1
|
Files MA, Gentles L, Kehoe L, Adler A, Lacombe K, Dickerson JA, Greninger A, Waghmare A, Fairlie T, Pringle K, Midgley CM, Hagen MB, Englund JA, Seshadri C. Kinetics and Durability of Antibody and T-Cell Responses to SARS-CoV-2 in Children. J Infect Dis 2024; 230:889-900. [PMID: 38838218 PMCID: PMC11481334 DOI: 10.1093/infdis/jiae301] [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: 01/17/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The kinetics and durability of T-cell responses to SARS-CoV-2 in children are not well characterized. We studied a cohort of children aged 6 months to 20 years with COVID-19 in whom peripheral blood mononuclear cells and sera were archived at approximately 1, 6, and 12 months after symptom onset. METHODS We compared antibody responses (n = 85) and T-cell responses (n = 30) to nucleocapsid (N) and spike (S) glycoprotein over time across 4 age strata: 6 months to 5 years and 5-9, 10-14, and 15-20 years. RESULTS N-specific antibody responses declined over time, becoming undetectable in 26 (81%) of 32 children by approximately 1 year postinfection. Functional breadth of anti-N CD4+ T-cell responses also declined over time and were positively correlated with N-antibody responses (Pearson r = .31, P = .008). CD4+ T-cell responses to S displayed greater functional breadth than N in unvaccinated children and, with neutralization titers, were stable over time and similar across age strata. Functional profiles of CD4+ T-cell responses against S were not significantly modulated by vaccination. CONCLUSIONS Our data reveal durable age-independent T-cell immunity to SARS-CoV-2 structural proteins in children over time following COVID-19 infection as well as S-antibody responses in comparison with declining antibody responses to N.
Collapse
Affiliation(s)
- Megan A Files
- Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Lauren Gentles
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Leanne Kehoe
- Division of Pediatric Infectious Diseases, Seattle Children's Research Institute, Seattle, Washington
| | - Amanda Adler
- Division of Pediatric Infectious Diseases, Seattle Children's Research Institute, Seattle, Washington
| | - Kirsten Lacombe
- Division of Pediatric Infectious Diseases, Seattle Children's Research Institute, Seattle, Washington
| | - Jane A Dickerson
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, Washington
- Department of Laboratories, Seattle Children's Hospital, Seattle, Washington, USA
| | - Alexander Greninger
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, Washington
| | - Alpana Waghmare
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Center for Clinical and Translational Research, Seattle Children's Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Tarayn Fairlie
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kimberly Pringle
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Claire M Midgley
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Melissa Briggs Hagen
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Janet A Englund
- Division of Pediatric Infectious Diseases, Seattle Children's Research Institute, Seattle, Washington
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Chetan Seshadri
- Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
2
|
Qui M, Hariharaputran S, Hang SK, Zhang J, Tan CW, Chong CY, Low J, Wang L, Bertoletti A, Yung CF, Le Bert N. T cell hybrid immunity against SARS-CoV-2 in children: a longitudinal study. EBioMedicine 2024; 105:105203. [PMID: 38896919 PMCID: PMC11237860 DOI: 10.1016/j.ebiom.2024.105203] [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: 01/26/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Hybrid immunity to SARS-CoV-2, resulting from both vaccination and natural infection, remains insufficiently understood in paediatric populations, despite increasing rates of breakthrough infections among vaccinated children. METHODS We conducted a prospective longitudinal study to investigate the magnitude, specificity, and cytokine profile of antigen-specific T cell responses elicited by breakthrough SARS-CoV-2 infection in a cohort of mRNA-vaccinated children (n = 29) aged 5-11. This longitudinal analysis involved six distinct time points spanning a 16-month period post-vaccination, during which we analysed a total of 159 blood samples. All children who were followed for at least 12 months (n = 26) experienced a breakthrough infection. We conducted cytokine release assays using minimal blood samples, and we verified the cellular origin of these responses through intracellular cytokine staining. FINDINGS After breakthrough infection, children who had received mRNA vaccines showed enhanced Th1 responses specific to Spike peptides. Additionally, their Spike-specific T cells exhibited a distinctive enrichment of CD4+ IFN-γ+IL10+ cells, a characteristic akin to adults with hybrid immunity. Importantly, vaccination did not impede the development of multi-specific T cell responses targeting Membrane, Nucleoprotein, and ORF3a/7/8 antigens. INTERPRETATION Children, previously primed with a Spike-based mRNA vaccine and experiencing either symptomatic or asymptomatic breakthrough infection, retained the ability to enhance and diversify Th1/IL-10 antigen-specific T cell responses against multiple SARS-CoV-2 proteins. These findings mirror characteristics associated with hybrid cellular immunity in adults, known to confer resistance against severe COVID-19. FUNDING This study was funded by the National Medical Research Council (NMRC) Singapore (COVID19RF-0019, MOH-000019, MOH-000535, OFLCG19May-0034 and MOH-OFYIRG19nov-0002).
