1
|
Buhre JS, Pongracz T, Geisen UM, Schubert M, Wang W, Nouta J, Obara M, Lehrian S, Rahmöller J, Petry J, Tran F, Schreiber S, Sümbül M, Berner D, Gerdes S, Schirmer J, Longardt AC, Hoff P, Kalinke U, Ludwig RJ, Bartsch YC, Hoyer BF, Wuhrer M, Ehlers M. Anti-TNF therapy impairs both short- and long-term IgG responses after repeated vaccination. Allergy 2025; 80:423-439. [PMID: 39049686 PMCID: PMC11804311 DOI: 10.1111/all.16241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Recently, it has been questioned whether vaccination of patients with inflammatory (auto)immune diseases under anti-tumor necrosis factor (TNF) treatment leads to impaired vaccine-induced immune responses and protection against breakthrough infections. However, the effects of TNF blockade on short- and long-term immune responses after repeated vaccination remain unclear. Vaccination studies have shown that initial short-term IgG antibodies (Abs) carry highly galactosylated and sialylated Fc glycans, whilst long-term IgG Abs have low levels of galactosylation and sialylation and are most likely generated by long-lived plasma cells (PCs) derived primarily from the germinal center (GC) response. Thus, IgG Fc glycosylation patterns may be applicable to distinguish short- and long-term vaccine responses after repeated vaccination under the influence of anti-TNF treatment. METHODS We used COVID-19 vaccination as a model to investigate vaccine-induced IgG subclass levels and Fc glycosylation patterns, B cell subsets, and effector functions of short- and long-term Ab responses after up to three vaccinations in patients on anti-TNF or other immunosuppressive treatments and in healthy individuals. Using TriNetX, a global healthcare database, we determined the risk of SARS-CoV-2 breakthrough infections in vaccinated patients treated with anti-TNF or other immunosuppressive drugs. RESULTS Anti-TNF treatment reduced the long-term abundance of all anti-S IgG subclasses with low levels of galactosylation and sialylation. Re-activation of potential memory B cells initially generated highly galactosylated and sialylated IgG antibodies, which were progressively reduced after each booster dose in anti-TNF-treated patients, especially in the elderly. The reduced short- and long-term IgG (1) levels in anti-TNF-treated patients correlated with diminished functional activity and an increased risk for the development of COVID-19. CONCLUSIONS The data suggest that anti-TNF treatment reduces both GC-dependent long-lived PCs and GC-dependent memory B cell-derived short-lived PCs, hence both the long- and short-term IgG subclass responses, respectively, after repeated vaccination. We propose that anti-TNF therapy, especially in the elderly, reduces the benefit of booster vaccination.
Collapse
Affiliation(s)
- Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional MedicineUniversity of Lübeck and University Medical Center Schleswig‐HolsteinLübeckGermany
| | - Tamas Pongracz
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Ulf Martin Geisen
- Medical Department 1, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Mareike Schubert
- Laboratory of Anti‐viral antibody‐omicsTWINCORE—Institute for Experimental Infection Research, Helmholtz Center for Infection Research (HZI) and Medical School Hannover (MHH)HannoverGermany
| | - Wenjun Wang
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Jan Nouta
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Maureen Obara
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical SchoolHannoverGermany
| | - Selina Lehrian
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional MedicineUniversity of Lübeck and University Medical Center Schleswig‐HolsteinLübeckGermany
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional MedicineUniversity of Lübeck and University Medical Center Schleswig‐HolsteinLübeckGermany
| | - Janina Petry
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional MedicineUniversity of Lübeck and University Medical Center Schleswig‐HolsteinLübeckGermany
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian‐Albrecht University of KielKielGermany
- Department for Internal Medicine IUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian‐Albrecht University of KielKielGermany
- Department for Internal Medicine IUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Melike Sümbül
- Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Dennis Berner
- Medical Department 1, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Sascha Gerdes
- Department for DermatologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Jan Schirmer
- Medical Department 1, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | | | - Paula Hoff
- Department of RheumatologyEndokrinologikum‐GruppeBerlinGermany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical SchoolHannoverGermany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental DermatologyUniversity of LübeckLübeckGermany
- Department of DermatologyUniversity Medical Center Schleswig‐HolsteinLübeckGermany
| | - Yannic C. Bartsch
- Laboratory of Anti‐viral antibody‐omicsTWINCORE—Institute for Experimental Infection Research, Helmholtz Center for Infection Research (HZI) and Medical School Hannover (MHH)HannoverGermany
| | - Bimba F. Hoyer
- Medical Department 1, Rheumatology and Clinical ImmunologyUniversity Medical Center Schleswig‐HolsteinKielGermany
| | - Manfred Wuhrer
- Center for Proteomics and MetabolomicsLeiden University Medical CenterLeidenThe Netherlands
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional MedicineUniversity of Lübeck and University Medical Center Schleswig‐HolsteinLübeckGermany
- Airway Research Center North (ARCN)University of Lübeck, German Center for Lung Research (DZL)LübeckGermany
| |
Collapse
|
2
|
Chou CY, Cheng CY, Lee CH, Kuro-O M, Chen TH, Wang SY, Chuang YK, Yang YJ, Lin YH, Tsai IL. Unveiling unique effector function-related bulk antibody profiles in long-term hemodialysis patients following COVID-19 mRNA booster vaccination. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:27-37. [PMID: 39395903 DOI: 10.1016/j.jmii.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2024] [Accepted: 09/27/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Hemodialysis patients exhibit a reduced response to vaccination and have different vaccine dose regimens. Vaccines induce antibodies and affect the inflammatory balance through antibody glycosylation and effector functions. Therefore, we aimed to analyze the antibody glycosylation profiles in hemodialysis patients who were vaccinated against severe acute respiratory syndrome coronavirus 2, infected with the virus, or both, and compare them with those of dialysis patients in a control group. METHODS Plasma samples from 112 hemodialysis patients were assigned to four groups: control, infected, vaccinated, and post-vaccine-infected. Paired plasma samples from 47 people with vaccination (vaccinees) were analyzed before and after the booster dose. The same analytical approach was applied to the four groups for a cross-sectional comparison. RESULTS Our study found that both vaccination and infection groups showed decreased fucosylation of IgG1, which is associated with a proinflammatory biosignature. However, vaccination also leads to increased galactosylation and bisection of IgG antibodies, which are associated with anti-inflammatory effects and the additional regulation of immune responses. In contrast, infection led to an additional decrease in the fucosylation of IgG2 and IgA, demonstrating a more intense proinflammatory biosignature than vaccination. CONCLUSIONS Our findings emphasize the proinflammatory biosignature of afucosylation in both vaccination and infection groups. Additionally, we uncovered further regulated profiles related to galactosylation in vaccinees. These findings suggest that antibody investigation for vaccination or infection should not solely focus on neutralization but should also consider effector function-related glycosylation profiling. This comprehensive information can be valuable for fine-tuning vaccine development in the future.
Collapse
Affiliation(s)
- Chia-Yi Chou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan
| | - Chung-Yi Cheng
- Taipei Medical University Research Center of Urology and Kidney, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsin Lee
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Tso-Hsiao Chen
- Taipei Medical University Research Center of Urology and Kidney, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - San-Yuan Wang
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kun Chuang
- Master Program in Food Safety, College of Nutrition, Taipei Medical University, Taiwan
| | - Yun-Jung Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan
| | - Yun-Hsuan Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan
| | - I-Lin Tsai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan; Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
3
|
Váradi C. The Glycosylation of Serum IgG Antibodies in Post-COVID-19 and Post-Vaccination Patients. Int J Mol Sci 2025; 26:807. [PMID: 39859521 PMCID: PMC11765615 DOI: 10.3390/ijms26020807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
The signature of human serum IgG glycosylation is critical in the defense against pathogens. Alterations of IgG N-glycome were associated with COVID-19 (Coronavirus disease 2019) severity, although knowledge on the response to vaccination is limited. IgG N-glycome was analyzed in this study in post-COVID-19 and post-vaccination patients to reveal potential glycosylation-based alterations using hydrophilic interaction liquid chromatography (HILIC-UPLC) with fluorescence (FLR) and mass-spectrometric (MS) detection. IgG antibodies were purified from serum samples through protein G affinity chromatography followed by PNGase F digestion-based deglycosylation. The released glycans were fluorescently derivatized by procainamide labeling and purified via solid-phase extraction. Higher levels of sialylation and afucosylation were identified in post-COVID-19 patients, which was further expanded by vaccination, but only in those who were previously SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) infected.
Collapse
Affiliation(s)
- Csaba Váradi
- Institute of Chemistry, Faculty of Materials and Chemical Engineering, University of Miskolc, 3515 Miskolc, Hungary
| |
Collapse
|
4
|
Oliva-Ariza G, Criado I, Fuentes-Herrero B, Carbonell C, Sánchez-Gallego JI, López-Bernús A, Gutiérrez ML, Rolo-Ramírez A, Bernal-Ribes M, Almenara-Morales Y, Lecrevisse Q, van Dongen JJM, Marcos M, Almeida J, Orfao A. Early Immune Cell and Antibody Kinetics Following SARS-CoV-2 Vaccination in Healthy Adults and Low-Count Monoclonal B-Cell Lymphocytosis. Int J Mol Sci 2025; 26:681. [PMID: 39859394 PMCID: PMC11765611 DOI: 10.3390/ijms26020681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The early immune kinetics after SARS-CoV-2 vaccination remain poorly understood, particularly among individuals with low-count monoclonal B-cell lymphocytosis (MBLlo). We investigated the cellular and humoral kinetics in the blood of 50 non-MBL healthy donors (HD) vs. 16 MBLlo subjects after SARS-CoV-2 vaccination, who were subclassified according to their history of previous exposure to SARS-CoV-2 into SARS-CoV-2 naïve and previously infected subjects. Overall, we found decreased neutrophil and lymphocyte counts at day +4 following each dose in non-MBL HD, together with an earlier and higher increase in plasma cell (PC) counts and SARS-CoV-2-specific antibody levels after the first vaccine in previously infected non-MBL HD. MBLlo subjects showed a similar profile, except for lower B-cell and higher PC counts after vaccination, and a trend towards a higher (but delayed) antibody response. In summary, we found different cell-kinetic profiles following vaccination in SARS-CoV-2 naïve vs. previously infected non-MBL HD (earlier PC and antibody responses in the latter group); additionally, MBLlo subjects had significantly lower B-cell and higher PC counts after vaccination, and a delayed SARS-CoV-2-specific antibody response.
Collapse
Affiliation(s)
- Guillermo Oliva-Ariza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ignacio Criado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Blanca Fuentes-Herrero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Carbonell
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - José Ignacio Sánchez-Gallego
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Amparo López-Bernús
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
- Department of Infectious Diseases, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), 37007 Salamanca, Spain
| | - María Laura Gutiérrez
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alejandro Rolo-Ramírez
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Marta Bernal-Ribes
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Yolimar Almenara-Morales
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jacques J. M. van Dongen
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
| | - Miguel Marcos
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.R.-R.); (Y.A.-M.)
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, Instituto de Biología Molecular y Celular del Cáncer, CSIC—University of Salamanca), Cytometry Service, NUCLEUS, 37007 Salamanca, Spain; (G.O.-A.); (I.C.); (B.F.-H.); (J.I.S.-G.); (M.L.G.); (M.B.-R.); (Q.L.); (J.J.M.v.D.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain; (C.C.); (A.L.-B.); (M.M.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Radovani B, Nimmerjahn F. IgG Glycosylation: Biomarker, Functional Modulator, and Structural Component. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1573-1584. [PMID: 39556784 DOI: 10.4049/jimmunol.2400447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 11/20/2024]
Abstract
The family of IgG Abs is a crucial component of adaptive immunity. Glycosylation of IgG maintains its structural integrity and modulates its effector functions. In this review, we discuss IgG glycosylation covering cell biological as well as therapeutic and disease-related aspects, focusing on the glycan structures in distinct IgG regions (Fab versus Fc). We also cover the impact of IgG glycosylation on disease modulation and therapeutic outcomes, alongside the potential for development of vaccines designed to induce Ag-specific IgG with glycoforms for optimal immune responses. Overall, we emphasize the significance of studying glycosylation to enhance our understanding of the dynamics and functional impacts of IgG glycosylation. These insights could be beneficial for advancing future research and clinical applications.
