had higher median titers of anti-IFN-γ autoantibodies (5.01) compared with non-severe patients (1.33) or HC (0.44). The immunoblotting assay could verify the detectable anti-IFN-γ autoantibodies and revealed more effective inhibition of signal transducer and activator of transcription (STAT1) phosphorylation on THP-1 cells treated with serum samples from anti-IFN-γ autoantibodies-positive patients compared with those from HC (2.21 ± 0.33 versus 4.47 ± 1.64, p < 0.05). In flow-cytometry analysis, sera from autoantibodies-positive patients could also significantly more effectively suppress the STAT1 phosphorylation (median,67.28%, interquartile range [IQR] 55.2–78.0%) compared with serum from HC (median,106.7%, IQR 100.0–117.8%, p < 0.05) or autoantibodies-negative patients (median,105.9%, IQR 85.5–116.3%, p < 0.05). Multivariate analysis revealed that the positivity and titers of anti-IFN-γ autoantibodies were significant predictors of severe/critical COVID-19. Compared with non-severe COVID-19 patients, we reveal that a significantly higher proportion of severe/critical COVID-19 patients are positive for anti-IFN-γ autoantibodies with neutralizing capacity.

Conclusion

Our results would add COVID-19 to the list of diseases with the presence of neutralizing anti-IFN-γ autoAbs. Anti-IFN-γ autoantibodies positivity is a potential predictor of severe/critical COVID-19.
Appendix
Available only for authorised users
Literature
1.
go back to reference Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 2007;96:41–101.PubMedCrossRef Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 2007;96:41–101.PubMedCrossRef
2.
go back to reference Schroder K, Hertzog PJ, Ravasi T, et al. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–89.PubMedCrossRef Schroder K, Hertzog PJ, Ravasi T, et al. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–89.PubMedCrossRef
3.
go back to reference Patel SY, Ding L, Brown MR, et al. Anti-IFN-gamma autoantibodies in disseminated nontuberculous mycobacterial infections. J Immunol. 2005;175:4769–76.PubMedCrossRef Patel SY, Ding L, Brown MR, et al. Anti-IFN-gamma autoantibodies in disseminated nontuberculous mycobacterial infections. J Immunol. 2005;175:4769–76.PubMedCrossRef
4.
go back to reference Wipasa J, Chaiwarith R, Chawansuntati K, et al. Characterization of anti-interferon-γ antibodies in HIV-negative immunodeficient patients infected with unusual intracellular microorganisms. Exp Biol Med (Maywood). 2018;243:621–6.PubMedCrossRef Wipasa J, Chaiwarith R, Chawansuntati K, et al. Characterization of anti-interferon-γ antibodies in HIV-negative immunodeficient patients infected with unusual intracellular microorganisms. Exp Biol Med (Maywood). 2018;243:621–6.PubMedCrossRef
5.
go back to reference Haverkamp MH, van Dissel JT, Holland SM. Human host genetic factors in nontuberculous mycobacterial infection: lessons from single gene disorders affecting innate and adaptive immunity and lessons from molecular defects in interferon-gamma-dependent signaling. Microbes Infect. 2006;8:1157–66.PubMedCrossRef Haverkamp MH, van Dissel JT, Holland SM. Human host genetic factors in nontuberculous mycobacterial infection: lessons from single gene disorders affecting innate and adaptive immunity and lessons from molecular defects in interferon-gamma-dependent signaling. Microbes Infect. 2006;8:1157–66.PubMedCrossRef
7.
go back to reference Chi CY, Lin CH, Ho MW, et al. Clinical manifestations, course, and outcome of patients with neutralizing anti-interferon-γ autoantibodies and disseminated nontuberculous mycobacterial infections. Medicine (Baltimore). 2016;95:e3927.PubMedCrossRef Chi CY, Lin CH, Ho MW, et al. Clinical manifestations, course, and outcome of patients with neutralizing anti-interferon-γ autoantibodies and disseminated nontuberculous mycobacterial infections. Medicine (Baltimore). 2016;95:e3927.PubMedCrossRef
8.