Collapse
Affiliation(s)
- Martin Qui
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Shou Kit Hang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jinyan Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chia Yin Chong
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jenny Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore General Hospital, Department of Infectious Diseases, Singapore
| | - Linfa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore Immunology Network, A∗STAR, Singapore
| | - Chee Fu Yung
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.
| |
Collapse
|
3
|
Menon T, Illing PT, Chaurasia P, McQuilten HA, Shepherd C, Rowntree LC, Petersen J, Littler DR, Khuu G, Huang Z, Allen LF, Rockman S, Crowe J, Flanagan KL, Wakim LM, Nguyen THO, Mifsud NA, Rossjohn J, Purcell AW, van de Sandt CE, Kedzierska K. CD8 + T-cell responses towards conserved influenza B virus epitopes across anatomical sites and age. Nat Commun 2024; 15:3387. [PMID: 38684663 PMCID: PMC11059233 DOI: 10.1038/s41467-024-47576-y] [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: 09/05/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Influenza B viruses (IBVs) cause substantive morbidity and mortality, and yet immunity towards IBVs remains understudied. CD8+ T-cells provide broadly cross-reactive immunity and alleviate disease severity by recognizing conserved epitopes. Despite the IBV burden, only 18 IBV-specific T-cell epitopes restricted by 5 HLAs have been identified currently. A broader array of conserved IBV T-cell epitopes is needed to develop effective cross-reactive T-cell based IBV vaccines. Here we identify 9 highly conserved IBV CD8+ T-cell epitopes restricted to HLA-B*07:02, HLA-B*08:01 and HLA-B*35:01. Memory IBV-specific tetramer+CD8+ T-cells are present within blood and tissues. Frequencies of IBV-specific CD8+ T-cells decline with age, but maintain a central memory phenotype. HLA-B*07:02 and HLA-B*08:01-restricted NP30-38 epitope-specific T-cells have distinct T-cell receptor repertoires. We provide structural basis for the IBV HLA-B*07:02-restricted NS1196-206 (11-mer) and HLA-B*07:02-restricted NP30-38 epitope presentation. Our study increases the number of IBV CD8+ T-cell epitopes, and defines IBV-specific CD8+ T-cells at cellular and molecular levels, across tissues and age.
Collapse
Affiliation(s)
- Tejas Menon
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Patricia T Illing
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Priyanka Chaurasia
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Chloe Shepherd
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Jan Petersen
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Dene R Littler
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Grace Khuu
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ziyi Huang
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Steve Rockman
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
- CSL Seqirus Ltd, Parkville, VIC, Australia
| | - Jane Crowe
- Deepdene Surgery, Deepdene, VIC, Australia
| | - Katie L Flanagan
- Tasmanian Vaccine Trial Centre, Launceston General Hospital, Launceston, TAS, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, VIC, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Nicole A Mifsud
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Anthony W Purcell
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| |
Collapse
|
4
|
Kim M, Cheng WA, Congrave-Wilson Z, Marentes Ruiz CJ, Turner L, Mendieta S, Jumarang J, Del Valle J, Lee Y, Fabrizio T, Allen EK, Thomas PG, Webby R, Gordon A, Pannaraj PS. Comparisons of Pediatric and Adult SARS-CoV-2-Specific Antibodies up to 6 Months after Infection, Vaccination, or Hybrid Immunity. J Pediatric Infect Dis Soc 2024; 13:91-99. [PMID: 38016076 PMCID: PMC10824260 DOI: 10.1093/jpids/piad107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Characterization of longitudinal SARS-CoV-2-specific antibody responses in children following infection and vaccination is needed to inform SARS-CoV-2 vaccine policy decisions for children, which may differ from adults. METHODS We enrolled individuals at the time of SARS-CoV-2 infection or vaccination for longitudinal serological testing and compared SARS-CoV-2-spike-specific IgG and neutralization activity in children and adults stratified by infection and vaccination status using enzyme-linked immunosorbent and virus neutralization assays. RESULTS Between June 2020 and December 2022, we collected sera from 669 participants aged 40 days to 55 years, including 330 unvaccinated individuals with laboratory-confirmed SARS-CoV-2 infection, 180 vaccinated SARS-CoV-2-naïve individuals, and 159 vaccinated previously infected individuals. Half (n = 330, 49.3%) were children. SARS-CoV-2-specific IgG and neutralization activity in children < 12 years old in response to infection persisted at higher levels than those of adults through at least 6 months (spike-specific IgG levels, 2.05 [95% CI: 1.4-3.1] times higher than adults; neutralizing activity, median 88.8 vs 75.2%, respectively, p = .04). In addition, all pediatric participants had significantly higher IgG levels compared with adults at 6 months following infection or vaccination, regardless of prior infection status. Vaccine-induced SARS-CoV-2-specific IgG responses in previously infected individuals persisted at higher levels than those from infection alone at 6 months (median AUC, children 5-11 years old, 9115 vs 368; adolescents 3613 vs 475; adults 1956 vs 263, all p < .001). CONCLUSIONS These data demonstrate the robust and persistent immunologic response of SARS-CoV-2 vaccination in children and emphasize the benefit of vaccination after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Minjun Kim