Collapse
Affiliation(s)
- Barbara Radovani
- Faculty of Biotechnology and Drug Development, University of Rijeka, Rijeka, Croatia
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Profile Center Immunomedicine, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
6
|
Yaugel-Novoa M, Noailly B, Jospin F, Pizzorno A, Traversier A, Pozzetto B, Waeckel L, Longet S, Pillet S, Botelho-Nevers E, Rosa-Calatrava M, Bourlet T, Paul S. Impaired mucosal IgA response in patients with severe COVID-19. Emerg Microbes Infect 2024; 13:2401940. [PMID: 39358866 PMCID: PMC11451292 DOI: 10.1080/22221751.2024.2401940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Several studies have investigated the antibody response to SARS-CoV-2, focusing particularly on the systemic humoral immune response and the production of immunoglobulin G (IgG) antibodies. IgA antibodies play a crucial role in protecting against respiratory viral infections but have also been associated with the pathophysiology of COVID-19. We performed a prospective study of 169 COVID-19 patients - 50 with critical/severe (ICU), 47 with moderate (Non-ICU), and 72 with asymptomatic COVID-19 - to explore the humoral immune response to SARS-CoV-2 infection. We found that the early systemic IgA response strongly induced in patients with severe disease did not block IgG neutralization functions and activated FcRs more effectively than IgG. However, even if SIgA levels were high, mucosal IgA antibodies could not control the infection effectively in patients with severe disease. Our findings highlight the complexity of the immune response to SARS-CoV-2 exhibiting high systemic levels of IgA with strong neutralizing capacity in severe cases, together with higher levels of IgA-FcR activation than in asymptomatic patients. They also suggest the need for further research to fully understand the role of IgA and its structural alterations in mucosal tissues in cases of severe disease and the impact of these antibodies on disease progression.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Blandine Noailly
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Fabienne Jospin
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Andrés Pizzorno
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aurélien Traversier
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Bruno Pozzetto
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Louis Waeckel
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphanie Longet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Sylvie Pillet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Diseases Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Manuel Rosa-Calatrava
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Thomas Bourlet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Lead contact
| |
Collapse
|
7
|
Brook B, Checkervarty AK, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The BNT162b2 mRNA vaccine demonstrates reduced age-associated T H1 support in vitro and in vivo. iScience 2024; 27:111055. [PMID: 39569372 PMCID: PMC11576392 DOI: 10.1016/j.isci.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
mRNA vaccines demonstrate impaired immunogenicity and durability in vulnerable older populations. We hypothesized that human in vitro modeling and proteomics could elucidate age-specific mRNA vaccine actions. BNT162b2-stimulation changed the plasma proteome of blood samples from young (18-50Y) and older adult (≥60Y) participants, assessed by mass spectrometry, proximity extension assay, and multiplex. Young adult up-regulation (e.g., PSMC6, CPN1) contrasted reduced induction in older adults (e.g., TPM4, APOF, APOC2, CPN1, PI16). 30-85% lower TH1-polarizing cytokines and chemokines were induced in elderly blood (e.g., IFNγ, CXCL10). Analytes lower in older adult samples included human in vivo mRNA immunogenicity biomarkers (e.g., IFNγ, CXCL10, CCL4, IL-1RA). BNT162b2 also demonstrated reduced CD4+ TH1 responses in aged vs. young adult mice. Our study demonstrates the utility of human in vitro platforms modeling age-specific mRNA vaccine immunogenicity, highlights impaired support of TH1 polarization in older adults, and provides a rationale for precision mRNA vaccine adjuvantation to induce greater immunogenicity.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy C Sherman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine- Chair of Pediatrics, University of Rome, 00133 Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dheeraj Soni
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
8
|
Sherman JD, Karmali V, Kumar B, Simon TW, Bechnak S, Panjwani A, Ciric CR, Wang D, Huerta C, Johnson B, Anderson EJ, Rouphael N, Collins MH, Rostad CA, Azadi P, Scherer EM. Altered Spike Immunoglobulin G Fc N-Linked Glycans Are Associated With Hyperinflammatory State in Adult Coronavirus Disease 2019 and Multisystem Inflammatory Syndrome in Children. Open Forum Infect Dis 2024; 11:ofae626. [PMID: 39494457 PMCID: PMC11528514 DOI: 10.1093/ofid/ofae626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Background Severe coronavirus disease 2019 (COVID-19) and multisystem inflammatory syndrome (MIS-C) are characterized by excessive inflammatory cytokines/chemokines. In adults, disease severity is associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific immunoglobulin G (IgG) Fc afucosylation, which induces proinflammatory cytokine secretion from innate immune cells. This study aimed to define spike IgG Fc glycosylation following SARS-CoV-2 infection in adults and children and following SARS-CoV-2 vaccination in adults and the relationships between glycan modifications and cytokines/chemokines. Methods We analyzed longitudinal (n = 146) and cross-sectional (n = 49) serum/plasma samples from adult and pediatric COVID-19 patients, MIS-C patients, adult vaccinees, and adult and pediatric controls. We developed methods for characterizing bulk and spike IgG Fc glycosylation by capillary electrophoresis and measured levels of 10 inflammatory cytokines/chemokines by multiplexed enzyme-linked immunosorbent assay. Results Spike IgG was more afucosylated than bulk IgG during acute adult COVID-19 and MIS-C. We observed an opposite trend following vaccination, but it was not significant. Spike IgG was more galactosylated and sialylated and less bisected than bulk IgG during adult COVID-19, with similar trends observed during pediatric COVID-19/MIS-C and following SARS-CoV-2 vaccination. Spike IgG glycosylation changed with time following adult COVID-19 or vaccination. Afucosylated spike IgG exhibited inverse and positive correlations with inflammatory markers in MIS-C and following vaccination, respectively; galactosylated and sialylated spike IgG inversely correlated with proinflammatory cytokines in adult COVID-19 and MIS-C; and bisected spike IgG positively correlated with inflammatory cytokines/chemokines in multiple groups. Conclusions We identified previously undescribed relationships between spike IgG glycan modifications and inflammatory cytokines/chemokines that expand our understanding of IgG glycosylation changes that may impact COVID-19 and MIS-C immunopathology.
Collapse
Affiliation(s)
- Jacob D Sherman
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vinit Karmali
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Trevor W Simon
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sarah Bechnak
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anusha Panjwani
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Caroline R Ciric
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dongli Wang
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christopher Huerta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brandi Johnson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Evan J Anderson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Matthew H Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christina A Rostad
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Erin M Scherer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Pérez-Latorre L, Ramilo O. SARS-CoV-2 infection vs. vaccination during pregnancy: the placenta leads the way. Curr Opin Infect Dis 2024; 37:402-406. [PMID: 39105669 DOI: 10.1097/qco.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
PURPOSE OF REVIEW To understand the characteristics and determinants of transplacental antibody transfer against SARS-CoV-2 and to compare the differences between SARS-CoV-2 infection and vaccination. RECENT FINDINGS The need for information during the COVID-19 pandemic and the exclusion of pregnant women from randomized clinical trials have led to a vast amount of clinical data primarily based on observational studies with diverse design and sample analyses that yield variable results. This review aims to critically and comprehensively integrate the relevant knowledge related to transplacental transfer of antibodies against SARS-CoV-2, emphasizing the differences between infection and vaccination. SUMMARY Passive immunization is key to conferring protection to the infant during their first months of life. Understanding the mechanisms of transplacental antibody transfer during SARS-CoV-2 infection and vaccination, and their associated protection will allow optimizing the implementation of well tolerated and effective preventive strategies for both pregnant women and infants.
Collapse
Affiliation(s)
- Leire Pérez-Latorre
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañon, Madrid
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Spain
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
10
|
Sherman JD, Karmali V, Kumar B, Simon TW, Bechnak S, Panjwani A, Ciric CR, Wang D, Huerta C, Johnson B, Anderson EJ, Rouphael N, Collins MH, Rostad CA, Azadi P, Scherer EM. Altered spike IgG Fc N-linked glycans are associated with hyperinflammatory state in adult COVID and Multisystem Inflammatory Syndrome in Children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310381. [PMID: 39040211 PMCID: PMC11261911 DOI: 10.1101/2024.07.14.24310381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Background Severe COVID and multisystem inflammatory syndrome (MIS-C) are characterized by excessive inflammatory cytokines/chemokines. In adults, disease severity is associated with SARS-CoV-2-specific IgG Fc afucosylation, which induces pro-inflammatory cytokine secretion from innate immune cells. This study aimed to define spike IgG Fc glycosylation following SARS-CoV-2 infection in adults and children and following SARS-CoV-2 vaccination in adults and the relationships between glycan modifications and cytokine/chemokine levels. Methods We analyzed longitudinal (n=146) and cross-sectional (n=49) serum/plasma samples from adult and pediatric COVID patients, MIS-C patients, adult vaccinees, and adult and pediatric healthy controls. We developed methods for characterizing bulk and spike IgG Fc glycosylation by capillary electrophoresis (CE) and measured levels of ten inflammatory cytokines/chemokines by multiplexed ELISA. Results Spike IgG were more afucosylated than bulk IgG during acute adult COVID and MIS-C. We observed an opposite trend following vaccination, but it was not significant. Spike IgG were more galactosylated and sialylated and less bisected than bulk IgG during adult COVID, with similar trends observed during pediatric COVID/MIS-C and following SARS-CoV-2 vaccination. Spike IgG glycosylation changed with time following adult COVID or vaccination. Afucosylated spike IgG exhibited inverse and positive correlations with inflammatory markers in MIS-C and following vaccination, respectively; galactosylated and sialylated spike IgG inversely correlated with pro-inflammatory cytokines in adult COVID and MIS-C; and bisected spike IgG positively correlated with inflammatory cytokines/chemokines in multiple groups. Conclusions We identified previously undescribed relationships between spike IgG glycan modifications and inflammatory cytokines/chemokines that expand our understanding of IgG glycosylation changes that may impact COVID and MIS-C immunopathology.
Collapse
Affiliation(s)
- Jacob D. Sherman
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Vinit Karmali
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Trevor W. Simon
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sarah Bechnak
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anusha Panjwani
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Caroline R. Ciric
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dongli Wang
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chris Huerta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Brandi Johnson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Matthew H. Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Christina A. Rostad
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Erin M. Scherer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
11
|
Rubio-Casillas A, Cowley D, Raszek M, Uversky VN, Redwan EM. Review: N1-methyl-pseudouridine (m1Ψ): Friend or foe of cancer? Int J Biol Macromol 2024; 267:131427. [PMID: 38583833 DOI: 10.1016/j.ijbiomac.2024.131427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/09/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Due to the health emergency created by SARS-CoV-2, the virus that causes the COVID-19 disease, the rapid implementation of a new vaccine technology was necessary. mRNA vaccines, being one of the cutting-edge new technologies, attracted significant interest and offered a lot of hope. The potential of these vaccines in preventing admission to hospitals and serious illness in people with comorbidities has recently been called into question due to the vaccines' rapidly waning immunity. Mounting evidence indicates that these vaccines, like many others, do not generate sterilizing immunity, leaving people vulnerable to recurrent infections. Additionally, it has been discovered that the mRNA vaccines inhibit essential immunological pathways, thus impairing early interferon signaling. Within the framework of COVID-19 vaccination, this inhibition ensures an appropriate spike protein synthesis and a reduced immune activation. Evidence is provided that adding 100 % of N1-methyl-pseudouridine (m1Ψ) to the mRNA vaccine in a melanoma model stimulated cancer growth and metastasis, while non-modified mRNA vaccines induced opposite results, thus suggesting that COVID-19 mRNA vaccines could aid cancer development. Based on this compelling evidence, we suggest that future clinical trials for cancers or infectious diseases should not use mRNA vaccines with a 100 % m1Ψ modification, but rather ones with the lower percentage of m1Ψ modification to avoid immune suppression.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico.
