1
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore IN, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. PLoS Pathog 2025; 21:e1012456. [PMID: 39847599 DOI: 10.1371/journal.ppat.1012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather than the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n = 6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kirti A Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Kathryn L Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Matthew R Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stacey A Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mark C Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Sarah E Scheuermann
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jennifer S Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Elizabeth H Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Amit A Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Ian N Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Daniel C Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas R Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, United States of America
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Steven E Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
2
|
Menezes A, Razafimahatratra SL, Wariri O, Graham AL, Metcalf CJE. Strengthening serological studies: the need for greater geographical diversity, biobanking, and data-accessibility. Trends Microbiol 2025:S0966-842X(24)00322-6. [PMID: 39818508 DOI: 10.1016/j.tim.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Serological studies uniquely strengthen infectious disease surveillance, expanding prevalence estimates to encompass asymptomatic infections, and revealing the otherwise inapparent landscape of immunity, including who is and is not susceptible to infection. They are thus a powerful complement to often incomplete epidemiological and public health measures (administrative measures of vaccination coverage, incidence estimates, etc.). The recent surge in the deployment of serological surveys globally (in part due to the SARS-CoV-2 pandemic), alongside the development of new assays and new inference methods, means that the time is ripe to interrogate areas to strengthen future serosurveillance efforts. We identify three themes warranting attention: first, expanding the geographical diversity of these studies; second, investing globally in infrastructure for storage of blood samples (biobanking), opening the way to future analyses; and third, establishing protocols to increase data accessibility and to facilitate data usage for current and future studies. We conclude that strengthening serological studies is necessary and achievable through thoughtful sampling design, wide-scale sample storage, and thorough reporting practices.
Collapse
Affiliation(s)
- Arthur Menezes
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA.
| | | | - Oghenebrume Wariri
- Vaccines and Immunity Theme, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, London, Banjul, The Gambia; Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA; Santa Fe Institute, Santa Fe, NM, USA
| | - C Jessica E Metcalf
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA; Princeton School of Public and International Affairs, Princeton University, Princeton, NJ, USA
| |
Collapse
|
3
|
Williams E, Echeverri Tribin F, Carreño JM, Krammer F, Hoffer M, Pallikkuth S, Pahwa S. Proteomic signatures of vaccine-induced and breakthrough infection-induced host responses to SARS-CoV-2. Vaccine 2025; 43:126484. [PMID: 39520894 DOI: 10.1016/j.vaccine.2024.126484] [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: 08/27/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
The severity of SARS-CoV-2 illness is influenced by factors including age, sex, pre-existing health conditions, and individual immune responses. However, the mechanisms conferring immunity following antigenic challenge have not been fully elucidated. There are currently no studies evaluating longitudinal proteomic changes in individuals following vaccination and breakthrough, limiting our understanding of the underlying mechanisms driving conferred immunity. In this work, we evaluated the differential protein expression in individuals with (CoV-P) or without (CoV-N) prior SARS-CoV-2 infection following primary vaccination and after breakthrough infection (CoV-BT). Overall, we found that individuals receiving primary vaccination relied on innate immune mechanisms, including complement and coagulation cascades, and natural killer cell-mediated cytotoxicity, while conversely, breakthrough infection immune mechanisms relied on T cell-mediated immunity. These mechanistic differences may help explain heterogeneity associated with vaccine-induced and breakthrough infection-related outcomes.
Collapse
Affiliation(s)
- Erin Williams
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, Florida, 33136, USA
| | | | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, NY, New York, 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, NY, New York, 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Michael Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA; Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, Florida, 33136, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, 33146, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, 33146, USA
| |
Collapse
|
4
|
Sakakibara S, Liu YC, Ishikawa M, Edahiro R, Shirai Y, Haruna S, El Hussien MA, Xu Z, Li S, Yamaguchi Y, Murakami T, Morita T, Kato Y, Hirata H, Takeda Y, Sugihara F, Naito Y, Motooka D, Tsai CY, Ono C, Matsuura Y, Wing JB, Matsumoto H, Ogura H, Okada M, Kumanogoh A, Okada Y, Standley DM, Kikutani H, Okuzaki D. Clonal landscape of autoantibody-secreting plasmablasts in COVID-19 patients. Life Sci Alliance 2024; 7:e202402774. [PMID: 39288992 PMCID: PMC11408605 DOI: 10.26508/lsa.202402774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Whereas severe COVID-19 is often associated with elevated autoantibody titers, the underlying mechanism behind their generation has remained unclear. Here we report clonal composition and diversity of autoantibodies in humoral response to SARS-CoV-2. Immunoglobulin repertoire analysis and characterization of plasmablast-derived monoclonal antibodies uncovered clonal expansion of plasmablasts producing cardiolipin (CL)-reactive autoantibodies. Half of the expanded CL-reactive clones exhibited strong binding to SARS-CoV-2 antigens. One such clone, CoV1804, was reactive to both CL and viral nucleocapsid (N), and further showed anti-nucleolar activity in human cells. Notably, antibodies sharing genetic features with CoV1804 were identified in COVID-19 patient-derived immunoglobulins, thereby constituting a novel public antibody. These public autoantibodies had numerous mutations that unambiguously enhanced anti-N reactivity, when causing fluctuations in anti-CL reactivity along with the acquisition of additional self-reactivities, such as anti-nucleolar activity, in the progeny. Thus, potentially CL-reactive precursors may have developed multiple self-reactivities through clonal selection, expansion, and somatic hypermutation driven by viral antigens. Our results revealed the nature of autoantibody production during COVID-19 and provided novel insights into the origin of virus-induced autoantibodies.
Collapse
Affiliation(s)
- Shuhei Sakakibara
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yu-Chen Liu
- Laboratory of Human Immunology (Single Cell Genomics), Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masakazu Ishikawa
- Laboratory of Human Immunology (Single Cell Genomics), Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Ryuya Edahiro
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yuya Shirai
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Soichiro Haruna
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Marwa Ali El Hussien
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Zichang Xu
- Laboratory of Systems Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Songling Li
- Laboratory of Systems Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Teruaki Murakami
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Naito
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Motooka
- Laboratory of Human Immunology (Single Cell Genomics), Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Chao-Yuan Tsai
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Chikako Ono
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Yoshiharu Matsuura
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - James B Wing
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Human Single Cell Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Hisatake Matsumoto
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Ogura
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masato Okada
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - Yukinari Okada
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Wakō, japan
| | - Daron M Standley
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory of Systems Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Hitoshi Kikutani
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| |
Collapse
|
5
|
Singh T, Macintyre AN, Burke TW, Anderson J, Petzold E, Stover EL, French MJ, Oguin TH, Demarco T, McClain MT, Ko ER, Park LP, Denny T, Sempowski GD, Woods CW. Dynamics of cytokine and antibody responses in community versus hospital SARS-CoV-2 infections. Front Immunol 2024; 15:1468871. [PMID: 39650666 PMCID: PMC11621060 DOI: 10.3389/fimmu.2024.1468871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Dysregulated host cytokine responses to SARS-CoV-2 infection are a primary cause of progression to severe disease, whereas early neutralizing antibody responses are considered protective. However, there are gaps in understanding the early temporal dynamics of these immune responses, and the profile of productive immune responses generated by non-hospitalized people with mild infections in the community. Methods Here we conducted a prospective cohort study of people with suspected infections/exposures in the US state of North Carolina, before vaccine availability. We recruited participants not only in hospitals/clinics, but also in their homes. With serial sampling, we compared virologic and immunologic factors in 258 community cases versus 114 hospital cases of COVID-19 to define factors associated with severity. Results We found that high early neutralizing antibodies were associated with lower nasal viral load, but not protection from hospitalization. Cytokine responses were evaluated in 125 cases, with subsets at first versus second week of illness to assess for time-dependent trajectories. The hospital group demonstrated a higher magnitude of serum IL-6, IL-1R antagonist, IP-10, and MIG; prolonged upregulation of IL-17; and lesser downregulation of GROα, IL-1R antagonist, and MCP1, in comparison to the community group suggesting that these factors may contribute to immunopathology. In the second week of illness, 2-fold increases in IL-6, IL-1R antagonist, and IP-10 were associated with 2.2, 1.8, and 10-fold higher odds of hospitalization respectively, whereas a 2-fold increase in IL-10 was associated with 63% reduction in odds of hospitalization (p<0.05). Moreover, antibody responses at 3-6 months post mild SARS-CoV-2 infections in the community revealed long-lasting antiviral IgM and IgA antibodies as well as a stable set point of neutralizing antibodies that were not waning. Discussion Our data provide valuable temporal cytokine benchmarks to track the progression of immunopathology in COVID-19 patients and guide improvements in immunotherapies.
Collapse
Affiliation(s)
- Tulika Singh
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
- Duke Global Health Institute, Durham, NC, United States
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Thomas W. Burke
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Jack Anderson
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Elizabeth Petzold
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Erica L. Stover
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Matthew J. French
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thomas H. Oguin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Todd Demarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Micah T. McClain
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| | - Emily R. Ko
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| | - Lawrence P. Park
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Thomas Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- RTI International, Research Triangle Park, NC, United States
| | - Christopher W. Woods
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| |
Collapse
|
6
|
Boongird S, Srithongkul T, Sethakarun S, Bruminhent J, Kiertiburanakul S, Nongnuch A, Kitiyakara C, Sritippayawan S. Tixagevimab-cilgavimab for preventing breakthrough COVID-19 in dialysis patients: a prospective study. Clin Kidney J 2024; 17:sfae309. [PMID: 39539359 PMCID: PMC11558061 DOI: 10.1093/ckj/sfae309] [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: 06/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background The effectiveness of tixagevimab-cilgavimab as pre-exposure prophylaxis (PrEP) against breakthrough coronavirus disease 2019 (COVID-19) in dialysis patients remains uncertain due to limited data. Methods In this multicenter prospective study, we enrolled vaccinated dialysis patients and divided them into two groups: a tixagevimab-cilgavimab group (received a 150 mg/150 mg intramuscular dose of tixagevimab-cilgavimab) and a control group (age-matched patients not receiving tixagevimab-cilgavimab). The primary outcome was the breakthrough COVID-19 rate at 6 months, whereas secondary outcomes included COVID-19-related hospitalization, intensive care unit admission, endotracheal intubation and mortality. The safety of tixagevimab-cilgavimab was assessed. Results Two hundred participants were enrolled, with equal numbers in each group (n = 100 each). Baseline characteristics were comparable between groups, except for a higher number of COVID-19 vaccine doses in the tixagevimab-cilgavimab group [median (IQR) 4 (3-5) vs. 3 (3-4); P = .01]. At 6 months, the breakthrough COVID-19 rates were comparable between the tixagevimab-cilgavimab (17%) and control (15%) groups (P = .66). However, the median (IQR) time to diagnosis of breakthrough infections tended to be longer in the tixagevimab-cilgavimab group [4.49 (2.81-4.98) vs 1.96 (1.65-2.91) months; P = .08]. Tixagevimab-cilgavimab significantly reduced COVID-19-related hospitalization rates (5.9% vs 40.0%; P = .02) among participants with breakthrough infections. All tixagevimab-cilgavimab-related adverse events were mild. Conclusion The use of tixagevimab-cilgavimab as PrEP in vaccinated dialysis patients during the Omicron surge did not prevent breakthrough infections but significantly reduced COVID-19-related hospitalizations. Further research should prioritize alternative strategies.
Collapse
Affiliation(s)
- Sarinya Boongird
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thatsaphan Srithongkul
- Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Jackrapong Bruminhent
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasisopin Kiertiburanakul
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arkom Nongnuch
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suchai Sritippayawan
- Division of Nephrology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
7
|
Song C, Li H, Han Y, Luo J, Zhao Y, Zhou C, Zhang A, Wang H. Host restriction factor Rab11a limits porcine epidemic diarrhea virus invasion of cells via fusion peptide-mediated membrane fusion. Int J Biol Macromol 2024; 279:135299. [PMID: 39233171 DOI: 10.1016/j.ijbiomac.2024.135299] [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: 07/20/2024] [Revised: 08/11/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes enormous economic losses to the pork industry, and its extensive cell tropism poses a substantial challenge to public health and safety. However, the invasion mechanisms and relevant host factors of PEDV remain poorly understood. In this study, we identified 422 differentially expressed genes related to PEDV infection through transcriptome analysis. Among these, Annexin A2 (ANXA2), Prohibitin-2 (PHB2), and Caveolin-2 (CAV2) were identified through screening and verifying as having a specific interaction with the PEDV S protein, and positive regulation of PEDV internalization was validated by siRNA and overexpression tests. Subsequently, using host membrane protein interaction networks and co-immunoprecipitation analysis, we found that ANXA2 PHB2 or CAV2 directly interact with Rab11a. Next, we constructed a pseudovirus model (LV-PEDV S-GFP) to further confirm that the downregulation of Rab11a could promote PEDV invasion. In detail, ANXA2, PHB2, or CAV2 promoted PEDV invasion via downregulating Rab11a. Furthermore, we showed that the S-protein fusion peptide (FP) was sufficient for S-protein interaction with ANXA2, PHB2, CAV2, and Rab11a, and the addition of exogenous GTP could regulate the efficiency of PEDV invasion. Collectively, ANXA2, PHB2, or CAV2 influenced the membrane fusion of PEDV with host cells through the host restriction factor Rab11a. This study could be targeted for future research to develop strategies for the control of PEDV.