go back to reference Hase I, Morimoto K, Sakagami T, et al. Patient ethnicity and causative species determine the manifestations of anti-interferon-gamma autoantibody-associated nontuberculous mycobacterial disease: a review. Diagn Microbiol Infect Dis. 2017;88:308–15.PubMedCrossRef Hase I, Morimoto K, Sakagami T, et al. Patient ethnicity and causative species determine the manifestations of anti-interferon-gamma autoantibody-associated nontuberculous mycobacterial disease: a review. Diagn Microbiol Infect Dis. 2017;88:308–15.PubMedCrossRef
9.
go back to reference Hanitsch LG, Lobel M, Muller-Redetzky H, et al. Late-onset disseminated Mycobacterium avium intracellulare complex infection (MAC), cerebral toxoplasmosis and salmonella sepsis in a German Caucasian patient with unusual anti-interferon-gamma IgG1 autoantibodies. J Clin Immunol. 2015;35:361–5.PubMedCrossRef Hanitsch LG, Lobel M, Muller-Redetzky H, et al. Late-onset disseminated Mycobacterium avium intracellulare complex infection (MAC), cerebral toxoplasmosis and salmonella sepsis in a German Caucasian patient with unusual anti-interferon-gamma IgG1 autoantibodies. J Clin Immunol. 2015;35:361–5.PubMedCrossRef
10.
go back to reference Hong GH, Ortega-Villa AM, Hunsberger S, et al. Natural history and evolution of anti-interferon-γ autoantibody-associated immunodeficiency syndrome in Thailand and the United States. Clin Infect Dis. 2020;71:53–62.PubMedCrossRef Hong GH, Ortega-Villa AM, Hunsberger S, et al. Natural history and evolution of anti-interferon-γ autoantibody-associated immunodeficiency syndrome in Thailand and the United States. Clin Infect Dis. 2020;71:53–62.PubMedCrossRef
11.
12.
go back to reference Mahajan S, Caraballo C, Li SX, et al. SARS-CoV-2 infection hospitalization rate and infection fatality rate among the non-congregate population in Connecticut. Am J Med. 2021;134:812-16.e2.PubMedPubMedCentralCrossRef Mahajan S, Caraballo C, Li SX, et al. SARS-CoV-2 infection hospitalization rate and infection fatality rate among the non-congregate population in Connecticut. Am J Med. 2021;134:812-16.e2.PubMedPubMedCentralCrossRef
13.
14.
go back to reference Tan KT, Hsu BC, Chen DY. Autoimmune and rheumatic manifestations associated with COVID-19 in adults: an updated systematic review. Front Immunol. 2021;12:5013. Tan KT, Hsu BC, Chen DY. Autoimmune and rheumatic manifestations associated with COVID-19 in adults: an updated systematic review. Front Immunol. 2021;12:5013.
15.
go back to reference Dotan A, Muller S, Kanduc D, et al. The SARS-CoV-2 as an instrumental trigger of autoimmunity. Autoimmunity Rev. 2021;20:102792.CrossRef Dotan A, Muller S, Kanduc D, et al. The SARS-CoV-2 as an instrumental trigger of autoimmunity. Autoimmunity Rev. 2021;20:102792.CrossRef
16.
go back to reference Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370:6515.CrossRef Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370:6515.CrossRef
18.
go back to reference Guan WJ, Ni ZY, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–20.PubMedCrossRef Guan WJ, Ni ZY, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–20.PubMedCrossRef
20.
go back to reference Krisnawati DI, Liu YC, Lee YJ, et al. Functional neutralization of anti-IFN-gamma autoantibody in patients with non-tuberculous mycobacteria infection. Sci Rep. 2019;9:5682.PubMedPubMedCentralCrossRef Krisnawati DI, Liu YC, Lee YJ, et al. Functional neutralization of anti-IFN-gamma autoantibody in patients with non-tuberculous mycobacteria infection. Sci Rep. 2019;9:5682.PubMedPubMedCentralCrossRef
21.