| | - David Cowley
- University of Lincoln, Brayford Pool, Lincoln, Lincolnshire LN6 7TS, United Kingdom
| | - Mikolaj Raszek
- Merogenomics (Genomic Sequencing Consulting), Edmonton, AB T5J 3R8, Canada
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt.
| |
Collapse
|
12
|
Bermejo-Jambrina M, van der Donk LE, van Hamme JL, Wilflingseder D, de Bree G, Prins M, de Jong M, Nieuwkerk P, van Gils MJ, Kootstra NA, Geijtenbeek TB. Control of complement-induced inflammatory responses to SARS-CoV-2 infection by anti-SARS-CoV-2 antibodies. EMBO J 2024; 43:1135-1163. [PMID: 38418557 PMCID: PMC10987522 DOI: 10.1038/s44318-024-00061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024] Open
Abstract
Dysregulated immune responses contribute to the excessive and uncontrolled inflammation observed in severe COVID-19. However, how immunity to SARS-CoV-2 is induced and regulated remains unclear. Here, we uncover the role of the complement system in the induction of innate and adaptive immunity to SARS-CoV-2. Complement rapidly opsonizes SARS-CoV-2 particles via the lectin pathway. Complement-opsonized SARS-CoV-2 efficiently induces type-I interferon and pro-inflammatory cytokine responses via activation of dendritic cells, which are inhibited by antibodies against the complement receptors (CR) 3 and 4. Serum from COVID-19 patients, or monoclonal antibodies against SARS-CoV-2, attenuate innate and adaptive immunity induced by complement-opsonized SARS-CoV-2. Blocking of CD32, the FcγRII antibody receptor of dendritic cells, restores complement-induced immunity. These results suggest that opsonization of SARS-CoV-2 by complement is involved in the induction of innate and adaptive immunity to SARS-CoV-2 in the acute phase of infection. Subsequent antibody responses limit inflammation and restore immune homeostasis. These findings suggest that dysregulation of the complement system and FcγRII signaling may contribute to severe COVID-19.
Collapse
Affiliation(s)
- Marta Bermejo-Jambrina
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Lieve Eh van der Donk
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - John L van Hamme
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Godelieve de Bree
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
- Department of Internal Medicine, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Prins
- Department of Internal Medicine, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, The Netherlands
| | - Menno de Jong
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Pythia Nieuwkerk
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, The Netherlands
- Department of Medical Psychology (J3-2019-1), Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Teunis Bh Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Lasrado N, Collier ARY, Miller J, Hachmann NP, Liu J, Anand T, A. Bondzie E, Fisher JL, Mazurek CR, Patio RC, Rodrigues SL, Rowe M, Surve N, Ty DM, Wu C, Chicz TM, Tong X, Korber B, McNamara RP, Barouch DH. Waning immunity and IgG4 responses following bivalent mRNA boosting. SCIENCE ADVANCES 2024; 10:eadj9945. [PMID: 38394195 PMCID: PMC10889350 DOI: 10.1126/sciadv.adj9945] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Messenger RNA (mRNA) vaccines were highly effective against the ancestral SARS-CoV-2 strain, but the efficacy of bivalent mRNA boosters against XBB variants was substantially lower. Here, we show limited durability of neutralizing antibody (NAb) responses against XBB variants and isotype switching to immunoglobulin G4 (IgG4) responses following bivalent mRNA boosting. Bivalent mRNA boosting elicited modest XBB.1-, XBB.1.5-, and XBB.1.16-specific NAbs that waned rapidly within 3 months. In contrast, bivalent mRNA boosting induced more robust and sustained NAbs against the ancestral WA1/2020 strain, suggesting immune imprinting. Following bivalent mRNA boosting, serum antibody responses were primarily IgG2 and IgG4 responses with poor Fc functional activity. In contrast, a third monovalent mRNA immunization boosted all isotypes including IgG1 and IgG3 with robust Fc functional activity. These data show substantial immune imprinting for the ancestral spike and isotype switching to IgG4 responses following bivalent mRNA boosting, with important implications for future booster designs and boosting strategies.
Collapse
Affiliation(s)
- Ninaad Lasrado
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ai-ris Y. Collier
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Miller
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nicole P. Hachmann
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jinyan Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Trisha Anand
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Esther A. Bondzie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jana L. Fisher
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Camille R. Mazurek
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert C. Patio
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Marjorie Rowe
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nehalee Surve
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Darren M. Ty
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cindy Wu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Taras M. Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Xin Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Bette Korber
- Los Alamos National Laboratory and New Mexico Consortium, Los Alamos, NM, USA
| | | | - Dan H. Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
14
|
Monroe JM, Quach HQ, Punia S, Enninga EAL, Fedyshyn Y, Girsch JH, Fedyshyn B, Lemens M, Littlefield D, Behl S, Sintim-Aboagye E, Mejia Plazas MC, Yamaoka S, Ebihara H, Pandey A, Correia C, Ung CY, Li H, Vassallo R, Sun J, Johnson EL, Olson JE, Theel ES, Badley AD, Kennedy RB, Theiler RN, Chakraborty R. Vertical Transmission of SARS-CoV-2-Specific Antibodies and Cytokine Profiles in Pregnancy. J Infect Dis 2024; 229:473-484. [PMID: 37786979 PMCID: PMC11491822 DOI: 10.1093/infdis/jiad399] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
Despite intensive characterization of immune responses after COVID-19 infection and vaccination, research examining protective correlates of vertical transmission in pregnancy are limited. Herein, we profiled humoral and cellular characteristics in pregnant women infected or vaccinated at different trimesters and in their corresponding newborns. We noted a significant correlation between spike S1-specific IgG antibody and its RBD-ACE2 blocking activity (receptor-binding domain-human angiotensin-converting enzyme 2) in maternal and cord plasma (P < .001, R > 0.90). Blocking activity of spike S1-specific IgG was significantly higher in pregnant women infected during the third trimester than the first and second trimesters. Elevated levels of 28 cytokines/chemokines, mainly proinflammatory, were noted in maternal plasma with infection at delivery, while cord plasma with maternal infection 2 weeks before delivery exhibited the emergence of anti-inflammatory cytokines. Our data support vertical transmission of protective SARS-CoV-2-specific antibodies. This vertical antibody transmission and the presence of anti-inflammatory cytokines in cord blood may offset adverse outcomes of inflammation in exposed newborns.
Collapse
Affiliation(s)
| | | | - Sohan Punia
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | | | - Yaroslav Fedyshyn
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | - James H Girsch
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
- Graduate School of Biomedical Sciences
| | | | - Maureen Lemens
- Division of Obstetrics, Department of Obstetrics and Gynecology
| | - Dawn Littlefield
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | - Supriya Behl
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | - Elise Sintim-Aboagye
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | - Maria C Mejia Plazas
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| | | | | | - Akhilesh Pandey
- Division of Clinical Biochemistry and Immunology, Department of Laboratory Medicine and Pathology, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Bangalore
- Department of Community Medicine, Manipal Academy of Higher Education, Manipal, India
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics
| | - Robert Vassallo
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Department of Immunology, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota
| | - Jie Sun
- Thoracic Diseases Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Department of Immunology, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota
- Carter Immunology Center, School of Medicine, University of Virginia, Charlottesville
| | - Erica L Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia
| | | | - Elitza S Theel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology
| | - Andrew D Badley
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Regan N Theiler
- Division of Obstetrics, Department of Obstetrics and Gynecology
| | - Rana Chakraborty
- Children Research Center, Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine
| |
Collapse
|
15
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging Maternal and Cord Antibody Functions From SARS-CoV-2 Infection and Vaccination in Pregnancy. J Infect Dis 2024; 229:462-472. [PMID: 37815524 PMCID: PMC10873180 DOI: 10.1093/infdis/jiad421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/27/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023] Open
Abstract
Maternal immunity impacts the infant, but how is unclear. To understand the implications of the immune exposures of vaccination and infection in pregnancy for neonatal immunity, we evaluated antibody functions in paired peripheral maternal and cord blood. We compared those who in pregnancy received mRNA coronavirus disease 2019 (COVID-19) vaccine, were infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the combination. We found that vaccination enriched a subset of neutralizing activities and Fc effector functions that was driven by IgG1 and was minimally impacted by antibody glycosylation in maternal blood. In paired cord blood, maternal vaccination also enhanced IgG1. However, Fc effector functions compared to neutralizing activities were preferentially transferred. Moreover, changes in IgG posttranslational glycosylation contributed more to cord than peripheral maternal blood antibody functional potency. These differences were enhanced with the combination of vaccination and infection as compared to either alone. Thus, Fc effector functions and antibody glycosylation highlight underexplored maternal opportunities to safeguard newborns.
Collapse
Affiliation(s)
- Emily H Adhikari
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health, Dallas Texas, USA
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ye Jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ann R McDonald
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica E Pruszynski
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy A Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Savannah K McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Fikadu G Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Lenette L Lu
- Parkland Health, Dallas Texas, USA
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
Reinig S, Shih SR. Non-neutralizing functions in anti-SARS-CoV-2 IgG antibodies. Biomed J 2024; 47:100666. [PMID: 37778697 PMCID: PMC10825350 DOI: 10.1016/j.bj.2023.100666] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Most individuals infected with or vaccinated against COVID-19 develop antigenic neutralizing immunoglobulin G (IgG) antibodies against the SARS-CoV-2 spike protein. Although neutralizing antibodies are biomarkers of the adaptive immune response, their mere presence is insufficient to explain the protection afforded against the disease or its pathology. IgG exhibits other secondary effector functions that activate innate immune components, including complement, natural killer cells, and macrophages. The affinity for effector cells depends on the isotypes and glycosylation of IgG antibodies. The anti-spike IgG titer should be sufficient to provide significant Fc-mediated effects in severe COVID-19, mRNA, and protein subunit vaccinations. In combination with aberrant effector cells, pro-inflammatory afucosylated IgG1 and IgG3 may be detrimental in severe COVID-19. The antibody response of mRNA vaccines leads to higher fucosylation and a less inflammatory IgG profile, with a long-term shift to IgG4, which is correlated with protection from disease.
Collapse
Affiliation(s)
- Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
17
|
Lauritsen CJ, Trinh IV, Desai SP, Clancey E, Murrell AE, Rambaran S, Chandra S, Elliott DH, Smira AR, Mo Z, Stone AE, Agbodji A, Dugas CM, Satou R, Pridjian G, Longo S, Ley SH, Robinson JE, Norton EB, Piedimonte G, Gunn BM. Passive antibody transfer from pregnant women to their fetus are maximized after SARS-CoV-2 vaccination irrespective of prior infection. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100189. [PMID: 38268538 PMCID: PMC10805668 DOI: 10.1016/j.jacig.2023.100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/19/2023] [Accepted: 08/14/2023] [Indexed: 01/26/2024]
Abstract
Background Pregnancy is associated with a higher risk of adverse symptoms and outcomes for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection for both mother and neonate. Antibodies can provide protection against SARS-CoV-2 infection and are induced in pregnant women after vaccination or infection. Passive transfer of these antibodies from mother to fetus in utero may provide protection to the neonate against infection. However, it is unclear whether the magnitude or quality and kinetics of maternally derived fetal antibodies differs in the context of maternal infection or vaccination. Objective We aimed to determine whether antibodies transferred from maternal to fetus differed in quality or quantity between infection- or vaccination-induced humoral immune responses. Methods We evaluated 93 paired maternal and neonatal umbilical cord blood plasma samples collected between October 2020 and February 2022 from a birth cohort of pregnant women from New Orleans, Louisiana, with histories of SARS-CoV-2 infection and/or vaccination. Plasma was profiled for the levels of spike-specific antibodies and induction of antiviral humoral immune functions, including neutralization and Fc-mediated innate immune effector functions. Responses were compared between 4 groups according to maternal infection and vaccination. Results We found that SARS-CoV-2 vaccination or infection during pregnancy increased the levels of antiviral antibodies compared to naive subjects. Vaccinated mothers and cord samples had the highest anti-spike antibody levels and antiviral function independent of the time of vaccination during pregnancy. Conclusions These results show that the most effective passive transfer of functional antibodies against SARS-CoV-2 in utero is achieved through vaccination, highlighting the importance of vaccination in pregnant women.