Collapse
Affiliation(s)
- Cailiang Song
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hao Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yun Han
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Jinchao Luo
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yu Zhao
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Changyu Zhou
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Anyun Zhang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
8
|
Zhang Z, Wang J, Niu Q, Guan G, Yin H, Yang J. An immunoassay based on bioluminescent sensors for rapid detection of African swine fever virus antibodies. J Clin Microbiol 2024; 62:e0046324. [PMID: 39235247 PMCID: PMC11481549 DOI: 10.1128/jcm.00463-24] [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: 03/23/2024] [Accepted: 08/15/2024] [Indexed: 09/06/2024] Open
Abstract
Serological assays for antibody detection have contributed significantly to the diagnosis and control of infectious diseases. African swine fever is the most devastating infectious disease of domestic pigs and wild boars, severely threatening the global pig industry in recent years. Here, we developed a rapid, simple, and sensitive immunoassay based on the split-luciferase system to detect IgG antibodies against African swine fever virus (ASFV). In this assay, the p30 protein of ASFV was genetically coupled to the LgBiT and SmBiT subunits of nanoluciferase, which were used as fusion probes for specific antibodies. Target engagement of the probes results in the reconstitution of a functional nanoluciferase, which further catalyzes bioluminescent reactions. Different orientations of the LgBiT and SmBiT-p30 fusion sensors were designed and investigated, and N-LgBiT/p30 and N-SmBiT/p30 were identified as a promising sensor pair for reforming active nanoluciferase in the presence of specific antibodies. After optimization, this split-luciferase complementation assay showed high sensitivity and specificity for the detection of ASFV antibodies. The analytical sensitivity of the assay was 16 times greater than that of the blocking enzyme-linked immunosorbent assay (ELISA) by the detection of serial dilutions of serum, and no cross-reaction was observed with other swine pathogens. As demonstrated in clinical samples, its performance is highly consistent with that of a commercial ELISA kit, with a concordance rate of 98.19%. This assay is simple and easy to perform, providing a more flexible and efficient approach for the measurement of ASFV antibodies in clinical applications. IMPORTANCE The study is about a homogeneous split-luciferase assay for antibody detection. Split nanoluciferase biosensors for the detection of ASFV antibodies were designed. This sensor platform enables the sensitive and specific detection of antibodies. The split-luciferase assay is simple, rapid, and easy to use.
Collapse
Affiliation(s)
- Zhonghui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jinming Wang
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Qingli Niu
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jifei Yang
- State Key Laboratory for Animal Disease Control and Prevention, African Swine Fever Regional Laboratory of China (Lanzhou), Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Lu X, Gao L, Qian SJ, Dai LMJ, Zhou ZY, Qiu TL, Xia Y, Miao Y, Qin SC, Fan L, Xu W, Li JY, Zhu HY. [Single-center study of COVID-19 in patients with chronic lymphocytic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:923-930. [PMID: 39622756 PMCID: PMC11579760 DOI: 10.3760/cma.j.cn121090-20240621-00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Indexed: 12/06/2024]
Abstract
Objective: To investigate the vaccination status, characteristics and prognosis of patients suffering from a combination of COVID-19 and chronic lymphocytic anemia (CLL) in China. Methods: Clinical data of 328 patients with chronic lymphocytic leukemia (CLL) who were first diagnosed with COVID-19 and treated in the Department of Hematology of Jiangsu Provincial People's Hospital between November 2022 and February 2023 were retrospectively analyzed. Univariate and multivariate analysis of data of patients with severe/critical COVID-19 were conducted by applying the binary logistic regression model. Results: The median age of the CLL patients was 60 (24-87) years. 23.5% (77/328) of these patients suffered from severe/critical COVID-19 infection. Univariate analysis of the data demonstrated that a combination of factors including age >67 years (OR=2.15, 95% CI 1.24- 3.73, P=0.006), diabetes (OR=2.09, 95% CI 1.05-4.20, P=0.037), chronic hepatitis B (OR=2.91, 95% CI 1.30-6.51, P=0.010), CLL progressive (OR=3.79, 95% CI 1.57-9.15, P=0.003) and CD20 antibody-based treatments within three months prior to the COVID-19 infection (OR=2.79, 95% CI 1.35-5.77, P=0.006) were the risk factors for severe/critical COVID-19. According to the multivariate analysis, CLL progressive (OR=2.98, 95% CI 1.10-8.10, P=0.033) was an independent risk factor for severe/critical COVID-19 and administration of the BTK (Bruton tyrosine kinase) inhibitor monotherapy might exert a protective effect and influence a positive outcome of the COVID-19 infection (OR=0.38, 95% CI 0.16-0.90, P=0.028). Among the 242 patients who were followed up until October 2023, 9.1% (22/242) had multiple subsequent COVID-19 infections (≥3), and 2.1% (5/242) had persistent COVID-19 infections (patients with persistent positive test for the SARS-CoV-2 antigen testing until missing follow-up for any reason). The peak value of the anti-SARS-CoV-2-IgG titres was observed between three and four months post symptom onset (median: 3.511 S/CO vs 1.047 S/CO, P<0.05). The levels of immunoglobulin A gradually decreased following infection with COVID-19, and its trough levels were attained between two to four weeks post infection (median: 0.30 g/L vs 0.74 g/L, P<0.05). According to this study the mortality of patients suffering from a combination of COVID-19 infection and CLL was 2.7% (9/328), and the main reason for their death was respiratory failure and heart failure. Conclusions: A low rate of COVID-19 vaccination and a high rate of severe/critical COVID-19 infection was observed in the CLL patients. CLL progressive was associated with severe/critical COVID-19. Anti-CD20-based treatments received within the past three months might be a risk factor for exacerbation of COVID-19 infection, whereas a monotherapy with BTK inhibitors exert a protective effect and improve outcome of COVID-19 infection.
Collapse
Affiliation(s)
- X Lu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - L Gao
- Department of Hematology, The Affiliate Suqian First People's Hospital of Nanjing Medical University, Suqian Branch of Jiangsu Provincial People's Hospital, Suqian 223999, China
| | - S J Qian
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - L M J Dai
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Z Y Zhou
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - T L Qiu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Y Xia
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - Y Miao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - S C Qin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - L Fan
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - W Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - J Y Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China
| | - H Y Zhu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China Department of Hematology, The Affiliate Suqian First People's Hospital of Nanjing Medical University, Suqian Branch of Jiangsu Provincial People's Hospital, Suqian 223999, China
| |
Collapse
|
10
|
Walker MR, Underwood A, Björnsson KH, Raghavan SSR, Bassi MR, Binderup A, Pham LV, Ramirez S, Pinholt M, Dagil R, Knudsen AS, Idorn M, Soegaard M, Wang K, Ward AB, Salanti A, Bukh J, Barfod L. Broadly potent spike-specific human monoclonal antibodies inhibit SARS-CoV-2 Omicron sub-lineages. Commun Biol 2024; 7:1239. [PMID: 39354108 PMCID: PMC11445456 DOI: 10.1038/s42003-024-06951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
The continuous emergence of SARS-CoV-2 variants of concern has rendered many therapeutic monoclonal antibodies (mAbs) ineffective. To date, there are no clinically authorized therapeutic antibodies effective against the recently circulating Omicron sub-lineages BA.2.86 and JN.1. Here, we report the isolation of broad and potent neutralizing human mAbs (HuMabs) from a healthcare worker infected with SARS-CoV-2 early in the pandemic. These include a genetically unique HuMab, named K501SP6, which can neutralize different Omicron sub-lineages, including BQ.1, XBB.1, BA.2.86 and JN.1, by targeting a highly conserved epitope on the N terminal domain, as well as an RBD-specific HuMab (K501SP3) with high potency towards earlier circulating variants that was escaped by the more recent Omicron sub-lineages through spike F486 and E484 substitutions. Characterizing SARS-CoV-2 spike-specific HuMabs, including broadly reactive non-RBD-specific HuMabs, can give insight into the immune mechanisms involved in neutralization and immune evasion, which can be a valuable addition to already existing SARS-CoV-2 therapies.
Collapse
Affiliation(s)
- Melanie R Walker
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Underwood
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper H Björnsson
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sai Sundar Rajan Raghavan
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Maria R Bassi
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alekxander Binderup
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Long V Pham
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Pinholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Robert Dagil
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne S Knudsen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Manja Idorn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Kaituo Wang
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Barfod
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Joseph JO, Ylade M, Daag JV, Aogo R, Crisostomo MV, Mpingabo P, Premkumar L, Deen J, Katzelnick LC. High transmission of endemic human coronaviruses before and during the COVID-19 pandemic in adolescents in Cebu, Philippines. BMC Infect Dis 2024; 24:1042. [PMID: 39333882 PMCID: PMC11430261 DOI: 10.1186/s12879-024-09672-8] [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: 11/08/2023] [Accepted: 07/25/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND SARS-CoV-2, the causative agent of COVID-19, is a betacoronavirus belonging to the same genus as endemic human coronaviruses (hCoVs) OC43 and HKU1 and is distinct from alpha hCoVs 229E and NL63. In a study of adolescents in the Philippines, we evaluated seroprevalence to the hCoVs, whether pre-pandemic hCoV immunity modulated subsequent risk of SARS-CoV-2 infection, and if SARS-CoV-2 infection affected the transmission of the hCoVs. METHODS From 499 individuals screened in 2021 for SARS-CoV-2 receptor binding domain (RBD) antibodies by enzyme-linked immunosorbent assay (ELISA), we randomly selected 59 SARS-CoV-2 negative and 61 positive individuals for further serological evaluation. We measured RBD and spike antibodies to the four hCoVs and SARS-CoV-2 by ELISA in samples from the same participants collected pre-pandemic (2018-2019) and mid-pandemic (2021), before COVID-19 vaccination. RESULTS We observed over 72% seropositivity to the four hCoVs pre-pandemic. Binding antibodies increased with age to 229E and OC43, suggesting endemic circulation, while antibody levels was flat across ages for HKU1 and NL63. During the COVID-19 pandemic, antibodies increased significantly to the RBDs of OC43, NL63, and 229E and spikes of all four hCoVs in both SARS-CoV-2 negative and positive adolescents. Those aged 12-15 years old in 2021 had higher antibodies to RBD and spike of OC43, NL63, and 229E than adolescents the same age in 2019, further demonstrating intense transmission of the hCoVs during the pandemic. CONCLUSIONS We observe a limited impact of the COVID-19 pandemic on endemic hCoV transmission. This study provides insight into co-circulation of hCoVs and SARS-CoV-2.
Collapse
Affiliation(s)
- Janet O Joseph
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michelle Ylade
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines-Manila, Manila, Philippines
| | - Jedas Veronica Daag
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines-Manila, Manila, Philippines
| | - Rosemary Aogo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Vinna Crisostomo
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines-Manila, Manila, Philippines
| | - Patrick Mpingabo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jacqueline Deen
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines-Manila, Manila, Philippines
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
12
|
Pang X, Lee H, Rong J, Zhu Q, Xu S. Self-Thermal Management in Filtered Selenium-Terminated MXene Films for Flexible Safe Batteries. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309580. [PMID: 38705865 DOI: 10.1002/smll.202309580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/04/2024] [Indexed: 05/07/2024]
Abstract
Li-ion batteries with superior interior thermal management are crucial to prevent thermal runaway and ensure safe, long-lasting operation at high temperatures or during rapid discharging and charging. Typically, such thermal management is achieved by focusing on the separator and electrolyte. Here, the study introduces a Se-terminated MXene free-standing electrode with exceptional electrical conductivity and low infrared emissivity, synergistically combining high-rate capacity with reduced heat radiation for safe, large, and fast Li+ storage. This is achieved through a one-step organic Lewis acid-assisted gas-phase reaction and vacuum filtration. The Se-terminated Nb2Se2C outperformed conventional disordered O/OH/F-terminated materials, enhancing Li+-storage capacity by ≈1.5 times in the fifth cycle (221 mAh·g-1 at 1 A·g-1) and improving mid-infrared adsorption with low thermal radiation. These benefits result from its superior electrical conductivity, excellent structural stability, and high permittivity in the infrared region. Calculations further reveal that increased permittivity and conductivity along the z-direction can reduce heat radiation from electrodes. This work highlights the potential of surface groups-terminated layered material-based free-standing flexible electrodes with self-thermal management ability for safe, fast energy storage.
Collapse
Affiliation(s)
- Xin Pang
- State Key Lab of High-Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hyunjin Lee
- Department of Biomedical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Jingzhi Rong
- State Key Lab of High-Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Qiaoyu Zhu
- State Key Lab of High-Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Shumao Xu
- Department of Biomedical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
13
|
McCoy KM, Ackerman ME, Grigoryan G. A comparison of antibody-antigen complex sequence-to-structure prediction methods and their systematic biases. Protein Sci 2024; 33:e5127. [PMID: 39167052 PMCID: PMC11337930 DOI: 10.1002/pro.5127] [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: 03/15/2024] [Revised: 06/24/2024] [Accepted: 07/14/2024] [Indexed: 08/23/2024]
Abstract
The ability to accurately predict antibody-antigen complex structures from their sequences could greatly advance our understanding of the immune system and would aid in the development of novel antibody therapeutics. There have been considerable recent advancements in predicting protein-protein interactions (PPIs) fueled by progress in machine learning (ML). To understand the current state of the field, we compare six representative methods for predicting antibody-antigen complexes from sequence, including two deep learning approaches trained to predict PPIs in general (AlphaFold-Multimer and RoseTTAFold), two composite methods that initially predict antibody and antigen structures separately and dock them (using antibody-mode ClusPro), local refinement in Rosetta (SnugDock) of globally docked poses from ClusPro, and a pipeline combining homology modeling with rigid-body docking informed by ML-based epitope and paratope prediction (AbAdapt). We find that AlphaFold-Multimer outperformed other methods, although the absolute performance leaves considerable room for improvement. AlphaFold-Multimer models of lower quality display significant structural biases at the level of tertiary motifs (TERMs) toward having fewer structural matches in non-antibody-containing structures from the Protein Data Bank (PDB). Specifically, better models exhibit more common PDB-like TERMs at the antibody-antigen interface than worse ones. Importantly, the clear relationship between performance and the commonness of interfacial TERMs suggests that the scarcity of interfacial geometry data in the structural database may currently limit the application of ML to the prediction of antibody-antigen interactions.