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Wesley A Cheng
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Zion Congrave-Wilson
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | | | - Lauren Turner
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Shirley Mendieta
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Jaycee Jumarang
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Jennifer Del Valle
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Yesun Lee
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Thomas Fabrizio
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richard Webby
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Pia S Pannaraj
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Division of Infectious Diseases, Rady Children’s Hospital, San Diego, CA, USA
| |
Collapse
|
5
|
Benede N, Tincho MB, Walters A, Subbiah V, Ngomti A, Baguma R, Butters C, Hahnle L, Mennen M, Skelem S, Adriaanse M, Facey-Thomas H, Scott C, Day J, Spracklen TF, van Graan S, Balla SR, Moyo-Gwete T, Moore PL, MacGinty R, Botha M, Workman L, Johnson M, Goldblatt D, Zar HJ, Ntusi NA, Zühlke L, Webb K, Riou C, Burgers WA, Keeton RS. Distinct T cell polyfunctional profile in SARS-CoV-2 seronegative children associated with endemic human coronavirus cross-reactivity. iScience 2024; 27:108728. [PMID: 38235336 PMCID: PMC10792240 DOI: 10.1016/j.isci.2023.108728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
SARS-CoV-2 infection in children typically results in asymptomatic or mild disease. There is a paucity of studies on SARS-CoV-2 antiviral immunity in African children. We investigated SARS-CoV-2-specific T cell responses in 71 unvaccinated asymptomatic South African children who were seropositive or seronegative for SARS-CoV-2. SARS-CoV-2-specific CD4+ T cell responses were detectable in 83% of seropositive and 60% of seronegative children. Although the magnitude of the CD4+ T cell response did not differ significantly between the two groups, their functional profiles were distinct, with SARS-CoV-2 seropositive children exhibiting a higher proportion of polyfunctional T cells compared to their seronegative counterparts. The frequency of SARS-CoV-2-specific CD4+ T cells in seronegative children was associated with the endemic human coronavirus (HCoV) HKU1 IgG response. Overall, the presence of SARS-CoV-2-responding T cells in seronegative children may result from cross-reactivity to endemic coronaviruses and could contribute to the relative protection from disease observed in SARS-CoV-2-infected children.
Collapse
Affiliation(s)
- Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Marius B. Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Avril Walters
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Vennesa Subbiah
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Claire Butters
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Lina Hahnle
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Heidi Facey-Thomas
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Jonathan Day
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Timothy F. Spracklen
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Strauss van Graan
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Sashkia R. Balla
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L. Moore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Rae MacGinty
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Maresa Botha
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Lesley Workman
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Heather J. Zar
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Ntobeko A.B. Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Liesl Zühlke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Kate Webb
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
- Crick African Network, The Francis Crick Institute, London, UK
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Roanne S. Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| |
Collapse
|
6
|
Horlenko OM, Hechko K, Prylypko LB, Hechko M, Horlenko FV, Tomey AI, Lenchenko AV. Analysis and interpretation of Coronavirus infection children's incidence, contributing factors, risks of complications and their relationship. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:484-490. [PMID: 38691790 DOI: 10.36740/wlek202403116] [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: 05/03/2024]
Abstract
OBJECTIVE Aim: To study and investigate the incidence of Coronavirus infection in children, the course of the disease, the risks of complications and their interrelationships. PATIENTS AND METHODS Materials and Methods: Study included the analysis and observation of children (n=55, aged 14.36±3.62 years) with confirmed Coronerovirus infection, who were observed in the CNE ≪CMCH≫ in Uzhgorod in outpatient conditions. A study of clinical presentations, a clinical and laboratory examination followed by a mathematical analysis of the symptoms data in children with an identified Coronavirus infection and in the dynamics up to week 30 (with survey intervals in 3 weeks) from the diagnosis verification was carried out. RESULTS Results: A dynamic analysis of the clinical manifestation of symptoms in children with an identified Corona virus infection and within 30 weeks (with survey intervals of 3 weeks) from the beginning of the diagnosis