Collapse
Affiliation(s)
- Cody J. Lauritsen
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Ivy V. Trinh
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Srushti P. Desai
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Erin Clancey
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Amelie E. Murrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Saraswatie Rambaran
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Sruti Chandra
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Debra H. Elliott
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Ashley R. Smira
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Zhiyin Mo
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Addison E. Stone
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Ayitevi Agbodji
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Courtney M. Dugas
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, La
| | - Gabriella Pridjian
- Department of Obstetrics and Gynecology, Tulane University School of Medicine, New Orleans, La
| | | | - Sylvia H. Ley
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, La
| | - Giovanni Piedimonte
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| |
Collapse
|
18
|
Mayer L, Weskamm LM, Fathi A, Kono M, Heidepriem J, Krähling V, Mellinghoff SC, Ly ML, Friedrich M, Hardtke S, Borregaard S, Hesterkamp T, Loeffler FF, Volz A, Sutter G, Becker S, Dahlke C, Addo MM. MVA-based vaccine candidates encoding the native or prefusion-stabilized SARS-CoV-2 spike reveal differential immunogenicity in humans. NPJ Vaccines 2024; 9:20. [PMID: 38278816 PMCID: PMC10817990 DOI: 10.1038/s41541-023-00801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 01/28/2024] Open
Abstract
In response to the COVID-19 pandemic, multiple vaccines were developed using platforms such as viral vectors and mRNA technology. Here, we report humoral and cellular immunogenicity data from human phase 1 clinical trials investigating two recombinant Modified Vaccinia virus Ankara vaccine candidates, MVA-SARS-2-S and MVA-SARS-2-ST, encoding the native and the prefusion-stabilized SARS-CoV-2 spike protein, respectively. MVA-SARS-2-ST was more immunogenic than MVA-SARS-2-S, but both were less immunogenic compared to licensed mRNA- and ChAd-based vaccines in SARS-CoV-2 naïve individuals. In heterologous vaccination, previous MVA-SARS-2-S vaccination enhanced T cell functionality and MVA-SARS-2-ST boosted the frequency of T cells and S1-specific IgG levels when used as a third vaccination. While the vaccine candidate containing the prefusion-stabilized spike elicited predominantly S1-specific responses, immunity to the candidate with the native spike was skewed towards S2-specific responses. These data demonstrate how the spike antigen conformation, using the same viral vector, directly affects vaccine immunogenicity in humans.
Collapse
Affiliation(s)
- Leonie Mayer
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| | - Leonie M Weskamm
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Anahita Fathi
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- First Department of Medicine, Division of Infectious Diseases, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Maya Kono
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Jasmin Heidepriem
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Verena Krähling
- Institute for Virology, Philipps University Marburg, Marburg, Germany
- German Centre for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Sibylle C Mellinghoff
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Centre for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), German CLL Group (GCLLSG), University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - My Linh Ly
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Monika Friedrich
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Svenja Hardtke
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | | | - Thomas Hesterkamp
- German Centre for Infection Research, Translational Project Management Office, Brunswick, Germany
| | - Felix F Loeffler
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
- German Centre for Infection Research, Partner Site Hannover-Brunswick, Hanover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Institute for Infectious Diseases and Zoonoses, LMU Munich, Munich, Germany
- German Centre for Infection Research, Partner Site Munich, Munich, Germany
| | - Stephan Becker
- Institute for Virology, Philipps University Marburg, Marburg, Germany
- German Centre for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Christine Dahlke
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
19
|
Balinsky CA, Jiang L, Jani V, Cheng Y, Zhang Z, Belinskaya T, Qiu Q, Long TK, Schilling MA, Jenkins SA, Corson KS, Martin NJ, Letizia AG, Hontz RD, Sun P. Antibodies to S2 domain of SARS-CoV-2 spike protein in Moderna mRNA vaccinated subjects sustain antibody-dependent NK cell-mediated cell cytotoxicity against Omicron BA.1. Front Immunol 2023; 14:1266829. [PMID: 38077368 PMCID: PMC10702584 DOI: 10.3389/fimmu.2023.1266829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
Vaccination with the primary two-dose series of SARS-CoV-2 mRNA protects against infection with the ancestral strain, and limits the presentation of severe disease after re-infection by multiple variants of concern (VOC), including Omicron, despite the lack of a strong neutralizing response to these variants. We compared antibody responses in serum samples collected from mRNA-1273 (Moderna) vaccinated subjects to identify mechanisms of immune escape and cross-protection. Using pseudovirus constructs containing domain-specific amino acid changes representative of Omicron BA.1, combined with domain competition and RBD-antibody depletion, we showed that RBD antibodies were primarily responsible for virus neutralization and variant escape. Antibodies to NTD played a less significant role in antibody neutralization but acted along with RBD to enhance neutralization. S2 of Omicron BA.1 had no impact on neutralization escape, suggesting it is a less critical domain for antibody neutralization; however, it was as capable as S1 at eliciting IgG3 responses and NK-cell mediated, antibody-dependent cell cytotoxicity (ADCC). Antibody neutralization and ADCC activities to RBD, NTD, and S1 were all prone to BA.1 escape. In contrast, ADCC activities to S2 resisted BA.1 escape. In conclusion, S2 antibodies showed potent ADCC function and resisted Omicron BA.1 escape, suggesting that S2 contributes to cross-protection against Omicron BA.1. In line with its conserved nature, S2 may hold promise as a vaccine target against future variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Corey A. Balinsky
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Le Jiang
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Vihasi Jani
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | | | - Zhiwen Zhang
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Tatyana Belinskaya
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Qi Qiu
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | | | - Megan A. Schilling
- Virology and Emerging Infectious Department, U.S. Naval Medical Research Unit SOUTH, Lima, Peru
| | - Sarah A. Jenkins
- Diagnostics and Surveillance Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Karen S. Corson
- US Naval Medical Research Unit-INDO PACIFIC, Singapore, Singapore
| | | | | | - Robert D. Hontz
- US Naval Medical Research Unit-INDO PACIFIC, Singapore, Singapore
| | - Peifang Sun
- Diagnostics and Surveillance Department, Naval Medical Research Command, Silver Spring, MD, United States
| |
Collapse
|
20
|
Capuano C, De Federicis D, Ciuti D, Turriziani O, Angeloni A, Anastasi E, Giannini G, Belardinilli F, Molfetta R, Alvaro D, Palmieri G, Galandrini R. Impact of SARS-CoV-2 vaccination on FcγRIIIA/CD16 dynamics in Natural Killer cells: relevance for antibody-dependent functions. Front Immunol 2023; 14:1285203. [PMID: 38045702 PMCID: PMC10693335 DOI: 10.3389/fimmu.2023.1285203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Natural Killer (NK) cells contribute to the protective effects of vaccine-induced antibodies thanks to the low affinity receptor for IgG, FcγRIIIA/CD16, whose aggregation leads to the killing of infected cells and IFNγ release, through which they potentiate adaptive immune responses. Methods Forty-seven healthy young individuals undergoing either homologous (ChAdOx1-S/ChAdOx1-S) or heterologous (ChAdOx1-S/BNT162B2) SARS-CoV-2 vaccination settings were recruited. Peripheral blood samples were collected immediately prior to vaccination and 8 weeks after the booster dose. The phenotypic and functional profile of NK cells was evaluated by flow cytometry at both time points. Serum samples were tested to evaluate circulating anti-Spike IgG levels and cytomegalovirus serostatus. CD16 F158V polymorphism was assessed by sequencing analysis. Results The downregulation of CD16 and the selective impairment of antibody-dependent cytotoxicity and IFNγ production in CD56dim NK population, persisting 8 weeks after boosting, were observed in heterologous, but not in homologous SARS-CoV-2 vaccination scheme. While the magnitude of CD16-dependent functions of the global CD56dim pool correlated with receptor levels before and after vaccination, the responsivity of NKG2C+ subset, that displays amplified size and functionality in HCMV+ individuals, resulted intrinsically insensitive to CD16 levels. Individual CD16 responsiveness was also affected by CD16F158V polymorphism; F/F low affinity individuals, characterized by reduced CD16 levels and functions independently of vaccination, did not show post-vaccinal functional impairment with respect to intermediate and high affinity ones, despite a comparable CD16 downregulation. Further, CD16 high affinity ligation conditions by means of afucosylated mAb overcame vaccine-induced and genotype-dependent functional defects. Finally, the preservation of CD16 expression directly correlated with anti-Spike IgG titer, hinting that the individual magnitude of receptor-dependent functions may contribute to the amplification of the vaccinal response. Conclusion This study demonstrates a durable downmodulation of CD16 levels and Ab-dependent NK functions after SARS-CoV-2 heterologous vaccination, and highlights the impact of genetic and environmental host-related factors in modulating NK cell susceptibility to post-vaccinal Fc-dependent functional impairment.
Collapse
Affiliation(s)
- Cristina Capuano
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Davide De Federicis
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniel Ciuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Gabriella Palmieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
21
|
Van Coillie J, Pongracz T, Šuštić T, Wang W, Nouta J, Le Gars M, Keijzer S, Linty F, Cristianawati O, Keijser JB, Visser R, van Vught LA, Slim MA, van Mourik N, Smit MJ, Sander A, Schmidt DE, Steenhuis M, Rispens T, Nielsen MA, Mordmüller BG, Vlaar AP, Ellen van der Schoot C, Roozendaal R, Wuhrer M, Vidarsson G. Comparative analysis of spike-specific IgG Fc glycoprofiles elicited by adenoviral, mRNA, and protein-based SARS-CoV-2 vaccines. iScience 2023; 26:107619. [PMID: 37670790 PMCID: PMC10475480 DOI: 10.1016/j.isci.2023.107619] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
IgG antibodies are important mediators of vaccine-induced immunity through complement- and Fc receptor-dependent effector functions. Both are influenced by the composition of the conserved N-linked glycan located in the IgG Fc domain. Here, we compared the anti-Spike (S) IgG1 Fc glycosylation profiles in response to mRNA, adenoviral, and protein-based COVID-19 vaccines by mass spectrometry (MS). All vaccines induced a transient increase of antigen-specific IgG1 Fc galactosylation and sialylation. An initial, transient increase of afucosylated IgG was induced by membrane-encoding S protein formulations. A fucose-sensitive ELISA for antigen-specific IgG (FEASI) exploiting FcγRIIIa affinity for afucosylated IgG was used as an orthogonal method to confirm the LC-MS-based afucosylation readout. Our data suggest that vaccine-induced anti-S IgG glycosylation is dynamic, and although variation is seen between different vaccine platforms and individuals, the evolution of glycosylation patterns display marked overlaps.
Collapse
Affiliation(s)
- Julie Van Coillie
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Tonći Šuštić
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Sofie Keijzer
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Federica Linty
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Olvi Cristianawati
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Jim B.D. Keijser
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Remco Visser
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
- Department of Intensive Care, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marleen A. Slim
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
- Department of Intensive Care, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Niels van Mourik
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
- Department of Intensive Care, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Merel J. Smit
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Adam Sander
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- AdaptVac Aps, Copenhagen, Denmark
| | - David E. Schmidt
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Morten A. Nielsen
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin G. Mordmüller
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alexander P.J. Vlaar
- Department of Intensive Care, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Laboratory of Experimental Intensive Care and Anaesthesiology, L.E.I.C.A., Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
22
|
St. Germain R, Bossard EL, Corey L, Sholukh AM. Serum concentration of antigen-specific IgG can substantially bias interpretation of antibody-dependent phagocytosis assay readout. iScience 2023; 26:107527. [PMID: 37664583 PMCID: PMC10469534 DOI: 10.1016/j.isci.2023.107527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/21/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
Because virus neutralization cannot solely explain vaccine-induced, antibody-mediated protection, antibody effector functions are being considered as a potential correlate of protection (CoP). However, measuring effector functions at a fixed serum dilution for high throughput purposes makes it difficult to distinguish between the effect of serum antibody concentration and antibody properties such as epitopes, subclass, and glycosylation. To address this issue, we evaluated antibody-dependent cellular phagocytosis (ADCP) assay against SARS-CoV-2 spike. Adjustment of serum samples to the same concentration of antigen-specific IgG prior to the ADCP assay revealed concentration-independent differences in ADCP after mRNA vaccination in subjects with and without prior SARS-CoV-2 infection not detectable in assay performed with fixed serum dilution. Phagocytosis measured at different concentrations of spike-specific IgG strongly correlated with the area under the curve (AUC) indicating that ADCP assay can be performed at a standardized antibody concentration for the high throughput necessary for vaccine trial analyses.