Collapse
Affiliation(s)
- Katherine Maia McCoy
- Molecular and Cell Biology Graduate ProgramDartmouth CollegeHanoverNew HampshireUSA
| | - Margaret E. Ackerman
- Molecular and Cell Biology Graduate ProgramDartmouth CollegeHanoverNew HampshireUSA
- Thayer School of EngineeringDartmouth CollegeHanoverNew HampshireUSA
| | - Gevorg Grigoryan
- Molecular and Cell Biology Graduate ProgramDartmouth CollegeHanoverNew HampshireUSA
- Department of Computer ScienceDartmouth CollegeHanoverNew HampshireUSA
| |
Collapse
|
14
|
Leon-Rojas JE, Arias-Erazo F, Jiménez-Arias P, Recalde-Navarrete R, Guevara A, Coloma J, Martin M, Chis Ster I, Cooper P, Romero-Sandoval N. COVID-19 IgG seropositivity and its determinants in occupational groups of varying infection risks in two Andean cities of Ecuador before mass vaccination. PLoS One 2024; 19:e0309466. [PMID: 39208200 PMCID: PMC11361580 DOI: 10.1371/journal.pone.0309466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has caused over 68.7 million infections and 1.35 million deaths in South America. There are limited data on SARS-CoV-2 seropositivity and its determinants from Andean countries prior to mass vaccinations against COVID-19. OBJECTIVE To estimate SARS-CoV-2 seropositivity and its determinants before vaccination in occupational groups of adults presumed to have different levels of exposure and associations with potential symptomatology. METHODS We measured seropositivity of anti-SARS-CoV-2 IgG antibodies in a cross-sectional study of vaccine-naïve adults aged 18 years and older, recruited within three occupational risk groups (defined as low [LR], moderate [MR], and high [HR]) between January and September 2021 in two Andean cities in Ecuador. Associations with risk factors were estimated using logistic regression. RESULTS In a sample of 882 adults, IgG seropositivity for the three different occupational risk groups was 39.9% (CI 95% 35.3-44.6), 74.6% (CI 95% 66.4-81.4), and 39.0% (CI 95% 34.0-44.4) for the HR, MR, and LR groups, respectively. History of an illness with loss of taste and/or smell was significantly associated with seropositivity in all occupational groups, with adjusted ORs of 14.31 (95%CI, 5.83-35.12; p<0.001), 14.34 (95%CI 3.01-68.42; p<0.001), and 8.79 (95%CI 2.69-28.72; p<0.001), for the HR, MR, and LR groups, respectively; while fever was significant for the LR group with an adjusted OR of 1.24 (95%CI, 1.11-4.57; p = 0.025) and myalgia for the HR group with an adjusted OR of 2.07 (95%CI, 1.13-3.81; p = 0.019). CONCLUSION Notable proportions of seropositivity were seen in all occupational groups between January and September 2021 prior to mass vaccination. Loss of taste and/or smell was strongly associated with presence of anti-SARS-CoV-2 IgG antibodies irrespective of presumed occupational exposure risk.
Collapse
Affiliation(s)
- Jose E. Leon-Rojas
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
| | - Fernanda Arias-Erazo
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
- Grupo de Investigación en Sanidad Animal y Humana (GISAH) ESPE, Quito, Ecuador
| | - Patricia Jiménez-Arias
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- Grupo de Investigación en Sanidad Animal y Humana (GISAH) ESPE, Quito, Ecuador
- Departamento de Ciencias de la Vida y de la Agricultura, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Ricardo Recalde-Navarrete
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- Medical School, Universidad Tecnica de Ambato, Ambato, Ecuador
| | | | - Josefina Coloma
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States of America
| | - Miguel Martin
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Philip Cooper
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
- Institute of Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Natalia Romero-Sandoval
- Research Network Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL), Universidad Internacional del Ecuador, Quito, Ecuador
- School of Medicine, Universidad Internacional del Ecuador, Quito, Ecuador
| | | |
Collapse
|
15
|
Póvoa da Costa F, de Sarges KML, da Silva R, dos Santos EF, do Nascimento MH, Rodrigues AM, Cantanhede MHD, Rodrigues FBB, Viana MDNDSDA, Leite MDM, de Oliveira CF, Neves PFMD, Pereira Neto GDS, de Brito MTFM, da Silva ALS, Henriques DF, Quaresma JAS, Falcão LFM, Queiroz MAF, Vallinoto IMVC, Vallinoto ACR, Viana GMR, dos Santos EJM. Genetic, Clinical, Epidemiological, and Immunological Profiling of IgG Response Duration after SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:8740. [PMID: 39201427 PMCID: PMC11354850 DOI: 10.3390/ijms25168740] [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: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
The IgG response against SARS-CoV-2 infection can persist for over six months (long response; LR). However, among 30% of those infected, the duration can be as short as three months or less (short response; SR). The present study assembled serological data on the anti-SARS-CoV-2 IgG response duration of two previous studies and integrated these results with the plasmatic cytokine levels and genetic profile of 10 immune-relevant SNPs that were also previously published, along with the plasmatic total IgG, IgA, and IgM levels, allowing for the genetic, clinical, immunological, and epidemiological aspects of the post-COVID-19 IgG response duration to be understood. The SR was associated with previous mild acute COVID-19 and with an SNP (rs2228145) in IL6R related to low gene expression. Additionally, among the SR subgroup, no statistically significant Spearman correlations were observed between the plasma levels of IL-17A and the Th17 regulatory cytokines IFN-γ (rs = 0.2399; p = 0.1043), IL-4 (rs = 0.0273; p = 0.8554), and IL-2 (rs = 0.2204; p = 0.1365), while among the LR subgroup, weaker but statistically significant Spearman correlations were observed between the plasma levels of IL-17A and IFN-γ (rs = 0.3873; p = 0.0016), IL-4 (rs = 0.2671; p = 0.0328), and IL-2 (rs = 0.3959; p = 0.0012). These results suggest that the Th17 response mediated by the IL-6 pathway has a role in the prolonged IgG response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Flávia Póvoa da Costa
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Kevin Matheus Lima de Sarges
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Rosilene da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Erika Ferreira dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Matheus Holanda do Nascimento
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Alice Maciel Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Marcos Henrique Damasceno Cantanhede
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Fabíola Brasil Barbosa Rodrigues
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| | - Camille Ferreira de Oliveira
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Pablo Fabiano Moura das Neves
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Gabriel dos Santos Pereira Neto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
| | - Daniele Freitas Henriques
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil; (C.F.d.O.); (D.F.H.)
| | - Juarez Antônio Simões Quaresma
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil; (P.F.M.d.N.); (J.A.S.Q.); (L.F.M.F.)
| | - Maria Alice Freitas Queiroz
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Izaura Maria Vieira Cayres Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Giselle Maria Rachid Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
- Malaria Basic Research Laboratory, Parasitology Section, Evandro Chagas Institute, Health Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 67000-000, Pará, Brazil
| | - Eduardo José Melo dos Santos
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Pará, Brazil; (F.P.d.C.); (K.M.L.d.S.); (R.d.S.); (E.F.d.S.); (M.H.d.N.); (A.M.R.); (M.H.D.C.); (F.B.B.R.); (M.d.N.d.S.d.A.V.); (M.d.M.L.); (M.T.F.M.d.B.); (A.L.S.d.S.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Pará, Brazil; (G.d.S.P.N.); (M.A.F.Q.); (I.M.V.C.V.); (A.C.R.V.); (G.M.R.V.)
| |
Collapse
|
16
|
Roohi A, Gharagozlou S. Vitamin D supplementation and calcium: Many-faced gods or nobody in fighting against Corona Virus Disease 2019. Clin Nutr ESPEN 2024; 62:172-184. [PMID: 38901939 DOI: 10.1016/j.clnesp.2024.05.015] [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: 11/15/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
In December 2019, Corona Virus Disease 2019 (COVID-19) was first identified and designated as a pandemic in March 2020 due to rapid spread of the virus globally. At the beginning of the pandemic, only a few treatment options, mainly focused on supportive care and repurposing medications, were available. Due to its effects on immune system, vitamin D was a topic of interest during the pandemic, and researchers investigated its potential impact on COVID-19 outcomes. However, the results of studies about the impact of vitamin D on the disease are inconclusive. In the present narrative review, different roles of vitamin D regarding the COVID-19 have been discussed to show that vitamin D supplementation should be recommended carefully.
Collapse
Affiliation(s)
- Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
17
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605768. [PMID: 39109178 PMCID: PMC11302620 DOI: 10.1101/2024.07.30.605768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, Minnesota 55356, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ian Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Vanamudhu A, Devi Arumugam R, Nancy A, Selvaraj N, Moiden K, Hissar S, Ranganathan UD, Bethunaickan R, Babu S, Kumar NP. Elucidating the Immune Response to SARS-CoV-2: Natural Infection versus Covaxin/Covishield Vaccination in a South Indian Population. Viruses 2024; 16:1178. [PMID: 39205152 PMCID: PMC11360806 DOI: 10.3390/v16081178] [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: 06/04/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
A natural infection or a vaccination can initially prime the immune system to form immunological memory. The immunity engendered by vaccination against COVID-19 versus natural infection with SARS-CoV-2 has not been well studied in the Indian population. In this study, we compared the immunity conferred by COVID-19 vaccines to naturally acquired immunity to SARS-CoV-2 in a South Indian population. We examined binding and neutralizing antibody (NAb) levels against the ancestral and variant lineages and assessed the ex vivo cellular parameters of memory T cells, memory B cells, and monocytes and finally measured the circulating cytokine response. COVID-19 vaccination stimulates heightened levels of IgG antibodies against the original strain of SARS-CoV-2, as well as increased binding to the spike protein and neutralizing antibody levels. This enhanced response extends to variant lineages such as B.1.617.2 (Delta, India), B.1.1.529 (Omicron, India), B.1.351 (Beta, South Africa), and B.1.1.7 (Alpha, UK). COVID-19 vaccination differs from SARS-CoV-2 infection by having increased frequencies of classical memory B cells, activated memory B and plasma cells, CD4/CD8 T cells of effector memory, effector cells, stem cell-like memory T cells, and classical and intermediate monocytes and diminished frequencies of CD4/CD8 T cells of central memory and non-classical monocytes in vaccinated individuals in comparison to those with natural infection. Thus, COVID-19 vaccination is characterized by enhanced humoral responses and robust activation of innate and memory T cell responses in comparison to natural infection in a South Indian population.
Collapse
Affiliation(s)
- Agalya Vanamudhu
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Renuka Devi Arumugam
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Arul Nancy
- National Institutes of Health, National Institute for Research in Tuberculosis, International Center for Excellence in Research, Chennai 600031, India
| | - Nandhini Selvaraj
- National Institutes of Health, National Institute for Research in Tuberculosis, International Center for Excellence in Research, Chennai 600031, India
| | - Kadar Moiden
- National Institutes of Health, National Institute for Research in Tuberculosis, International Center for Excellence in Research, Chennai 600031, India
| | - Syed Hissar
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Uma Devi Ranganathan
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Ramalingam Bethunaickan
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Subash Babu
- National Institutes of Health, National Institute for Research in Tuberculosis, International Center for Excellence in Research, Chennai 600031, India
| | - Nathella Pavan Kumar
- Department of Immunology, ICMR, National Institute for Research in Tuberculosis, Chennai 600031, India
| |
Collapse
|
19
|
Heiskanen A, Galipeau Y, Little J, Langlois M, Cooper CL. Reduced seasonal coronavirus incidence in high-risk population groups during the COVID-19 pandemic. Immun Inflamm Dis 2024; 12:e1342. [PMID: 39023424 PMCID: PMC11256882 DOI: 10.1002/iid3.1342] [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: 01/02/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Epidemiological data on seasonal coronaviruses (sCoVs) may provide insight on transmission patterns and demographic factors that favor coronaviruses (CoVs) with greater disease severity. This study describes the incidence of CoVs in several high-risk groups in Ottawa, Canada, from October 2020 to March 2022. METHODS Serological assays quantified IgG and IgM antibodies to SARS-CoV-2, HCoV-OC43, HCoV-NL63, HCoV-HKU1, and HCoV-229E. Incident infections were compared between four population groups: individuals exposed to children, transit users, immunocompromised, and controls. Associations between antibody prevalence indicative of natural infection and demographic variables were assessed using regression analyses. RESULTS Transit users and those exposed to children were at no greater risk of infection compared to the control group. Fewer infections were detected in the immunocompromised group (p = .03). SARS-CoV-2 seroprevalence was greater in individuals with low income and within ethnic minorities. CONCLUSIONS Our findings suggest that nonpharmaceutical interventions intended to reduce SAR-CoV-2 transmission protected populations at high risk of exposure. The re-emergence of sCoVs and other common respiratory viruses alongside SARS-CoV-2 may alter infection patterns and increase the risk in vulnerable populations.
Collapse
Affiliation(s)
- Aliisa Heiskanen
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Julian Little
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Marc‐André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Centre for Infection, Immunity and Inflammation (CI3)University of OttawaOttawaOntarioCanada
| | - Curtis L. Cooper
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
| |
Collapse
|
20
|
McCoy KM, Ackerman ME, Grigoryan G. A Comparison of Antibody-Antigen Complex Sequence-to-Structure Prediction Methods and their Systematic Biases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585121. [PMID: 38979267 PMCID: PMC11230293 DOI: 10.1101/2024.03.15.585121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The ability to accurately predict antibody-antigen complex structures from their sequences could greatly advance our understanding of the immune system and would aid in the development of novel antibody therapeutics. There have been considerable recent advancements in predicting protein-protein interactions (PPIs) fueled by progress in machine learning (ML). To understand the current state of the field, we compare six representative methods for predicting antibody-antigen complexes from sequence, including two deep learning approaches trained to predict PPIs in general (AlphaFold-Multimer, RoseTTAFold), two composite methods that initially predict antibody and antigen structures separately and dock them (using antibody-mode ClusPro), local refinement in Rosetta (SnugDock) of globally docked poses from ClusPro, and a pipeline combining homology modeling with rigid-body docking informed by ML-based epitope and paratope prediction (AbAdapt). We find that AlphaFold-Multimer outperformed other methods, although the absolute performance leaves considerable room for improvement. AlphaFold-Multimer models of lower-quality display significant structural biases at the level of tertiary motifs (TERMs) towards having fewer structural matches in non-antibody containing structures from the Protein Data Bank (PDB). Specifically, better models exhibit more common PDB-like TERMs at the antibody-antigen interface than worse ones. Importantly, the clear relationship between performance and the commonness of interfacial TERMs suggests that scarcity of interfacial geometry data in the structural database may currently limit application of machine learning to the prediction of antibody-antigen interactions.
