1
|
Sayama Y, Lo C, Tomizawa H, Saito M, Okamoto M, Ohmiya S, Nishimura H, Oshitani H. Serological analyses against endemic human coronaviruses and SARS-CoV-2 in children and adults using samples collected before the COVID-19 pandemic. IJID REGIONS 2024; 13:100485. [PMID: 39659746 PMCID: PMC11629223 DOI: 10.1016/j.ijregi.2024.100485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024]
Abstract
Objectives Four endemic human coronaviruses (HCoVs), HCoV-229E, HCoV-NL63, HCoV-HKU1, and HCoV-OC43, infect humans during childhood and cause the common cold. COVID-19 caused by SARS-CoV-2 leads to mild symptoms in children, possibly owing to the protection conferred by immunity developed during a previous HCoV infection. This study analyzed the seroreactivity of four endemic HCoVs and SARS-CoV-2 in children and adults. Methods A total of 747 serum samples (from individuals aged 6 months to 69 years) were collected from 2015 to 2019 before the COVID-19 pandemic in Japan. The samples were tested for immunoglobulin G antibodies against the four endemic HCoVs and SARS-CoV-2 wild-type spike ectodomain proteins using enzyme-linked immunosorbent assay. Results The seroprevalence of endemic HCoVs (except HCoV-229E) showed 90% positivity by 3-4 years old, whereas HCoV-229E seroprevalence was observed at 8 years old. Approximately 35% of the samples showed reactivity to SARS-CoV-2 and did not change with age. However, the children's group presented higher antibody levels than the adult group. The sample reactivity against SARS-CoV-2 did not confirm neutralization capability. Conclusions The reactive samples against SARS-CoV-2 showed varying antibody levels among different age groups. These findings may contribute to a deeper understanding of the clinical symptoms of COVID-19 and coronavirus diseases.
Collapse
Affiliation(s)
- Yusuke Sayama
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chuan Lo
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Tomizawa
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiko Okamoto
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Suguru Ohmiya
- Virus Research Center, Clinical Research Division, Sendai Medical Center, Sendai, Japan
| | - Hidekazu Nishimura
- Virus Research Center, Clinical Research Division, Sendai Medical Center, Sendai, Japan
| | - Hitoshi Oshitani
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
2
|
Johnson NV, Wall SC, Kramer KJ, Holt CM, Periasamy S, Richardson SI, Manamela NP, Suryadevara N, Andreano E, Paciello I, Pierleoni G, Piccini G, Huang Y, Ge P, Allen JD, Uno N, Shiakolas AR, Pilewski KA, Nargi RS, Sutton RE, Abu-Shmais AA, Parks R, Haynes BF, Carnahan RH, Crowe JE, Montomoli E, Rappuoli R, Bukreyev A, Ross TM, Sautto GA, McLellan JS, Georgiev IS. Discovery and characterization of a pan-betacoronavirus S2-binding antibody. Structure 2024; 32:1893-1909.e11. [PMID: 39326419 PMCID: PMC11560675 DOI: 10.1016/j.str.2024.08.022] [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: 02/05/2024] [Revised: 06/12/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
The continued emergence of deadly human coronaviruses from animal reservoirs highlights the need for pan-coronavirus interventions for effective pandemic preparedness. Here, using linking B cell receptor to antigen specificity through sequencing (LIBRA-seq), we report a panel of 50 coronavirus antibodies isolated from human B cells. Of these, 54043-5 was shown to bind the S2 subunit of spike proteins from alpha-, beta-, and deltacoronaviruses. A cryoelectron microscopy (cryo-EM) structure of 54043-5 bound to the prefusion S2 subunit of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike defined an epitope at the apex of S2 that is highly conserved among betacoronaviruses. Although non-neutralizing, 54043-5 induced Fc-dependent antiviral responses in vitro, including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). In murine SARS-CoV-2 challenge studies, protection against disease was observed after introduction of Leu234Ala, Leu235Ala, and Pro329Gly (LALA-PG) substitutions in the Fc region of 54043-5. Together, these data provide new insights into the protective mechanisms of non-neutralizing antibodies and define a broadly conserved epitope within the S2 subunit.
Collapse
MESH Headings
- Humans
- SARS-CoV-2/immunology
- SARS-CoV-2/metabolism
- SARS-CoV-2/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- Antibodies, Viral/chemistry
- Mice
- COVID-19/immunology
- COVID-19/virology
- Cryoelectron Microscopy
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/metabolism
- Models, Molecular
- Protein Binding
- Epitopes/immunology
- Epitopes/chemistry
- Antibody-Dependent Cell Cytotoxicity
Collapse
Affiliation(s)
- Nicole V Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Steven C Wall
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA
| | - Kevin J Kramer
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA
| | - Clinton M Holt
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sivakumar Periasamy
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Simone I Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Service, 2131 Johannesburg, South Africa; South African MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Nelia P Manamela
- National Institute for Communicable Diseases of the National Health Laboratory Service, 2131 Johannesburg, South Africa; South African MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, 53100 Siena, Italy
| | - Ida Paciello
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, 53100 Siena, Italy
| | - Giulio Pierleoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, 53100 Siena, Italy
| | | | - Ying Huang
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Pan Ge
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - James D Allen
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Naoko Uno
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44196, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Andrea R Shiakolas
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA
| | - Kelsey A Pilewski
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA
| | - Rachel S Nargi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel E Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alexandria A Abu-Shmais
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA; Departments of Medicine and Immunology, Duke University, Durham, NC 27710, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Emanuele Montomoli
- VisMederi Research S.r.l., 53100 Siena, Italy; VisMederi S.r.l, 53100 Siena, Italy; Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Rino Rappuoli
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, 53100 Siena, Italy; Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Ted M Ross
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44196, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Giuseppe A Sautto
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA.
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Ivelin S Georgiev
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 73232, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Computer Science, Vanderbilt University, Nashville, TN 37232, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA; Program in Computational Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
3
|
Fathi P, Alfonso AL, Yek C, Putman Z, Drew M, Esposito D, Zaidi I, Chea S, Ly S, Sath R, Lon C, Chea H, Leang R, Huy R, Ly S, Seng H, Tan CW, Zhu F, Wang LF, Oliveira F, Sadtler K, Manning J. Humoral Immunity Profiling to Pandemic and Bat-Derived Coronavirus Variants: A Geographical Comparison. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403503. [PMID: 39471070 DOI: 10.1002/advs.202403503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/05/2024] [Indexed: 11/01/2024]
Abstract
Dynamic pathogen exposure may impact the immunological response to SARS-CoV-2 (SCV2). One potential explanation for the lack of severe SCV2-related morbidity and mortality in Southeast Asia is prior exposure to related betacoronaviruses. Recent discoveries of SCV2-related betacoronaviruses from horseshoe bats (Rhinolophus sinicus) in Thailand, Laos, and Cambodia suggest the potential for bat-to-human spillover exposures in the region. In this work, serum antibodies to protein constructs from SCV2 and a representative bat coronavirus isolated in Cambodia (RshSTT182) are measured in pre-pandemic Cambodian human sera using ELISA assays. Of 293 Cambodian samples tested (N = 131 with acute malaria, n = 162 with acute undifferentiated febrile illness), 32 (10.9%) are seropositive for SCV2 based on established Spike and receptor-binding domain (RBD) cutoffs. Within SCV2 seropositive samples, 16 (50%) have higher antibody levels to antigens from the representative virus RshSTT182 versus SCV2 antigens; competitive binding ELISA assays demonstrate inhibition of reactivity to SCV2 Spike after pre-incubation with RshSTT182 Spike. Surrogate virus neutralization tests demonstrate that 8/30 (26.7%) SCV2 ELISA positive pre-pandemic Cambodian samples have neutralizing activity against SCV2, while 14/30 (46.7%) have activity against other SCV2-related betacoronaviruses. These data suggest that exposure to related betacoronaviruses may elicit cross-reactive immunity to SCV2 prior to the global pandemic.
Collapse
Affiliation(s)
- Parinaz Fathi
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, 20892, USA
| | - Andrea Lucia Alfonso
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, 20892, USA
| | - Christina Yek
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20892, USA
| | - Zoe Putman
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Matthew Drew
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Dominic Esposito
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20892, USA
| | - Sophana Chea
- International Center of Excellence in Research Cambodia, National Institute of Allergy and Infectious Diseases, Phnom Penh, 120801, Cambodia
| | - Sokna Ly
- International Center of Excellence in Research Cambodia, National Institute of Allergy and Infectious Diseases, Phnom Penh, 120801, Cambodia
| | - Rathanak Sath
- International Center of Excellence in Research Cambodia, National Institute of Allergy and Infectious Diseases, Phnom Penh, 120801, Cambodia
| | - Chanthap Lon
- International Center of Excellence in Research Cambodia, National Institute of Allergy and Infectious Diseases, Phnom Penh, 120801, Cambodia
| | - Huch Chea
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, 120801, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, 120801, Cambodia
| | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, 120801, Cambodia
| | - Sovann Ly
- Cambodian Center for Disease Control, Ministry of Health, Phnom Penh, 120407, Cambodia
| | - Heng Seng
- Cambodian Center for Disease Control, Ministry of Health, Phnom Penh, 120407, Cambodia
| | - Chee Wah Tan
- Programme for Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore, Singapore
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore, Singapore
| | - Feng Zhu
- Programme for Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore, Singapore
| | - Lin-Fa Wang
- Programme for Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore, Singapore
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20892, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, 20892, USA
| | - Jessica Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20892, USA
- International Center of Excellence in Research Cambodia, National Institute of Allergy and Infectious Diseases, Phnom Penh, 120801, Cambodia
| |
Collapse
|
4
|
Callery EL, Morais CLM, Taylor JV, Challen K, Rowbottom AW. Investigation of Long-Term CD4+ T Cell Receptor Repertoire Changes Following SARS-CoV-2 Infection in Patients with Different Severities of Disease. Diagnostics (Basel) 2024; 14:2330. [PMID: 39451653 PMCID: PMC11507081 DOI: 10.3390/diagnostics14202330] [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: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The difference in the immune response to severe acute respiratory syndrome coro-navirus 2 (SARS-CoV-2) in patients with mild versus severe disease remains poorly understood. Recent scientific advances have recognised the vital role of both B cells and T cells; however, many questions remain unanswered, particularly for T cell responses. T cells are essential for helping the generation of SARS-CoV-2 antibody responses but have also been recognised in their own right as a major factor influencing COVID-19 disease outcomes. The examination of T cell receptor (TCR) family differences over a 12-month period in patients with varying COVID-19 disease severity is crucial for understanding T cell responses to SARS-CoV-2. METHODS We applied a machine learning approach to analyse TCR vb family responses in COVID-19 patients (n = 151) across multiple timepoints and disease severities alongside SARS-CoV-2 infection-naïve (healthy control) individ-uals (n = 62). RESULTS Blood samples from hospital in-patients with moderate, severe, or critical disease could be classified with an accuracy of 94%. Furthermore, we identified significant variances in TCR vb family specificities between disease and control subgroups. CONCLUSIONS Our findings suggest advantageous and disadvantageous TCR repertoire patterns in relation to disease severity. Following validation in larger cohorts, our methodology may be useful in detecting protective immunity and the assessment of long-term outcomes, particularly as we begin to unravel the immunological mechanisms leading to post-COVID complications.
Collapse
Affiliation(s)
- Emma L. Callery
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Camilo L. M. Morais
- Institute of Chemistry, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil;
| | - Jemma V. Taylor
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Kirsty Challen
- Department of Emergency Medicine, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Anthony W. Rowbottom
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| |
Collapse
|
5
|
Tong X, Kellman B, Avendano MJ, Mendu M, Hsiao JC, Serrano E, Garcia-Salum T, Muena N, Pardo-Roa C, Morales M, Levican J, Salinas E, Cardenas-Cáceres S, Riquelme A, Tischler ND, Lauffenburger DA, Alter G, McNamara RP, Medina RA. Humoral waning kinetics against SARS-CoV-2 is dictated by disease severity and vaccine platform. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.17.24315607. [PMID: 39484236 PMCID: PMC11527045 DOI: 10.1101/2024.10.17.24315607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
SARS-CoV-2 vaccine-acquired immunity provides robust cross-variant recognition, while infection-acquired immunity can be heterogenous, with disease severity often modulating post-recovery responses. We assessed antibody waning dynamics between infection- and vaccination-acquired immunity across variants of concern (VOC). mRNA vaccination induced potent, cross-VOC Spike recognition and functional responses, but waned more rapidly for Omicron Spike. Hospitalized individuals developed more durable functional responses with lower peaks compared to mRNA vaccination, while outpatients exhibited slower decay than inactivated vaccine recipients. Humoral decay for the receptor binding domain tracked with neutralizing antibody titers, while S2-directed responses tracked with antibody-dependent myeloid cellular phagocytosis. Boosting the recovered patients with mRNA or inactivated vaccines expanded humoral breadth, durability, and restored functional responses, eliminating the severity- and platform-associated decay differences. Therefore, post-recovery hybrid immunization compensates for this distinction and broadens humoral breadth, highlighting the value of boosting immunity in previously infected individuals.