Collapse
Affiliation(s)
- Russell St. Germain
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Emily L. Bossard
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence Corey
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Anton M. Sholukh
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
23
|
Xue VW, Wong SCC, Li B, Cho WCS. The discovery and development of mRNA vaccines for the prevention of SARS-CoV-2 infection. Expert Opin Drug Discov 2023; 18:769-780. [PMID: 37237360 DOI: 10.1080/17460441.2023.2218083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
INTRODUCTION COVID-19 pandemic is one of the most serious public health events of this century. There have been more than 670 million confirmed cases and more than 6 million deaths worldwide. From the emergence of the Alpha variant to the later rampant Omicron variant, the high transmissibility and pathogenicity of SARS-CoV-2 accelerate the research and development of effective vaccines. Against this background, mRNA vaccines stepped onto the historical stage and became an important tool for COVID-19 prevention. AREAS COVERED This article introduces the characteristics of different mRNA vaccines in the prevention of COVID-19, including antigen selection, therapeutic mRNA design and modification, and different delivery systems of mRNA molecules. It also summarizes and discusses the mechanisms, safety, effectiveness, side effects, and limitations of current COVID-19 mRNA vaccines. EXPERT OPINION Therapeutic mRNA molecules have plenty of advantages, including flexible design, rapid production, sufficient immune activation, safety without the risk of genome insertion in the host cells, and no viral vectors or particles involved, making them an important tool to fight diseases in the future. However, the application of COVID-19 mRNA vaccines also faces many challenges, such as storage and transportation, mass production, and nonspecific immunity.
Collapse
Affiliation(s)
- Vivian Weiwen Xue
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, China
| | - Sze Chuen Cesar Wong
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Bo Li
- School of Textile Science and Engineering, Xi'an Polytechnic University, Xi'an, China
| | | |
Collapse
|
24
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging maternal and infant cord antibody functions from SARS-CoV-2 infection and vaccination in pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538955. [PMID: 37205338 PMCID: PMC10187183 DOI: 10.1101/2023.05.01.538955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunization in pregnancy is a critical tool that can be leveraged to protect the infant with an immature immune system but how vaccine-induced antibodies transfer to the placenta and protect the maternal-fetal dyad remains unclear. Here, we compare matched maternal-infant cord blood from individuals who in pregnancy received mRNA COVID-19 vaccine, were infected by SARS-CoV-2, or had the combination of these two immune exposures. We find that some but not all antibody neutralizing activities and Fc effector functions are enriched with vaccination compared to infection. Preferential transport to the fetus of Fc functions and not neutralization is observed. Immunization compared to infection enriches IgG1-mediated antibody functions with changes in antibody post-translational sialylation and fucosylation that impact fetal more than maternal antibody functional potency. Thus, vaccine enhanced antibody functional magnitude, potency and breadth in the fetus are driven more by antibody glycosylation and Fc effector functions compared to maternal responses, highlighting prenatal opportunities to safeguard newborns as SARS-CoV-2 becomes endemic.
Collapse
Affiliation(s)
- Emily H. Adhikari
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
- Parkland Health, Dallas TX
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ye jin Kang
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ann R. McDonald
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Jessica E. Pruszynski
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
| | - Timothy A. Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Savannah K. McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Fikadu G. Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Lenette L. Lu
- Parkland Health, Dallas TX
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
- Department of Immunology, UTSW Medical Center, Dallas, TX
| |
Collapse
|
26
|
Zelek WM, Harrison RA. Complement and COVID-19: Three years on, what we know, what we don't know, and what we ought to know. Immunobiology 2023; 228:152393. [PMID: 37187043 PMCID: PMC10174470 DOI: 10.1016/j.imbio.2023.152393] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus was identified in China in 2019 as the causative agent of COVID-19, and quickly spread throughout the world, causing over 7 million deaths, of which 2 million occurred prior to the introduction of the first vaccine. In the following discussion, while recognising that complement is just one of many players in COVID-19, we focus on the relationship between complement and COVID-19 disease, with limited digression into directly-related areas such as the relationship between complement, kinin release, and coagulation. Prior to the 2019 COVID-19 outbreak, an important role for complement in coronavirus diseases had been established. Subsequently, multiple investigations of patients with COVID-19 confirmed that complement dysregulation is likely to be a major driver of disease pathology, in some, if not all, patients. These data fuelled evaluation of many complement-directed therapeutic agents in small patient cohorts, with claims of significant beneficial effect. As yet, these early results have not been reflected in larger clinical trials, posing questions such as who to treat, appropriate time to treat, duration of treatment, and optimal target for treatment. While significant control of the pandemic has been achieved through a global scientific and medical effort to comprehend the etiology of the disease, through extensive SARS-CoV-2 testing and quarantine measures, through vaccine development, and through improved therapy, possibly aided by attenuation of the dominant strains, it is not yet over. In this review, we summarise complement-relevant literature, emphasise its main conclusions, and formulate a hypothesis for complement involvement in COVID-19. Based on this we make suggestions as to how any future outbreak might be better managed in order to minimise impact on patients.
Collapse
Affiliation(s)
- Wioleta M Zelek
- Dementia Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
27
|
Wang J, Mai X, He Y, Zhu C, Zhou D. IgG1-Dominant Antibody Response Induced by Recombinant Trimeric SARS-CoV-2 Spike Protein with PIKA Adjuvant. Vaccines (Basel) 2023; 11:vaccines11040827. [PMID: 37112739 PMCID: PMC10144704 DOI: 10.3390/vaccines11040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Recombinant trimeric SARS-CoV-2 Spike protein with PIKA (polyI:C) adjuvant induces potent and durable neutralizing antibodies that protect against multiple SARS-CoV-2 variants. The immunoglobulin subclasses of viral-specific antibodies remain unknown, as do their glycosylation status on Fc regions. In this study, we analyzed immunoglobulins adsorbed by plate-bound recombinant trimeric SARS-CoV-2 Spike protein from serum of Cynomolgus monkey immunized by recombinant trimeric SARS-CoV-2 Spike protein with PIKA (polyI:C) adjuvant. The results showed that IgG1 was the dominant IgG subclass as revealed by ion mobility mass spectrometry. The average percentage of Spike protein-specific IgG1 increased to 88.3% as compared to pre-immunization. Core fucosylation for Fc glycopeptide of Spike protein-specific IgG1 was found to be higher than 98%. These results indicate that a unique Th1-biased, IgG1-dominant antibody response was responsible for the effectiveness of PIKA (polyI:C) adjuvant. Vaccine-induced core-fucosylation of IgG1 Fc region may reduce incidence of severe COVID-19 disease associated with overstimulation of FCGR3A by afucosylated IgG1.
Collapse
Affiliation(s)
- Jingxia Wang
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Xinjia Mai
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Yu He
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Chenxi Zhu
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| | - Dapeng Zhou
- Department of Immunology and Pathogen Biology, Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, China
| |
Collapse
|
28
|
Poolchanuan P, Matsee W, Sengyee S, Siripoon T, Dulsuk A, Phunpang R, Pisutsan P, Piyaphanee W, Luvira V, Chantratita N. Dynamics of Different Classes and Subclasses of Antibody Responses to Severe Acute Respiratory Syndrome Coronavirus 2 Variants after Coronavirus Disease 2019 and CoronaVac Vaccination in Thailand. mSphere 2023; 8:e0046522. [PMID: 36688637 PMCID: PMC9942573 DOI: 10.1128/msphere.00465-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023] Open
Abstract
The humoral immune response plays a key role in protecting the population from SARS-CoV-2 transmission. Patients who recovered from COVID-19 as well as fully vaccinated individuals have elevated levels of antibodies. The dynamic levels of the classes and subclasses of antibody responses to new variants that occur in different populations remain unclear. We prospectively recruited 60 participants, including COVID-19 patients and CoronaVac-vaccinated individuals, in Thailand from May to August 2021. Plasma samples were collected on day 0, day 14, and day 28 to determine the dynamic levels of the classes and subclasses of plasma antibodies against the receptor-binding domain (RBD) in the spike protein (S) of four SARS-CoV-2 strains (Wuhan, Alpha, Delta, and Omicron) via enzyme-linked immunosorbent assay. Our results indicated that the patients with SARS-CoV-2 infections had broader class and subclass profiles as well as higher levels of anti-S RBD antibodies to the Wuhan, Alpha, and Delta strains than did the CoronaVac-vaccinated individuals. The median antibody levels increased and subsequently declined in a month in the COVID-19 patients and in the vaccinated group. Correlations of the classes and subclasses of antibodies were observed in the COVID-19 patients but not in the vaccinated individuals. The levels of all of the anti-S RBD antibodies against the Omicron variant were low in the patients and in the vaccinated individuals. Our study revealed distinct antibody profiles between the two cohorts, suggesting different pathways of immune activation. This could have an impact on protection from infections by new variants of concern (VOC). IMPORTANCE The antibody responses to new SARS-CoV-2 variants that occur in different populations remain unclear. In this study, we recruited 60 participants, including COVID-19 patients and CoronaVac-vaccinated individuals, in Thailand and determined the dynamic levels of the IgG, IgA, IgM, and IgG subclasses of antibodies against the spike protein (S) of four SARS-CoV-2 strains. Our results showed that the patients with SARS-CoV-2 infections had broader profiles and higher levels of antibodies to the Wuhan, Alpha, and Delta strains than did the CoronaVac-vaccinated individuals. The antibody levels of both groups increased and subsequently decreased within 1 month. Higher and functional correlations of these antibodies were observed in the COVID-19 patients. The levels of all anti-S RBD antibodies against the Omicron variant were low in patients and vaccinated individuals. Our study revealed distinct antibody responses between the two groups, suggesting different pathways of immune response, which may have an impact on protection from infections by new SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Prapassorn Poolchanuan
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wasin Matsee
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sineenart Sengyee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tanaya Siripoon
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Adul Dulsuk
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rungnapa Phunpang
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phimphan Pisutsan
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Watcharapong Piyaphanee
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Irrgang P, Gerling J, Kocher K, Lapuente D, Steininger P, Habenicht K, Wytopil M, Beileke S, Schäfer S, Zhong J, Ssebyatika G, Krey T, Falcone V, Schülein C, Peter AS, Nganou-Makamdop K, Hengel H, Held J, Bogdan C, Überla K, Schober K, Winkler TH, Tenbusch M. Class switch toward noninflammatory, spike-specific IgG4 antibodies after repeated SARS-CoV-2 mRNA vaccination. Sci Immunol 2023; 8:eade2798. [PMID: 36548397 PMCID: PMC9847566 DOI: 10.1126/sciimmunol.ade2798] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RNA vaccines are efficient preventive measures to combat the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. High levels of neutralizing SARS-CoV-2 antibodies are an important component of vaccine-induced immunity. Shortly after the initial two mRNA vaccine doses, the immunoglobulin G (IgG) response mainly consists of the proinflammatory subclasses IgG1 and IgG3. Here, we report that several months after the second vaccination, SARS-CoV-2-specific antibodies were increasingly composed of noninflammatory IgG4, which were further boosted by a third mRNA vaccination and/or SARS-CoV-2 variant breakthrough infections. IgG4 antibodies among all spike-specific IgG antibodies rose, on average, from 0.04% shortly after the second vaccination to 19.27% late after the third vaccination. This induction of IgG4 antibodies was not observed after homologous or heterologous SARS-CoV-2 vaccination with adenoviral vectors. Single-cell sequencing and flow cytometry revealed substantial frequencies of IgG4-switched B cells within the spike-binding memory B cell population [median of 14.4%; interquartile range (IQR) of 6.7 to 18.1%] compared with the overall memory B cell repertoire (median of 1.3%; IQR of 0.9 to 2.2%) after three immunizations. This class switch was associated with a reduced capacity of the spike-specific antibodies to mediate antibody-dependent cellular phagocytosis and complement deposition. Because Fc-mediated effector functions are critical for antiviral immunity, these findings may have consequences for the choice and timing of vaccination regimens using mRNA vaccines, including future booster immunizations against SARS-CoV-2.