Collapse
Affiliation(s)
- Katherine Maia McCoy
- Molecular and Cell Biology Graduate Program, Dartmouth College, Hanover, New Hampshire, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Molecular and Cell Biology Graduate Program, Dartmouth College, Hanover, New Hampshire, USA
| | - Gevorg Grigoryan
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, USA
- Molecular and Cell Biology Graduate Program, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
21
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic immunity from prior SARS-CoV-2 infection but not COVID-19 vaccination associates with lower endemic coronavirus incidence. Sci Transl Med 2024; 16:eado7588. [PMID: 38865483 PMCID: PMC11565543 DOI: 10.1126/scitranslmed.ado7588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Immune responses from prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19 vaccination mitigate disease severity, but they do not fully prevent subsequent infections, especially from genetically divergent strains. We examined the incidence of and immune differences against human endemic coronaviruses (eCoVs) as a proxy for response against future genetically heterologous coronaviruses (CoVs). We assessed differences in symptomatic eCoV and non-CoV respiratory disease incidence among those with known prior SARS-CoV-2 infection or previous COVID-19 vaccination but no documented SARS-CoV-2 infection or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not previous COVID-19 vaccination alone, associates with a lower incidence of subsequent symptomatic eCoV infection. There was no difference in non-CoV incidence, implying that the observed difference was eCoV specific. In a second cohort where both cellular and humoral immunity were measured, those with prior SARS-CoV-2 spike protein exposure had lower eCoV-directed neutralizing antibodies, suggesting that neutralization is not responsible for the observed decreased eCoV disease. The three groups had similar cellular responses against the eCoV spike protein and nucleocapsid antigens. However, CD8+ T cell responses to the nonstructural eCoV proteins nsp12 and nsp13 were higher in individuals with previous SARS-CoV-2 infection as compared with the other groups. This association between prior SARS-CoV-2 infection and decreased incidence of eCoV disease may therefore be due to a boost in CD8+ T cell responses against eCoV nsp12 and nsp13, suggesting that incorporation of nonstructural viral antigens in a future pan-CoV vaccine may improve vaccine efficacy.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
22
|
Filippatos F, Tatsi EB, Dourdouna MM, Zoumakis E, Margeli A, Syriopoulou V, Michos A. SARS-CoV-2 Seroepidemiology and Antibody Levels in Children during BA.5 Predominance Period. Diagnostics (Basel) 2024; 14:1039. [PMID: 38786337 PMCID: PMC11120608 DOI: 10.3390/diagnostics14101039] [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: 04/13/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
This is a SARS-CoV-2 seroepidemiological study in a pediatric population (0-16 years) during the BA.5 Omicron predominance period in the Athens metropolitan area. Serum samples were tested for SARS-CoV-2 nucleocapsid antibodies (Abs-N), representing natural infection during three periods of BA.5 predominance: 1 May 2022-31 August 2022 (period A), 1 September 2022-31 December 2022 (period B), and July 2023 (period C). Εpidemiological data were also collected. Additionally, in period C, Abs-N-seronegative samples were tested for SARS-CoV-2 spike antibodies (Abs-S). A total of 878 children were tested (males: 52.6%), with a median age (IQR) of 96 (36-156) months; the number of cases of seropositivity during the three periods were as follows: A: 292/417 (70%), B: 288/356 (80.9%), and C: 89/105 (84.8%), with p < 0.001. SARS-CoV-2 seropositivity increased from period A to C for children 0-1 year (p = 0.044), >1-4 years (p = 0.028), and >6-12 years (p = 0.003). Children > 6-12 years had the highest seropositivity rates in all periods (A: 77.3%, B: 91.4%, and C: 95.8%). A significant correlation of monthly median Abs-N titers with monthly seropositivity rates was detected (rs: 0.812, p = 0.008). During period C, 12/105 (11.4%) Abs-S-seropositive and Abs-N-seronegative samples were detected and total seropositivity was estimated at 96.2% (101/105). The findings of this study indicate a high SARS-CoV-2 exposure rate of children during the BA.5 predominance period and suggest that in future seroepidemiological studies, both antibodies should be tested in Abs-N-seronegative populations.
Collapse
Affiliation(s)
- Filippos Filippatos
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| | - Elizabeth-Barbara Tatsi
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| | - Maria-Myrto Dourdouna
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| | - Emmanouil Zoumakis
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| | - Alexandra Margeli
- Department of Clinical Biochemistry, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Vasiliki Syriopoulou
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| | - Athanasios Michos
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (F.F.); (E.-B.T.); (M.-M.D.); (E.Z.); (A.M.)
| |
Collapse
|
23
|
Wietschel KA, Fechtner K, Antileo E, Abdurrahman G, Drechsler CA, Makuvise MK, Rose R, Voß M, Krumbholz A, Michalik S, Weiss S, Ulm L, Franikowski P, Fickenscher H, Bröker BM, Raafat D, Holtfreter S. Non-cross-reactive epitopes dominate the humoral immune response to COVID-19 vaccination - kinetics of plasma antibodies, plasmablasts and memory B cells. Front Immunol 2024; 15:1382911. [PMID: 38807606 PMCID: PMC11130424 DOI: 10.3389/fimmu.2024.1382911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction COVID-19 vaccines are highly effective in inducing protective immunity. While the serum antibody response to COVID-19 vaccination has been studied in depth, our knowledge of the underlying plasmablast and memory B cell (Bmem) responses is still incomplete. Here, we determined the antibody and B cell response to COVID-19 vaccination in a naïve population and contrasted it with the response to a single influenza vaccination in a primed cohort. In addition, we analyzed the antibody and B cell responses against the four endemic human coronaviruses (HCoVs). Methods Measurement of specific plasma IgG antibodies was combined with functional analyses of antibody-secreting plasmablasts and Bmems. SARS-CoV-2- and HCoV-specific IgG antibodies were quantified with an in-house bead-based multiplexed immunoassay. Results The antibody and B cell responses to COVID-19 vaccination reflected the kinetics of a prime-boost immunization, characterized by a slow and moderate primary response and a faster and stronger secondary response. In contrast, the influenza vaccinees possessed robust immune memory for the vaccine antigens prior to vaccination, and the recall vaccination moderately boosted antibody production and Bmem responses. Antibody levels and Bmem responses waned several months after the 2nd COVID-19 vaccination, but were restored upon the 3rd vaccination. The COVID-19 vaccine-induced antibodies mainly targeted novel, non-cross-reactive S1 epitopes of the viral spike protein, while cross-reactive S2 epitopes were less immunogenic. Booster vaccination not only strongly enhanced neutralizing antibodies against an original SARS-CoV-2 strain, but also induced neutralizing antibodies against the Omicron BA.2 variant. We observed a 100% plasma antibody prevalence against the S1 subunits of HCoVs, which was not affected by vaccination. Discussion Overall, by complementing classical serology with a functional evaluation of plasmablasts and memory B cells we provide new insights into the specificity of COVID-19 vaccine-induced antibody and B cell responses.
Collapse
Affiliation(s)
- Kilian A. Wietschel
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Kevin Fechtner
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Elmer Antileo
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Chiara A. Drechsler
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Ruben Rose
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Mathias Voß
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Andi Krumbholz
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
- Labor Dr. Krause und Kollegen MVZ GmbH, Kiel, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Lena Ulm
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Philipp Franikowski
- Institute for Educational Quality Improvement, Humboldt University of Berlin, Berlin, Germany
| | - Helmut Fickenscher
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
24
|
Lugo-Trampe A, López-Cifuentes D, Mendoza-Pérez P, Tafurt-Cardona Y, Joo-Domínguez ADJ, Rios-Ibarra CP, Espinoza-Ruiz M, Chang-Rueda C, Rodriguez-Sanchez IP, Martinez-Fierro ML, Delgado-Enciso I, Trujillo-Murillo KDC. Nine-Month Trend of IgG Antibody Persistence and Associated Symptoms Post-SARS-CoV-2 Infection. Healthcare (Basel) 2024; 12:948. [PMID: 38727505 PMCID: PMC11083704 DOI: 10.3390/healthcare12090948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Between 2 and 8.5% of patients who recover from COVID-19 do not develop antibodies, and the durability of IgG antibodies is under scrutiny. Therefore, the presence and persistence of IgM and IgG antibodies were evaluated in a group of patients diagnosed with SARS-CoV-2 from May to August 2020. Out of 2199 suspected COVID-19 cases, 1264 were confirmed for SARS-CoV-2 by rRT-PCR; 328 consented to participate in the study, with 220 participants followed for 9 months, including 124 men (56%) and 96 women (44%). The primary symptoms were headache, dry cough, and fever. IgG antibodies developed in 95% of patients within 4 weeks post-diagnosis, and a second evaluation at 9 months showed that 72.7% still had detectable IgG antibodies. The presence of IgM in one individual (0.45%) suggested the possibility of reinfection.
Collapse
Affiliation(s)
- Angel Lugo-Trampe
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
- Genodiagnóstica SA de CV, Tapachula, Chiapas 30700, Mexico
| | - Daniel López-Cifuentes
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Paúl Mendoza-Pérez
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Yaliana Tafurt-Cardona
- Faculty of Human Medicine, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | | | - Clara Patricia Rios-Ibarra
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco (CIATEJ), Guadalajara 44270, Mexico
| | - Marisol Espinoza-Ruiz
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Consuelo Chang-Rueda
- Faculty of Chemistry Sciences, Campus IV, Universidad Autónoma de Chiapas, Tapachula 30700, Mexico
| | - Iram Pablo Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Iván Delgado-Enciso
- School of Medicine, University of Colima, Colima 28040, Mexico
- Colima Cancerology State Institute, IMSS-Bienestar, Colima 28085, Mexico
| | | |
Collapse
|
25
|
Yadav PD, Patil DY, Sahay RR, Shete AM, Mohandas S, Nair V. The impact of Omicron on the COVID-19 vaccines: A review. VACUNAS 2024; 25:274-284. [DOI: 10.1016/j.vacun.2024.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
26
|
Gardner BJ, Kilpatrick AM. Predicting Vaccine Effectiveness for Hospitalization and Symptomatic Disease for Novel SARS-CoV-2 Variants Using Neutralizing Antibody Titers. Viruses 2024; 16:479. [PMID: 38543844 PMCID: PMC10975673 DOI: 10.3390/v16030479] [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: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 05/23/2024] Open
Abstract
The emergence of new virus variants, including the Omicron variant (B.1.1.529) of SARS-CoV-2, can lead to reduced vaccine effectiveness (VE) and the need for new vaccines or vaccine doses if the extent of immune evasion is severe. Neutralizing antibody titers have been shown to be a correlate of protection for SARS-CoV-2 and other pathogens, and could be used to quickly estimate vaccine effectiveness for new variants. However, no model currently exists to provide precise VE estimates for a new variant against severe disease for SARS-CoV-2 using robust datasets from several populations. We developed predictive models for VE against COVID-19 symptomatic disease and hospitalization across a 54-fold range of mean neutralizing antibody titers. For two mRNA vaccines (mRNA-1273, BNT162b2), models fit without Omicron data predicted that infection with the BA.1 Omicron variant increased the risk of hospitalization 2.8-4.4-fold and increased the risk of symptomatic disease 1.7-4.2-fold compared to the Delta variant. Out-of-sample validation showed that model predictions were accurate; all predictions were within 10% of observed VE estimates and fell within the model prediction intervals. Predictive models using neutralizing antibody titers can provide rapid VE estimates, which can inform vaccine booster timing, vaccine design, and vaccine selection for new virus variants.
Collapse
Affiliation(s)
- Billy J. Gardner
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| | - A. Marm Kilpatrick
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| |
Collapse
|
27
|
Mazzara C, Bassi J, Silacci-Fregni C, Muoio F, Passini N, Corti D, Simonetti GD, Vanoni F, Kottanattu L, Piccoli L. Increased breadth and neutralization of antibodies against SARS-CoV-2 variants after infection and vaccination: A serosurveillance study in pediatric patients of Southern Switzerland. Eur J Pediatr 2024; 183:1425-1434. [PMID: 38175262 DOI: 10.1007/s00431-023-05400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Little information is available about the nature of the immune response in children after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. The aim of this study is to define the seroprevalence and the features of the antibody response in children of Southern Switzerland during the different waves of Coronavirus Disease 2019 (COVID-19) pandemic. By analyzing 756 sera collected from children aged 0 to 16 years admitted to the Institute of Pediatrics of Southern Switzerland during the prepandemic period (before March 2020) and the first four pandemic waves (between March 2020 and June 2022), we investigated binding titers, cross-reactivity, and neutralizing properties of the serum antibodies against SARS-CoV-2 variants. Seroprevalence varied from 6% during the first wave to 14% and 17% during the second and third waves, respectively, peaking at 39% during the fourth wave. The 96 seropositive cases were mostly asymptomatic (42.7%) or showed mild (20.8%) to moderate (32.3%) symptoms. Moderate symptoms and close contact with COVID-19-positive individuals were associated with a higher infection risk (P < 0.001). The antibody response was mainly driven by IgG directed to the receptor-binding domain (RBD) of Wuhan-1 SARS-CoV-2 Spike (S). Children infected in the first three waves produced antibodies with up to 11-fold and 5.5-fold reduction in binding and neutralizing titers, respectively, against different SARS-CoV-2 variants, including Beta, Delta, and Omicron BA.1, BA.2, and BA.5. Such reductions were less pronounced in children infected during the fourth wave, who showed the highest frequency and titers of neutralizing antibodies against the same variants. Compared to infection, vaccination with a Wuhan-1-based messenger RNA (mRNA) vaccine induced higher and heterogenous levels of antibodies cross-reacting to the different SARS-CoV-2 variants analyzed. Conclusions: Despite the high burden of COVID-19 in Southern Switzerland, we observed an initial low seroprevalence of SARS-CoV-2 in children, which increased in the later waves. The antibody response was poor in the first three waves and improved in the fourth wave, when children produced higher levels of neutralizing antibodies after vaccination or infection with Delta and/or Omicron variants. What is Known: • Children were marginally affected by the initial SARS-CoV-2 variants. • The number of infected and hospitalized children increased after the appearance of the Omicron variants. What is New: • Seroprevalence of SARS-CoV-2 in children of Southern Switzerland increased overtime. • Children produced higher levels of neutralizing antibodies after vaccination or infection with Delta and/or Omicron variants in the fourth wave compared to children infected in the first three waves.