Collapse
Affiliation(s)
- Xin Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Benjamin Kellman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Maria-Jose Avendano
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Maanasa Mendu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Jeff C. Hsiao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Massachusetts, MA, 02139, USA
| | - Eileen Serrano
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Tamara Garcia-Salum
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Nicolas Muena
- Laboratorio de Virología Molecular, Fundación Ciencia and Vida, Santiago 8581151, Chile
| | - Catalina Pardo-Roa
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Department of Child and Adolescent Health, School of Nursing, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Mauricio Morales
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jorge Levican
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Erick Salinas
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | | | - Arnoldo Riquelme
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8331010, Chile
| | - Nicole D. Tischler
- Laboratorio de Virología Molecular, Fundación Ciencia and Vida, Santiago 8581151, Chile
- Escuela de Bioquímica, Facultad de Salud y Ciencia, Universidad San Sebastián, Santiago 7510156, Chile
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Massachusetts, MA, 02139, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Rafael A. Medina
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Center for Research on Influenza Pathogenesis and Transmission (CRIPT) Center of Excellence of Influenza Research and Response (CEIRR), New York, New York, USA
| |
Collapse
|
6
|
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
|
7
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
8
|
Meltzer HC, Goodwin JL, Fowler LA, Britt TW, Pirrallo RG, Grier JT. Severe acute respiratory syndrome coronavirus 2-reactive salivary antibody detection in South Carolina emergency healthcare workers, September 2019-March 2020. Epidemiol Infect 2024; 152:e102. [PMID: 39320488 PMCID: PMC11427973 DOI: 10.1017/s0950268824000967] [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: 09/26/2024] Open
Abstract
On 19 January 2020, the first case of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection was identified in the United States, with the first cases in South Carolina confirmed on 06 March 2020. Due to initial limited testing capabilities and potential for asymptomatic transmission, it is possible that SARS-CoV-2 may have been present earlier than previously thought, while the immune status of at-risk populations was unknown. Saliva from 55 South Carolina emergency healthcare workers (EHCWs) was collected from September 2019 to March 2020, pre- and post-healthcare shifts, and stored frozen. To determine the presence of SARS-CoV-2-reactive antibodies, saliva-acquired post-shift was analysed by enzyme-linked immunosorbent assay (ELISA) with a repeat of positive or inconclusive results and follow-up testing of pre-shift samples. Two participants were positive for SARS-CoV-2 N/S1-reactive IgG, confirmed by follow-up testing, with S1 receptor binding domain (RBD)-specific IgG present in one individual. Positive samples were collected from medical students working in emergency medical services (EMSs) in October or November 2019. The presence of detectable anti-SARS-CoV-2 antibodies in 2019 suggests that immune responses to the virus existed in South Carolina, and the United States, in a small percentage of EHCWs prior to the earliest documented coronavirus disease 2019 (COVID-19) cases. These findings suggest the feasibility of saliva as a noninvasive tool for surveillance of emerging outbreaks, and EHCWs represent a high-risk population that should be the focus of infectious disease surveillance.
Collapse
Affiliation(s)
- Haley C Meltzer
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| | - Jane L Goodwin
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| | - Lauren A Fowler
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Charlotte, NC, USA
| | - Thomas W Britt
- Department of Psychology, Clemson University, Clemson, SC, USA
| | - Ronald G Pirrallo
- Department of Emergency Medicine, University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| | - Jennifer T Grier
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| |
Collapse
|
9
|
Thimmiraju SR, Villar MJ, Kimata JT, Strych U, Bottazzi ME, Hotez PJ, Pollet J. Optimization of Cellular Transduction by the HIV-Based Pseudovirus Platform with Pan-Coronavirus Spike Proteins. Viruses 2024; 16:1492. [PMID: 39339968 PMCID: PMC11437443 DOI: 10.3390/v16091492] [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: 08/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past three years, new SARS-CoV-2 variants have continuously emerged, evolving to a point where an immune response against the original vaccine no longer provided optimal protection against these new strains. During this time, high-throughput neutralization assays based on pseudoviruses have become a valuable tool for assessing the efficacy of new vaccines, screening updated vaccine candidates against emerging variants, and testing the efficacy of new therapeutics such as monoclonal antibodies. Lentiviral vectors derived from HIV-1 are popular for developing pseudo and chimeric viruses due to their ease of use, stability, and long-term transgene expression. However, the HIV-based platform has lower transduction rates for pseudotyping coronavirus spike proteins than other pseudovirus platforms, necessitating more optimized methods. As the SARS-CoV-2 virus evolved, we produced over 18 variants of the spike protein for pseudotyping with an HIV-based vector, optimizing experimental parameters for their production and transduction. In this article, we present key parameters that were assessed to improve such technology, including (a) the timing and method of collection of pseudovirus supernatant; (b) the timing of host cell transduction; (c) cell culture media replenishment after pseudovirus adsorption; and (d) the centrifugation (spinoculation) parameters of the host cell+ pseudovirus mix, towards improved transduction. Additionally, we found that, for some pseudoviruses, the addition of a cationic polymer (polybrene) to the culture medium improved the transduction process. These findings were applicable across variant spike pseudoviruses that include not only SARS-CoV-2 variants, but also SARS, MERS, Alpha Coronavirus (NL-63), and bat-like coronaviruses. In summary, we present improvements in transduction efficiency, which can broaden the dynamic range of the pseudovirus titration and neutralization assays.
Collapse
Affiliation(s)
- Syamala Rani Thimmiraju
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Jose Villar
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
10
|
Li Z, Obraztsova A, Shang F, Oludada OE, Malapit J, Busch K, van Straaten M, Stebbins E, Murugan R, Wardemann H. Affinity-independent memory B cell origin of the early antibody-secreting cell response in naive individuals upon SARS-CoV-2 vaccination. Immunity 2024; 57:2191-2201.e5. [PMID: 39168129 DOI: 10.1016/j.immuni.2024.07.023] [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: 12/18/2023] [Revised: 02/02/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024]
Abstract
Memory B cells (MBCs) formed over the individual's lifetime constitute nearly half of the circulating B cell repertoire in humans. These pre-existing MBCs dominate recall responses to their cognate antigens, but how they respond to recognition of novel antigens is not well understood. Here, we tracked the origin and followed the differentiation paths of MBCs in the early anti-spike (S) response to mRNA vaccination in SARS-CoV-2-naive individuals on single-cell and monoclonal antibody levels. Pre-existing, highly mutated MBCs showed no signs of germinal center re-entry and rapidly developed into mature antibody-secreting cells (ASCs). By contrast, and despite similar levels of S reactivity, naive B cells showed strong signs of antibody affinity maturation before differentiating into MBCs and ASCs. Thus, pre-existing human MBCs differentiate into ASCs in response to novel antigens, but the quality of the humoral and cellular anti-S response improved through the clonal selection and affinity maturation of naive precursors.
Collapse
Affiliation(s)
- Zhe Li
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Anna Obraztsova
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany; Faculty of Biosciences, University of Heidelberg, Heidelberg 69120, Germany.
| | - Fuwei Shang
- Cellular Immunology, German Cancer Research Center, Heidelberg 69120, Germany; Faculty of Medicine, University of Heidelberg, Heidelberg 69120, Germany
| | - Opeyemi Ernest Oludada
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany; Faculty of Biosciences, University of Heidelberg, Heidelberg 69120, Germany
| | - Joshua Malapit
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany; Faculty of Biosciences, University of Heidelberg, Heidelberg 69120, Germany
| | - Katrin Busch
- Cellular Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Monique van Straaten
- Structural Biology of Infection and Immunity, German Cancer Research Center, Heidelberg 69120, Germany
| | - Erec Stebbins
- Structural Biology of Infection and Immunity, German Cancer Research Center, Heidelberg 69120, Germany
| | - Rajagopal Murugan
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Hedda Wardemann
- B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany.
| |
Collapse
|
11
|
Markov NS, Ren Z, Senkow KJ, Grant RA, Gao CA, Malsin ES, Sichizya L, Kihshen H, Helmin KA, Jovisic M, Arnold JM, Pérez-Leonor XG, Abdala-Valencia H, Swaminathan S, Nwaezeapu J, Kang M, Rasmussen L, Ozer EA, Lorenzo-Redondo R, Hultquist JF, Simons LM, Rios-Guzman E, Misharin AV, Wunderink RG, Budinger GRS, Singer BD, Morales-Nebreda L. Distinctive evolution of alveolar T cell responses is associated with clinical outcomes in unvaccinated patients with SARS-CoV-2 pneumonia. Nat Immunol 2024; 25:1607-1622. [PMID: 39138384 PMCID: PMC11490290 DOI: 10.1038/s41590-024-01914-w] [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: 01/15/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Abstract
The evolution of T cell molecular signatures in the distal lung of patients with severe pneumonia is understudied. Here, we analyzed T cell subsets in longitudinal bronchoalveolar lavage fluid samples from 273 patients with severe pneumonia, including unvaccinated patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or with respiratory failure not linked to pneumonia. In patients with SARS-CoV-2 pneumonia, activation of interferon signaling pathways, low activation of the NF-κB pathway and preferential targeting of spike and nucleocapsid proteins early after intubation were associated with favorable outcomes, whereas loss of interferon signaling, activation of NF-κB-driven programs and specificity for the ORF1ab complex late in disease were associated with mortality. These results suggest that in patients with severe SARS-CoV-2 pneumonia, alveolar T cell interferon responses targeting structural SARS-CoV-2 proteins characterize individuals who recover, whereas responses against nonstructural proteins and activation of NF-κB are associated with poor outcomes.
Collapse
Affiliation(s)
- Nikolay S Markov
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyou Ren
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karolina J Senkow
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A Grant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catherine A Gao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth S Malsin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lango Sichizya
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hermon Kihshen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn A Helmin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Milica Jovisic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason M Arnold
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xóchitl G Pérez-Leonor
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Suchitra Swaminathan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julu Nwaezeapu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mengjia Kang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Luke Rasmussen
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Egon A Ozer
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Judd F Hultquist
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lacy M Simons
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Estefany Rios-Guzman
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Richard G Wunderink
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
12
|
Serdyuk YV, Zornikova KV, Dianov DV, Ivanova NO, Davydova VD, Fefelova EI, Nenasheva TA, Sheetikov SA, Bogolyubova AV. T-Cell Receptors Cross-Reactive to Coronaviral Epitopes Homologous to the SPR Peptide. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1631-1642. [PMID: 39418521 DOI: 10.1134/s0006297924090098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024]
Abstract
The COVID-19 pandemic caused by the rapid spread of the novel coronavirus SARS-CoV-2, has promoted an interest in studying the T-cell immune response. It was found that the polyclonal and cross-reactive T-cell response against seasonal coronaviruses and other SARS-CoV-2 strains reduced disease severity. We investigated the immunodominant T-cell epitope SPRWYFYYYL from the nucleocapsid protein of SARS-CoV-2. The immune response to this epitope is characterized by the formation of highly homologous (convergent) receptors that have been found in the T-cell receptor (TCR) repertoires of different individuals. This epitope belongs to a group of highly conserved peptides that are rarely mutated in novel SARS-CoV-2 strains and are homologous to the epitopes of seasonal coronaviruses. It has been suggested that the cross-reactive response to homologous peptides contributes to the reduction of COVID-19 severity. However, some investigators have questioned this hypothesis, suggesting that the low affinity of the cross-reactive receptors reduces the strength of the immune response. The aim of this study was to evaluate the effect of amino acid substitutions in the SPR epitope on its binding affinity to specific TCRs. For this, we performed antigen-dependent cellular expansions were performed using samples from four COVID-19-transfected donors and sequenced their TCR repertoires. The resulting SPR-specific repertoire of β-chains in TCRs had a greater sequence diversity than the repertoire of α-chains. However, the TCR repertoires of all four donors contained public receptors, three of which were cloned and used to generate the Jurkat E6-1 TPR cell line. Only one of these receptors was activated by the SPR peptide and recognized with the same affinity by its mutant homologue LPRWYFYYY from seasonal coronaviruses. This indicates that the presence of the mutation did not affect the strength of the immune response, which may explain why the cross-reactive response to the SPR epitope is so frequent and contributes positively to COVID-19 infection.
Collapse
Affiliation(s)
- Yana V Serdyuk
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ksenia V Zornikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Dmitry V Dianov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nataliia O Ivanova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Vassa D Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina I Fefelova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Tatiana A Nenasheva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Saveliy A Sheetikov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Apollinariya V Bogolyubova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| |
Collapse
|
13
|
Kuthning D, Raafat D, Holtfreter S, Gramenz J, Wittmann N, Bröker BM, Meyer-Bahlburg A. Variant-specific antibody profiling for tracking SARS-CoV-2 variant infections in children and adolescents. Front Immunol 2024; 15:1434291. [PMID: 39257574 PMCID: PMC11384586 DOI: 10.3389/fimmu.2024.1434291] [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: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Monitoring the seroprevalence of SARS-CoV-2 in children and adolescents can provide valuable information for effective SARS-CoV-2 surveillance, and thus guide vaccination strategies. In this study, we quantified antibodies against the spike S1 domains of several SARS-CoV-2 variants (wild-type, Alpha, Delta, and Omicron variants) as well as endemic human coronaviruses (HCoVs) in 1,309 children and adolescents screened between December 2020 and March 2023. Their antibody binding profiles were compared with those of 22 pre-pandemic samples from children and adolescents using an in-house Luminex®-based Corona Array (CA). The primary objectives of this study were to (i) monitor SARS-CoV-2-specific antibodies in children and adolescents, (ii) evaluate whether the S1-specific antibody response can identify the infecting variant of concern (VoC), (iii) estimate the prevalence of silent infections, and (iv) test whether vaccination or infection with SARS-CoV-2 induce HCoV cross-reactive antibodies. Both SARS-CoV-2 infection and vaccination induced a robust antibody response against the S1 domain of WT and VoCs in children and adolescents. Antibodies specific for the S1 domain were able to distinguish between SARS-CoV-2 VoCs in infected children. The serologically identified VoC was typically the predominant VoC at the time of infection. Furthermore, our highly sensitive CA identified more silent SARS-CoV-2 infections than a commercial ELISA (12.1% vs. 6.3%, respectively), and provided insights into the infecting VoC. Seroconversion to endemic HCoVs occurred in early childhood, and vaccination or infection with SARS-CoV-2 did not induce HCoV S1 cross-reactive antibodies. In conclusion, the antibody response to the S1 domain of the spike protein of SARS-CoV-2 is highly specific, providing information about the infecting VoC and revealing clinically silent infections.