verification was carried out. Complaints from the respiratory system were prevailed. The most long-lasting complaint was observed ≪changes in the sense of taste and smell≫ (from 35(63.6%) to 6(10.9%) up to 18 weeks inclusive. Other complaints ≪Cough, Rhinitis, Shortness of breath, Pain in the chest≫ was observed for 6 weeks. Sore throat, muscular and joint pain were persisted for 3 weeks. Fever was not identified at week 3. Illness with other viral respiratory diseases started at week 9 and was observed until week 30 (from 10(18.2%) to 19(34.5%)) with varying levels. A decrease in cases of IgM identification was observed within 6 weeks (from 55, 100% to 20, 36, 4%). On the 9th week, the presence of IgM was not established. There is also an increase in the number of cases of detection of IgG in patients with a level maximum of 6 weeks. CONCLUSION Conclusions: There is a positive effect of the CRP level on the occurrence of symptoms of cough, rhinitis, shortness of breath, chest pain, change in taste and smell, muscle and joint pain (r=0.33-0.55), with the most significant data for the symptom of pain in chest (p=0.00001). Ferritin level interactions mostly had a negative direction (r=-0.35-0.48, p=0.02-0.00001) on the development of symptoms, with the exception of rhinorrhea (r=0.48, p=0.00002) and chest pains (r=0.39, 0.003). According to multiple logistic regression analysis the chance of the symptom of a change in taste and smell increases due to an increasing in the level of Procalcitonin in 1.48 times. The chance of the symptom of shortness of breath increased due to an increasing in the Ferritin level in 1.025 times.
Collapse
|
7
|
Bellocchio L, Dipalma G, Inchingolo AM, Inchingolo AD, Ferrante L, Del Vecchio G, Malcangi G, Palermo A, Qendro A, Inchingolo F. COVID-19 on Oral Health: A New Bilateral Connection for the Pandemic. Biomedicines 2023; 12:60. [PMID: 38255167 PMCID: PMC10813615 DOI: 10.3390/biomedicines12010060] [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: 11/27/2023] [Revised: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission are generally known to be produced by respiratory droplets and aerosols from the oral cavity (O.C.) of infected subjects, as stated by the World Health Organization. Saliva also retains the viral particles and aids in the spread of COVID-19. Angiotensin-converting enzyme Type 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are two of the numerous factors that promote SARS-CoV-2 infection, expressed by O.C. structures, various mucosa types, and the epithelia of salivary glands. A systemic SARS-CoV-2 infection might result from viral replication in O.C. cells. On the other hand, cellular damage of different subtypes in the O.C. might be associated with various clinical signs and symptoms. Factors interfering with SARS-CoV-2 infection potential might represent fertile ground for possible local pharmacotherapeutic interventions, which may confine SARS-CoV-2 virus entry and transmission in the O.C., finally representing a way to reduce COVID-19 incidence and severity.
Collapse
Affiliation(s)
- Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, University of Bordeaux, 33063 Bordeaux, France;
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Gaetano Del Vecchio
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Andis Qendro
- Faculty of Dental Medicine, University of Medicine, 1005 Tirana, Albania;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Study “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (L.F.); (G.D.V.); (F.I.)
| |
Collapse
|
8
|
Castro VT, Chardin H, Amorim dos Santos J, Barra GB, Castilho GR, Souza PM, Magalhães PDO, Acevedo AC, Guerra ENS. Detection of anti-SARS-CoV-2 salivary antibodies in vaccinated adults. Front Immunol 2023; 14:1296603. [PMID: 38022522 PMCID: PMC10661372 DOI: 10.3389/fimmu.2023.1296603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Since the introduction of efficient anti-SARS-CoV-2 vaccines, the detection of antibodies becomes useful for immunological monitoring and COVID-19 control. Therefore, this longitudinal study aimed to evaluate the detection of SARS-CoV-2 antibodies in the serum and saliva of COVID-19-vaccinated adults. The study included 13 not vaccinated and 35 vaccinated participants with two doses of CoronaVac (Sinovac/Butantan) vaccine who subsequently received BNT162b2 (Pfizer-BioNTech) vaccine as a booster dose. Vaccinated participants donated saliva and serum in three different time points. Enzyme-linked immunosorbent assay was used for antibody detection. In our results, the serum neutralizing antibodies (NAb) were detected in 34/35 samples after second dose and in 35/35 samples one and five months after the booster dose. In saliva, NAb were detected in 30/35 samples after second dose and in 35/35 of samples one and five months after the booster dose. IgA was detected in 19/34 saliva samples after second dose, in 18/35 one month after the booster and in 30/35 five months after. IgG in saliva was detected in 1/34 samples after second dose, 33/35 samples one month after the booster dose and in 20/35 five months after. A strong correlation was found between IgG and neutralizing activity in saliva, and salivary IgA would be a sign of recent exposure to the virus. In conclusion, saliva can be suitable for monitoring antibodies anti-SARS-CoV-2 after vaccination. Heterologous vaccination contributed to increase anti-SARS-CoV-2 antibodies in the Brazilian health context. Complementary studies with large groups are mandatory to conclude the interest in following mucosal immunity.