Collapse
Affiliation(s)
- Calogero Mazzara
- Institute of Pediatrics of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Jessica Bassi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Francesco Muoio
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nadia Passini
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Giacomo D Simonetti
- Institute of Pediatrics of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Federica Vanoni
- Institute of Pediatrics of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Lisa Kottanattu
- Institute of Pediatrics of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland.
| |
Collapse
|
28
|
Leon-Rojas JE, Veloz T, Teran J, Perez M, Arias-Erazo F, Villacis L, Velez J, Recalde R, Jiménez P, Martin M, Chis Ster I, Cooper P, Romero N. The dynamics and determinants of specific systemic and mucosal antibody responses to SARS-CoV-2 in three adult cohorts in the Ecuadorian Andes: a study protocol. F1000Res 2024; 11:1392. [PMID: 38434000 PMCID: PMC10905138 DOI: 10.12688/f1000research.126577.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction There are limited longitudinal data on the systemic and mucosal antibody responses to SARS-CoV-2 from Latin America, a region severely affected by COVID-19, and where vaccine strategies have been implemented during the evolving pandemic. Objective To evaluate determinants of seroprevalence and changes in levels of anti-SARS-CoV-2 antibodies longitudinally in adults with different levels of exposure to SARS-CoV-2 (defined a priori as low, medium, and high based on presumed occupational risk), in two Andean cities in Ecuador. Methods Longitudinal cohort study of 1,000 adults aged 18 years and older with questionnaire data and sample collection done at 0, 3, 6, and 12 months during the period 2020-2023. Observations collected included WHO-ISARIC questionnaire and peripheral blood and saliva samples for measurement of IgG and IgA antibodies, respectively. Planned analyses are tailored to the longitudinal nature of the outcomes defined by participants' antibody levels and aim at estimating their average trends with time since infection in each of the occupational groups, adjusted for demographics and calendar-time levels of SARS-CoV-2 infection in the general population. The latter reflect the impact of the national control measures such as vaccinations and movement restrictions. Importance Understanding the duration and the dynamics of waning immunity to SARS-CoV-2, in the context of exposures to emerging virus variants and immunization, will inform the implementation of targeted public health strategies in the Latin American region. Ethics and Dissemination This study will observe the bioethical principles of the Declaration of Helsinki. Informed written consent will be obtained. Samples from participants will be stored for up to three years after which they will be destroyed. The study protocol was approved by the Ecuadorian Ministry of Public Health Ethics Committee for COVID-19 Research. Antibody results will be provided to participants and participating institutions and to the national health authorities.
Collapse
Affiliation(s)
- Jose E. Leon-Rojas
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Red Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL),, Quito, Ecuador
| | - Tatiana Veloz
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Jair Teran
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Monica Perez
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Fernanda Arias-Erazo
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
- Departamento de Ciencias de la Vida y de la Agricultura, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Lizet Villacis
- Medical School, Universidad Técnica de Ambato, Ambato, Ecuador
| | - Jorge Velez
- Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
| | - Ricardo Recalde
- Red Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL),, Quito, Ecuador
- Medical School, Universidad Técnica de Ambato, Ambato, Ecuador
| | - Patricia Jiménez
- Departamento de Ciencias de la Vida y de la Agricultura, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
- Grupo de Investigación en Sanidad Animal y Humana (GISAH), Quito, Ecuador
| | - Miguel Martin
- Departamento de Pediatría, Obstetricia y Ginecología y Medicina Preventiva, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
- Red Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL),, Quito, Ecuador
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
| | | | - Philip Cooper
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
- St George's University of London, London, UK
| | - Natalia Romero
- Red Grups de Recerca d’Amèrica i Àfrica Llatines (GRAAL),, Quito, Ecuador
- Medical School, Universidad Internacional del Ecuador, Quito, Ecuador
| |
Collapse
|
29
|
McKendry R, Lemm NM, Papargyris L, Chiu C. Human Challenge Studies with Coronaviruses Old and New. Curr Top Microbiol Immunol 2024; 445:69-108. [PMID: 35181805 DOI: 10.1007/82_2021_247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus infections have been known to cause disease in animals since as early as the 1920s. However, only seven coronaviruses capable of causing human disease have been identified thus far. These Human Coronaviruses (HCoVs) include the causes of the common cold, but more recent coronaviruses that have emerged (i.e. SARS-CoV, MERS-CoV and SARS-CoV-2) are associated with much greater morbidity and mortality. HCoVs have been relatively under-studied compared to other common respiratory infections, as historically they have presented with mild symptoms. This has led to a relatively limited understanding of their animal reservoirs, transmission and determinants of immune protection. To address this, human infection challenge studies with HCoVs have been performed that enable a detailed clinical and immunological analysis of the host response at specific time points under controlled conditions with standardised viral inocula. Until recently, all such human challenge studies were conducted with common cold HCoVs, with the study of SARS-CoV and MERS-CoV unacceptable due to their greater pathogenicity. However, with the emergence of SARS-CoV-2 and the COVID-19 pandemic during which severe outcomes in young healthy adults have been rare, human challenge studies with SARS-CoV-2 are now being developed. Two SARS-CoV-2 human challenge studies in the UK studying individuals with and without pre-existing immunity are underway. As well as providing a platform for testing of antivirals and vaccines, such studies will be critical for understanding the factors associated with susceptibility to SARS-CoV-2 infection and thus developing improved strategies to tackle the current as well as future HCoV pandemics. Here, we summarise the major questions about protection and pathogenesis in HCoV infection that human infection challenge studies have attempted to answer historically, as well as the knowledge gaps that aim to be addressed with contemporary models.
Collapse
Affiliation(s)
- Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
30
|
Fung CYJ, Scott M, Lerner-Ellis J, Taher J. Applications of SARS-CoV-2 serological testing: impact of test performance, sample matrices, and patient characteristics. Crit Rev Clin Lab Sci 2024; 61:70-88. [PMID: 37800891 DOI: 10.1080/10408363.2023.2254390] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
Laboratory testing has been a key tool in managing the SARS-CoV-2 global pandemic. While rapid antigen and PCR testing has proven useful for diagnosing acute SARS-CoV-2 infections, additional testing methods are required to understand the long-term impact of SARS-CoV-2 infections on immune response. Serological testing, a well-documented laboratory practice, measures the presence of antibodies in a sample to uncover information about host immunity. Although proposed applications of serological testing for clinical use have previously been limited, current research into SARS-CoV-2 has shown growing utility for serological methods in these settings. To name a few, serological testing has been used to identify patients with past infections and long-term active disease and to monitor vaccine efficacy. Test utility and result interpretation, however, are often complicated by factors that include poor test sensitivity early in infection, lack of immune response in some individuals, overlying infection and vaccination responses, lack of standardization of antibody titers/levels between instruments, unknown titers that confer immune protection, and large between-individual biological variation following infection or vaccination. Thus, the three major components of this review will examine (1) factors that affect serological test utility: test performance, testing matrices, seroprevalence concerns and viral variants, (2) patient factors that affect serological response: timing of sampling, age, sex, body mass index, immunosuppression and vaccination, and (3) informative applications of serological testing: identifying past infection, immune surveillance to guide health practices, and examination of protective immunity. SARS-CoV-2 serological testing should be beneficial for clinical care if it is implemented appropriately. However, as with other laboratory developed tests, use of SARS-CoV-2 serology as a testing modality warrants careful consideration of testing limitations and evaluation of its clinical utility.
Collapse
Affiliation(s)
- Chun Yiu Jordan Fung
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Mackenzie Scott
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Lerner-Ellis
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Taher
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Stern D, Meyer TC, Treindl F, Mages HW, Krüger M, Skiba M, Krüger JP, Zobel CM, Schreiner M, Grossegesse M, Rinner T, Peine C, Stoliaroff-Pépin A, Harder T, Hofmann N, Michel J, Nitsche A, Stahlberg S, Kneuer A, Sandoni A, Kubisch U, Schlaud M, Mankertz A, Schwarz T, Corman VM, Müller MA, Drosten C, de la Rosa K, Schaade L, Dorner MB, Dorner BG. A bead-based multiplex assay covering all coronaviruses pathogenic for humans for sensitive and specific surveillance of SARS-CoV-2 humoral immunity. Sci Rep 2023; 13:21846. [PMID: 38071261 PMCID: PMC10710470 DOI: 10.1038/s41598-023-48581-9] [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: 08/29/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Serological assays measuring antibodies against SARS-CoV-2 are key to describe the epidemiology, pathobiology or induction of immunity after infection or vaccination. Of those, multiplex assays targeting multiple antigens are especially helpful as closely related coronaviruses or other antigens can be analysed simultaneously from small sample volumes, hereby shedding light on patterns in the immune response that would otherwise remain undetected. We established a bead-based 17-plex assay detecting antibodies targeting antigens from all coronaviruses pathogenic for humans: SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV strains 229E, OC43, HKU1, and NL63. The assay was validated against five commercial serological immunoassays, a commercial surrogate virus neutralisation test, and a virus neutralisation assay, all targeting SARS-CoV-2. It was found to be highly versatile as shown by antibody detection from both serum and dried blot spots and as shown in three case studies. First, we followed seroconversion for all four endemic HCoV strains and SARS-CoV-2 in an outbreak study in day-care centres for children. Second, we were able to link a more severe clinical course to a stronger IgG response with this 17-plex-assay, which was IgG1 and IgG3 dominated. Finally, our assay was able to discriminate recent from previous SARS-CoV-2 infections by calculating the IgG/IgM ratio on the N antigen targeting antibodies. In conclusion, due to the comprehensive method comparison, thorough validation, and the proven versatility, our multiplex assay is a valuable tool for studies on coronavirus serology.
Collapse
Affiliation(s)
- Daniel Stern
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany.
| | - Tanja C Meyer
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Fridolin Treindl
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Hans Werner Mages
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Maren Krüger
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Martin Skiba
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Jan Philipp Krüger
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Berlin, Berlin, Germany
| | - Christian M Zobel
- Department of Internal Medicine, Bundeswehr Hospital Berlin, Berlin, Germany
| | | | - Marica Grossegesse
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Thomas Rinner
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Caroline Peine
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Anna Stoliaroff-Pépin
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Thomas Harder
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Natalie Hofmann
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Janine Michel
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Andreas Nitsche
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Silke Stahlberg
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Antje Kneuer
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Anna Sandoni
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Ulrike Kubisch
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Martin Schlaud
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Annette Mankertz
- Measles, Mumps, Rubella, and Viruses Affecting Immunocompromised Patients (FG 12), Robert Koch Institute, 13353, Berlin, Germany
| | - Tatjana Schwarz
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Corporate Member, Freie Universität Berlin, 10117, Berlin, Germany
- Corporate Member, Humboldt-Universität zu Berlin, 14195, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Kathrin de la Rosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
- Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Lars Schaade
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Martin B Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Brigitte G Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany.
| |
Collapse
|
32
|
Tong MZW, Sng JDJ, Carney M, Cooper L, Brown S, Lineburg KE, Chew KY, Collins N, Ignacio K, Airey M, Burr L, Joyce BA, Jayasinghe D, McMillan CLD, Muller DA, Adhikari A, Gallo LA, Dorey ES, Barrett HL, Gras S, Smith C, Good‐Jacobson K, Short KR. Elevated BMI reduces the humoral response to SARS-CoV-2 infection. Clin Transl Immunology 2023; 12:e1476. [PMID: 38050635 PMCID: PMC10693902 DOI: 10.1002/cti2.1476] [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: 09/17/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
Objective Class III obesity (body mass index [BMI] ≥ 40 kg m-2) significantly impairs the immune response to SARS-CoV-2 vaccination. However, the effect of an elevated BMI (≥ 25 kg m-2) on humoral immunity to SARS-CoV-2 infection and COVID-19 vaccination remains unclear. Methods We collected blood samples from people who recovered from SARS-CoV-2 infection approximately 3 and 13 months of post-infection (noting that these individuals were not exposed to SARS-CoV-2 or vaccinated in the interim). We also collected blood samples from people approximately 5 months of post-second dose COVID-19 vaccination (the majority of whom did not have a prior SARS-CoV-2 infection). We measured their humoral responses to SARS-CoV-2, grouping individuals based on a BMI greater or less than 25 kg m-2. Results Here, we show that an increased BMI (≥ 25 kg m-2), when accounting for age and sex differences, is associated with reduced antibody responses after SARS-CoV-2 infection. At 3 months of post-infection, an elevated BMI was associated with reduced antibody titres. At 13 months of post-infection, an elevated BMI was associated with reduced antibody avidity and a reduced percentage of spike-positive B cells. In contrast, no significant association was noted between a BMI ≥ 25 kg m-2 and humoral immunity to SARS-CoV-2 at 5 months of post-secondary vaccination. Conclusions Taken together, these data showed that elevated BMI is associated with an impaired humoral immune response to SARS-CoV-2 infection. The impairment of infection-induced immunity in individuals with a BMI ≥ 25 kg m-2 suggests an added impetus for vaccination rather than relying on infection-induced immunity.