Collapse
Affiliation(s)
- Daniela Kuthning
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, 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
| | - Jana Gramenz
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nico Wittmann
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
14
|
Sumi T, Harada K. Vaccine and antiviral drug promise for preventing post-acute sequelae of COVID-19, and their combination for its treatment. Front Immunol 2024; 15:1329162. [PMID: 39185419 PMCID: PMC11341427 DOI: 10.3389/fimmu.2024.1329162] [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: 10/28/2023] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Most healthy individuals recover from acute SARS-CoV-2 infection, whereas a remarkable number continues to suffer from unexplained symptoms, known as Long COVID or post-acute COVID-19 syndrome (PACS). It is therefore imperative that methods for preventing and treating the onset of PASC be investigated with the utmost urgency. Methods A mathematical model of the immune response to vaccination and viral infection with SARS-CoV-2, incorporating immune memory cells, was developed. Results and discussion Similar to our previous model, persistent infection was observed by the residual virus in the host, implying the possibility of chronic inflammation and delayed recovery from tissue injury. Pre-infectious vaccination and antiviral medication administered during onset can reduce the acute viral load; however, they show no beneficial effects in preventing persistent infection. Therefore, the impact of these treatments on the PASC, which has been clinically observed, is mainly attributed to their role in preventing severe tissue damage caused by acute viral infections. For PASC patients with persistent infection, vaccination was observed to cause an immediate rapid increase in viral load, followed by a temporary decrease over approximately one year. The former was effectively suppressed by the coadministration of antiviral medications, indicating that this combination is a promising treatment for PASC.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, Okayama, Japan
- Department of Chemistry, Faculty of Science, Okayama University, Okayama, Japan
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Toyohashi, Aichi, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Toyohashi, Aichi, Japan
| |
Collapse
|
15
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [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: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
16
|
Kim W. Germinal Center Response to mRNA Vaccination and Impact of Immunological Imprinting on Subsequent Vaccination. Immune Netw 2024; 24:e28. [PMID: 39246619 PMCID: PMC11377948 DOI: 10.4110/in.2024.24.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 09/10/2024] Open
Abstract
Vaccines are the most effective intervention currently available, offering protective immunity against targeted pathogens. The emergence of the coronavirus disease 2019 pandemic has prompted rapid development and deployment of lipid nanoparticle encapsulated, mRNA-based vaccines. While these vaccines have demonstrated remarkable immunogenicity, concerns persist regarding their ability to confer durable protective immunity to continuously evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. This review focuses on human B cell responses induced by SARS-CoV-2 mRNA vaccination, with particular emphasis on the crucial role of germinal center reactions in shaping enduring protective immunity. Additionally, we explored observations of immunological imprinting and dynamics of recalled pre-existing immunity following variants of concern-based booster vaccination. Insights from this review contribute to comprehensive understanding B cell responses to mRNA vaccination in humans, thereby refining vaccination strategies for optimal and sustained protection against evolving coronavirus variants.
Collapse
Affiliation(s)
- Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
17
|
Yan X, Zhao X, Du Y, Wang H, Liu L, Wang Q, Liu J, Wei S. Dynamics of anti-SARS-CoV-2 IgG antibody responses following breakthrough infection and the predicted protective efficacy: A longitudinal community-based population study in China. Int J Infect Dis 2024; 145:107075. [PMID: 38697605 DOI: 10.1016/j.ijid.2024.107075] [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: 01/14/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES To assess the dynamics of the anti-SARS-CoV-2 IgG antibody levels and their efficacy against COVID-19. METHODS We conducted a longitudinal serological analysis of 852 breakthrough COVID-19 infections among the community-based population in Yichang, China. Anti-SARS-CoV-2 IgG levels were measured by chemiluminescence at approximately 3, 4, and 9 months after infection. A linear mixed model predicted IgG antibody decline over 18 months. The effectiveness of antibodies in preventing symptomatic and severe infections was determined using an existing meta-regression model. RESULTS IgG antibodies slowly declined after breakthrough infections. Initially high at around 3 months (339.44 AU/mL, IQR: 262.78-382.95 AU/mL), levels remained significant at 9 months (297.74 AU/mL, IQR: 213.22-360.62 AU/mL). The elderly (≥60 years) had lower antibody levels compared to the young (<20 years) (P < 0.001). The protective efficacy of antibodies against symptomatic and severe infections was lower in the elderly (≥60 years) (78.34% and 86.33%) compared to the young (<20 years) (96.56% and 98.75%) after 1 year. CONCLUSION The study indicated a slow decline in anti-SARS-CoV-2 IgG antibodies, maintaining considerable efficacy for over 1 year. However, lower levels in the elderly suggest reduced protective effects, underscoring the need for age-specific vaccination strategies.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhao
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Yin Du
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Hao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Liu
- Center for Disease Control and Prevention, Yichang, Hubei, China
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
18
|
Wilson R, Kovacs D, Crosby M, Ho A. Global Epidemiology and Seasonality of Human Seasonal Coronaviruses: A Systematic Review. Open Forum Infect Dis 2024; 11:ofae418. [PMID: 39113828 PMCID: PMC11304597 DOI: 10.1093/ofid/ofae418] [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: 04/03/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Background We characterized the global epidemiology and seasonality of human coronaviruses (HCoVs) OC43, NL63, 229E, and HKU1. Methods In this systematic review, we searched MEDLINE, EMBASE, Web of Science, SCOPUS, CINAHL, and backward citations for studies published until 1 September 2023. We included studies with ≥12 months of consecutive data and tested for ≥1 HCoV species. Case reports, review articles, animal studies, studies focusing on SARS-CoV-1, SARS-CoV-2, and/or Middle East respiratory syndrome, and those including <100 cases were excluded. Study quality and risk of bias were assessed using Joanna Briggs Institute Critical Appraisal Checklist tools. We reported the prevalence of all HCoVs and individual species. Seasonality was reported for studies that included ≥100 HCoVs annually. This study is registered with PROSPERO, CRD42022330902. Results A total of 201 studies (1 819 320 samples) from 68 countries were included. A high proportion were from China (19.4%; n = 39), whereas the Southern Hemisphere was underrepresented. Most were case series (77.1%, n = 155) with samples from secondary care (74.1%, n = 149). Seventeen (8.5%) studies included asymptomatic controls, whereas 76 (37.8%) reported results for all 4 HCoV species. Overall, OC43 was the most prevalent HCoV. Median test positivity of OC43 and NL63 was higher in children, and 229E and HKU1 in adults. Among 18 studies that described seasonality (17 from the Northern Hemisphere), circulation of all HCoVs mostly peaked during cold months. Conclusions In our comprehensive review, few studies reported the prevalence of individual HCoVs or seasonality. Further research on the burden and circulation of HCoVs is needed, particularly from Africa, South Asia, and Central/South America.
Collapse
Affiliation(s)
- Rory Wilson
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Dory Kovacs
- College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mairi Crosby
- College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Antonia Ho
- Medical Research Council-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Kozawa K, Higashimoto Y, Kawamura Y, Miura H, Hattori F, Mihara Y, Nakai H, Nishimura N, Ozaki T, Ihira M, Yoshikawa T. Temporal patterns of asymptomatic SARS-CoV-2 infection among pediatric population in Japan: A 3-year analysis. J Med Virol 2024; 96:e29847. [PMID: 39105394 DOI: 10.1002/jmv.29847] [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: 04/02/2024] [Revised: 07/02/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
To elucidate the seroprevalence and rate of asymptomatic infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Japanese children, serological analysis was performed using serum samples collected from March 2020 to February 2023. A total of 1493 serum samples were collected during the first study period (March 2020 to February 2021). None of the serum samples was positive for SARS-CoV-2 antibody. In the second period (March 2021 to February 2022), seven of the 1055 patients (0.7%) experienced SARS-CoV-2 infection. The third period (March 2022 to February 2023) was divided into three terms: from March to June 30, 2022; from July to October 2022; and from November 2022 to February 2023. The seroprevalence gradually increased throughout this period, with rates of 6.0%, 18.6%, and 30.4% in the three terms, respectively. Pediatric cases of asymptomatic SARS-CoV-2 infection occurred after the surge of Omicron variants. Since none of the SARS-CoV-2 antibody-positive patients had a previous history of coronavirus disease 2019, the seroprevalence rate in this study may represent the rate of asymptomatic infection.
Collapse
Affiliation(s)
- Kei Kozawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yuki Higashimoto
- Department of Clinical Microbiology, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yoshiki Kawamura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Miura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Fumihiko Hattori
- Department of Pediatrics, Toyokawa City Hospital, Toyokawa, Aichi, Japan
| | - Yuka Mihara
- Department of Pediatrics, Kariya Toyota General Hospital, Kariya, Aichi, Japan
| | - Hidetaka Nakai
- Department of Pediatrics, Toyokawa City Hospital, Toyokawa, Aichi, Japan
| | - Naoko Nishimura
- Department of Pediatrics, Konan Kosei Hospital, Konan, Aichi, Japan
| | - Takao Ozaki
- Department of Pediatrics, Konan Kosei Hospital, Konan, Aichi, Japan
| | - Masaru Ihira
- Department of Clinical Science for Biological Monitoring, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
20
|
Du P, Li N, Tang S, Zhou Z, Liu Z, Wang T, Li J, Zeng S, Chen J. Development and evaluation of vaccination strategies for addressing the continuous evolution SARS-CoV-2 based on recombinant trimeric protein technology: Potential for cross-neutralizing activity and broad coronavirus response. Heliyon 2024; 10:e34492. [PMID: 39148990 PMCID: PMC11324815 DOI: 10.1016/j.heliyon.2024.e34492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 08/17/2024] Open
Abstract
Given the significant decline in vaccine efficacy against Omicron, the development of novel vaccines with specific or broad-spectrum effectiveness is paramount. In this study, we formulated four monovalent vaccines based on recombinant spike trimer proteins, along with three bivalent vaccines, and five monovalent vaccines based on recombinant spike proteins. We evaluated the efficacy of different vaccination regimens in eliciting neutralizing antibodies in mice through pseudovirus neutralization assays. Following two doses of primary immunization with D614G, mice received subsequent immunizations with Omicron (BA.1, BA.2, BA.4/5) boosters individually, which led to the generation of broader and more potent cross-neutralizing activity compared to D614G boosters. Notably, the BA.4/5 booster exhibited superior efficacy. Following two doses of primary immunization with Omicron (BA.1, BA.2, BA.4/5), mice were subsequently immunized with one dose of D614G booster which resulted in broader neutralizing activity compared to one dose of Omicron (BA.1, BA.2, or BA.4/5). In unvaccinated mice, full-course immunization with different bivalent vaccines induced broad neutralizing activity against Omicron and pre-Omicron variants, with D614G&BA.4/5 demonstrating superior efficacy. However, compared to other variants, the neutralizing activity against XBB.1.5/1.9.1 is notably reduced. This observation emphasizes the necessity of timely updates to the vaccine antigen composition. Based on these findings and existing studies, we propose a vaccination strategy aimed at preserving the epitope repertoire to its maximum potential: (1) Individuals previously vaccinated or infected with pre-Omicron variants should inoculate a monovalent vaccine containing Omicron components; (2) Individuals who have only been vaccinated or infected with Omicron should be inoculated a monovalent vaccine containing pre-Omicron variants components; (3) Individuals without SARS-CoV-2 infection and vaccination should inoculate a bivalent vaccine comprising both pre-Omicron and Omicron components for primary immunization. Additionally, through cross-inoculation of SARS-CoV-2 D614G spike trimer protein and SARS-CoV-1 spike protein in mice, we preliminarily demonstrated the possibility of cross-reaction between different coronavirus vaccines to produce resistance to the pan-coronavirus.
Collapse
Affiliation(s)
- Peng Du
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Ning Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Shengjun Tang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Zhongcheng Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Zhihai Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Taorui Wang
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Jiahui Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Simiao Zeng
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Juan Chen
- Reproductive Medicine Center, Guangdong Second Provincial General Hospital, #466 Xin-Gang-Zhong-Lu, Haizhu District, Guangzhou, 510317, China
| |
Collapse
|
21
|
Lechuga GC, Temerozo JR, Napoleão-Pêgo P, Carvalho JPRS, Gomes LR, Bou-Habib DC, Morel CM, Provance DW, Souza TML, De-Simone SG. Enhanced Assessment of Cross-Reactive Antigenic Determinants within the Spike Protein. Int J Mol Sci 2024; 25:8180. [PMID: 39125749 PMCID: PMC11311977 DOI: 10.3390/ijms25158180] [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: 06/05/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Despite successful vaccination efforts, the emergence of new SARS-CoV-2 variants poses ongoing challenges to control COVID-19. Understanding humoral responses regarding SARS-CoV-2 infections and their impact is crucial for developing future vaccines that are effective worldwide. Here, we identified 41 immunodominant linear B-cell epitopes in its spike glycoprotein with an SPOT synthesis peptide array probed with a pool of serum from hospitalized COVID-19 patients. The bioinformatics showed a restricted set of epitopes unique to SARS-CoV-2 compared to other coronavirus family members. Potential crosstalk was also detected with Dengue virus (DENV), which was confirmed by screening individuals infected with DENV before the COVID-19 pandemic in a commercial ELISA for anti-SARS-CoV-2 antibodies. A high-resolution evaluation of antibody reactivity against peptides representing epitopes in the spike protein identified ten sequences in the NTD, RBD, and S2 domains. Functionally, antibody-dependent enhancement (ADE) in SARS-CoV-2 infections of monocytes was observed in vitro with pre-pandemic Dengue-positive sera. A significant increase in viral load was measured compared to that of the controls, with no detectable neutralization or considerable cell death, suggesting its role in viral entry. Cross-reactivity against peptides from spike proteins was observed for the pre-pandemic sera. This study highlights the importance of identifying specific epitopes generated during the humoral response to a pathogenic infection to understand the potential interplay of previous and future infections on diseases and their impact on vaccinations and immunodiagnostics.