Collapse
Affiliation(s)
- Pascal Irrgang
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Juliane Gerling
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Katharina Kocher
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Philipp Steininger
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Katharina Habenicht
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Monika Wytopil
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Stephanie Beileke
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Simon Schäfer
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - Jahn Zhong
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany
| | - George Ssebyatika
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck; Luebeck, Germany
| | - Thomas Krey
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck; Luebeck, Germany
| | - Valeria Falcone
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg; Freiburg, Germany
| | - Christine Schülein
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Antonia Sophia Peter
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany
| | - Krystelle Nganou-Makamdop
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Hartmut Hengel
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg; Freiburg, Germany
| | - Jürgen Held
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Klaus Überla
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Wasserturmstr. 3/5, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| | - Thomas H. Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU); Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg; Schlossgarten 4, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054 Erlangen, Germany.,Corresponding author. (K.S.); (T.H.W.); (M.T.)
| |
Collapse
|
30
|
Buhre JS, Pongracz T, Künsting I, Lixenfeld AS, Wang W, Nouta J, Lehrian S, Schmelter F, Lunding HB, Dühring L, Kern C, Petry J, Martin EL, Föh B, Steinhaus M, von Kopylow V, Sina C, Graf T, Rahmöller J, Wuhrer M, Ehlers M. mRNA vaccines against SARS-CoV-2 induce comparably low long-term IgG Fc galactosylation and sialylation levels but increasing long-term IgG4 responses compared to an adenovirus-based vaccine. Front Immunol 2023; 13:1020844. [PMID: 36713457 PMCID: PMC9877300 DOI: 10.3389/fimmu.2022.1020844] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/09/2022] [Indexed: 01/15/2023] Open
Abstract
Background The new types of mRNA-containing lipid nanoparticle vaccines BNT162b2 and mRNA-1273 and the adenovirus-based vaccine AZD1222 were developed against SARS-CoV-2 and code for its spike (S) protein. Several studies have investigated short-term antibody (Ab) responses after vaccination. Objective However, the impact of these new vaccine formats with unclear effects on the long-term Ab response - including isotype, subclass, and their type of Fc glycosylation - is less explored. Methods Here, we analyzed anti-S Ab responses in blood serum and the saliva of SARS-CoV-2 naïve and non-hospitalized pre-infected subjects upon two vaccinations with different mRNA- and adenovirus-based vaccine combinations up to day 270. Results We show that the initially high mRNA vaccine-induced blood and salivary anti-S IgG levels, particularly IgG1, markedly decrease over time and approach the lower levels induced with the adenovirus-based vaccine. All three vaccines induced, contrary to the short-term anti-S IgG1 response with high sialylation and galactosylation levels, a long-term anti-S IgG1 response that was characterized by low sialylation and galactosylation with the latter being even below the corresponding total IgG1 galactosylation level. Instead, the mRNA, but not the adenovirus-based vaccines induced long-term IgG4 responses - the IgG subclass with inhibitory effector functions. Furthermore, salivary anti-S IgA levels were lower and decreased faster in naïve as compared to pre-infected vaccinees. Predictively, age correlated with lower long-term anti-S IgG titers for the mRNA vaccines. Furthermore, higher total IgG1 galactosylation, sialylation, and bisection levels correlated with higher long-term anti-S IgG1 sialylation, galactosylation, and bisection levels, respectively, for all vaccine combinations. Conclusion In summary, the study suggests a comparable "adjuvant" potential of the newly developed vaccines on the anti-S IgG Fc glycosylation, as reflected in relatively low long-term anti-S IgG1 galactosylation levels generated by the long-lived plasma cell pool, whose induction might be driven by a recently described TH1-driven B cell response for all three vaccines. Instead, repeated immunization of naïve individuals with the mRNA vaccines increased the proportion of the IgG4 subclass over time which might influence the long-term Ab effector functions. Taken together, these data shed light on these novel vaccine formats and might have potential implications for their long-term efficacy.
Collapse
Affiliation(s)
- Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Inga Künsting
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Anne S. Lixenfeld
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Selina Lehrian
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Franziska Schmelter
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hanna B. Lunding
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Lara Dühring
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Carsten Kern
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Janina Petry
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Emily L. Martin
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Bandik Föh
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Moritz Steinhaus
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Vera von Kopylow
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Tobias Graf
- Medical Department 2, University Heart Center of Schleswig-Holstein, Lübeck, Germany
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Manfred Wuhrer, ; Marc Ehlers,
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany,Airway Research Center North (ARCN), University of Lübeck, German Center for Lung Research (DZL), Lübeck, Germany,*Correspondence: Manfred Wuhrer, ; Marc Ehlers,
| |
Collapse
|
31
|
Yu P, Liu Z, Zhu Z, Yang J, Deng M, Chen M, Lai C, Kong W, Xiong S, Wan L, Mai W, Chen L, Lei Y, Khan SA, Ruan J, Kang A, Guo X, Zhou Q, Li W, Chen Z, Liang Y, Li P, Zhang L, Ji T. Omicron variants breakthrough infection elicited higher specific memory immunity than third dose booster in healthy vaccinees. Virol Sin 2023; 38:233-243. [PMID: 36603767 PMCID: PMC10176432 DOI: 10.1016/j.virs.2022.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Homologous booster, heterologous booster, and Omicron variants breakthrough infection (OBI) could improve the humoral immunity against Omicron variants. Questions concerning about memory B cells (MBCs) and T cells immunity against Omicron variants, features of long-term immunity, after booster and OBI, needs to be explored. Here, comparative analysis demonstrate antibody and T cell immunity against ancestral strain, Delta and Omicron variants in Omicron breakthrough infected patients (OBIPs) are comparable to that in Ad5-nCoV boosted healthy volunteers (HVs), higher than that in inactivated vaccine (InV) boosted HVs. However, memory B cells (MBCs) immunity against Omicron variants was highest in OBIPs, followed by Ad5-nCoV boosted and InV boosted HVs. OBIPs and Ad5-nCoV boosted HVs have higher classical MBCs and activated MBCs, and lower naïve MBCs and atypical MBCs relative to both vaccine boosted HVs. Collectively, these data indicate Omicron breakthrough infection elicit higher MBCs and T cells against SARS-CoV-2 especially Omicron variants relative to homologous InV booster and heterologous Ad5-nCoV booster.
Collapse
Affiliation(s)
- Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Zijian Liu
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China
| | - Zhuoqi Zhu
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Jiaqing Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shilong Xiong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Li Wan
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shahzad Akbar Khan
- Laboratory of Pathology, Department of Pathobiology, University of Poonch Rawalakot Azad Kashmir Pakistan 12350, Pakistan
| | - Jianfeng Ruan
- Hospital Infection-Control Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - An Kang
- Medical Examination Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xuguang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Qiang Zhou
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Zheng Chen
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| | - Yuemei Liang
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China.
| | - Pingchao Li
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China.
| | - Lei Zhang
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Department of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, 510630, China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
32
|
Brook B, Fatou B, Kumar Checkervarty A, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The mRNA vaccine BNT162b2 demonstrates impaired T H1 immunogenicity in human elders in vitro and aged mice in vivo. RESEARCH SQUARE 2022:rs.3.rs-2395118. [PMID: 36597547 PMCID: PMC9810224 DOI: 10.21203/rs.3.rs-2395118/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
mRNA vaccines have been key to addressing the SARS-CoV-2 pandemic but have impaired immunogenicity and durability in vulnerable older populations. We evaluated the mRNA vaccine BNT162b2 in human in vitro whole blood assays with supernatants from adult (18-50 years) and elder (≥60 years) participants measured by mass spectrometry and proximity extension assay proteomics. BNT162b2 induced increased expression of soluble proteins in adult blood (e.g., C1S, PSMC6, CPN1), but demonstrated reduced proteins in elder blood (e.g., TPM4, APOF, APOC2, CPN1, and PI16), including 30-85% lower induction of TH1-polarizing cytokines and chemokines (e.g., IFNγ, and CXCL10). Elder TH1 impairment was validated in mice in vivo and associated with impaired humoral and cellular immunogenicity. Our study demonstrates the utility of a human in vitro platform to model age-specific mRNA vaccine activity, highlights impaired TH1 immunogenicity in older adults, and provides rationale for developing enhanced mRNA vaccines with greater immunogenicity in vulnerable populations.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC, Canada
| | - Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Amy C Sherman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Bambino Gesù Children's Hospital, Rome, Italy
- Chair of Pediatrics, University of Rome, Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
33
|
Beyond neutralization: Fc-dependent antibody effector functions in SARS-CoV-2 infection. Nat Rev Immunol 2022:10.1038/s41577-022-00813-1. [PMID: 36536068 PMCID: PMC9761659 DOI: 10.1038/s41577-022-00813-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Neutralizing antibodies are known to have a crucial role in protecting against SARS-CoV-2 infection and have been suggested to be a useful correlate of protection for vaccine clinical trials and for population-level surveys. In addition to neutralizing virus directly, antibodies can also engage immune effectors through their Fc domains, including Fc receptor-expressing immune cells and complement. The outcome of these interactions depends on a range of factors, including antibody isotype-Fc receptor combinations, Fc receptor-bearing cell types and antibody post-translational modifications. A growing body of evidence has shown roles for these Fc-dependent antibody effector functions in determining the outcome of SARS-CoV-2 infection. However, measuring these functions is more complicated than assays that measure antibody binding and virus neutralization. Here, we examine recent data illuminating the roles of Fc-dependent antibody effector functions in the context of SARS-CoV-2 infection, and we discuss the implications of these data for the development of next-generation SARS-CoV-2 vaccines and therapeutics.
Collapse
|
34
|
Anti-SARS-Cov-2 S-RBD IgG Formed after BNT162b2 Vaccination Can Bind C1q and Activate Complement. J Immunol Res 2022; 2022:7263740. [PMID: 36573216 PMCID: PMC9789906 DOI: 10.1155/2022/7263740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/19/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background The ability of vaccine-induced antibodies to bind C1q could affect pathogen neutralization. In this study, we investigated C1q binding and subsequent complement activation by anti-spike (S) protein receptor-binding domain (RBD) specific antibodies produced following vaccination with either the mRNA vaccine BNT162b2 or the inactivated vaccine BBIBP-CorV. Methods Serum samples were collected in the period of July 2021-March 2022. Participants' demographic data, type of vaccine, date of vaccination, as well as adverse effects of the vaccine were recorded. The serum samples were incubated with S protein RBD-coated plates. Levels of human IgG, IgA, IgM, C1q, and mannose-binding lectin (MBL) that were bound to the plate, as well as formed C3d, and C5b-9 were compared between different groups of participants. Results A total of 151 samples were collected from vaccinated (n = 116) and nonvaccinated (n = 35) participants. Participants who received either one or two doses of BNT162b2 formed higher levels of anti-RBD IgG and IgA than participants who received BBIBP-CorV. The anti-RBD IgG formed following either vaccine bound C1q, but significantly more C1q binding was observed in participants who received BNT162b2. Subsequently, C5b-9 formation was significantly higher in participants who received BNT162b2, while no significant difference in C5b-9 formation was found between the nonvaccinated and BBIBP-CorV groups. The formation of C5b-9 was strongly correlated to C1q binding and not to MBL binding, additionally, the ratio of formed C5b-9/bound C1q was significantly higher in the BNT162b2 group. Conclusion Anti-RBD IgG formed following vaccination can bind C1q with subsequent complement activation, and the degree of terminal complement pathway activation differed between vaccines, which could play a role in the protection offered by COVID-19 vaccines. Further investigation into the correlation between vaccine protection and vaccine-induced antibodies' ability to activate complement is required.