Collapse
Affiliation(s)
- Vitória Tavares Castro
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Hélène Chardin
- Department of Analytical, Bioanalytical Sciences and Miniaturization, École Supérieure de Physique et de Chimie Industrielles (ESPCI) de la Ville de Paris, Paris, France
- Unité de Formation et de Recherche d’Odontologie, Université Paris Cité, Paris, France
| | - Juliana Amorim dos Santos
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | | | | | - Paula Monteiro Souza
- Laboratory of Natural Products, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | | | - Ana Carolina Acevedo
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| |
Collapse
|
9
|
Soriano-Arandes A, Brett A, Buonsenso D, Emilsson L, de la Fuente Garcia I, Gkentzi D, Helve O, Kepp KP, Mossberg M, Muka T, Munro A, Papan C, Perramon-Malavez A, Schaltz-Buchholzer F, Smeesters PR, Zimmermann P. Policies on children and schools during the SARS-CoV-2 pandemic in Western Europe. Front Public Health 2023; 11:1175444. [PMID: 37564427 PMCID: PMC10411527 DOI: 10.3389/fpubh.2023.1175444] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/26/2023] [Indexed: 08/12/2023] Open
Abstract
During the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), mitigation policies for children have been a topic of considerable uncertainty and debate. Although some children have co-morbidities which increase their risk for severe coronavirus disease (COVID-19), and complications such as multisystem inflammatory syndrome and long COVID, most children only get mild COVID-19. On the other hand, consistent evidence shows that mass mitigation measures had enormous adverse impacts on children. A central question can thus be posed: What amount of mitigation should children bear, in response to a disease that is disproportionally affecting older people? In this review, we analyze the distinct child versus adult epidemiology, policies, mitigation trade-offs and outcomes in children in Western Europe. The highly heterogenous European policies applied to children compared to adults did not lead to significant measurable differences in outcomes. Remarkably, the relative epidemiological importance of transmission from school-age children to other age groups remains uncertain, with current evidence suggesting that schools often follow, rather than lead, community transmission. Important learning points for future pandemics are summarized.
Collapse
Affiliation(s)
- Antoni Soriano-Arandes
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ana Brett
- Infectious Diseases Unit and Emergency Service, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Milan, Italy
| | - Louise Emilsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna, Sweden
- Department of General Practice, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Isabel de la Fuente Garcia
- Pediatric Infectious Diseases, National Pediatric Center, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Despoina Gkentzi
- Department of Paediatrics, Patras Medical School, Patras, Greece
| | - Otto Helve
- Department of Health Security, Institute for Health and Welfare, Helsinki, Finland
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Kasper P. Kepp
- Section of Biophysical and Biomedicinal Chemistry, DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Maria Mossberg
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Epistudia, Bern, Switzerland
| | - Alasdair Munro
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Faculty of Medicine, Institute of Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Cihan Papan
- Institute for Hygiene and Public Health, University Hospital Bonn, Bonn, Germany
| | - Aida Perramon-Malavez
- Computational Biology and Complex Systems (BIOCOM-SC) Group, Department of Physics, Universitat Politècnica de Catalunya (UPC·BarcelonaTech), Barcelona, Spain
| | | | - Pierre R. Smeesters
- Department of Pediatrics, University Hospital Brussels, Academic Children’s Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
- Molecular Bacteriology Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Petra Zimmermann
- Department of Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
| |
Collapse
|
10
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Maniu I, Maniu GC, Antonescu E, Duica L, Grigore N, Totan M. SARS-CoV-2 Antibody Responses in Pediatric Patients: A Bibliometric Analysis. Biomedicines 2023; 11:biomedicines11051455. [PMID: 37239126 DOI: 10.3390/biomedicines11051455] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The characteristics, dynamics and mechanisms/determinants of the immune response to SARS-CoV-2 infection are not fully understood. We performed a bibliometric review of studies that have assessed SARS-CoV-2 antibody responses in the pediatric population using Web of Science online databases, VOSviewer and Bibliometrix tools. The analysis was conducted on 84 publications, from 310 institutions located in 29 countries and published in 57 journals. The results showed the collaboration of scientists and organizations, international research interactions and summarized the findings on (i) the measured titers of antibodies (total antibody and/or individual antibody classes IgG, IgM, IgA) against different antigens (C-terminal region of N (N CT), full-length N protein (N FL), RBD, RBD Alpha, RBD Beta, RBD Gamma, RBD Delta, spike (S), S1, S2) in the case of different clinical forms of the disease; and (ii) the correlations between SARS-CoV-2 antibodies and cytokines, chemokines, neutrophils, C-reactive protein, ferritin, and the erythrocyte sedimentation rate. The presented study offers insights regarding research directions to be explored in the studied field and may provide a starting point for future research.