Collapse
Affiliation(s)
- Guilherme C. Lechuga
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Jairo R. Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (J.R.T.); (D.C.B.-H.)
- National Institute for Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Paloma Napoleão-Pêgo
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
| | - João P. R. S. Carvalho
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
- Graduate Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Fluminense Federal University, Niterói 24220-900, Brazil
| | - Larissa R. Gomes
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (J.R.T.); (D.C.B.-H.)
- National Institute for Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Carlos M. Morel
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
| | - David W. Provance
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
| | - Thiago M. L. Souza
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Salvatore G. De-Simone
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswald Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (G.C.L.); (P.N.-P.); (L.R.G.); (C.M.M.); (T.M.L.S.)
- Graduate Program in Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Fluminense Federal University, Niterói 24220-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
22
|
Planchais C, Fernández I, Chalopin B, Bruel T, Rosenbaum P, Beretta M, Dimitrov JD, Conquet L, Donati F, Prot M, Porrot F, Planas D, Staropoli I, Guivel-Benhassine F, Baquero E, van der Werf S, Haouz A, Simon-Lorière E, Montagutelli X, Maillère B, Rey FA, Guardado-Calvo P, Nozach H, Schwartz O, Mouquet H. Broad sarbecovirus neutralization by combined memory B cell antibodies to ancestral SARS-CoV-2. iScience 2024; 27:110354. [PMID: 39071888 PMCID: PMC11277385 DOI: 10.1016/j.isci.2024.110354] [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: 04/25/2024] [Revised: 05/27/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Antibodies play a pivotal role in protecting from SARS-CoV-2 infection, but their efficacy is challenged by the continuous emergence of viral variants. In this study, we describe two broadly neutralizing antibodies cloned from the memory B cells of a single convalescent individual after infection with ancestral SARS-CoV-2. Cv2.3194, a resilient class 1 anti-RBD antibody, remains active against Omicron sub-variants up to BA.2.86. Cv2.3132, a near pan-Sarbecovirus neutralizer, targets the heptad repeat 2 membrane proximal region. When combined, Cv2.3194 and Cv2.3132 form a complementary SARS-CoV-2 neutralizing antibody cocktail exhibiting a local dose-dependent synergy. Thus, remarkably robust neutralizing memory B cell antibodies elicited in response to ancestral SARS-CoV-2 infection can withstand viral evolution and immune escape. The cooperative effect of such antibody combination may confer a certain level of protection against the latest SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Cyril Planchais
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, 75015 Paris, France
| | - Ignacio Fernández
- Institut Pasteur, Université Paris Cité, Structural Virology Unit, 75015 Paris, France
- CNRS UMR3569, 75015 Paris, France
| | - Benjamin Chalopin
- CEA, INRAE, Medicines and Healthcare Technologies Department, SIMoS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Timothée Bruel
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Pierre Rosenbaum
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, 75015 Paris, France
| | - Maxime Beretta
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, 75015 Paris, France
| | - Jordan D. Dimitrov
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Laurine Conquet
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, 75015 Paris, France
| | - Flora Donati
- Institut Pasteur, Université Paris Cité, G5 Evolutionary Genomics of RNA Viruses, 75015 Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, 75015 Paris, France
| | - Matthieu Prot
- Institut Pasteur, Université Paris Cité, G5 Evolutionary Genomics of RNA Viruses, 75015 Paris, France
| | - Françoise Porrot
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Delphine Planas
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Isabelle Staropoli
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Florence Guivel-Benhassine
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Eduard Baquero
- Institut Pasteur, Université Paris Cité, Structural Virology Unit, 75015 Paris, France
- CNRS UMR3569, 75015 Paris, France
| | - Sylvie van der Werf
- CNRS UMR3569, 75015 Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Molecular Genetics of RNA Viruses, 75015 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Cristallography Platform-C2RT, 75015 Paris, France
| | - Etienne Simon-Lorière
- Institut Pasteur, Université Paris Cité, G5 Evolutionary Genomics of RNA Viruses, 75015 Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, 75015 Paris, France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, 75015 Paris, France
| | - Bernard Maillère
- CEA, INRAE, Medicines and Healthcare Technologies Department, SIMoS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Félix A. Rey
- Institut Pasteur, Université Paris Cité, Structural Virology Unit, 75015 Paris, France
- CNRS UMR3569, 75015 Paris, France
| | - Pablo Guardado-Calvo
- Institut Pasteur, Université Paris Cité, Structural Virology Unit, 75015 Paris, France
- CNRS UMR3569, 75015 Paris, France
| | - Hervé Nozach
- CEA, INRAE, Medicines and Healthcare Technologies Department, SIMoS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Olivier Schwartz
- CNRS UMR3569, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Virus & Immunity Unit, 75015 Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, 75015 Paris, France
| |
Collapse
|
23
|
Fernandez SA, Pelaez-Prestel HF, Fiyouzi T, Gomez-Perosanz M, Reiné J, Reche PA. Tetanus-diphtheria vaccine can prime SARS-CoV-2 cross-reactive T cells. Front Immunol 2024; 15:1425374. [PMID: 39091504 PMCID: PMC11291333 DOI: 10.3389/fimmu.2024.1425374] [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: 04/29/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Vaccines containing tetanus-diphtheria antigens have been postulated to induce cross-reactive immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which could protect against coronavirus disease (COVID-19). In this work, we investigated the capacity of Tetanus-diphtheria (Td) vaccine to prime existing T cell immunity to SARS-CoV-2. To that end, we first collected known SARS-CoV-2 specific CD8+ T cell epitopes targeted during the course of SARS-CoV-2 infection in humans and identified as potentially cross-reactive with Td vaccine those sharing similarity with tetanus-diphtheria vaccine antigens, as judged by Levenshtein edit distances (≤ 20% edits per epitope sequence). As a result, we selected 25 potentially cross-reactive SARS-CoV-2 specific CD8+ T cell epitopes with high population coverage that were assembled into a synthetic peptide pool (TDX pool). Using peripheral blood mononuclear cells, we first determined by intracellular IFNγ staining assays existing CD8+ T cell recall responses to the TDX pool and to other peptide pools, including overlapping peptide pools covering SARS-CoV-2 Spike protein and Nucleocapsid phosphoprotein (NP). In the studied subjects, CD8+ T cell recall responses to Spike and TDX peptide pools were dominant and comparable, while recall responses to NP peptide pool were less frequent and weaker. Subsequently, we studied responses to the same peptides using antigen-inexperienced naive T cells primed/stimulated in vitro with Td vaccine. Priming stimulations were carried out by co-culturing naive T cells with autologous irradiated peripheral mononuclear cells in the presence of Td vaccine, IL-2, IL-7 and IL-15. Interestingly, naive CD8+ T cells stimulated/primed with Td vaccine responded strongly and specifically to the TDX pool, not to other SARS-CoV-2 peptide pools. Finally, we show that Td-immunization of C57BL/6J mice elicited T cells cross-reactive with the TDX pool. Collectively, our findings support that tetanus-diphtheria vaccines can prime SARS-CoV-2 cross-reactive T cells and likely contribute to shape the T cell responses to the virus.
Collapse
Affiliation(s)
- Sara Alonso Fernandez
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | - Hector F. Pelaez-Prestel
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | - Tara Fiyouzi
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | - Marta Gomez-Perosanz
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| | - Jesús Reiné
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Pedro A. Reche
- Department of Immunology & O2, Faculty of Medicine, Complutense University of Madrid, Ciudad Universitaria, Madrid, Spain
| |
Collapse
|
24
|
Wu X, Li W, Rong H, Pan J, Zhang X, Hu Q, Shi ZL, Zhang XE, Cui Z. A Nanoparticle Vaccine Displaying Conserved Epitopes of the Preexisting Neutralizing Antibody Confers Broad Protection against SARS-CoV-2 Variants. ACS NANO 2024; 18:17749-17763. [PMID: 38935412 DOI: 10.1021/acsnano.4c03075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The rapid development of the SARS-CoV-2 vaccine has been used to prevent the spread of coronavirus 2019 (COVID-19). However, the ongoing and future pandemics caused by SARS-CoV-2 variants and mutations underscore the need for effective vaccines that provide broad-spectrum protection. Here, we developed a nanoparticle vaccine with broad protection against divergent SARS-CoV-2 variants. The corresponding conserved epitopes of the preexisting neutralizing (CePn) antibody were presented on a self-assembling Helicobacter pylori ferritin to generate the CePnF nanoparticle. Intranasal immunization of mice with CePnF nanoparticles induced robust humoral, cellular, and mucosal immune responses and a long-lasting immunity. The CePnF-induced antibodies exhibited cross-reactivity and neutralizing activity against different coronaviruses (CoVs). CePnF vaccination significantly inhibited the replication and pathology of SARS-CoV-2 Delta, WIV04, and Omicron strains in hACE2 transgenic mice and, thus, conferred broad protection against these SARS-CoV-2 variants. Our constructed nanovaccine targeting the conserved epitopes of the preexisting neutralizing antibodies can serve as a promising candidate for a universal SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Xuefan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Heng Rong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jingdi Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Zheng-Li Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
| | - Xian-En Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
25
|
Awad H, El-Brolossy TA, Abdallah T, Osman A, Negm S, Mansour OI, Girgis SA, Hafez HM, Zaki AM, Talaat H. Accurate and reliable surface-enhanced Raman spectroscopy assay for early detection of SARS-CoV-2 RNA with exceptional sensitivity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124184. [PMID: 38608556 DOI: 10.1016/j.saa.2024.124184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/28/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024]
Abstract
This research proposes a highly sensitive and simple surface-enhanced Raman spectroscopy (SERS) assay for the detection of SARS-CoV-2 RNA using suitably designed probes specific for RdRp and N viral genes attached to a Raman marker. The sensitivity of the assay was optimized through precise adjustments to the conditions of immobilization and hybridization processes of the target RNA, including modifications to factors such as time and temperature. The assay achieved a remarkable sensitivity down to 58.39 copies/mL, comparable to or lower than the sensitivities reported for commercial fluorescent polymerase chain reaction (PCR) based methods. It has good selectivity in discriminating SARS-CoV-2 RNA against other respiratory viruses, respiratory syncytial virus (RSV), and influenza A virus. The reliability of the assay was validated by testing 24 clinical samples, including 12 positive samples with varying cycle threshold (Ct) values and 12 negative samples previously tested using real-time PCR. The assay consistently predicted true results that were in line with the PCR results for all samples. Furthermore, the assay demonstrated a notable limit of detection (LOD) of Ct (38 for RdRp gene and 37.5 for N-gene), indicating its capability to detect low concentrations of the target analyte and potentially facilitating early detection of the pathogen.
Collapse
Affiliation(s)
- Hend Awad
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Tamer Abdallah
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ahmed Osman
- Institute of Basic and Applied Science - Egpt-Japan University of Science and Technology (E-JUST), Egypt
| | - Sohair Negm
- Department of Physics and Mathematics, Banha University, Banha, Egypt
| | | | | | - Hala M Hafez
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali M Zaki
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hassan Talaat
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
26
|
Khan S, Partuk EO, Chiaravalli J, Kozer N, Shurrush KA, Elbaz-Alon Y, Scher N, Giraud E, Tran-Rajau J, Agou F, Barr HM, Avinoam O. High-throughput screening identifies broad-spectrum Coronavirus entry inhibitors. iScience 2024; 27:110019. [PMID: 38883823 PMCID: PMC11176637 DOI: 10.1016/j.isci.2024.110019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
The COVID-19 pandemic highlighted the need for antivirals against emerging coronaviruses (CoV). Inhibiting spike (S) glycoprotein-mediated viral entry is a promising strategy. To identify small molecule inhibitors that block entry downstream of receptor binding, we established a high-throughput screening (HTS) platform based on pseudoviruses. We employed a three-step process to screen nearly 200,000 small molecules. First, we identified hits that inhibit pseudoviruses bearing the SARS-CoV-2 S glycoprotein. Counter-screening against pseudoviruses with the vesicular stomatitis virus glycoprotein (VSV-G), yielded sixty-five SARS-CoV-2 S-specific inhibitors. These were further tested against pseudoviruses bearing the MERS-CoV S glycoprotein, which uses a different receptor. Out of these, five compounds, which included the known broad-spectrum inhibitor Nafamostat, were subjected to further validation and tested against pseudoviruses bearing the S glycoprotein of the Alpha, Delta, and Omicron variants as well as bona fide SARS-CoV-2. This rigorous approach revealed an unreported inhibitor and its derivative as potential broad-spectrum antivirals.