go back to reference Nithichanon A, Chetchotisakd P, Matsumura T, et al. Diagnosis of NTM active infection in lymphadenopathy patients with anti-interferon-gamma autoantibody using inhibitory ELISA vs. indirect ELISA. Sci Rep. 2020;10:8968.PubMedPubMedCentralCrossRef Nithichanon A, Chetchotisakd P, Matsumura T, et al. Diagnosis of NTM active infection in lymphadenopathy patients with anti-interferon-gamma autoantibody using inhibitory ELISA vs. indirect ELISA. Sci Rep. 2020;10:8968.PubMedPubMedCentralCrossRef
22.
go back to reference Lin CH, Lewinski MK, Pache L, et al. Identification of a major epitope by anti-interferon-gamma autoantibodies in patients with mycobacterial disease. Nat Med. 2016;22:994–1001.PubMedCrossRef Lin CH, Lewinski MK, Pache L, et al. Identification of a major epitope by anti-interferon-gamma autoantibodies in patients with mycobacterial disease. Nat Med. 2016;22:994–1001.PubMedCrossRef
23.
go back to reference Bhaskar S, Sinha A, Banach M, Mittoo S, Weissert R, Kass JS, et al. Cytokine storm in COVID-19-immunopathological mechanisms, clinical considerations, and therapeutic approaches: The REPROGRAM Consortium Position Paper. Front Immunol. 2020;11:1648.PubMedPubMedCentralCrossRef Bhaskar S, Sinha A, Banach M, Mittoo S, Weissert R, Kass JS, et al. Cytokine storm in COVID-19-immunopathological mechanisms, clinical considerations, and therapeutic approaches: The REPROGRAM Consortium Position Paper. Front Immunol. 2020;11:1648.PubMedPubMedCentralCrossRef
24.
25.
go back to reference Chen R, Lan Z, Ye J, et al. Cytokine storm: the primary determinant for the pathophysiological evolution of COVID-19 deterioration. Front Immunol. 2021;12:589095.PubMedPubMedCentralCrossRef Chen R, Lan Z, Ye J, et al. Cytokine storm: the primary determinant for the pathophysiological evolution of COVID-19 deterioration. Front Immunol. 2021;12:589095.PubMedPubMedCentralCrossRef
26.
go back to reference Zawawi A, Naser AY, Alwafi H, Minshawi F. Profile of circulatory cytokines and chemokines in human coronaviruses: a systemic review and meta-analysis. Front Immunol. 2021;12:666223.PubMedPubMedCentralCrossRef Zawawi A, Naser AY, Alwafi H, Minshawi F. Profile of circulatory cytokines and chemokines in human coronaviruses: a systemic review and meta-analysis. Front Immunol. 2021;12:666223.PubMedPubMedCentralCrossRef
27.
go back to reference Carlos WG, Dela Cruz CS, Cao B, et al. Novel Wuhan (2019-nCoV) coronavirus. Am J Respir Crit Care Med. 2020;201:P7–8.PubMedCrossRef Carlos WG, Dela Cruz CS, Cao B, et al. Novel Wuhan (2019-nCoV) coronavirus. Am J Respir Crit Care Med. 2020;201:P7–8.PubMedCrossRef
28.
go back to reference Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369:eabc8511.PubMedPubMedCentralCrossRef Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369:eabc8511.PubMedPubMedCentralCrossRef
29.
go back to reference Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory immune responses in severe COVID-19 patients. Science. 2020;369:718–24.PubMedPubMedCentralCrossRef Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory immune responses in severe COVID-19 patients. Science. 2020;369:718–24.PubMedPubMedCentralCrossRef
31.
go back to reference Bonjardim CA. Interferons (IFNs) are key cytokines in both innate and adaptive antiviral immune responses—and viruses counteract IFN action. Microbes Infect. 2005;7:569–78.PubMedCrossRef Bonjardim CA. Interferons (IFNs) are key cytokines in both innate and adaptive antiviral immune responses—and viruses counteract IFN action. Microbes Infect. 2005;7:569–78.PubMedCrossRef
32.
go back to reference Ruetsch C, Brglez V, Crémoni M, et al. Functional exhaustion of both types of IFNs production in severe COVID-19 patients. Front Immunol. 2021;7:603961. Ruetsch C, Brglez V, Crémoni M, et al. Functional exhaustion of both types of IFNs production in severe COVID-19 patients. Front Immunol. 2021;7:603961.