Collapse
Affiliation(s)
- Ionela Maniu
- Mathematics and Informatics Department, Research Center in Informatics and Information Technology, Faculty of Sciences, "Lucian Blaga" University, 5-7 Ion Ratiu Str., 550025 Sibiu, Romania
- Pediatric Research Team, Clinical Pediatric Hospital, 2-4 Pompeiu Onofreiu Str., 550166 Sibiu, Romania
| | - George Constantin Maniu
- Mathematics and Informatics Department, Research Center in Informatics and Information Technology, Faculty of Sciences, "Lucian Blaga" University, 5-7 Ion Ratiu Str., 550025 Sibiu, Romania
| | - Elisabeta Antonescu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Lavinia Duica
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Nicolae Grigore
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- County Clinical Emergency Hospital, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Maria Totan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
- Clinical Laboratory, Clinical Pediatric Hospital, 2-4 Pompeiu Onofreiu Str., 550166 Sibiu, Romania
| |
Collapse
|
12
|
Nakayama EE, Shioda T. SARS-CoV-2 Related Antibody-Dependent Enhancement Phenomena In Vitro and In Vivo. Microorganisms 2023; 11:microorganisms11041015. [PMID: 37110438 PMCID: PMC10145615 DOI: 10.3390/microorganisms11041015] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies produced in the body after infection or vaccination may enhance subsequent viral infections in vitro and in vivo. Although rare, symptoms of viral diseases are also enhanced by ADE following infection or vaccination in vivo. This is thought to be due to the production of antibodies with low neutralizing activity that bind to the virus and facilitate viral entry, or antigen-antibody complexes that cause airway inflammation, or a predominance of T-helper 2 cells among the immune system cells which leads to excessive eosinophilic tissue infiltration. Notably, ADE of infection and ADE of disease are different phenomena that overlap. In this article, we will describe the three types of ADE: (1) Fc receptor (FcR)-dependent ADE of infection in macrophages, (2) FcR-independent ADE of infection in other cells, and (3) FcR-dependent ADE of cytokine production in macrophages. We will describe their relationship to vaccination and natural infection, and discuss the possible involvement of ADE phenomena in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
13
|
Li J, Chen Y, Ye W, Zhang M, Zhu J, Zhi W, Cheng Q. Molecular breast cancer subtype identification using photoacoustic spectral analysis and machine learning at the biomacromolecular level. PHOTOACOUSTICS 2023; 30:100483. [PMID: 37063308 PMCID: PMC10090435 DOI: 10.1016/j.pacs.2023.100483] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
Breast cancer threatens the health of women worldwide, and its molecular subtypes largely determine the therapy and prognosis of patients. However, an uncomplicated and accurate method to identify subtypes is currently lacking. This study utilized photoacoustic spectral analysis (PASA) based on the partial least squares discriminant algorithm (PLS-DA) to identify molecular breast cancer subtypes at the biomacromolecular level in vivo. The area of power spectrum density (APSD) was extracted to semi-quantify the biomacromolecule content. The feature wavelengths were obtained via the variable importance in projection (VIP) score and the selectivity ratio (Sratio), to identify the biomarkers. The PASA achieved an accuracy of 84%. Most of the feature wavelengths fell into the collagen-dominated absorption waveband, which was consistent with the histopathological results. This paper proposes a successful method for identifying molecular breast cancer subtypes and proves that collagen can be treated as a biomarker for molecular breast cancer subtyping.