Collapse
|
35
|
Van Coillie J, Pongracz T, Rahmöller J, Chen HJ, Geyer CE, van Vught LA, Buhre JS, Šuštić T, van Osch TLJ, Steenhuis M, Hoepel W, Wang W, Lixenfeld AS, Nouta J, Keijzer S, Linty F, Visser R, Larsen MD, Martin EL, Künsting I, Lehrian S, von Kopylow V, Kern C, Lunding HB, de Winther M, van Mourik N, Rispens T, Graf T, Slim MA, Minnaar RP, Bomers MK, Sikkens JJ, Vlaar AP, van der Schoot CE, den Dunnen J, Wuhrer M, Ehlers M, Vidarsson G, Fatebenefratelli-Sacco Infectious Diseases Physicians group AntinoriSpinelloBassoliCinziaBestettiGiovannaCorbellinoMarioCovizziAliceLupoAngelicaMilazzoLauraSchiumaMarcoTorreAlessandro, UMC COVID-19 S3/HCW study group AppelmanBrentBeek van deDiederikBomersMarije K.Brabander deJustinBrouwerMatthijs C.BuisDavid T.P.ChekrouniNoraGils vanMarit J.Jong deMenno D.LavellAyesha H.A.Mourik vanNielsOlieSabine E.PetersEdgar J.G.ReijndersTom D.Y.SchinkelMichielSchuurmanAlex R.SikkensJonne J.SlimMarleen A.SmuldersYvo M.VlaarAlexander P.J.Vught vanLonneke A.WiersingaJoost W.. The BNT162b2 mRNA SARS-CoV-2 vaccine induces transient afucosylated IgG1 in naive but not in antigen-experienced vaccinees. EBioMedicine 2022; 87:104408. [PMID: 36529104 PMCID: PMC9756879 DOI: 10.1016/j.ebiom.2022.104408] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Afucosylated IgG1 responses have only been found against membrane-embedded epitopes, including anti-S in SARS-CoV-2 infections. These responses, intrinsically protective through enhanced FcγRIIIa binding, can also trigger exacerbated pro-inflammatory responses in severe COVID-19. We investigated if the BNT162b2 SARS-CoV-2 mRNA also induced afucosylated IgG responses. METHODS Blood from vaccinees during the first vaccination wave was collected. Liquid chromatography-Mass spectrometry (LC-MS) was used to study anti-S IgG1 Fc glycoprofiles. Responsiveness of alveolar-like macrophages to produce proinflammatory cytokines in presence of sera and antigen was tested. Antigen-specific B cells were characterized and glycosyltransferase levels were investigated by Fluorescence-Activated Cell Sorting (FACS). FINDINGS Initial transient afucosylated anti-S IgG1 responses were found in naive vaccinees, but not in antigen-experienced ones. All vaccinees had increased galactosylated and sialylated anti-S IgG1. Both naive and antigen-experienced vaccinees showed relatively low macrophage activation potential, as expected, due to the low antibody levels for naive individuals with afucosylated IgG1, and low afucosylation levels for antigen-experienced individuals with high levels of anti-S. Afucosylation levels correlated with FUT8 expression in antigen-specific plasma cells in naive individuals. Interestingly, low fucosylation of anti-S IgG1 upon seroconversion correlated with high anti-S IgG levels after the second dose. INTERPRETATION Here, we show that BNT162b2 mRNA vaccination induces transient afucosylated anti-S IgG1 responses in naive individuals. This observation warrants further studies to elucidate the clinical context in which potent afucosylated responses would be preferred. FUNDING LSBR1721, 1908; ZonMW10430012010021, 09150161910033, 10430012010008; DFG398859914, 400912066, 390884018; PMI; DOI4-Nr. 3; H2020-MSCA-ITN 721815.
Collapse
Affiliation(s)
- Julie Van Coillie
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Johann Rahmöller
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany,Department of Anesthesiology and Intensive Care, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Chiara Elisabeth Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Jana Sophia Buhre
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Tonći Šuštić
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Thijs Luc Junior van Osch
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Maurice Steenhuis
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Willianne Hoepel
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands,Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Amsterdam Rheumatology and Immunology Center, Amsterdam, the Netherlands
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne Sophie Lixenfeld
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sofie Keijzer
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Federica Linty
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Remco Visser
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Mads Delbo Larsen
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Emily Lara Martin
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Inga Künsting
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Selina Lehrian
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Vera von Kopylow
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Carsten Kern
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Hanna Bele Lunding
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany
| | - Menno de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Niels van Mourik
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Theo Rispens
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - Tobias Graf
- Medical Department 2, University Heart Center of Schleswig-Holstein, Lübeck, Germany
| | - Marleen Adriana Slim
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands,Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Marije Kristianne Bomers
- Department of Internal Medicine, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Jonne Jochum Sikkens
- Department of Internal Medicine, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - Alexander P.J. Vlaar
- Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - C. Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands,Corresponding author.
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute of Nutritional Medicine, University of Lübeck and University Medical Center of Schleswig-Holstein, Lübeck, Germany,Airway Research Center North, University of Lübeck, German Center for Lung Research (DZL), Lübeck, Germany,Corresponding author.
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands,Corresponding author.
| | | | | |
Collapse
|
36
|
Ash MK, Bhimalli PP, Cho BK, Mattamana BB, Gambut S, Tarhoni I, Fhied CL, Reyes AF, Welninski SJ, Arivalagan J, Negrão F, Goel R, Beck TL, Hope TJ, Sha BE, Goo YA, Al-Harthi L, Mamede JI, Borgia JA, Kelleher NL, Schneider JR. Bulk IgG glycosylation predicts COVID-19 severity and vaccine antibody response. Cell Rep 2022; 41:111799. [PMID: 36493786 PMCID: PMC9678812 DOI: 10.1016/j.celrep.2022.111799] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 10/13/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
Although vaccination efforts have expanded, there are still gaps in our understanding surrounding the immune response to SARS-CoV-2. Measuring IgG Fc glycosylation provides insight into an infected individual's inflammatory state, among other functions. We set out to interrogate bulk IgG glycosylation changes from SARS-CoV-2 infection and vaccination, using plasma from mild or hospitalized COVID-19 patients, and from vaccinated individuals. Inflammatory glycans are elevated in hospitalized COVID-19 patients and increase over time, while mild patients have anti-inflammatory glycans that increase over time, including increased sialic acid correlating with RBD antibody levels. Vaccinated individuals with low RBD antibody levels and low neutralization have the same IgG glycan traits as hospitalized COVID-19 patients. In addition, a small vaccinated cohort reveals a decrease in inflammatory glycans associated with peak IgG concentrations and neutralization. This report characterizes the bulk IgG glycome associated with COVID-19 severity and vaccine responsiveness and can help guide future studies into SARS-CoV-2 protective immunity.
Collapse
Affiliation(s)
- Michelle K. Ash
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Pavan P. Bhimalli
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Byoung-Kyu Cho
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | | | - Stéphanie Gambut
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Imad Tarhoni
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Cristina L. Fhied
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Anjelica F. Reyes
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Samantha J. Welninski
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Jaison Arivalagan
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Fernanda Negrão
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Renu Goel
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA,Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Todd L. Beck
- Statistics Core, Rush University Medical Center, Chicago, IL, USA
| | - Thomas J. Hope
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA,Department of Cell & Developmental Biology, Northwestern University, Chicago, IL, USA,Department of Obstetrics & Gynecology, Northwestern University, Chicago, IL, USA,Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,Chemistry of Life Processes Institute, Northwestern University, Chicago, IL, USA
| | - Beverly E. Sha
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL, USA
| | - Young Ah Goo
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Lena Al-Harthi
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - João I. Mamede
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA
| | - Jeffrey A. Borgia
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA,Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Neil L. Kelleher
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA,Department of Chemistry, Northwestern University, Chicago, IL, USA,Department of Molecular Biosciences, Northwestern University, Chicago, IL, USA,Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,Chemistry of Life Processes Institute, Northwestern University, Chicago, IL, USA
| | - Jeffrey R. Schneider
- Department of Microbial Pathogens & Immunity, Rush University Medical Center, Cohn Research Building, 1735 W. Harrison St., Chicago, IL 60612, USA,Corresponding author
| |
Collapse
|
37
|
Yaugel-Novoa M, Bourlet T, Paul S. Role of the humoral immune response during COVID-19: guilty or not guilty? Mucosal Immunol 2022; 15:1170-1180. [PMID: 36195658 PMCID: PMC9530436 DOI: 10.1038/s41385-022-00569-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/07/2022] [Accepted: 09/19/2022] [Indexed: 02/04/2023]
Abstract
Systemic and mucosal humoral immune responses are crucial to fight respiratory viral infections in the current pandemic of COVID-19 caused by the SARS-CoV-2 virus. During SARS-CoV-2 infection, the dynamics of systemic and mucosal antibody infections are affected by patient characteristics, such as age, sex, disease severity, or prior immunity to other human coronaviruses. Patients suffering from severe disease develop higher levels of anti-SARS-CoV-2 antibodies in serum and mucosal tissues than those with mild disease, and these antibodies are detectable for up to a year after symptom onset. In hospitalized patients, the aberrant glycosylation of anti-SARS-CoV-2 antibodies enhances inflammation-associated antibody Fc-dependent effector functions, thereby contributing to COVID-19 pathophysiology. Current vaccines elicit robust humoral immune responses, principally in the blood. However, they are less effective against new viral variants, such as Delta and Omicron. This review provides an overview of current knowledge about the humoral immune response to SARS-CoV-2, with a particular focus on the protective and pathological role of humoral immunity in COVID-19 severity. We also discuss the humoral immune response elicited by COVID-19 vaccination and protection against emerging viral variants.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Bourlet
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France
| | - Stéphane Paul
- CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Université Claude Bernard Lyon 1, Lyon, France,CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
38
|
Bates TA, Lu P, Kang YJ, Schoen D, Thornton M, McBride SK, Park C, Kim D, Messer WB, Curlin ME, Tafesse FG, Lu LL. BNT162b2-induced neutralizing and non-neutralizing antibody functions against SARS-CoV-2 diminish with age. Cell Rep 2022; 41:111544. [PMID: 36252569 PMCID: PMC9533669 DOI: 10.1016/j.celrep.2022.111544] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 08/12/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022] Open
Abstract
Each severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant renews concerns about decreased vaccine neutralization weakening efficacy. However, while prevention of infection varies, protection from disease remains and implicates immunity beyond neutralization in vaccine efficacy. Polyclonal antibodies function through Fab domains that neutralize virus and Fc domains that induce non-neutralizing responses via engagement of Fc receptors on immune cells. To understand how vaccines promote protection, we leverage sera from 51 SARS-CoV-2 uninfected individuals after two doses of the BNT162b2 mRNA vaccine. We show that neutralizing activities against clinical isolates of wild-type and five SARS-CoV-2 variants, including Omicron BA.2, link to FcγRIIIa/CD16 non-neutralizing effector functions. This is associated with post-translational afucosylation and sialylation of vaccine-specific antibodies. Further, polyfunctional neutralizing and non-neutralizing breadth, magnitude, and coordination diminish with age. Thus, studying Fc functions in addition to Fab-mediated neutralization provides greater insight into vaccine efficacy for vulnerable populations, such as the elderly, against SARS-CoV-2 and novel variants.
Collapse
Affiliation(s)
- Timothy A Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ye Jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Devin Schoen
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Micah Thornton
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Savannah K McBride
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Chanhee Park
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daehwan Kim
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Marcel E Curlin
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR 97239, USA.
| | - Fikadu G Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA.
| | - Lenette L Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Parkland Health & Hospital System, Dallas, TX 75235, USA.
| |
Collapse
|
39
|
Emmenegger M, Fiedler S, Brugger SD, Devenish SR, Morgunov AS, Ilsley A, Ricci F, Malik AY, Scheier T, Batkitar L, Madrigal L, Rossi M, Meisl G, Lynn AK, Saleh L, von Eckardstein A, Knowles TP, Aguzzi A. Both COVID-19 infection and vaccination induce high-affinity cross-clade responses to SARS-CoV-2 variants. iScience 2022; 25:104766. [PMID: 35875683 PMCID: PMC9288251 DOI: 10.1016/j.isci.2022.104766] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/06/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
The B.1.1.529 (omicron) variant has rapidly supplanted most other SARS-CoV-2 variants. Using microfluidics-based antibody affinity profiling (MAAP), we have characterized affinity and IgG concentration in the plasma of 39 individuals with multiple trajectories of SARS-CoV-2 infection and/or vaccination. Antibody affinity was similar against the wild-type, delta, and omicron variants (K A ranges: 122 ± 155, 159 ± 148, 211 ± 307 μM-1, respectively), indicating a surprisingly broad and mature cross-clade immune response. Postinfectious and vaccinated subjects showed different IgG profiles, with IgG3 (p-value = 0.002) against spike being more prominent in the former group. Lastly, we found that the ELISA titers correlated linearly with measured concentrations (R = 0.72) but not with affinity (R = 0.29). These findings suggest that the wild-type and delta spike induce a polyclonal immune response capable of binding the omicron spike with similar affinity. Changes in titers were primarily driven by antibody concentration, suggesting that B-cell expansion, rather than affinity maturation, dominated the response after infection or vaccination.