Collapse
Affiliation(s)
- Suman Khan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Efrat Ozer Partuk
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jeanne Chiaravalli
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Noga Kozer
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Khriesto A Shurrush
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Elbaz-Alon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nadav Scher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Emilie Giraud
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Jaouen Tran-Rajau
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Fabrice Agou
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Haim Michael Barr
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
27
|
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
|
28
|
He D, Artzy-Randrup Y, Musa SS, Gräf T, Naveca F, Stone L. Modelling the unexpected dynamics of COVID-19 in Manaus, Brazil. Infect Dis Model 2024; 9:557-568. [PMID: 38545442 PMCID: PMC10966176 DOI: 10.1016/j.idm.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 11/11/2024] Open
Abstract
In late March 2020, SARS-CoV-2 arrived in Manaus, Brazil, and rapidly developed into a large-scale epidemic that collapsed the local health system and resulted in extreme death rates. Several key studies reported that ∼76% of residents of Manaus were infected (attack rate AR≃76%) by October 2020, suggesting protective herd immunity had been reached. Despite this, an unexpected second wave of COVID-19 struck again in November and proved to be larger than the first, creating a catastrophe for the unprepared population. It has been suggested that this could be possible if the second wave was driven by reinfections. However, it is widely reported that reinfections were at a low rate (before the emergence of Omicron), and reinfections tend to be mild. Here, we use novel methods to model the epidemic from mortality data without considering reinfection-caused deaths and evaluate the impact of interventions to explain why the second wave appeared. The method fits a "flexible" reproductive number R 0 ( t ) that changes over the epidemic, and it is demonstrated that the method can successfully reconstruct R 0 ( t ) from simulated data. For Manaus, the method finds AR≃34% by October 2020 for the first wave, which is far less than required for herd immunity yet in-line with seroprevalence estimates. The work is complemented by a two-strain model. Using genomic data, the model estimates transmissibility of the new P.1 virus lineage as 1.9 times higher than that of the non-P.1. Moreover, an age class model variant that considers the high mortality rates of older adults show very similar results. These models thus provide a reasonable explanation for the two-wave dynamics in Manaus without the need to rely on large reinfection rates, which until now have only been found in negligible to moderate numbers in recent surveillance efforts.
Collapse
Affiliation(s)
- Daihai He
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yael Artzy-Randrup
- Department of Theoretical and Computational Ecology, IBED, University of Amsterdam, Amsterdam, Netherlands
| | - Salihu S. Musa
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong SAR, China
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Mathematics, Aliko Dangote University of Science and Technology, Kano, Nigeria
| | - Tiago Gräf
- Instituto Gonçalo Moniz, Fiocruz, Salvador, Bahia, Brazil
| | - Felipe Naveca
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
| | - Lewi Stone
- Mathematical Sciences, School of Science, RMIT University, Melbourne, Australia
- Biomathematics Unit, School of Zoology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
29
|
Almanzar G, Koosha K, Vogt T, Stein A, Ziegler L, Asam C, Weps M, Schwägerl V, Richter L, Hepp N, Fuchs A, Wagenhäuser I, Reusch J, Krone M, Geldmacher C, Protzer U, Steininger P, Überla K, Wagner R, Liese J, Prelog M. Hybrid immunity by two COVID-19 mRNA vaccinations and one breakthrough infection provides a robust and balanced cellular immune response as basic immunity against severe acute respiratory syndrome coronavirus 2. J Med Virol 2024; 96:e29739. [PMID: 38899449 DOI: 10.1002/jmv.29739] [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: 01/09/2024] [Revised: 04/22/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
This longitudinal prospective controlled multicenter study aimed to monitor immunity generated by three exposures caused by breakthrough infections (BTI) after COVID-19-vaccination considering pre-existing cell-mediated immunity to common-corona-viruses (CoV) which may impact cellular reactivity against SARS-CoV-2. Anti-SARS-CoV-2-spike-IgG antibodies (anti-S-IgG) and cellular reactivity against Spike-(S)- and nucleocapsid-(N)-proteins were determined in fully-vaccinated (F) individuals who either experienced BTI (F+BTI) or had booster vaccination (F+Booster) compared to partially vaccinated (P+BTI) and unvaccinated (U) from 1 to 24 weeks post PCR-confirmed infection. High avidity anti-S-IgG were found in F+BTI compared to U, the latter exhibiting increased long-lasting pro-inflammatory cytokines to S-stimulation. CoV was associated with higher cellular reactivity in U, whereas no association was seen in F. The study illustrates the induction of significant S-specific cellular responses in F+BTI building-up basic immunity by three exposures. Only U seem to benefit from pre-existing CoV immunity but demonstrated inflammatory immune responses compared to F+BTI who immunologically benefit from enhanced humoral and cellular immunity after BTI. This study demonstrates that individuals with hybrid immunity from COVID-19-vaccination and BTI acquire a stable humoral and cellular immune response that is maintained for at least 6 months. Our findings corroborate recommendations by health authorities to build on basic immunity by three S-protein exposures.
Collapse
Affiliation(s)
- Giovanni Almanzar
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Kimia Koosha
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Tim Vogt
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Astrid Stein
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Lars Ziegler
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Claudia Asam
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Manuela Weps
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Valeria Schwägerl
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Lorena Richter
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Nicola Hepp
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Andre Fuchs
- Internal Medicine III-Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Isabell Wagenhäuser
- Institute for Hygiene and Microbiology, Julius-Maximilian-Universität Würzburg, Würzburg, Germany
| | - Julia Reusch
- Institute for Hygiene and Microbiology, Julius-Maximilian-Universität Würzburg, Würzburg, Germany
| | - Manuel Krone
- Institute for Hygiene and Microbiology, Julius-Maximilian-Universität Würzburg, Würzburg, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ulrike Protzer
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
- German Center for Infection Research, Institute of Virology, Helmholtz Munich, Munich Partner Site, Munich, Germany
| | - Philipp Steininger
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf Wagner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Johannes Liese
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| | - Martina Prelog
- Department of Pediatrics, Pediatric Rheumatology/Special Immunology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
30
|
Chen X, Mohapatra A, Nguyen HTV, Schimanski L, Kit Tan T, Rijal P, Chen CP, Cheng SH, Lee WH, Chou YC, Townsend AR, Ma C, Huang KYA. The presence of broadly neutralizing anti-SARS-CoV-2 RBD antibodies elicited by primary series and booster dose of COVID-19 vaccine. PLoS Pathog 2024; 20:e1012246. [PMID: 38857264 PMCID: PMC11192315 DOI: 10.1371/journal.ppat.1012246] [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: 12/01/2023] [Revised: 06/21/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
Antibody-mediated immunity plays a key role in protection against SARS-CoV-2. We characterized B-cell-derived anti-SARS-CoV-2 RBD antibody repertoires from vaccinated and infected individuals and elucidate the mechanism of action of broadly neutralizing antibodies and dissect antibodies at the epitope level. The breadth and clonality of anti-RBD B cell response varies among individuals. The majority of neutralizing antibody clones lose or exhibit reduced activities against Beta, Delta, and Omicron variants. Nevertheless, a portion of anti-RBD antibody clones that develops after a primary series or booster dose of COVID-19 vaccination exhibit broad neutralization against emerging Omicron BA.2, BA.4, BA.5, BQ.1.1, XBB.1.5 and XBB.1.16 variants. These broadly neutralizing antibodies share genetic features including a conserved usage of the IGHV3-53 and 3-9 genes and recognize three clustered epitopes of the RBD, including epitopes that partially overlap the classically defined set identified early in the pandemic. The Fab-RBD crystal and Fab-Spike complex structures corroborate the epitope grouping of antibodies and reveal the detailed binding mode of broadly neutralizing antibodies. Structure-guided mutagenesis improves binding and neutralization potency of antibody with Omicron variants via a single amino-substitution. Together, these results provide an immunological basis for partial protection against severe COVID-19 by the ancestral strain-based vaccine and indicate guidance for next generation monoclonal antibody development and vaccine design.
Collapse
Affiliation(s)
- Xiaorui Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Hong Thuy Vy Nguyen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Lisa Schimanski
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Pramila Rijal
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Cheng-Pin Chen
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shu-Hsing Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Wen-Hsin Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Alain R. Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuan-Ying A. Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Immunology and Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
31
|
Adami FL, de Castro MV, Almeida BDS, Daher IP, Yamamoto MM, Souza Santos K, Zatz M, Naslavsky MS, Rosa DS, Cunha-Neto E, de Oliveira VL, Kalil J, Boscardin SB. Anti-RBD IgG antibodies from endemic coronaviruses do not protect against the acquisition of SARS-CoV-2 infection among exposed uninfected individuals. Front Immunol 2024; 15:1396603. [PMID: 38846944 PMCID: PMC11153698 DOI: 10.3389/fimmu.2024.1396603] [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: 03/06/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Background The Coronaviridae family comprises seven viruses known to infect humans, classified into alphacoronaviruses (HCoV-229E and HCoV-NL63) and betacoronaviruses (HCoV-OC43 and HCoV-HKU1), which are considered endemic. Additionally, it includes SARS-CoV (severe acute respiratory syndrome), MERS-CoV (Middle East respiratory syndrome), and the novel coronavirus SARS-CoV-2, responsible for COVID-19. SARS-CoV-2 induces severe respiratory complications, particularly in the elderly, immunocompromised individuals and those with underlying diseases. An essential question since the onset of the COVID-19 pandemic has been to determine whether prior exposure to seasonal coronaviruses influences immunity or protection against SARS-CoV-2. Methods In this study, we investigated a cohort of 47 couples (N=94), where one partner tested positive for SARS-CoV-2 infection via real-time PCR while the other remained negative. Plasma samples, collected at least 30 days post-PCR reaction, were assessed using indirect ELISA and competition assays to measure specific antibodies against the receptor-binding domain (RBD) portion of the Spike (S) protein from SARS-CoV-2, HCoV-229E, HCoV-NL63, HCoV-OC43, and HCoV-HKU1. Results IgG antibody levels against the four endemic coronavirus RBD proteins were similar between the PCR-positive and PCR-negative individuals, suggesting that IgG against endemic coronavirus RBD regions was not associated with protection from infection. Moreover, we found no significant IgG antibody cross-reactivity between endemic coronaviruses and SARS-CoV-2 RBDs. Conclusions Taken together, results suggest that anti-RBD antibodies induced by a previous infection with endemic HCoVs do not protect against acquisition of COVID-19 among exposed uninfected individuals.
Collapse
Affiliation(s)
- Flávia Lopes Adami
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Mateus Vidigal de Castro
- Centro de Estudos do Genoma Humano e Células Tronco, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Bianca da Silva Almeida
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Isabela Pazotti Daher
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Márcio Massao Yamamoto
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Keity Souza Santos
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Departamento de Clínica Médica, Disciplina de Alergia e Imunologia Clínica, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo, Brazil
| | - Mayana Zatz
- Centro de Estudos do Genoma Humano e Células Tronco, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Michel Satya Naslavsky
- Centro de Estudos do Genoma Humano e Células Tronco, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Disciplina de Imunologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Departamento de Clínica Médica, Disciplina de Alergia e Imunologia Clínica, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo, Brazil
| | - Vivian Leite de Oliveira
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Jorge Kalil
- Laboratório de Imunologia, LIM19, Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Departamento de Clínica Médica, Disciplina de Alergia e Imunologia Clínica, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Investigação em Imunologia-Instituto Nacional de Ciências e Tecnologia (iii-INCT), São Paulo, Brazil
| |
Collapse
|
32
|
Stanley AM, Aksyuk AA, Wilkins D, Green JA, Lan D, Shoemaker K, Tieu HV, Sobieszczyk ME, Falsey AR, Kelly EJ. Seasonal human coronavirus humoral responses in AZD1222 (ChaAdOx1 nCoV-19) COVID-19 vaccinated adults reveal limited cross-immunity. Front Immunol 2024; 15:1401728. [PMID: 38827749 PMCID: PMC11143795 DOI: 10.3389/fimmu.2024.1401728] [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: 03/15/2024] [Accepted: 04/30/2024] [Indexed: 06/04/2024] Open
Abstract
Background Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now widespread; however, the degree of cross-immunity between SARS-CoV-2 and endemic, seasonal human coronaviruses (HCoVs) remains unclear. Methods SARS-CoV-2 and HCoV cross-immunity was evaluated in adult participants enrolled in a US sub-study in the phase III, randomized controlled trial (NCT04516746) of AZD1222 (ChAdOx1 nCoV-19) primary-series vaccination for one-year. Anti-HCoV spike-binding antibodies against HCoV-229E, HCoV-HKU1, HCoV-OC43, and HCoV-NL63 were evaluated in participants following study dosing and, in the AZD1222 group, after a non-study third-dose booster. Timing of SARS-CoV-2 seroconversion (assessed via anti-nucleocapsid antibody levels) and incidence of COVID-19 were evaluated in those who received AZD1222 primary-series by baseline anti-HCoV titers. Results We evaluated 2,020/21,634 participants in the AZD1222 group and 1,007/10,816 in the placebo group. At the one-year data cutoff (March 11, 2022) mean duration of follow up was 230.9 (SD: 106.36, range: 1-325) and 94.3 (74.12, 1-321) days for participants in the AZD1222 (n = 1,940) and placebo (n = 962) groups, respectively. We observed little elevation in anti-HCoV humoral titers post study-dosing or post-boosting, nor evidence of waning over time. The occurrence and timing of SARS-CoV-2 seroconversion and incidence of COVID-19 were not largely impacted by baseline anti-HCoV titers. Conclusion We found limited evidence for cross-immunity between SARS-CoV-2 and HCoVs following AZD1222 primary series and booster vaccination. Susceptibility to future emergence of novel coronaviruses will likely persist despite a high prevalence of SARS-CoV-2 immunity in global populations.