Collapse
Affiliation(s)
- Jiayan Li
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Yingna Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Wanli Ye
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Mengjiao Zhang
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Jingtao Zhu
- School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Wenxiang Zhi
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China
| |
Collapse
|
14
|
Swadling L, Maini MK. Can T Cells Abort SARS-CoV-2 and Other Viral Infections? Int J Mol Sci 2023; 24:4371. [PMID: 36901802 PMCID: PMC10002440 DOI: 10.3390/ijms24054371] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Despite the highly infectious nature of the SARS-CoV-2 virus, it is clear that some individuals with potential exposure, or even experimental challenge with the virus, resist developing a detectable infection. While a proportion of seronegative individuals will have completely avoided exposure to the virus, a growing body of evidence suggests a subset of individuals are exposed, but mediate rapid viral clearance before the infection is detected by PCR or seroconversion. This type of "abortive" infection likely represents a dead-end in transmission and precludes the possibility for development of disease. It is, therefore, a desirable outcome on exposure and a setting in which highly effective immunity can be studied. Here, we describe how early sampling of a new pandemic virus using sensitive immunoassays and a novel transcriptomic signature can identify abortive infections. Despite the challenges in identifying abortive infections, we highlight diverse lines of evidence supporting their occurrence. In particular, expansion of virus-specific T cells in seronegative individuals suggests abortive infections occur not only after exposure to SARS-CoV-2, but for other coronaviridae, and diverse viral infections of global health importance (e.g., HIV, HCV, HBV). We discuss unanswered questions related to abortive infection, such as: 'Are we just missing antibodies? Are T cells an epiphenomenon? What is the influence of the dose of viral inoculum?' Finally, we argue for a refinement of the current paradigm that T cells are only involved in clearing established infection; instead, we emphasise the importance of considering their role in terminating early viral replication by studying abortive infections.
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K. Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| |
Collapse
|
15
|
Khoo WH, Jackson K, Phetsouphanh C, Zaunders JJ, Alquicira-Hernandez J, Yazar S, Ruiz-Diaz S, Singh M, Dhenni R, Kyaw W, Tea F, Merheb V, Lee FXZ, Burrell R, Howard-Jones A, Koirala A, Zhou L, Yuksel A, Catchpoole DR, Lai CL, Vitagliano TL, Rouet R, Christ D, Tang B, West NP, George S, Gerrard J, Croucher PI, Kelleher AD, Goodnow CG, Sprent JD, Powell JE, Brilot F, Nanan R, Hsu PS, Deenick EK, Britton PN, Phan TG. Tracking the clonal dynamics of SARS-CoV-2-specific T cells in children and adults with mild/asymptomatic COVID-19. Clin Immunol 2023; 246:109209. [PMID: 36539107 PMCID: PMC9758763 DOI: 10.1016/j.clim.2022.109209] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/28/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Children infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop less severe coronavirus disease 2019 (COVID-19) than adults. The mechanisms for the age-specific differences and the implications for infection-induced immunity are beginning to be uncovered. We show by longitudinal multimodal analysis that SARS-CoV-2 leaves a small footprint in the circulating T cell compartment in children with mild/asymptomatic COVID-19 compared to adult household contacts with the same disease severity who had more evidence of systemic T cell interferon activation, cytotoxicity and exhaustion. Children harbored diverse polyclonal SARS-CoV-2-specific naïve T cells whereas adults harbored clonally expanded SARS-CoV-2-specific memory T cells. A novel population of naïve interferon-activated T cells is expanded in acute COVID-19 and is recruited into the memory compartment during convalescence in adults but not children. This was associated with the development of robust CD4+ memory T cell responses in adults but not children. These data suggest that rapid clearance of SARS-CoV-2 in children may compromise their cellular immunity and ability to resist reinfection.