33.
go back to reference Bastard P, Gervais A, Le Voyere T, et al. Autoantibodies neutralizing type I IFNs are present in ~4% of uninfected individuals over 70 years old and account for ~20% of COVID-19 deaths. Sci Immunol. 2021;6:eabl4340.PubMedPubMedCentralCrossRef Bastard P, Gervais A, Le Voyere T, et al. Autoantibodies neutralizing type I IFNs are present in ~4% of uninfected individuals over 70 years old and account for ~20% of COVID-19 deaths. Sci Immunol. 2021;6:eabl4340.PubMedPubMedCentralCrossRef
34.
go back to reference Goncalves D, Mezidi M, Bastard P, et al. Antibodies against type I interferon: detection and association with severe clinical outcome in COVID-19 patients. Clin Transl Immunol. 2021;10:e1327.CrossRef Goncalves D, Mezidi M, Bastard P, et al. Antibodies against type I interferon: detection and association with severe clinical outcome in COVID-19 patients. Clin Transl Immunol. 2021;10:e1327.CrossRef
35.
go back to reference Troya J, Bastard P, Planas-Serra L, et al. Neutralizing autoantibodies to type IFNs in >10% of patients with severe COVID-19 pneumonia hospitalized in Madrid, Spain. J Clin Immunol. 2021;41:914–22.PubMedPubMedCentralCrossRef Troya J, Bastard P, Planas-Serra L, et al. Neutralizing autoantibodies to type IFNs in >10% of patients with severe COVID-19 pneumonia hospitalized in Madrid, Spain. J Clin Immunol. 2021;41:914–22.PubMedPubMedCentralCrossRef
36.
go back to reference Chauvineau-Grenier A, Bastard P, Servajea A, et al. Autoantibodies neutralizing type I interferons in 20% of COVID-19 deaths in a French hospital. RES Sq. 2021;rs.3.rs-915062. Chauvineau-Grenier A, Bastard P, Servajea A, et al. Autoantibodies neutralizing type I interferons in 20% of COVID-19 deaths in a French hospital. RES Sq. 2021;rs.3.rs-915062.
37.
go back to reference Harada M, Furuhashi K, Karayama M, et al. Subcutaneous injection of interferon gamma therapy could be useful for anti-IFN-γ autoantibody associated disseminated nontuberculous mycobacterial infection. J Infect Chemother. 2021;27:373–8.PubMedCrossRef Harada M, Furuhashi K, Karayama M, et al. Subcutaneous injection of interferon gamma therapy could be useful for anti-IFN-γ autoantibody associated disseminated nontuberculous mycobacterial infection. J Infect Chemother. 2021;27:373–8.PubMedCrossRef
38.
go back to reference Darazam IA, Shokouhi S, Pourhoseingholi MA, et al. Role of interferon therapy in severe COVID19: the COVIFERON randomized controlled trial. Sci Rep. 2021;11:8059.CrossRef Darazam IA, Shokouhi S, Pourhoseingholi MA, et al. Role of interferon therapy in severe COVID19: the COVIFERON randomized controlled trial. Sci Rep. 2021;11:8059.CrossRef
40.
go back to reference Chi CY, Chu CC, Liu JP, et al. Anti-IFN-gamma autoantibodies in adults with disseminated nontuberculous mycobacterial infections are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02 and the reactivation of latent varicella-zoster virus infection. Blood. 2013;121:1357–66.PubMedCrossRef Chi CY, Chu CC, Liu JP, et al. Anti-IFN-gamma autoantibodies in adults with disseminated nontuberculous mycobacterial infections are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02 and the reactivation of latent varicella-zoster virus infection. Blood. 2013;121:1357–66.PubMedCrossRef
41.