Collapse
Affiliation(s)
- Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Anastasia A. Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Justin A. Green
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dongmei Lan
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Kathryn Shoemaker
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Hong-Van Tieu
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York-Presbyterian Columbia University Irving Medical Center, New York, NY, United States
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States
| | - Ann R. Falsey
- Department of Medicine, Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Infectious Disease, Rochester Regional Health, Rochester, NY, United States
| | - Elizabeth J. Kelly
- Formerly Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
33
|
Kulmala I, Taipale A, Sanmark E, Lastovets N, Sormunen P, Nuorti P, Saari S, Luoto A, Säämänen A. Estimated relative potential for airborne SARS-CoV-2 transmission in a day care centre. Heliyon 2024; 10:e30724. [PMID: 38756615 PMCID: PMC11096945 DOI: 10.1016/j.heliyon.2024.e30724] [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: 08/20/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
We estimated the hourly probability of airborne severe acute respiratory coronavirus 2 (SARS-CoV-2) transmission and further the estimated number of persons at transmission risk in a day care centre by calculating the inhaled dose for airborne pathogens based on their concentration, exposure time and activity. Information about the occupancy and activity of the rooms was collected from day care centre personnel and building characteristics were obtained from the design values. The generation rate of pathogens was calculated as a product of viral load of the respiratory fluids and the emission of the exhaled airborne particles, considering the prevalence of the disease and the activity of the individuals. A well-mixed model was used in the estimation of the concentration of pathogens in the air. The Wells-Riley model was used for infection probability. The approach presented in this study was utilised in the identification of hot spots and critical events in the day care centre. Large variation in the infection probabilities and estimated number of persons at transmission risk was observed when modelling a normal day at the centre. The estimated hourly infection probabilities between the worst hour in the worst room and the best hour in the best room varied in the ratio of 100:1. Similarly, the number of persons at transmission risk between the worst and best cases varied in the ratio 1000:1. Although there are uncertainties in the input values affecting the absolute risk estimates the model proved to be useful in ranking and identifying the hot spots and events in the building and implementing effective control measures.
Collapse
Affiliation(s)
- Ilpo Kulmala
- VTT Smart Energy and Built Environment, Visiokatu 4, PO Box 1300, FI-33101, Tampere, Finland
| | - Aimo Taipale
- VTT Smart Energy and Built Environment, Visiokatu 4, PO Box 1300, FI-33101, Tampere, Finland
| | - Enni Sanmark
- Helsinki University Hospital, Department of Otorhinolaryngology and Phoniatrics – Head and Neck Surgery, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | - Natalia Lastovets
- Tampere University, Faculty of Built Environment, Civil Engineering Unit, Korkeakoulunkatu 5D, FI-33720, Tampere, Finland
| | - Piia Sormunen
- Tampere University, Faculty of Built Environment, Civil Engineering Unit, Korkeakoulunkatu 5D, FI-33720, Tampere, Finland
| | - Pekka Nuorti
- Tampere University, Faculty of Social Sciences, Health Sciences Unit, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Sampo Saari
- Tampere University of Applied Sciences, Kuntokatu 3, 33520, Tampere, Finland
| | - Anni Luoto
- Granlund Oy, Malminkaari 21, 00700, Helsinki, Finland
| | - Arto Säämänen
- VTT Smart Energy and Built Environment, Visiokatu 4, PO Box 1300, FI-33101, Tampere, Finland
| |
Collapse
|
34
|
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
|
35
|
Yu Y, Zhang M, Huang L, Chen Y, Wu X, Li T, Li Y, Wang Y, Huang W. COVID-19 Serum Drives Spike-Mediated SARS-CoV-2 Variation. Viruses 2024; 16:763. [PMID: 38793644 PMCID: PMC11126028 DOI: 10.3390/v16050763] [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: 03/29/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Neutralizing antibodies targeting the spike (S) protein of SARS-CoV-2, elicited either by natural infection or vaccination, are crucial for protection against the virus. Nonetheless, the emergence of viral escape mutants presents ongoing challenges by contributing to breakthrough infections. To define the evolution trajectory of SARS-CoV-2 within the immune population, we co-incubated replication-competent rVSV/SARS-CoV-2/GFP chimeric viruses with sera from COVID-19 convalescents. Our findings revealed that the E484D mutation contributes to increased viral resistant against both convalescent and vaccinated sera, while the L1265R/H1271Y double mutation enhanced viral infectivity in 293T-hACE2 and Vero cells. These findings suggest that under the selective pressure of polyclonal antibodies, SARS-CoV-2 has the potential to accumulate mutations that facilitate either immune evasion or greater infectivity, facilitating its adaption to neutralizing antibody responses. Although the mutations identified in this study currently exhibit low prevalence in the circulating SARS-CoV-2 populations, the continuous and meticulous surveillance of viral mutations remains crucial. Moreover, there is an urgent necessity to develop next-generation antibody therapeutics and vaccines that target diverse, less mutation-prone antigenic sites to ensure more comprehensive and durable immune protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Yuanling Yu
- Changping Laboratory, Beijing 102206, China; (Y.Y.); (L.H.)
| | - Mengyi Zhang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (M.Z.)
- National Institutes for Food and Drug Control, Chinese Academy of Medical Science & Peking Union Medical College, No. 9 Dongdan Santiao, Dongcheng District, Beijing 100730, China
| | - Lan Huang
- Changping Laboratory, Beijing 102206, China; (Y.Y.); (L.H.)
| | - Yanhong Chen
- Changping Laboratory, Beijing 102206, China; (Y.Y.); (L.H.)
| | - Xi Wu
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (M.Z.)
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Tao Li
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (M.Z.)
| | - Yanbo Li
- Beijing Yunling Biotechnology Co., Ltd., Beijing 100176, China
| | - Youchun Wang
- Changping Laboratory, Beijing 102206, China; (Y.Y.); (L.H.)
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (M.Z.)
| |
Collapse
|
36
|
Lee R, Cheng S, Zhao J, Tsoi A, Lau K, Chan C, Li J, Hui D, Peiris M, Yen H. Cross-Reactive Antibody Responses to Coronaviruses Elicited by SARS-CoV-2 Infection or Vaccination. Influenza Other Respir Viruses 2024; 18:e13309. [PMID: 38725111 PMCID: PMC11082085 DOI: 10.1111/irv.13309] [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: 02/02/2024] [Revised: 03/28/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND The newly emerged SARS-CoV-2 possesses shared antigenic epitopes with other human coronaviruses. We investigated if COVID-19 vaccination or SARS-CoV-2 infection may boost cross-reactive antibodies to other human coronaviruses. METHODS Prevaccination and postvaccination sera from SARS-CoV-2 naïve healthy subjects who received three doses of the mRNA vaccine (BioNTech, BNT) or the inactivated vaccine (CoronaVac, CV) were used to monitor the level of cross-reactive antibodies raised against other human coronaviruses by enzyme-linked immunosorbent assay. In comparison, convalescent sera from COVID-19 patients with or without prior vaccination history were also tested. Pseudoparticle neutralization assay was performed to detect neutralization antibody against MERS-CoV. RESULTS Among SARS-CoV-2 infection-naïve subjects, BNT or CV significantly increased the anti-S2 antibodies against Betacoronaviruses (OC43 and MERS-CoV) but not Alphacoronaviruses (229E). The prevaccination antibody response to the common cold human coronaviruses did not negatively impact the postvaccination antibody response to SARS-CoV-2. Cross-reactive antibodies that binds to the S2 protein of MERS-CoV were similarly detected from the convalescent sera of COVID-19 patients with or without vaccination history. However, these anti-S2 antibodies do not possess neutralizing activity in MERS-CoV pseudoparticle neutralization tests. CONCLUSIONS Our results suggest that SARS-CoV-2 infection or vaccination may potentially modulate population immune landscape against previously exposed or novel human coronaviruses. The findings have implications for future sero-epidemiological studies on MERS-CoV.
Collapse
Affiliation(s)
- Richard S. H. Lee
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Samuel M. S. Cheng
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Jin Zhao
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Annie Y. S. Tsoi
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Kaman K. M. Lau
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - CoCo H. C. Chan
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - John K. C. Li
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - David S. C. Hui
- Department of Medicine and TherapeuticsCUHK, Prince of Wales HospitalHong KongChina
| | - Malik Peiris
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
- Centre for Immunology & InfectionHong Kong Science ParkHong KongChina
| | - Hui‐Ling Yen
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| |
Collapse
|
37
|
Simanavičius M, Kučinskaitė-Kodzė I, Kaselienė S, Sauliūnė S, Gudas D, Jančorienė L, Jasinskienė R, Vitkauskienė A, Žūtautienė R, Žvirblienė A, Stankūnas M. Prevalence of SARS-CoV-2-specific antibodies in a sample of the Lithuanian population-based study in Spring 2023. Heliyon 2024; 10:e29343. [PMID: 38681561 PMCID: PMC11053182 DOI: 10.1016/j.heliyon.2024.e29343] [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: 09/05/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 05/01/2024] Open
Abstract
Objectives Despite positive trends in SARS-CoV-2 epidemiology, seroprevalence surveys remain an important tool for estimating the magnitude of the COVID-19 pandemic. This study aimed to investigate the prevalence of IgG antibodies against SARS-CoV-2 nucleocapsid (N) and spike (S) proteins in a sample of the Lithuanian population (N = 517) and evaluate how the pattern of seropositivity correlates with the levels of SARS-CoV-2 infection and vaccination. Methods Study participants (aged 18-88 years) filled in the questionnaire self-reporting their demographic-social variables, health status, and SARS-CoV-2-related status. The anti-S and anti-N IgG levels were estimated using a microarray ELISA test. Results After several pandemic waves and vaccination campaign, the seroprevalence of SARS-CoV-2-specific IgG in the analyzed sample was 97.87 % by March-May 2023. We determined the 96.91 % prevalence of anti-S and 58.03 % prevalence of anti-N IgG. The majority of study participants (71.18 %) had hybrid immunity induced by vaccination and SARS-CoV-2 infection. 20.3 % of study participants were anti-N IgG positive without reporting any previous symptoms or a positive SARS-CoV-2 test. A decline of anti-N IgG positivity within 9 months after infection was observed. Conclusions This study demonstrates high total seroprevalence in March-May 2023 in all age groups indicating a widely established humoral immunity against SARS-CoV-2 in Lithuania.
Collapse
Affiliation(s)
- Martynas Simanavičius
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257, Vilnius, Lithuania
| | - Indrė Kučinskaitė-Kodzė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257, Vilnius, Lithuania
| | - Snieguolė Kaselienė
- Department of Health Management, Lithuanian University of Health Sciences, Tilžės g. 18, LT-47181, Kaunas, Lithuania
| | - Skirmantė Sauliūnė
- Department of Health Management, Lithuanian University of Health Sciences, Tilžės g. 18, LT-47181, Kaunas, Lithuania
| | - Dainius Gudas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257, Vilnius, Lithuania
| | - Ligita Jančorienė
- Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M. K. Čiurlionio g. 21, LT-03101, Vilnius, Lithuania
| | - Rūta Jasinskienė
- Faculty of Public Health, Lithuanian University of Health Sciences, Tilžės g. 18, LT-47181, Kaunas, Lithuania
| | - Astra Vitkauskienė
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, Eivenių g. 2, LT-50161, Kaunas, Lithuania
| | - Rasa Žūtautienė
- Department of Environmental and Occupational Medicine, Lithuanian University of Health Sciences, Tilžės g. 18, LT-47181, Kaunas, Lithuania
| | - Aurelija Žvirblienė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257, Vilnius, Lithuania
| | - Mindaugas Stankūnas
- Department of Health Management, Lithuanian University of Health Sciences, Tilžės g. 18, LT-47181, Kaunas, Lithuania
| |
Collapse
|
38
|
Pušnik J, Zorn J, Monzon-Posadas WO, Peters K, Osypchuk E, Blaschke S, Streeck H. Vaccination impairs de novo immune response to omicron breakthrough infection, a precondition for the original antigenic sin. Nat Commun 2024; 15:3102. [PMID: 38600072 PMCID: PMC11006949 DOI: 10.1038/s41467-024-47451-w] [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: 11/16/2023] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
Several studies have suggested the imprinting of SARS-CoV-2 immunity by original immune challenge without addressing the formation of the de novo response to successive antigen exposures. As this is crucial for the development of the original antigenic sin, we assessed the immune response against the mutated epitopes of omicron SARS-CoV-2 after vaccine breakthrough. Our data demonstrate a robust humoral response in thrice-vaccinated individuals following omicron breakthrough which is a recall of vaccine-induced memory. The humoral and memory B cell responses against the altered regions of the omicron surface proteins are impaired. The T cell responses to mutated epitopes of the omicron spike protein are present due to the high cross-reactivity of vaccine-induced T cells rather than the formation of a de novo response. Our findings, therefore, underpin the speculation that the imprinting of SARS-CoV-2 immunity by vaccination may lead to the development of original antigenic sin if future variants overcome the vaccine-induced immunity.
Collapse
Affiliation(s)
- Jernej Pušnik
- Institute of Virology, University Hospital Bonn, Bonn, Germany.
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany.
| | - Jasmin Zorn
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Werner O Monzon-Posadas
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
- Occupational Medicine Department, University Hospital Bonn, Bonn, Germany
| | - Kathrin Peters
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Emmanuil Osypchuk
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Sabine Blaschke
- Emergency Department, University Medical Center Goettingen, Goettingen, Germany
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| |
Collapse
|
39
|
Bartels M, Sala Solé E, Sauerschnig LM, Rijkers GT. Back to the Future: Immune Protection or Enhancement of Future Coronaviruses. Microorganisms 2024; 12:617. [PMID: 38543668 PMCID: PMC10975256 DOI: 10.3390/microorganisms12030617] [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: 02/05/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 11/12/2024] Open
Abstract
Before the emergence of SARS-CoV-1, MERS-CoV, and most recently, SARS-CoV-2, four other coronaviruses (the alpha coronaviruses NL63 and 229E and the beta coronaviruses OC43 and HKU1) had already been circulating in the human population. These circulating coronaviruses all cause mild respiratory illness during the winter seasons, and most people are already infected in early life. Could antibodies and/or T cells, especially against the beta coronaviruses, have offered some form of protection against (severe) COVID-19 caused by infection with SARS-CoV-2? Related is the question of whether survivors of SARS-CoV-1 or MERS-CoV would be relatively protected against SARS-CoV-2. More importantly, would humoral and cellular immunological memory generated during the SARS-CoV-2 pandemic, either by infection or vaccination, offer protection against future coronaviruses? Or rather than protection, could antibody-dependent enhancement have taken place, a mechanism by which circulating corona antibodies enhance the severity of COVID-19? Another related phenomenon, the original antigenic sin, would also predict that the effectiveness of the immune response to future coronaviruses would be impaired because of the reactivation of memory against irrelevant epitopes. The currently available evidence indicates that latter scenarios are highly unlikely and that especially cytotoxic memory T cells directed against conserved epitopes of human coronaviruses could at least offer partial protection against future coronaviruses.