Collapse
Affiliation(s)
- Marcus ZW Tong
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Julian DJ Sng
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Meagan Carney
- School of Mathematics and PhysicsThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Cooper
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Samuel Brown
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Katie E Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Keng Yih Chew
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Neve Collins
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Kirsten Ignacio
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Megan Airey
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Burr
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- Department of Respiratory MedicineMater HealthBrisbaneQLDAustralia
| | - Briony A Joyce
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Dhilshan Jayasinghe
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Christopher LD McMillan
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - David A Muller
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - Anurag Adhikari
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Linda A Gallo
- School of HealthUniversity of the Sunshine CoastPetrieQLDAustralia
| | - Emily S Dorey
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Helen L Barrett
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
- University of New South Wales MedicineKensingtonNSWAustralia
- Obstetric MedicineRoyal Hospital for WomenRandwickNSWAustralia
| | - Stephanie Gras
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Kim Good‐Jacobson
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Kirsty R Short
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
33
|
Wang M, Zhou B, Fan Q, Zhou X, Liao X, Lin J, Ma Z, Dong J, Wang H, Ge X, Ju B, Zhang Z. Omicron variants escape the persistent SARS-CoV-2-specific antibody response in 2-year COVID-19 convalescents regardless of vaccination. Emerg Microbes Infect 2023; 12:2151381. [PMID: 36444724 PMCID: PMC9809350 DOI: 10.1080/22221751.2022.2151381] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With the ongoing COVID-19 pandemic and the emergence of various SARS-CoV-2 variants, a comprehensive evaluation of long-term efficacy of antibody response in convalescent individuals is urgently needed. Several longitudinal studies had reported the antibody dynamics after SARS-CoV-2 acute infection, but the follow-up was mostly limited to 1 year or 18 months at the maximum. In this study, we investigated the durability, potency, and susceptibility to immune evasion of SARS-CoV-2-specific antibody in COVID-19 convalescents for 2 years after discharge. These results showed the persistent antibody-dependent immunity could protect against the WT and Delta variant to some extent. However, the Omicron variants (BA.1, BA.2, and BA.4/5) largely escaped this preexisting immunity in recovered individuals. Furthermore, we revealed that inactivated vaccines (BBIBP-CorV, CoronaVac, or KCONVAC) could improve the plasma neutralization and help to maintain the broadly neutralizing antibodies at a certain level. Notably, with the time-dependent decline of antibody, 1-dose or 2-dose vaccination strategy seemed not to be enough to provide immune protection against the emerging variants. Overall, these results facilitated our understanding of SARS-CoV-2-induced antibody memory, contributing to the development of immunization strategy against SARS-CoV-2 variants for such a large number of COVID-19 survivors.
Collapse
Affiliation(s)
- Miao Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Bing Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Qing Fan
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xinrong Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jingyan Lin
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Zhenghua Ma
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Jingke Dong
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Xiangyang Ge
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China,Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, People’s Republic of China, Bin Ju Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Zheng Zhang Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong518112, People’s Republic of China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China,Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, People’s Republic of China,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, People’s Republic of China, Bin Ju Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Zheng Zhang Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong518112, People’s Republic of China; Guangdong Key Laboratory for Anti-infection Drug Quality Evaluation, Shenzhen, Guangdong518112, People’s Republic of China; Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong518112, People’s Republic of China
| |
Collapse
|
34
|
Joseph JO, Ylade M, Daag JV, Aogo R, Crisostomo MV, Mpingabo P, Premkumar L, Deen J, Katzelnick L. High transmission of endemic human coronaviruses before and during the COVID-19 pandemic in adolescents in Cebu, Philippines. RESEARCH SQUARE 2023:rs.3.rs-3581033. [PMID: 38014070 PMCID: PMC10680936 DOI: 10.21203/rs.3.rs-3581033/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background SARS-CoV-2, the causative agent of COVID-19, is a betacoronavirus belonging to the same genus as endemic human coronaviruses (hCoVs) OC43 and HKU1 and is distinct from alpha hCoVs 229E and NL63. In a study of adolescents in the Philippines, we evaluated the seroprevalence to hCoVs, whether pre-pandemic hCoV immunity modulated subsequent risk of SARS-CoV-2 infection, and if SARS-CoV-2 infection affected the transmission of the hCoVs. Methods From 499 samples collected in 2021 and screened by SARS-CoV-2 receptor binding domain (RBD) enzyme-linked immunosorbent assay (ELISA), we randomly selected 59 SARS-CoV-2 negative and 61 positive individuals for further serological evaluation. We measured RBD and spike antibodies to the four hCoVs and SARS-CoV-2 by ELISA in samples from the same participants collected pre-pandemic (2018-2019) and mid-pandemic (2021), before COVID-19 vaccination. Results We observed over 72% seropositivity to the four hCoVs pre-pandemic. Binding antibodies increased with age to 229E and OC43, suggesting endemic circulation, while immunity was flat across ages for HKU1 and NL63. During the COVID-19 pandemic, antibody level increased significantly to the RBDs of OC43, NL63, and 229E and spikes of all four hCoVs in both SARS-CoV-2 negative and positive adolescents. Those aged 12-15 years old in 2021 had higher antibodies to RBD and spike of OC43, NL63, and 229E than adolescents the same age in 2019, further demonstrating intense transmission of the hCoVs during the pandemic. Conclusions We observe a limited impact of the COVID-19 pandemic on endemic hCoV transmission. This study provides insight into co-circulation of hCoVs and SARS-CoV-2.
Collapse
Affiliation(s)
| | - Michelle Ylade
- National Institutes of Health, University of the Philippines-Manila
| | | | | | | | | | | | - Jacqueline Deen
- National Institutes of Health, University of the Philippines-Manila
| | | |
Collapse
|
35
|
Dowell AC, Tut G, Begum J, Bruton R, Bentley C, Butler M, Uwenedi G, Zuo J, Powell AA, Brent AJ, Brent B, Baawuah F, Okike I, Beckmann J, Ahmad S, Aiano F, Garstang J, Ramsay ME, Moss P, Ladhani SN. Nasal mucosal IgA levels against SARS-CoV-2 and seasonal coronaviruses are low in children but boosted by reinfection. J Infect 2023; 87:403-412. [PMID: 37660754 DOI: 10.1016/j.jinf.2023.08.013] [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: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Repeated coronavirus infections in childhood drive progressive maturation of systemic immune responses into adulthood. Analyses of immune responses in children have focused primarily upon systemic assessment but the importance of mucosal immunity is increasingly recognised. We studied virus-specific antibody responses in contemporaneous nasal swabs and blood samples from 99 children (4-15 years) and 28 adults (22-56 years), all of whom had prior SARS-CoV-2 infection. Whilst mucosal IgA titres against Influenza and Respiratory Syncytial virus were comparable between children and adults, those against all coronaviruses, including SARS-CoV-2, were lower in children. Mucosal IgA antibodies demonstrated comparable relative neutralisation capacity in both groups and retained activity against recent omicron variants such as XBB.1 which are highly evasive of IgG neutralisation. SARS-CoV-2 reinfection preferentially enhanced mucosal IgA responses whilst the impact of vaccination was more modest. Nasal IgA levels against coronaviruses thus display a pattern of incremental response to reinfection which likely determines the natural history of reinfection. This highlights the particular significance of developing mucosal vaccines against coronaviruses in children.
Collapse
Affiliation(s)
- Alexander C Dowell
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gokhan Tut
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jusnara Begum
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Bruton
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christopher Bentley
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Megan Butler
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Grace Uwenedi
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jianmin Zuo
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Annabel A Powell
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom
| | - Andrew J Brent
- Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, United Kingdom; University of Oxford, Wellington Square, Oxford, United Kingdom
| | - Bernadette Brent
- Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, United Kingdom
| | - Frances Baawuah
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom
| | - Ifeanyichukwu Okike
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom; University Hospitals of Derby and Burton NHS Foundation Trust, Uttoxeter New Road, Derby, United Kingdom
| | - Joanne Beckmann
- East London NHS Foundation Trust, 9 Allie Street, London, United Kingdom
| | - Shazaad Ahmad
- Manchester University NHS Foundation Trust, Oxford Road, Manchester, United Kingdom
| | - Felicity Aiano
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom
| | - Joanna Garstang
- Birmingham Community Healthcare NHS Trust, Holt Street, Aston, United Kingdom
| | - Mary E Ramsay
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom
| | - Paul Moss
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.
| | - Shamez N Ladhani
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, United Kingdom.
| |
Collapse
|
36
|
Familiar-Macedo D, Vieira Damasco P, Fiestas Solórzano VE, Carnevale Rodrigues J, Sampaio de Lemos ER, Barreto Dos Santos F, Agudo Mendonça Teixeira de Siqueira M, Leal de Azeredo E, de-Oliveira-Pinto LM. Inflammatory and cytotoxic mediators in COVID-19 patients and in ChAdOx1 nCoV-19 (AZD1222) vaccine recipients. Cytokine 2023; 171:156350. [PMID: 37672863 DOI: 10.1016/j.cyto.2023.156350] [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: 02/22/2023] [Revised: 07/14/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
Immunological and cytotoxic mediators are induced in natural infection and are essential for the effectiveness of vaccination. Vaccination is useful to prevent the spread of SARS-CoV-2 and limit the morbidity/mortality of COVID-19. ChAdOx1 nCoV-19 is one of the most widespread vaccines in the world. We compared the detection of anti-S1 SARS-CoV2 IgG and the profile of inflammatory and cytotoxic responses of patients who developed different clinical outcomes of COVID-19 with individuals previously exposed or not to the virus received the first and booster doses of ChAdOx1 nCoV-19. Plasma from 35 patients with COVID-19 and 11 vaccinated were evaluated by multiplex assay. Here, no vaccinated subjects had serious adverse effects. Those vaccinated with a booster dose had higher anti-S1 IgG than mild/moderate and recovered patients. Critically ill and deceased patients had IgG levels like those immunized. By univariate analysis, IL-2, IL-17, and perforin do not differentiate between patients and vaccinated individuals. Granzyme A increased at dose 1, while patients had their levels reduced. High levels of granulysin, sFas, and IL-6 were detected in the deaths, but after vaccination, all were declined. The multivariate analysis supports the role of IL-6 and granulysin as associated and non-confounding variables related to the worst clinical outcome of COVID-19, but not sFas. Our data confirm the ability of the ChAdOx1 vaccine to produce specific antibody levels up to booster time. Furthermore, our data suggest that the vaccine can regulate both the hyper-production and the kinetics of the production of inflammatory and cytotoxic mediators involved in the cytokine storm, such as granulysin and IL-6.
Collapse
Affiliation(s)
- Débora Familiar-Macedo
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Paulo Vieira Damasco
- Rede Casa Hospital Rio Laranjeiras e Rio Botafogo, Rio de Janeiro 22240-000, Brazil
| | - Victor Edgar Fiestas Solórzano
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Joyce Carnevale Rodrigues
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil
| | - Elba Regina Sampaio de Lemos
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Flávia Barreto Dos Santos
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Marilda Agudo Mendonça Teixeira de Siqueira
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais (LVRE), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Elzinandes Leal de Azeredo
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Luzia Maria de-Oliveira-Pinto
- Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
37
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic responses against nsp12/nsp13 from prior SARS-CoV-2 infection associates with lower subsequent endemic coronavirus incidence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563621. [PMID: 37961343 PMCID: PMC10634759 DOI: 10.1101/2023.10.23.563621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Immune responses from prior SARS-CoV-2 infection and COVID-19 vaccination do not prevent re-infections and may not protect against future novel coronaviruses (CoVs). We examined the incidence of and immune differences against human endemic CoVs (eCoV) as a proxy for response against future emerging CoVs. Assessment was among those with known SARS-CoV-2 infection, COVID-19 vaccination but no documented SARS-CoV-2 infection, or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not COVID-19 vaccination alone, protects against subsequent symptomatic eCoV infection. CD8+ T cell responses to the non-structural eCoV proteins, nsp12 and nsp13, were significantly higher in individuals with previous SARS-CoV-2 infection as compared to the other groups. The three groups had similar cellular responses against the eCoV spike and nucleocapsid, and those with prior spike exposure had lower eCoV-directed neutralizing antibodies. Incorporation of non-structural viral antigens in a future pan-CoV vaccine may improve protection against future heterologous CoV infections.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| |
Collapse
|
38
|
Mishra KP, Singh M, Saraswat D, Singh S. Development of ELISA-Based Assay for Detection of SARS-CoV-2 Neutralizing Antibody. Viral Immunol 2023; 36:495-502. [PMID: 37643285 DOI: 10.1089/vim.2023.0058] [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] [Indexed: 08/31/2023] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the plasma B cells to secrete specific antibodies against the viral antigen. However, not all antibodies can prevent the virus from entering the cells. The subpopulation of antibodies which blocks the entry of the virus into host cells is termed neutralizing antibodies (NAbs). The gold standard test for the detection of NAbs is the viral plaque reduction and neutralization test; however, various other methods can also be utilized to detect NAbs. In this study, we have developed an Enzyme Linked Immunosobent Assay (ELISA)-based protocol for rapid detection of SARS CoV-2 NAb by inhibiting the binding of the spike protein receptor-binding domain to angiotensin converting enzyme 2 and compared it with cPASS neutralizing antibody kit, which was approved by the Food and Drug Administration (FDA). The results obtained suggest that the in-house ELISA developed for the detection of NAbs against SARS-CoV-2 is rapid and reliable. Compared to FDA-approved GenScript's cPass assay, the specificity and the sensitivity of the in-house-developed ELISA kit were 100% (95% confidence intervals of 69.15-100.00) and 96% (95% confidence intervals of 86.29-99.51), respectively. Thus, the ELISA protocol developed to test the neutralizing activities of antibodies is rapid, which requires a BSL-2 infrastructure facility and can be easily performed. It has very high potential applications in the rapid screening of NAb against SARS-CoV-2.
Collapse
Affiliation(s)
- K P Mishra
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Mrinalini Singh
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Deepika Saraswat
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Somnath Singh
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Delhi, India
| |
Collapse
|
39
|
Rottmayer K, Loeffler-Wirth H, Gruenewald T, Doxiadis I, Lehmann C. Individual Immune Response to SARS-CoV-2 Infection-The Role of Seasonal Coronaviruses and Human Leukocyte Antigen. BIOLOGY 2023; 12:1293. [PMID: 37887003 PMCID: PMC10603889 DOI: 10.3390/biology12101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
During the coronavirus pandemic, evidence is growing that the severity, susceptibility and host immune response to SARS-CoV-2 infection can be highly variable. Several influencing factors have been discussed. Here, we investigated the humoral immune response against SARS-CoV-2 spike, S1, S2, the RBD, nucleocapsid moieties and S1 of seasonal coronaviruses: hCoV-229E, hCoV-HKU1, hCoV-NL63 and hCoV-OC43, as well as MERS-CoV and SARS-CoV, in a cohort of 512 individuals. A bead-based multiplex assay allowed simultaneous testing for all the above antigens and the identification of different antibody patterns. Then, we correlated these patterns with 11 HLA loci. Regarding the seasonal coronaviruses, we found a moderate negative correlation between antibody levels against hCoV-229E, hCoV-HKU1 and hCoV-NL63 and the SARS-CoV-2 antigens. This could be an indication of the original immunological imprinting. High and low antibody response patterns were distinguishable, demonstrating the individuality of the humoral response towards the virus. An immunogenetical factor associated with a high antibody response (formation of ≥4 different antibodies) was the presence of HLA A*26:01, C*02:02 and DPB1*04:01 alleles, whereas the HLA alleles DRB3*01:01, DPB1*03:01 and DB1*10:01 were enriched in low responders. A better understanding of this variable immune response could enable more individualized protective measures.