go back to reference Ku CL, Lin CH, Chang SW, Chu CC, Chan JF, Kong XF, et al. Anti-IFN-gamma autoantibodies are strongly associated with HLA-DR*15:02/16:02 and HLA-DQ*05:01/05:02 across Southeast Asia. J Allergy Clin Immunol. 2016;137:945-8.e8.PubMedCrossRef Ku CL, Lin CH, Chang SW, Chu CC, Chan JF, Kong XF, et al. Anti-IFN-gamma autoantibodies are strongly associated with HLA-DR*15:02/16:02 and HLA-DQ*05:01/05:02 across Southeast Asia. J Allergy Clin Immunol. 2016;137:945-8.e8.PubMedCrossRef
42.
go back to reference Krisnawati DI, Liu YC, Lee YJ, et al. Blockade effects of anti-interferon-(IFN-) γ autoantibodies on IFN-γ-regulated antimicrobial immunity. J Immunol Res. 2019;2019:1629258.PubMedPubMedCentralCrossRef Krisnawati DI, Liu YC, Lee YJ, et al. Blockade effects of anti-interferon-(IFN-) γ autoantibodies on IFN-γ-regulated antimicrobial immunity. J Immunol Res. 2019;2019:1629258.PubMedPubMedCentralCrossRef
43.
go back to reference Kim MH, Salloum S, Wang JY, et al. Type I, II, and III interferon signatures correspond to COVID-19 disease severity. J Infect Dis. 2021;224:777–82.PubMedCrossRef Kim MH, Salloum S, Wang JY, et al. Type I, II, and III interferon signatures correspond to COVID-19 disease severity. J Infect Dis. 2021;224:777–82.PubMedCrossRef
44.
go back to reference Chen ZM, Yang XY, Li ZT, et al. Anti-interferon-γ autoantibodies impair T-lymphocyte responses in patients with Talaromyces marneffei infection. Infect Drug Resist. 2022;15:3381–93.PubMedPubMedCentralCrossRef Chen ZM, Yang XY, Li ZT, et al. Anti-interferon-γ autoantibodies impair T-lymphocyte responses in patients with Talaromyces marneffei infection. Infect Drug Resist. 2022;15:3381–93.PubMedPubMedCentralCrossRef
45.
go back to reference De Biasi S, Meschiari M, Gibellini L, et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID19 pneumonia. Nat Commun. 2020;11:3434.PubMedPubMedCentralCrossRef De Biasi S, Meschiari M, Gibellini L, et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID19 pneumonia. Nat Commun. 2020;11:3434.PubMedPubMedCentralCrossRef
47.
go back to reference Blanco-Melo D, Nilsson-Payant BE, Liu W-C, et al. Imbalanced host response to SARS-CoV2 drives development of COVID-19. Cell. 2020;81:1036–45.CrossRef Blanco-Melo D, Nilsson-Payant BE, Liu W-C, et al. Imbalanced host response to SARS-CoV2 drives development of COVID-19. Cell. 2020;81:1036–45.CrossRef
48.
go back to reference Hasselbalch HC, Skov V, Kjær L, et al. COVID-19 as a mediator of interferon deficiency and hyperinflammation: rationale for the use of JAK1/2 inhibitors in combination with interferon. Cytokine Growth Factor Rev. 2021;60:28–45.PubMedPubMedCentralCrossRef Hasselbalch HC, Skov V, Kjær L, et al. COVID-19 as a mediator of interferon deficiency and hyperinflammation: rationale for the use of JAK1/2 inhibitors in combination with interferon. Cytokine Growth Factor Rev. 2021;60:28–45.PubMedPubMedCentralCrossRef
Metadata
Title
The detectable anti-interferon-γ autoantibodies in COVID-19 patients may be associated with disease severity
Authors
Po-Ku Chen
Kai-Jieh Yeo
Shih-Hsin Chang
Tsai-Ling Liao
Chia-Hui Chou
Joung-Liang Lan
Ching-Kun Chang
Der-Yuan Chen
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Virology Journal / Issue 1/2023
Electronic ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-023-01989-1

Other articles of this Issue 1/2023

Virology Journal 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.