Collapse
Affiliation(s)
| | | | | | - Ger T. Rijkers
- Science and Engineering Department, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.B.); (E.S.S.); (L.M.S.)
| |
Collapse
|
40
|
Escalera A, Rojo-Fernandez A, Rombauts A, Abelenda-Alonso G, Carratalà J, García-Sastre A, Aydillo T. SARS-CoV-2 infection induces robust mucosal antibody responses in the upper respiratory tract. iScience 2024; 27:109210. [PMID: 38433913 PMCID: PMC10906537 DOI: 10.1016/j.isci.2024.109210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/15/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
Despite multiple research efforts to characterize coronavirus disease 2019 (COVID-19) in humans, there is no clear data on the specific role of mucosal immunity on COVID-19 disease. Here, we longitudinally profile the antibody response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and seasonal HCoV-OC43 S proteins in serum and nasopharyngeal swabs from COVID-19 patients. Results showed that specific antibody responses against SARS-CoV-2 and HCoV-OC43 S proteins can be detected in the upper respiratory tract. We found that COVID-19 patients mounted a robust mucosal antibody response against SARS-CoV-2 S with specific secretory immunoglobulin A (sIgA), IgA, IgG, and IgM antibody subtypes detected in the nasal swabs. Additionally, COVID-19 patients showed IgG, IgA, and sIgA responses against HCoV-OC43 S in the local mucosa, whereas no specific IgM was detected. Interestingly, mucosal antibody titers against SARS-CoV-2 peaked at day 7, whereas HCoV-OC43 titers peaked earlier at day 3 post-recruitment, suggesting an immune memory recall to conserved epitopes of beta-HCoVs in the upper respiratory tract.
Collapse
Affiliation(s)
- Alba Escalera
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaya Rojo-Fernandez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander Rombauts
- Department of Infectious Diseases, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Center for Biomedical Research in Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCII), 28029 Madrid, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Center for Biomedical Research in Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCII), 28029 Madrid, Spain
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
41
|
Kim SH, Kim Y, Jeon S, Park U, Kang JI, Jeon K, Kim HR, Oh S, Rhee JY, Choi JP, Park WB, Park SW, Yang JS, Lee JY, Kang J, Shin HS, Kim Y, Kim S, Kim YS, Lim DG, Cho NH. Rise in broadly cross-reactive adaptive immunity against human β-coronaviruses in MERS-recovered patients during the COVID-19 pandemic. SCIENCE ADVANCES 2024; 10:eadk6425. [PMID: 38416834 PMCID: PMC10901372 DOI: 10.1126/sciadv.adk6425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/23/2024] [Indexed: 03/01/2024]
Abstract
To develop a universal coronavirus (CoV) vaccine, long-term immunity against multiple CoVs, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, Middle East respiratory syndrome (MERS)-CoV, and future CoV strains, is crucial. Following the 2015 Korean MERS outbreak, we conducted a long-term follow-up study and found that although neutralizing antibodies and memory T cells against MERS-CoV declined over 5 years, some recovered patients exhibited increased antibody levels during the COVID-19 pandemic. This likely resulted from cross-reactive immunity induced by SARS-CoV-2 vaccines or infections. A significant correlation in antibody responses across various CoVs indicates shared immunogenic epitopes. Two epitopes-the spike protein's stem helix and intracellular domain-were highly immunogenic after MERS-CoV infection and after SARS-CoV-2 vaccination or infection. In addition, memory T cell responses, especially polyfunctional CD4+ T cells, were enhanced during the pandemic, correlating significantly with MERS-CoV spike-specific antibodies and neutralizing activity. Therefore, incorporating these cross-reactive and immunogenic epitopes into pan-CoV vaccine formulations may facilitate effective vaccine development.
Collapse
Affiliation(s)
- So-Hee Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Yuri Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
| | - Sangeun Jeon
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea
| | - Uni Park
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Il Kang
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
| | - Kyeongseok Jeon
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Hye-Ran Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Songhyeok Oh
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ji-Young Rhee
- Division of Infectious Diseases, Department of Medicine, Dankook University College of Medicine, Cheonan 31116, Republic of Korea
| | - Jae-Phil Choi
- Department of Internal Medicine, Seoul Medical Center, Seoul 02053, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Won Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jeong-Sun Yang
- Center for Emerging Virus Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Republic of Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Republic of Korea
| | - Jihye Kang
- Translational Research Center, Research Institute of Public Health, National Medical Center, Seoul 04564, Republic of Korea
| | - Hyoung-Shik Shin
- Division of Infectious Diseases, Department of Internal Medicine, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon 34824, Republic of Korea
| | - Yeonjae Kim
- Center for Infectious Diseases, National Medical Center, Seoul 04564, Republic of Korea
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea
| | - Yeon-Sook Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Dong-Gyun Lim
- Translational Research Center, Research Institute of Public Health, National Medical Center, Seoul 04564, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Institute of Endemic Disease, Seoul National University Medical Research, Seoul 03080, Republic of Korea
- Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do 13620, Republic of Korea
| |
Collapse
|
42
|
Collins CP, Longo DL, Murphy WJ. The immunobiology of SARS-CoV-2 infection and vaccine responses: potential influences of cross-reactive memory responses and aging on efficacy and off-target effects. Front Immunol 2024; 15:1345499. [PMID: 38469293 PMCID: PMC10925677 DOI: 10.3389/fimmu.2024.1345499] [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: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Immune responses to both SARS-CoV-2 infection and its associated vaccines have been highly variable within the general population. The increasing evidence of long-lasting symptoms after resolution of infection, called post-acute sequelae of COVID-19 (PASC) or "Long COVID," suggests that immune-mediated mechanisms are at play. Closely related endemic common human coronaviruses (hCoV) can induce pre-existing and potentially cross-reactive immunity, which can then affect primary SARS-CoV-2 infection, as well as vaccination responses. The influence of pre-existing immunity from these hCoVs, as well as responses generated from original CoV2 strains or vaccines on the development of new high-affinity responses to CoV2 antigenic viral variants, needs to be better understood given the need for continuous vaccine adaptation and application in the population. Due in part to thymic involution, normal aging is associated with reduced naïve T cell compartments and impaired primary antigen responsiveness, resulting in a reliance on the pre-existing cross-reactive memory cell pool which may be of lower affinity, restricted in diversity, or of shorter duration. These effects can also be mediated by the presence of down-regulatory anti-idiotype responses which also increase in aging. Given the tremendous heterogeneity of clinical data, utilization of preclinical models offers the greatest ability to assess immune responses under a controlled setting. These models should now involve prior antigen/viral exposure combined with incorporation of modifying factors such as age on immune responses and effects. This will also allow for mechanistic dissection and understanding of the different immune pathways involved in both SARS-CoV-2 pathogen and potential vaccine responses over time and how pre-existing memory responses, including potential anti-idiotype responses, can affect efficacy as well as potential off-target effects in different tissues as well as modeling PASC.
Collapse
Affiliation(s)
- Craig P. Collins
- Graduate Program in Immunology, University of California (UC) Davis, Davis, CA, United States
| | - Dan L. Longo
- Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - William J. Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), University of California (UC) Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
43
|
Teng S, Hu Y, Wang Y, Tang Y, Wu Q, Zheng X, Lu R, Pan D, Liu F, Xie T, Wu C, Li YP, Liu W, Qu X. SARS-CoV-2 spike-reactive naïve B cells and pre-existing memory B cells contribute to antibody responses in unexposed individuals after vaccination. Front Immunol 2024; 15:1355949. [PMID: 38420128 PMCID: PMC10899457 DOI: 10.3389/fimmu.2024.1355949] [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: 12/14/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Since December 2019, the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing coronavirus disease 2019 (COVID-19) has presented considerable public health challenges. Multiple vaccines have been used to induce neutralizing antibodies (nAbs) and memory B-cell responses against the viral spike (S) glycoprotein, and many essential epitopes have been defined. Previous reports have identified severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-reactive naïve B cells and preexisting memory B cells in unexposed individuals. However, the role of these spike-reactive B cells in vaccine-induced immunity remains unknown. Methods To elucidate the characteristics of preexisting SARS-CoV-2 S-reactive B cells as well as their maturation after antigen encounter, we assessed the relationship of spike-reactive B cells before and after vaccination in unexposed human individuals. We further characterized the sequence identity, targeting domain, broad-spectrum binding activity and neutralizing activity of these SARS-CoV-2 S-reactive B cells by isolating monoclonal antibodies (mAbs) from these B cells. Results The frequencies of both spike-reactive naïve B cells and preexisting memory B cells before vaccination correlated with the frequencies of spike-reactive memory B cells after vaccination. Isolated mAbs from spike-reactive naïve B cells before vaccination had fewer somatic hypermutations (SHMs) than mAbs isolated from spike-reactive memory B cells before and after vaccination, but bound SARS-CoV-2 spike in vitro. Intriguingly, these germline-like mAbs possessed broad binding profiles for SARS-CoV-2 and its variants, although with low or no neutralizing capacity. According to tracking of the evolution of IGHV4-4/IGKV3-20 lineage antibodies from a single donor, the lineage underwent SHMs and developed increased binding activity after vaccination. Discussion Our findings suggest that spike-reactive naïve B cells can be expanded and matured by vaccination and cocontribute to vaccine-elicited antibody responses with preexisting memory B cells. Selectively and precisely targeting spike-reactive B cells by rational antigen design may provide a novel strategy for next-generation SARS-CoV-2 vaccine development.
Collapse
Affiliation(s)
- Shishan Teng
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yabin Hu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - You Wang
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yinggen Tang
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Qian Wu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Xingyu Zheng
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Rui Lu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Dong Pan
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Fen Liu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Tianyi Xie
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Chanfeng Wu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
- Translational Medicine Institute, The First People’s Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yi-Ping Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Wenpei Liu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
| | - Xiaowang Qu
- School of Public Health & School of Basic Medicine Sciences, Hengyang Medical School & Ministry of Education Key Laboratory of Rare Pediatric Diseases, University of South China, Hengyang, China
| |
Collapse
|
44
|
Luo C, Chen W, Cai J, He Y. The mechanisms of milder clinical symptoms of COVID-19 in children compared to adults. Ital J Pediatr 2024; 50:28. [PMID: 38355623 PMCID: PMC10865718 DOI: 10.1186/s13052-024-01587-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/07/2024] [Indexed: 02/16/2024] Open
Abstract
In stark contrast to adult patients, children who contract Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) typically manifest milder symptoms or remain asymptomatic. However, the precise underlying mechanisms of this pathogenesis remain elusive. In this review, we primarily retrospect the clinical characteristics of SARS-CoV-2 infection in children, and explore the factors that may contribute to the typically milder clinical presentation in pediatric Coronavirus Disease 2019 (COVID-19) patients compare with adults patients with COVID-19. The pathophysiological mechanisms that mitigate lung injury in children are as follows: the expression level of ACE2 receptor in children is lower; the binding affinity between ACE2 receptors and viral spike proteins in children was weaker; children have strong pre-activated innate immune response and appropriate adaptive immune response; children have more natural lymphocytes; children with COVID-19 can produce higher levels of IgM, IgG and interferon; children infected with SARS-CoV-2 can produce lower levels of IL-6 and IL-10; children have fewer underlying diseases and the lower risk of worsening COVID-19; children are usually exposed to other respiratory viruses and have an enhanced cross-reactive immunity. Comprehending the relative contributions of these processes to the protective phenotype in the developing lungs can help in the diagnosis, treatment and research pertaining to children with COVID-19.