Collapse
Affiliation(s)
- Karla Rottmayer
- Laboratory for Transplantation Immunology, University Hospital Leipzig, Johannisallee 32, 04103 Leipzig, Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, IZBI, Leipzig University, Haertelstr. 16–18, 04107 Leipzig, Germany
| | - Thomas Gruenewald
- Clinic for Infectious Diseases and Tropical Medicine, Klinikum Chemnitz, Flemmingstraße 2, 09116 Chemnitz, Germany
| | - Ilias Doxiadis
- Laboratory for Transplantation Immunology, University Hospital Leipzig, Johannisallee 32, 04103 Leipzig, Germany
| | - Claudia Lehmann
- Laboratory for Transplantation Immunology, University Hospital Leipzig, Johannisallee 32, 04103 Leipzig, Germany
| |
Collapse
|
40
|
Hasan A, Rahim R, Nakayama EE, Uno K, Hasan N, Rahman M, Shioda T. Enhancement of IL-6 Production Induced by SARS-CoV-2 Nucleocapsid Protein and Bangladeshi COVID-19 Patients' Sera. Viruses 2023; 15:2018. [PMID: 37896795 PMCID: PMC10611338 DOI: 10.3390/v15102018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by severe acute respiratory syndrome coronavirus 2 that can have detrimental effects on multiple organs and accelerate patient mortality. This study, which encompassed 130 confirmed COVID-19 patients who were assessed at three different time points (i.e., 3, 7, and 12 days) after the onset of symptoms, investigated interleukin-6 (IL-6) enhancement induced by a viral nucleocapsid (N) protein from a myeloid cell line. Disease severity was categorized as mild, moderate, or severe. The severe cases were characterized as having significant elevations in serum IL-6, C-reactive protein, D-dimer, ferritin, creatinine, leukocytes, and neutrophil-to-lymphocyte ratio and decreased hemoglobin, hematocrit, and albumin levels compared with mild and moderate cases. To evaluate IL-6-inducing activity, heat-inactivated sera from these patients were incubated with and without the N protein. The findings showed a progressive increase in IL-6 production in severe cases upon N protein stimulation. There was a strong correlation between anti-N antibodies and levels of IL-6 secreted by myeloid cells in the presence of N protein and sera, indicating the crucial role that the anti-N antibody plays in inducing IL-6 production. Uncontrolled IL-6 production played a pivotal role in disease pathogenesis, exacerbating both disease severity and mortality. Efficiently targeting the N protein could potentially be employed as a therapeutic strategy for regulating the immune response and alleviating inflammation in severe cases.
Collapse
Affiliation(s)
- Abu Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Rummana Rahim
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Emi E. Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| | - Kazuko Uno
- IFN & Host-Defense Research Laboratory, Louis Pasteur Center for Medical Research, Kyoto 606-8225, Japan;
| | - Nazmul Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Mizanur Rahman
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| |
Collapse
|
41
|
Nurisyah S, Iyori M, Hasyim AA, Sakamoto A, Hashimoto H, Yamagata K, Yamauchi S, Amru K, Zainal KH, Idris I, Yoshida S, Djaharuddin I, Syafruddin D, Bukhari A, Asih PBS, Yusuf Y. Comparison between Neutralization Capacity of Antibodies Elicited by COVID-19 Natural Infection and Vaccination in Indonesia: A Prospective Cohort. Antibodies (Basel) 2023; 12:60. [PMID: 37753974 PMCID: PMC10526084 DOI: 10.3390/antib12030060] [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: 08/01/2023] [Revised: 08/21/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND To fight the COVID-19 pandemic, immunity against SARS-CoV-2 should be achieved not only through natural infection but also by vaccination. The effect of COVID-19 vaccination on previously infected persons is debatable. METHODS A prospective cohort was undergone to collect sera from unvaccinated survivors and vaccinated persons-with and without COVID-19 pre-infection. The sera were analyzed for the anti-receptor binding domain (RBD) titers by ELISA and for the capacity to neutralize the pseudovirus of the Wuhan-Hu-1 strain by luciferase assays. RESULTS Neither the antibody titers nor the neutralization capacity was significantly different between the three groups. However, the correlation between the antibody titers and the percentage of viral neutralization derived from sera of unvaccinated survivors was higher than that from vaccinated persons with pre-infection and vaccinated naïve individuals (Spearman correlation coefficient (r) = -0.8558; 95% CI, -0.9259 to -0.7288), p < 0.0001 vs. -0.7855; 95% CI, -0.8877 to -0.6096, p < 0.0001 and -0.581; 95% CI, -0.7679 to -0.3028, p = 0.0002, respectively), indicating the capacity to neutralize the virus is most superior by infection alone. CONCLUSIONS Vaccines induce anti-RBD titers as high as the natural infection with lower neutralization capacity, and it does not boost immunity in pre-infected persons.
Collapse
Affiliation(s)
- Sitti Nurisyah
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Dr. Tadjuddin Chalid Hospital, Makassar 90241, Indonesia
| | - Mitsuhiro Iyori
- Research Institute of Pharmaceutical Sciences, Musashino University, Nishitokyo, Tokyo 202-8585, Japan;
| | - Ammar Abdurrahman Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Hinata Hashimoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Kyouhei Yamagata
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Saya Yamauchi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Khaeriah Amru
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Dr. Tadjuddin Chalid Hospital, Makassar 90241, Indonesia
| | - Kartika Hardianti Zainal
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Irfan Idris
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Hasanuddin University Medical Research Centre, Makassar 90245, Indonesia
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University, Kanazawa 920-1192, Japan; (A.A.H.); (A.S.); (H.H.); (K.Y.); (S.Y.); (K.H.Z.); (S.Y.)
| | - Irawaty Djaharuddin
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Dr. Wahidin Soedirohusodo Hospital, Makassar 90425, Indonesia
| | - Din Syafruddin
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Hasanuddin University Medical Research Centre, Makassar 90245, Indonesia
| | - Agussalim Bukhari
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
| | - Puji Budi Setia Asih
- Badan Riset dan Inovasi Nasional (National Research and Innovation Agency), Jakarta 10340, Indonesia;
| | - Yenni Yusuf
- Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia; (S.N.); (K.A.); (I.I.); (I.D.); (D.S.); (A.B.)
- Hasanuddin University Medical Research Centre, Makassar 90245, Indonesia
| |
Collapse
|
42
|
Cantoni D, Wilkie C, Bentley EM, Mayora-Neto M, Wright E, Scott S, Ray S, Castillo-Olivares J, Heeney JL, Mattiuzzo G, Temperton NJ. Correlation between pseudotyped virus and authentic virus neutralisation assays, a systematic review and meta-analysis of the literature. Front Immunol 2023; 14:1184362. [PMID: 37790941 PMCID: PMC10544934 DOI: 10.3389/fimmu.2023.1184362] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Background The virus neutralization assay is a principal method to assess the efficacy of antibodies in blocking viral entry. Due to biosafety handling requirements of viruses classified as hazard group 3 or 4, pseudotyped viruses can be used as a safer alternative. However, it is often queried how well the results derived from pseudotyped viruses correlate with authentic virus. This systematic review and meta-analysis was designed to comprehensively evaluate the correlation between the two assays. Methods Using PubMed and Google Scholar, reports that incorporated neutralisation assays with both pseudotyped virus, authentic virus, and the application of a mathematical formula to assess the relationship between the results, were selected for review. Our searches identified 67 reports, of which 22 underwent a three-level meta-analysis. Results The three-level meta-analysis revealed a high level of correlation between pseudotyped viruses and authentic viruses when used in an neutralisation assay. Reports that were not included in the meta-analysis also showed a high degree of correlation, with the exception of lentiviral-based pseudotyped Ebola viruses. Conclusion Pseudotyped viruses identified in this report can be used as a surrogate for authentic virus, though care must be taken in considering which pseudotype core to use when generating new uncharacterised pseudotyped viruses.
Collapse
Affiliation(s)
- Diego Cantoni
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Craig Wilkie
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Emma M. Bentley
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Edward Wright
- Viral Pseudotype Unit, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Simon Scott
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Giada Mattiuzzo
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| |
Collapse
|
43
|
Hitchings MDT, Patel EU, Khan R, Srikrishnan AK, Anderson M, Kumar KS, Wesolowski AP, Iqbal SH, Rodgers MA, Mehta SH, Cloherty G, Cummings DAT, Solomon SS. A Mixture Model for Estimating SARS-CoV-2 Seroprevalence in Chennai, India. Am J Epidemiol 2023; 192:1552-1561. [PMID: 37084085 PMCID: PMC10472327 DOI: 10.1093/aje/kwad103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 12/01/2022] [Accepted: 04/18/2023] [Indexed: 04/22/2023] Open
Abstract
Serological assays used to estimate the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) often rely on manufacturers' cutoffs established on the basis of severe cases. We conducted a household-based serosurvey of 4,677 individuals in Chennai, India, from January to May 2021. Samples were tested for SARS-CoV-2 immunoglobulin G (IgG) antibodies to the spike (S) and nucleocapsid (N) proteins. We calculated seroprevalence, defining seropositivity using manufacturer cutoffs and using a mixture model based on measured IgG level. Using manufacturer cutoffs, there was a 5-fold difference in seroprevalence estimated by each assay. This difference was largely reconciled using the mixture model, with estimated anti-S and anti-N IgG seroprevalence of 64.9% (95% credible interval (CrI): 63.8, 66.0) and 51.5% (95% CrI: 50.2, 52.9), respectively. Age and socioeconomic factors showed inconsistent relationships with anti-S and anti-N IgG seropositivity using manufacturer cutoffs. In the mixture model, age was not associated with seropositivity, and improved household ventilation was associated with lower seropositivity odds. With global vaccine scale-up, the utility of the more stable anti-S IgG assay may be limited due to the inclusion of the S protein in several vaccines. Estimates of SARS-CoV-2 seroprevalence using alternative targets must consider heterogeneity in seroresponse to ensure that seroprevalence is not underestimated and correlates are not misinterpreted.
Collapse
Affiliation(s)
- Matt D T Hitchings
- Correspondence to Dr. Matt Hitchings, Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Clinical and Translational Research Building, 5th Floor, 2004 Mowry Road, Gainesville, FL 32603 (e-mail: )
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Takahashi S, Peluso MJ, Hakim J, Turcios K, Janson O, Routledge I, Busch MP, Hoh R, Tai V, Kelly JD, Martin JN, Deeks SG, Henrich TJ, Greenhouse B, Rodríguez-Barraquer I. SARS-CoV-2 Serology Across Scales: A Framework for Unbiased Estimation of Cumulative Incidence Incorporating Antibody Kinetics and Epidemic Recency. Am J Epidemiol 2023; 192:1562-1575. [PMID: 37119030 PMCID: PMC10472487 DOI: 10.1093/aje/kwad106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/29/2022] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Serosurveys are a key resource for measuring severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) population exposure. A growing body of evidence suggests that asymptomatic and mild infections (together making up over 95% of all infections) are associated with lower antibody titers than severe infections. Antibody levels also peak a few weeks after infection and decay gradually. We developed a statistical approach to produce estimates of cumulative incidence from raw seroprevalence survey results that account for these sources of spectrum bias. We incorporate data on antibody responses on multiple assays from a postinfection longitudinal cohort, along with epidemic time series to account for the timing of a serosurvey relative to how recently individuals may have been infected. We applied this method to produce estimates of cumulative incidence from 5 large-scale SARS-CoV-2 serosurveys across different settings and study designs. We identified substantial differences between raw seroprevalence and cumulative incidence of over 2-fold in the results of some surveys, and we provide a tool for practitioners to generate cumulative incidence estimates with preset or custom parameter values. While unprecedented efforts have been launched to generate SARS-CoV-2 seroprevalence estimates over this past year, interpretation of results from these studies requires properly accounting for both population-level epidemiologic context and individual-level immune dynamics.
Collapse
Affiliation(s)
- Saki Takahashi
- Correspondence to Dr. Saki Takahashi, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205 (e-mail: )
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kanis FM, Meier JP, Guldan H, Niller HH, Dahm M, Dansard A, Zander T, Struck F, Soutschek E, Deml L, Möbus S, Barabas S. Performance of T-Track ® SARS-CoV-2, an Innovative Dual Marker RT-qPCR-Based Whole-Blood Assay for the Detection of SARS-CoV-2-Reactive T Cells. Diagnostics (Basel) 2023; 13:2722. [PMID: 37685260 PMCID: PMC10486492 DOI: 10.3390/diagnostics13172722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
T-cell immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays a central role in the control of the virus. In this study, we evaluated the performance of T-Track® SARS-CoV-2, a novel CE-marked quantitative reverse transcription-polymerase chain reaction (RT-qPCR) assay, which relies on the combined evaluation of IFNG and CXCL10 mRNA levels in response to the S1 and NP SARS-CoV-2 antigens, in 335 participants with or without a history of SARS-CoV-2 infection and vaccination, respectively. Of the 62 convalescent donors, 100% responded to S1 and 88.7% to NP antigens. In comparison, of the 68 naïve donors, 4.4% were reactive to S1 and 19.1% to NP. Convalescent donors <50 and ≥50 years of age demonstrated a 100% S1 reactivity and an 89.1% and 87.5% NP reactivity, respectively. T-cell responses by T-Track® SARS-CoV-2 and IgG serology by recomLine SARS-CoV-2 IgG according to the time from the last immunisation (by vaccination or viral infection) were comparable. Both assays showed a persistent cellular and humoral response for at least 36 weeks post immunisation in vaccinated and convalescent donors. Our results demonstrate the very good performance of the T-Track® SARS-CoV-2 molecular assay and suggest that it might be suitable to monitor the SARS-CoV-2-specific T-cell response in COVID-19 vaccinations trials and cross-reactivity studies.