Collapse
Affiliation(s)
- Marc Emmenegger
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Sebastian Fiedler
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Silvio D. Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sean R.A. Devenish
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Alexey S. Morgunov
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Alison Ilsley
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Francesco Ricci
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Anisa Y. Malik
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Thomas Scheier
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Leyla Batkitar
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Lidia Madrigal
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Marco Rossi
- Department of Laboratory Medicine, University Hospital Zürich, 8091 Zurich, Switzerland
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Andrew K. Lynn
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
| | - Lanja Saleh
- Department of Laboratory Medicine, University Hospital Zürich, 8091 Zurich, Switzerland
| | | | - Tuomas P.J. Knowles
- Fluidic Analytics, Unit A, The Paddocks Business Centre, Cherry Hinton Road, Cambridge CB1 8DH, UK
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Ave, Cambridge CB3 0HE, UK
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
40
|
Bates TA, Lu P, Kang YJ, Schoen D, Thornton M, McBride SK, Park C, Kim D, Messer WB, Curlin ME, Tafesse FG, Lu LL. BNT162b2 induced neutralizing and non-neutralizing antibody functions against SARSCoV-2 diminish with age. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.08.12.22278726. [PMID: 36032979 PMCID: PMC9413715 DOI: 10.1101/2022.08.12.22278726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Each novel SARS-CoV-2 variant renews concerns about decreased vaccine efficacy caused by evasion of vaccine induced neutralizing antibodies. However, accumulating epidemiological data show that while vaccine prevention of infection varies, protection from severe disease and death remains high. Thus, immune responses beyond neutralization could contribute to vaccine efficacy. Polyclonal antibodies function through their Fab domains that neutralize virus directly, and Fc domains that induce non-neutralizing host responses via engagement of Fc receptors on immune cells. To understand how vaccine induced neutralizing and non-neutralizing activities synergize to promote protection, we leverage sera from 51 SARS-CoV-2 uninfected health-care workers after two doses of the BNT162b2 mRNA vaccine. We show that BNT162b2 elicits antibodies that neutralize clinical isolates of wildtype and five variants of SARS-CoV-2, including Omicron BA.2, and, critically, induce Fc effector functions. FcγRIIIa/CD16 activity is linked to neutralizing activity and associated with post-translational afucosylation and sialylation of vaccine specific antibodies. Further, neutralizing and non-neutralizing functions diminish with age, with limited polyfunctional breadth, magnitude and coordination observed in those ≥65 years old compared to <65. Thus, studying Fc functions in addition to Fab mediated neutralization provides greater insight into vaccine efficacy for vulnerable populations such as the elderly against SARS-CoV-2 and novel variants.
Collapse
Affiliation(s)
- Timothy A. Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Ye jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Devin Schoen
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR
| | - Micah Thornton
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Savannah K. McBride
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Chanhee Park
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Daehwan Kim
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Marcel E. Curlin
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR
| | - Fikadu G. Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Lenette L. Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX
- Parkland Health & Hospital System
| |
Collapse
|
41
|
Nguyen DC, Lamothe PA, Woodruff MC, Saini AS, Faliti CE, Sanz I, Lee FE. COVID-19 and plasma cells: Is there long-lived protection? Immunol Rev 2022; 309:40-63. [PMID: 35801537 PMCID: PMC9350162 DOI: 10.1111/imr.13115] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ankur S. Saini
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Caterina E. Faliti
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ignacio Sanz
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Frances Eun‐Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
42
|
Ravindran A, Nirmal D, Jebalin. I. V BK, Pinkymol KP, Prajoon P, Ajayan J. InGaAs based gratings for UV-VIS spectrometer in prospective mRNA vaccine research. OPTICAL AND QUANTUM ELECTRONICS 2022; 54:555. [PMID: 35912403 PMCID: PMC9321284 DOI: 10.1007/s11082-022-04002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
During the outbreak of the COVID-19 illness, mRNA (messenger RNA) injections proved to be effective vaccination. Among the presently available analytical techniques, UV/VIS spectrophotometry is a trustworthy and practical instrument that may provide information on the chemical components of the vaccine at the molecular level. In this paper, we will present a one-dimensional grating of InGaAs as a prospect grating structure for UV-VIS spectrometer that can be used for mRNA vaccine development. The main parameters and the wavelength region used in mRNA vaccine development lies in the range of 200 nm to 700 nm (UV-VIS Range). The incorporation of new materials that are excellent for cutting-edge semiconductor industry procedures for MEMS manufacture, as well as new optimal parameters, will improve the grating and spectrometer's performance which will enhance the mRNA vaccine development and manufacturing workflows enabled by UV-VIS spectroscopy. Hence we evaluated the feasibility of the materials, Si (Silicon), GaN (Gallium Nitride), InGaAs (Indium Gallium Arsenide) and InP (Indium Phosphide) as a grating material. Reflection spectrum of the proposed structure shows 48% increase compared to the grating made up of Silicon. In order to model wave propagation in one grating unit cell, electromagnetic waves frequency domain interface is used. The periodic constraints of floquet periodicity are used for simulation at both faces of the unit cell. The reflectance of grating with each material as functions of the angle of incidence was plotted. Also we evaluated the effect of grating thickness, groove density, spectral resolution and efficiency over different materials namely Si, GaN, InGaAs and InP. After optimizing geometric parameters, the designed InGaAs based grating achieved a efficiency of 87.45% and can be a reliable prospect for mRNA based vaccine development.
Collapse
Affiliation(s)
- Ajith Ravindran
- Karunya Institute of Technology and Sciences, Coimbatore and Saintgits College of Engineering, Kottayam, India
| | - D. Nirmal
- Karunya Institute of Technology and Sciences, Coimbatore, India
| | | | - K. P. Pinkymol
- National Institute of Technology, Tiruchirappalli, India
| | - P. Prajoon
- Jyothi Engineering College, Cheruthuruthy, Thrissur, India
| | - J. Ajayan
- SR University, Warangal, Telangana India
| |
Collapse
|
43
|
Sudden Onset of IgA Vasculitis Affecting Vital Organs in Adult Patients following SARS-CoV-2 Vaccines. Vaccines (Basel) 2022; 10:vaccines10060923. [PMID: 35746531 PMCID: PMC9229537 DOI: 10.3390/vaccines10060923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
IgA vasculitis is an immune complex-mediated small-vessel vasculitis that mainly occurs in children and is characterized by palpable purpura, arthralgia, abdominal pain, and glomerulonephritis. We report three cases of new-onset IgA vasculitis involving major organs in adult patients after they received either the ChAdOx1 viral vector (Oxford/AstraZeneca) vaccine or the messenger RNA-1273 (Moderna) vaccine. These cases suggest that COVID-19 vaccines have the potential to trigger IgA vasculitis and indicate that physicians need to monitor for this possible complication.
Collapse
|
44
|
Schwedler C, Grzeski M, Kappert K, Rust J, Heymann G, Hoppe B, Blanchard V. Coronavirus Disease 2019-Related Alterations of Total and Anti-Spike IgG Glycosylation in Relation to Age and Anti-Spike IgG Titer. Front Microbiol 2022; 13:775186. [PMID: 35495660 PMCID: PMC9051488 DOI: 10.3389/fmicb.2022.775186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/02/2022] [Indexed: 01/02/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been affecting the world since January 2020 and has caused millions of deaths. To gain a better insight into molecular changes underlying the COVID-19 disease, we investigated here the N-glycosylation of three immunoglobulin G (IgG) fractions isolated from plasma of 35 severe COVID-19 patients, namely total IgG1, total IgG2, and anti-Spike IgG, by means of MALDI-TOF-MS. All analyses were performed at the glycopeptide level to assure subclass- and site-specific information. For each COVID-19 patient, the analyses included three blood withdrawals at different time-points of hospitalization, which allowed profiling longitudinal alterations in IgG glycosylation. The COVID-19 patients presented altered IgG N-glycosylation profiles in all investigated IgG fractions. The most pronounced COVID-19-related changes were observed in the glycosylation profiles of antigen-specific anti-Spike IgG1. Anti-Spike IgG1 fucosylation and galactosylation showed the strongest variation during the disease course, with the difference in anti-Spike IgG1 fucosylation being significantly correlated with patients’ age. Decreases in anti-Spike IgG1 galactosylation and sialylation in the course of the disease were found to be significantly correlated with the difference in anti-Spike IgG plasma concentration. The present findings suggest that patients’ age and anti-S IgG abundance might influence IgG N-glycosylation alterations occurring in COVID-19.
Collapse
Affiliation(s)
- Christian Schwedler
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- *Correspondence: Christian Schwedler, ,
| | - Marta Grzeski
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai Kappert
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Labor Berlin – Charité Vivantes GmbH, Berlin, Germany
| | - Jörn Rust
- Department of Anaesthesiology, Critical Care, and Pain Medicine, BG Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Guido Heymann
- Institute of Laboratory Medicine, BG Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Berthold Hoppe
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Laboratory Medicine, BG Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Véronique Blanchard
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Véronique Blanchard,
| |
Collapse
|
45
|
SARS-CoV-2 Beta and Delta variants trigger Fc effector function with increased cross-reactivity. Cell Rep Med 2022; 3:100510. [PMID: 35233544 PMCID: PMC8761540 DOI: 10.1016/j.xcrm.2022.100510] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/19/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants of concern (VOCs) exhibit escape from neutralizing antibodies, causing concern about vaccine effectiveness. However, while non-neutralizing cytotoxic functions of antibodies are associated with improved disease outcome and vaccine protection, Fc effector function escape from VOCs is poorly defined. Furthermore, whether VOCs trigger Fc functions with altered specificity, as has been reported for neutralization, is unknown. Here, we demonstrate that the Beta VOC partially evades Fc effector activity in individuals infected with the original (D614G) variant. However, not all functions are equivalently affected, suggesting differential targeting by antibodies mediating distinct Fc functions. Furthermore, Beta and Delta infection trigger responses with significantly improved Fc cross-reactivity against global VOCs compared with D614G-infected or Ad26.COV2.S-vaccinated individuals. This suggests that, as for neutralization, the infecting spike sequence affects Fc effector function. These data have important implications for vaccine strategies that incorporate VOCs, suggesting these may induce broader Fc effector responses.
Collapse
|
46
|
Pongracz T, Vidarsson G, Wuhrer M. Antibody glycosylation in COVID-19. Glycoconj J 2022; 39:335-344. [PMID: 35091890 PMCID: PMC8799414 DOI: 10.1007/s10719-022-10044-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/06/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022]
Abstract
AbstractAntibody glycosylation has received considerable attention in coronavirus disease 2019 (COVID-19) infections and recently also in vaccination. Antibody glycosylation and in particular immunoglobulin G1 fucosylation levels influence effector functions and are therefore key parameters for assessing the efficacy and safety of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directed immune responses. This review article summarizes and interprets recent research into antibody glycosylation in COVID-19. Experimental approaches for analyzing the glycosylation of SARS-CoV-2-directed antibody responses are evaluated. The pronounced dynamics, effector functions, clinical utility, and regulation of antibody glycosylation in COVID-19 are assessed. Future research on the role of antibody glycosylation in COVID may cover the glycosylation of other antibody classes beyond immunoglobulin G, the regulation of antibody glycosylation, and the role of non-canonical antibody receptors in determining effector functions.
Graphical abstract
Collapse
Affiliation(s)
- Tamas Pongracz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, UMC, University of Amsterdam, AmsterdamAmsterdam, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|