Collapse
Affiliation(s)
- Caiyin Luo
- Department of Pharmacy, the First Affiliated Hospital of Guangzhou Medical University, 28 Qiaozhong Middle Road, Liwan District, 510120, Guangzhou, China
| | - Wanwen Chen
- Department of Pharmacy, the First Affiliated Hospital of Guangzhou Medical University, 28 Qiaozhong Middle Road, Liwan District, 510120, Guangzhou, China
| | - Junying Cai
- Department of Pharmacy, the First Affiliated Hospital of Guangzhou Medical University, 28 Qiaozhong Middle Road, Liwan District, 510120, Guangzhou, China
| | - Yuwen He
- Department of Pharmacy, the First Affiliated Hospital of Guangzhou Medical University, 28 Qiaozhong Middle Road, Liwan District, 510120, Guangzhou, China.
| |
Collapse
|
45
|
Abela IA, Schwarzmüller M, Ulyte A, Radtke T, Haile SR, Ammann P, Raineri A, Rueegg S, Epp S, Berger C, Böni J, Manrique A, Audigé A, Huber M, Schreiber PW, Scheier T, Fehr J, Weber J, Rusert P, Günthard HF, Kouyos RD, Puhan MA, Kriemler S, Trkola A, Pasin C. Cross-protective HCoV immunity reduces symptom development during SARS-CoV-2 infection. mBio 2024; 15:e0272223. [PMID: 38270455 PMCID: PMC10865973 DOI: 10.1128/mbio.02722-23] [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: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Numerous clinical parameters link to severe coronavirus disease 2019, but factors that prevent symptomatic disease remain unknown. We investigated the impact of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and endemic human coronavirus (HCoV) antibody responses on symptoms in a longitudinal children cohort (n = 2,917) and a cross-sectional cohort including children and adults (n = 882), all first exposed to SARS-CoV-2 (March 2020 to March 2021) in Switzerland. Saliva (n = 4,993) and plasma (n = 7,486) antibody reactivity to the four HCoVs (subunit S1 [S1]) and SARS-CoV-2 (S1, receptor binding domain, subunit S2 [S2], nucleocapsid protein) was determined along with neutralizing activity against SARS-CoV-2 Wuhan, Alpha, Delta, and Omicron (BA.2) in a subset of individuals. Inferred recent SARS-CoV-2 infection was associated with a strong correlation between mucosal and systemic SARS-CoV-2 anti-spike responses. Individuals with pre-existing HCoV-S1 reactivity exhibited significantly higher antibody responses to SARS-CoV-2 in both plasma (IgG regression coefficients = 0.20, 95% CI = [0.09, 0.32], P < 0.001) and saliva (IgG regression coefficient = 0.60, 95% CI = [0.088, 1.11], P = 0.025). Saliva neutralization activity was modest but surprisingly broad, retaining activity against Wuhan (median NT50 = 32.0, 1Q-3Q = [16.4, 50.2]), Alpha (median NT50 = 34.9, 1Q-3Q = [26.0, 46.6]), and Delta (median NT50 = 28.0, 1Q-3Q = [19.9, 41.7]). In line with a rapid mucosal defense triggered by cross-reactive HCoV immunity, asymptomatic individuals presented with higher pre-existing HCoV-S1 activity in plasma (IgG HKU1, odds ratio [OR] = 0.53, 95% CI = [0.29,0.97], P = 0.038) and saliva (total HCoV, OR = 0.55, 95% CI = [0.33, 0.91], P = 0.019) and higher SARS-CoV-2 reactivity in saliva (IgG S2 fold change = 1.26, 95% CI = [1.03, 1.54], P = 0.030). By investigating the systemic and mucosal immune responses to SARS-CoV-2 and HCoVs in a population without prior exposure to SARS-CoV-2 or vaccination, we identified specific antibody reactivities associated with lack of symptom development.IMPORTANCEKnowledge of the interplay between human coronavirus (HCoV) immunity and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection is critical to understanding the coexistence of current endemic coronaviruses and to building knowledge potential future zoonotic coronavirus transmissions. This study, which retrospectively analyzed a large cohort of individuals first exposed to SARS-CoV-2 in Switzerland in 2020-2021, revealed several key findings. Pre-existing HCoV immunity, particularly mucosal antibody responses, played a significant role in improving SARS-CoV-2 immune response upon infection and reducing symptoms development. Mucosal neutralizing activity against SARS-CoV-2, although low in magnitude, retained activity against SARS-CoV-2 variants underlining the importance of maintaining local mucosal immunity to SARS-CoV-2. While the cross-protective effect of HCoV immunity was not sufficient to block infection by SARS-CoV-2, the present study revealed a remarkable impact on limiting symptomatic disease. These findings support the feasibility of generating pan-protective coronavirus vaccines by inducing potent mucosal immune responses.
Collapse
Affiliation(s)
- Irene A. Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Agne Ulyte
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Thomas Radtke
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Sarah R. Haile
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Priska Ammann
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Alessia Raineri
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Sonja Rueegg
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Selina Epp
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | | - Jürg Böni
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Amapola Manrique
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Annette Audigé
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Peter W. Schreiber
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Scheier
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jan Fehr
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Jacqueline Weber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Peter Rusert
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roger D. Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Milo A. Puhan
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Susi Kriemler
- Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Zurich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Chloé Pasin
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Collegium Helveticum, Zurich, Switzerland
| |
Collapse
|
46
|
Zong K, Yuan P, Wang R, Luo Q, Yang Y, Zhang X, Song Q, Du H, Gao C, Song J, Zhan W, Zhang M, Wang Y, Lin Q, Yao H, Xie B, Han J. Characteristics of innate, humoral and cellular immunity in children with non-severe SARS-CoV-2 infection. J Infect 2024; 88:158-166. [PMID: 38101522 DOI: 10.1016/j.jinf.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
The symptoms of children infected with SARS-CoV-2 are mainly asymptomatic, mild, moderate, and a few severe cases. To understand the immune response characteristics of children infected with SARS-COV-2 who do not develop severe cases, 82 children infected with the SARS-CoV-2 delta strain were recruited in this study. Our results showed that high levels of IgG, IgM, and neutralization antibodies appeared in children infected with SARS-CoV-2. SARS-CoV-2 induced upregulation of both pro-inflammatory factors including TNF-α and anti-inflammatory factors including IL-4 and IL-13 in the children, even IL-10. The expression of INF-α in infected children also showed a significant increase compared to healthy children. However, IL-6, one of the important inflammatory factors, did not show an increase in infected children. It is worth noting that a large number of chemokines reduced in the SARS-CoV-2-infected children. Subsequently, TCR Repertoire, TCRβ bias, and preferential usage were analyzed on data of TCR next-generation sequencing from 8 SARS-CoV-2-infected children and 8 healthy controls. We found a significant decrease in TCR clonal diversity and a significant increase in TCR clonal expansion in SARS-CoV-2-infected children compared to healthy children. The most frequent V and J genes in SARS-CoV-2 children were TRBV28 and TRBJ2-1. The most frequently VβJ gene pairing in SARS-CoV-2 infected children was TRBV20-1-TRBJ2-1. The strong antiviral antibody levels, low expression of key pro-inflammatory factors, significant elevation of anti-inflammatory factors, and downregulation of many chemokines jointly determine that SARS-CoV-2-infected children rarely develop severe cases. Overall, our findings shed a light on the immune response of non-severe children infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Kexin Zong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Ping Yuan
- Fujian Provincial Key Laboratory of Zoonosis Research (Fujian Center for Disease Control and Prevention); The Practice Base on the School of Public Health, Fujian Medical University, Fuzhou, Fujian 350011, China
| | - Ruifang Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Qin Luo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Yanqing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Xiaohong Zhang
- Fujian Provincial Key Laboratory of Zoonosis Research (Fujian Center for Disease Control and Prevention); The Practice Base on the School of Public Health, Fujian Medical University, Fuzhou, Fujian 350011, China
| | - Qinqin Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Haijun Du
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Chen Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Juan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Weihua Zhan
- Putian Center for Disease Control and Prevention, Putian, Fujian 351106, China
| | - Mengjie Zhang
- Putian Center for Disease Control and Prevention, Putian, Fujian 351106, China
| | - Yanhai Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China
| | - Qunying Lin
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Hailan Yao
- Department of Biochemistry & Immunology, Capital Institute of Pediatrics, YaBao Rd, Beijing 100020, China.
| | - Baosong Xie
- Department of Pulmonary and Critical Care Medicine, Fujian Provincial Hospital; Fujian Shengli Medical College, Fujian Medical University, Fuzhou, Fujian 350001, China.
| | - Jun Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, China.
| |
Collapse
|
47
|
Cankat S, Demael MU, Swadling L. In search of a pan-coronavirus vaccine: next-generation vaccine design and immune mechanisms. Cell Mol Immunol 2024; 21:103-118. [PMID: 38148330 PMCID: PMC10805787 DOI: 10.1038/s41423-023-01116-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023] Open
Abstract
Members of the coronaviridae family are endemic to human populations and have caused several epidemics and pandemics in recent history. In this review, we will discuss the feasibility of and progress toward the ultimate goal of creating a pan-coronavirus vaccine that can protect against infection and disease by all members of the coronavirus family. We will detail the unmet clinical need associated with the continued transmission of SARS-CoV-2, MERS-CoV and the four seasonal coronaviruses (HCoV-OC43, NL63, HKU1 and 229E) in humans and the potential for future zoonotic coronaviruses. We will highlight how first-generation SARS-CoV-2 vaccines and natural history studies have greatly increased our understanding of effective antiviral immunity to coronaviruses and have informed next-generation vaccine design. We will then consider the ideal properties of a pan-coronavirus vaccine and propose a blueprint for the type of immunity that may offer cross-protection. Finally, we will describe a subset of the diverse technologies and novel approaches being pursued with the goal of developing broadly or universally protective vaccines for coronaviruses.
Collapse
Affiliation(s)
- S Cankat
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - M U Demael
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - L Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK.
| |
Collapse
|
48
|
Murphy WJ, Collins CP, Ashwood P. Potential role of anti-Idiotype responses on the neurological effects of post-acute sequelae of COVID-19 (PASC). Brain Behav Immun 2024; 116:317-320. [PMID: 38123027 DOI: 10.1016/j.bbi.2023.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Affiliation(s)
- William J Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), UC Davis School of Medicine, Sacramento, CA, United States.
| | - Craig P Collins
- Graduate Program in Immunology, UC Davis, Davis, CA, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, MIND Institute, UC Davis, Sacramento, CA, United States
| |
Collapse
|
49
|
Nguyen TTN, Choo EM, Nakamura Y, Suzuki R, Shiina T, Shin-I T, Fukuta M, Nguyen CT, Nguyen TTT, Nguyen LKH, Hoang VMP, Morita K, Dang DA, Hasebe F, Le TQM, Moi ML. Pre-existing cross-reactive neutralizing activity against SARS-CoV-2 and seasonal coronaviruses prior to the COVID-19 pandemic (2014-2019) with limited immunity against recent emerging SARS-CoV-2 variants, Vietnam. Int J Infect Dis 2024; 139:109-117. [PMID: 37984763 DOI: 10.1016/j.ijid.2023.11.008] [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: 07/25/2023] [Revised: 09/30/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
OBJECTIVES SARS-CoV-2 transmission and epidemic potential is related to the population's immunity levels. As such, assessing different regions' preexisting immune responses to SARS-CoV-2 is important to understand the transmission potential of emerging SARS-CoV-2 variants. DESIGN In 975 serum samples from Vietnam (2014 to 2019), anti-SARS-CoV-2 Immunoglobulin G levels were determined by enzyme-linked immunosorbent assay. Plaque reduction neutralization test (PRNT) was performed using Wuhan strain and variants of concern (VOCs). Cross-reactivity was confirmed by analyzing B-cell receptor (BCR) repertoire sequences and identifying BCR repertoire sequences-derived T-cell epitopes. RESULTS Overall, 20.9% (n = 76/364) and 9.2% (n = 7) demonstrated SARS-CoV-2 neutralizing activity (PRNT50) against the Wuhan and Alpha strain, respectively. Neutralizing activity against Beta, Gamma, and Delta strains was absent (PRNT50<5) in all samples. Cross-reactive epitopes against SARS-CoV-2 and other coronavirus spike proteins were detected in the N-terminal domain, S2, and receptor-binding domain regions. CONCLUSIONS Following BCR and major histocompatibility complex analysis, T-cell receptor-recognized epitope motif (TREM) among pathogenic coronaviruses and coronaviruses spike proteins were the top TREM peptide, suggesting that pre-existing immunity against SARS-CoV-2 in Vietnam was due to exposure to common cold coronaviruses. With limited immunity against emerging VOCs, further monitoring, and control of the epidemic, along with COVID-19 vaccine programs against VOCs, are necessary.
Collapse
Affiliation(s)
- Thi Thanh Ngan Nguyen
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, Nagasaki, Japan; Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan; National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Ee Mei Choo
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | | | - Ryuji Suzuki
- Repertoire Genesis Inc., Ibaraki, Japan; Sagamihara National Hospital, Kanagawa, Japan
| | | | | | - Mizuki Fukuta
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, Nagasaki, Japan; Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Co Thach Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | | | | | | | - Kouichi Morita
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, Nagasaki, Japan; Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Duc Anh Dang
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Futoshi Hasebe
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, Nagasaki, Japan; Vietnam Research Station, Center for Infectious Disease Research in Asia and Africa, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | | | - Meng Ling Moi
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, Nagasaki, Japan; Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan; Department of Developmental Medical Sciences, School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan.
| |
Collapse
|
50
|
Lapuente D, Winkler TH, Tenbusch M. B-cell and antibody responses to SARS-CoV-2: infection, vaccination, and hybrid immunity. Cell Mol Immunol 2024; 21:144-158. [PMID: 37945737 PMCID: PMC10805925 DOI: 10.1038/s41423-023-01095-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 prompted scientific, medical, and biotech communities to investigate infection- and vaccine-induced immune responses in the context of this pathogen. B-cell and antibody responses are at the center of these investigations, as neutralizing antibodies (nAbs) are an important correlate of protection (COP) from infection and the primary target of SARS-CoV-2 vaccine modalities. In addition to absolute levels, nAb longevity, neutralization breadth, immunoglobulin isotype and subtype composition, and presence at mucosal sites have become important topics for scientists and health policy makers. The recent pandemic was and still is a unique setting in which to study de novo and memory B-cell (MBC) and antibody responses in the dynamic interplay of infection- and vaccine-induced immunity. It also provided an opportunity to explore new vaccine platforms, such as mRNA or adenoviral vector vaccines, in unprecedented cohort sizes. Combined with the technological advances of recent years, this situation has provided detailed mechanistic insights into the development of B-cell and antibody responses but also revealed some unexpected findings. In this review, we summarize the key findings of the last 2.5 years regarding infection- and vaccine-induced B-cell immunity, which we believe are of significant value not only in the context of SARS-CoV-2 but also for future vaccination approaches in endemic and pandemic settings.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany.
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany
| |
Collapse
|