Collapse
Affiliation(s)
| | | | | | - Hans-Helmut Niller
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yin D, Han Z, Lang B, Li Y, Mai G, Chen H, Feng L, Chen YQ, Luo H, Xiong Y, Jing L, Du X, Shu Y, Sun C. Effect of seasonal coronavirus immune imprinting on the immunogenicity of inactivated COVID-19 vaccination. Front Immunol 2023; 14:1195533. [PMID: 37654488 PMCID: PMC10467281 DOI: 10.3389/fimmu.2023.1195533] [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: 03/28/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Background Pre-existing cross-reactive immunity among different coronaviruses, also termed immune imprinting, may have a comprehensive impact on subsequent SARS-CoV-2 infection and COVID-19 vaccination effectiveness. Here, we aim to explore the interplay between pre-existing seasonal coronaviruses (sCoVs) antibodies and the humoral immunity induced by COVID-19 vaccination. Methods We first collected serum samples from healthy donors prior to COVID-19 pandemic and individuals who had received COVID-19 vaccination post-pandemic in China, and the levels of IgG antibodies against sCoVs and SARS-CoV-2 were detected by ELISA. Wilcoxon rank sum test and chi-square test were used to compare the difference in magnitude and seropositivity rate between two groups. Then, we recruited a longitudinal cohort to collect serum samples before and after COVID-19 vaccination. The levels of IgG antibodies against SARS-CoV-2 S, S1, S2 and N antigen were monitored. Association between pre-existing sCoVs antibody and COVID-19 vaccination-induced antibodies were analyzed by Spearman rank correlation. Results 96.0% samples (339/353) showed the presence of IgG antibodies against at least one subtype of sCoVs. 229E and OC43 exhibited the highest seroprevalence rates at 78.5% and 72.0%, respectively, followed by NL63 (60.9%) and HKU1 (52.4%). The levels of IgG antibodies against two β coronaviruses (OC43 and HKU1) were significantly higher in these donors who had inoculated with COVID-19 vaccines compared to pre-pandemic healthy donors. However, we found that COVID-19 vaccine-induced antibody levels were not significant different between two groups with high levelor low level of pre-existing sCoVs antibody among the longitudinal cohort. Conclusion We found a high prevalence of antibodies against sCoVs in Chinese population. The immune imprinting by sCoVs could be reactivated by COVID-19 vaccination, but it did not appear to be a major factor affecting the immunogenicity of COVID-19 vaccine. These findings will provide insights into understanding the impact of immune imprinting on subsequent multiple shots of COVID-19 vaccines.
Collapse
Affiliation(s)
- Di Yin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Zirong Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Bing Lang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yanjun Li
- Emergency Manage Department, Foshan, China
| | - Guoqin Mai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Hongbiao Chen
- Department of Epidemiology and Infectious Disease Control, Shenzhen, China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, China
| | - Yao-qing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yaming Xiong
- Institute of Clinical Medicine, First People's Hospital of Foshan, Foshan, China
| | - Lin Jing
- Institute of Clinical Medicine, First People's Hospital of Foshan, Foshan, China
| | - Xiangjun Du
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
- National Health Commission of the People's Republic of China (NHC) Key Laboratory of System Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
47
|
Barrios MH, Nicholson S, Bull RA, Martinello M, Rawlinson W, Mina M, Post JJ, Hudson B, Gilroy N, Lloyd AR, Konecny P, Mordant F, Catton M, Subbarao K, Caly L, Druce J, Netter HJ. Comparative Longitudinal Serological Study of Anti-SARS-CoV-2 Antibody Profiles in People with COVID-19. Microorganisms 2023; 11:1985. [PMID: 37630545 PMCID: PMC10458948 DOI: 10.3390/microorganisms11081985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Serological diagnostic assays are essential tools for determining an individual's protection against viruses like SARS-CoV-2, tracking the spread of the virus in the community, and evaluating population immunity. To assess the diversity and quality of the anti-SARS-CoV-2 antibody response, we have compared the antibody profiles of people with mild, moderate, and severe COVID-19 using a dot blot assay. The test targeted the four major structural proteins of SARS-CoV-2, namely the nucleocapsid (N), spike (S) protein domains S1 and S2, and receptor-binding domain (RBD). Serum samples were collected from 63 participants at various time points for up to 300 days after disease onset. The dot blot assay revealed patient-specific differences in the anti-SARS-CoV-2 antibody profiles. Out of the 63 participants with confirmed SARS-CoV-2 infections and clinical COVID-19, 35/63 participants exhibited diverse and robust responses against the tested antigens, while 14/63 participants displayed either limited responses to a subset of antigens or no detectable antibody response to any of the antigens. Anti-N-specific antibody levels decreased within 300 days after disease onset, whereas anti-S-specific antibodies persisted. The dynamics of the antibody response did not change during the test period, indicating stable antibody profiles. Among the participants, 28/63 patients with restricted anti-S antibody profiles or undetectable anti-S antibody levels in the dot blot assay also exhibited weak neutralization activity, as measured by a surrogate virus neutralization test (sVNT) and a microneutralization test. These results indicate that in some cases, natural infections do not lead to the production of neutralizing antibodies. Furthermore, the study revealed significant serological variability among patients, regardless of the severity of their COVID-19 illness. These differences need to be carefully considered when evaluating the protective antibody status of individuals who have experienced primary SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Marilou H. Barrios
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Rowena A. Bull
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
| | - Marianne Martinello
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
| | - William Rawlinson
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
- Serology and Virology Division, Department of Microbiology, New South Wales Health Pathology, Randwick, Sydney, NSW 2031, Australia
- Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Michael Mina
- Northern Beaches Hospital, Frenchs Forest, NSW 2086, Australia;
| | - Jeffrey J. Post
- Prince of Wales Hospital, Sydney, NSW 2031, Australia;
- School of Clinical Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
| | - Bernard Hudson
- Royal North Shore Hospital, Sydney, NSW 2065, Australia;
| | | | - Andrew R. Lloyd
- The Kirby Institute, University of New South Wales (UNSW), Sydney, NSW 2052, Australia; (R.A.B.); (M.M.); (A.R.L.)
| | - Pamela Konecny
- School of Clinical Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia;
- St. George Hospital, Sydney, NSW 2217, Australia
| | - Francesca Mordant
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Mike Catton
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Kanta Subbarao
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia
- World Health Organization Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute, Melbourne, VIC 3000, Australia
| | - Leon Caly
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Julian Druce
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
| | - Hans J. Netter
- Victorian Infectious Diseases Reference Laboratory (VIDRL), The Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (M.H.B.); (S.N.); (M.C.); (L.C.); (J.D.)
- Peter Doherty Institute, University of Melbourne, Melbourne, VIC 3000, Australia; (F.M.); (K.S.)
- School of Science, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC 3001, Australia
| |
Collapse
|
48
|
Sendi P, Widmer N, Branca M, Thierstein M, Büchi AE, Güntensperger D, Blum MR, Baldan R, Tinguely C, Heg D, Theel ES, Berbari E, Tande AJ, Endimiani A, Gowland P, Niederhauser C. Do quantitative levels of antispike-IgG antibodies aid in predicting protection from SARS-CoV-2 infection? Results from a longitudinal study in a police cohort. J Med Virol 2023; 95:e28904. [PMID: 37386901 DOI: 10.1002/jmv.28904] [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: 03/25/2023] [Revised: 06/04/2023] [Accepted: 06/11/2023] [Indexed: 07/01/2023]
Abstract
In a COVID-19 sero-surveillance cohort study with predominantly healthy and vaccinated individuals, the objectives were (i) to investigate longitudinally the factors associated with the quantitative dynamics of antispike (anti-S1) IgG antibody levels, (ii) to evaluate whether the levels were associated with protection from SARS-CoV-2 infection, and (iii) to assess whether the association was different in the pre-Omicron compared with the Omicron period. The QuantiVac Euroimmun ELISA test was used to quantify anti-S1 IgG levels. The entire study period (16 months), the 11-month pre-Omicron period and the cross-sectional analysis before the Omicron surge included 3219, 2310, and 895 reactive serum samples from 949, 919, and 895 individuals, respectively. Mixed-effect linear, mixed-effect time-to-event, and logistic regression models were used to achieve the objectives. Age and time since infection or vaccination were the only factors associated with a decline of anti-S1 IgG levels. Higher antibody levels were significantly associated with protection from SARS-CoV-2 infection (0.89, 95% confidence interval [CI] 0.82-0.97), and the association was higher during the time period when Omicron was predominantly circulating compared with the ones when Alpha and Delta variants were predominant (adjusted hazard ratio for interaction 0.66, 95% CI 0.53-0.84). In a prediction model, it was estimated that >8000 BAU/mL anti-S1 IgG was required to reduce the risk of infection with Omicron variants by approximately 20%-30% for 90 days. Though, such high levels were only found in 1.9% of the samples before the Omicron surge, and they were not durable for 3 months. Anti-S1 IgG antibody levels are statistically associated with protection from SARS-CoV-2 infection. However, the prediction impact of the antibody level findings on infection protection is limited.
Collapse
Affiliation(s)
- Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Nadja Widmer
- Interregional Blood Transfusion Swiss Red Cross, Bern, Switzerland
| | | | - Marc Thierstein
- Division Operations, Cantonal Police Bern, Bern, Switzerland
| | - Annina Elisabeth Büchi
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Manuel Raphael Blum
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Rossella Baldan
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Dik Heg
- CTU Bern, University of Bern, Bern, Switzerland
| | - Elitza S Theel
- Division of Clinical Microbiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elie Berbari
- Division of Public Health, Infectious Diseases, and Occupational Medicine Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Tande
- Division of Public Health, Infectious Diseases, and Occupational Medicine Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Endimiani
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Peter Gowland
- Interregional Blood Transfusion Swiss Red Cross, Bern, Switzerland
| | - Christoph Niederhauser
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Interregional Blood Transfusion Swiss Red Cross, Bern, Switzerland
| |
Collapse
|
49
|
Xu J, Zheng J, Tan Y, Cai J, Xiang Y, Ling H, Li Z, Bai Q. Longitudinal Observation of Immune Response for 23 Months in COVID-19 Convalescent Patients After Infection and Vaccination. Viral Immunol 2023; 36:389-400. [PMID: 37276049 DOI: 10.1089/vim.2022.0111] [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] [Indexed: 06/07/2023] Open
Abstract
To better understand dynamic changes of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) immune response, a prospective, single-center, cohort study was conducted on longitudinal immune response in 34 COVID-19 convalescent patients over 23 months in Chongqing. Two blood samples from convalescent patients were collected, first sample collected during 10-13 months (M10-13) after infection (pre-SARS-CoV-2 vaccination) and second sample collected during 20-23 months (M20-23) after infection (post-SARS-CoV-2 vaccination). The SARS-CoV-2-specific humoral and cellular immunity were traced by testing total antibody (Ab), anti-nucleocapsid (NP) immunoglobulin M (IgM), anti-NP immunoglobulin G (IgG), and anti-spike (S) IgG Abs, lymphocyte subset count, and Th1 cytokines. Healthy donors (30) were also included in the study as the uninspected healthy controls. Our data showed significant change in mean titer of SARS-CoV-2-specific Ab response from M10-13 to M20-23 included, namely, SARS-CoV-2-specific total Ab as 219 AU/mL increasing to 750.9 AU/mL; anti-NP IgM as 3.5 AU/mL decreasing significantly (p < 0.001) to 0.6 AU/mL; anti-NP IgG as 7.9 AU/mL increasing to 87.1 AU/mL; and anti-S IgG as 499.0 RU/mL increasing to 1,802.3 RU/mL. Our observations suggested that one vaccine dose might have been sufficient for COVID-19 convalescent patients. Larger sample sizes are needed to compare better immune effect of protein subunit vaccine. Besides, compared to healthy donors, patients had decreased CD3+ and CD8+ T lymphocyte counts during two periods. Patients had most cytokines recovered normally within 2 years, but IL-6 level was significantly elevated; however, IL-6 was negatively correlated with IgM and positively correlated with IgG. Changes in cytokines might have been caused by SARS-CoV-2 infection or vaccination. Patients with comorbidities were associated with decreased CD3+ and CD8+ T lymphocytes and lower Ab titers following SARS-CoV-2 vaccination. Vaccination enormously increased humoral immunity beneficial in COVID-19 convalescent patients. Elderly COVID-19 convalescent patients with comorbidities needed more attention.
Collapse
Affiliation(s)
- Jingru Xu
- College of Public Health, Chongqing Medical University, Chongqing, China
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Juan Zheng
- Department of Neurology, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing, China
| | - Yan Tan
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Jiaojiao Cai
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Yao Xiang
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Hua Ling
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Zhifeng Li
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Qunhua Bai
- College of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Pan F, Gong J, Ma X, Tang X, Xing J, Sheng X, Chi H, Zhan W. Expression characteristics of non-virion protein of Hirame novirhabdovirus and its transfection induced response in hirame natural embryo cells. Int J Biol Macromol 2023; 242:124567. [PMID: 37100320 DOI: 10.1016/j.ijbiomac.2023.124567] [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: 12/31/2022] [Revised: 03/19/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023]
Abstract
The non-virion (NV) protein is the signature of genus Novirhabdovirus, which has been of considerable concern due to its potential role in viral pathogenicity. However, its expression characteristics and induced immune response remain limited. In the present work, it was demonstrated that Hirame novirhabdovirus (HIRRV) NV protein was only detected in the viral infected hirame natural embryo (HINAE) cells, but absent in the purified virions. Results showed that the transcription of NV gene could be stably detected in HIRRV-infected HINAE cells at 12 h post infection (hpi) and then reached the peak at 72 hpi. A similar expression trend of NV gene was also found in HIRRV-infected flounders. Subcellular localization analysis further exhibited that HIRRV-NV protein was predominantly localized in the cytoplasm. To elucidate the biological function of HIRRV-NV protein, NV eukaryotic plasmid was transfected into HINAE cells for RNA-seq. Compared to empty plasmid group, some key genes in RLR signaling pathway were significantly downregulated in NV-overexpressed HINAE cells, indicating that RLR signaling pathway was inhibited by HIRRV-NV protein. The interferon-associated genes were also significantly suppressed upon transfection of NV gene. This research would improve our understanding of expression characteristics and biological function of NV protein during HIRRV infection process.
Collapse
Affiliation(s)
- Fenghuang Pan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jiaojiao Gong
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xinbiao Ma
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China.
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| |
Collapse
|