Collapse
Affiliation(s)
- Weng Hua Khoo
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | | | | | - John J Zaunders
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - José Alquicira-Hernandez
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Seyhan Yazar
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia
| | | | - Mandeep Singh
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Rama Dhenni
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Wunna Kyaw
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Fiona Tea
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Vera Merheb
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Fiona X Z Lee
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Rebecca Burrell
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | | | - Archana Koirala
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Li Zhou
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Aysen Yuksel
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Daniel R Catchpoole
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Catherine L Lai
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | | | - Romain Rouet
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Benjamin Tang
- Department of Intensive Care Medicine, Nepean Hospital, Sydney, Australia; Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, Australia; Respiratory Tract Infection Research Node, Marie Bashir Institute for Infectious Diseases and Biosecurity, Sydney, Australia
| | - Nicholas P West
- Systems Biology and Data Science, Menzies Health Institute QLD, Griffith University, Parklands, Australia
| | - Shane George
- Departments of Emergency Medicine and Children's Critical Care, Gold Coast University Hospital, Southport, QLD, Australia; School of Medicine and Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - John Gerrard
- Department of Infectious Diseases and Immunology, Gold Coast University Hospital, Southport, QLD, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | | | - Christopher G Goodnow
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia; UNSW Cellular Genomics Futures Institute, UNSW Sydney, Sydney, Australia
| | - Jonathan D Sprent
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Joseph E Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia; UNSW Cellular Genomics Futures Institute, UNSW Sydney, Sydney, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia; Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia; Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Ralph Nanan
- Charles Perkins Centre Nepean, University of Sydney, Sydney, Australia
| | - Peter S Hsu
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Elissa K Deenick
- Garvan Institute of Medical Research, Sydney, Australia; Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Philip N Britton
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
16
|
Davis SK, Selva KJ, Lopez E, Haycroft ER, Lee WS, Wheatley AK, Juno JA, Adair A, Pymm P, Redmond SJ, Gherardin NA, Godfrey DI, Tham W, Kent SJ, Chung AW. Heterologous SARS-CoV-2 IgA neutralising antibody responses in convalescent plasma. Clin Transl Immunology 2022; 11:e1424. [PMID: 36299410 PMCID: PMC9588388 DOI: 10.1002/cti2.1424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/21/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Following infection with SARS-CoV-2, virus-specific antibodies are generated, which can both neutralise virions and clear infection via Fc effector functions. The importance of IgG antibodies for protection and control of SARS-CoV-2 has been extensively reported. By comparison, other antibody isotypes including IgA have been poorly characterised. METHODS Here, we characterised plasma IgA from 41 early convalescent COVID-19 subjects for neutralisation and Fc effector functions. RESULTS Convalescent plasma IgA from > 60% of the cohort had the capacity to inhibit the interaction between wild-type RBD and ACE2. Furthermore, a third of the cohort induced stronger IgA-mediated ACE2 inhibition than matched IgG when tested at equivalent concentrations. Plasma IgA and IgG from this cohort broadly recognised similar RBD epitopes and had similar capacities to inhibit ACE2 from binding to 22 of the 23 prevalent RBD mutations assessed. However, plasma IgA was largely incapable of mediating antibody-dependent phagocytosis in comparison with plasma IgG. CONCLUSION Overall, convalescent plasma IgA contributed to the neutralising antibody response of wild-type SARS-CoV-2 RBD and various RBD mutations. However, this response displayed large heterogeneity and was less potent than IgG.
Collapse
Affiliation(s)
- Samantha K Davis
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Kevin John Selva
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Ester Lopez
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Ebene R Haycroft
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Wen Shi Lee
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia,The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Jennifer A Juno
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Amy Adair
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Phillip Pymm
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Samuel J Redmond
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Nicholas A Gherardin
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Dale I Godfrey
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| | - Wai‐Hong Tham
- The Walter and Eliza Hall Institute of Medical ResearchMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia,Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Amy W Chung
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
17
|
胡 亚, 姚 开. Clinical characteristics and epidemiological significance of coronavirus disease 2019 in children and adolescents. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:846-852. [PMID: 36036120 PMCID: PMC9425860 DOI: 10.7499/j.issn.1008-8830.2205026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/09/2022] [Indexed: 11/05/2022]
Abstract
The epidemic of coronavirus disease 2019 (COVID-19) started in late December 2019, and spread rapidly throughout the world. In March 2020, the World Health Organization (WHO) declared global epidemic of COVID-19. According to the American Academy of Pediatrics, nearly 13 million children have been diagnosed with COVID-19 since the outbreak. In general, children and teens have milder symptoms and fewer deaths from COVID-19 than adults. Understanding the symptoms, infectivity, and transmission patterns of COVID-19 in children and adolescents is of great significance for timely identifying suspected patients and developing effective control measures. Considering that some children will not be vaccinated for quite some time in the future, it is more important to improve the understanding of the clinical and epidemiological significance of COVID-19 in children and adolescents. This article summarizes the current understanding of the clinical manifestations and epidemiological significance of COVID-19 in children and adolescents to provide a reference for clinical diagnosis and treatment and the formulation of epidemic prevention and control strategies in children's gathering institutions such as kindergartens and schools.
Collapse
|
18
|
Nathanielsz J, Toh ZQ, Do LAH, Mulholland K, Licciardi PV. SARS-CoV-2 infection in children and implications for vaccination. Pediatr Res 2022; 93:1177-1187. [PMID: 35970935 PMCID: PMC9376896 DOI: 10.1038/s41390-022-02254-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/20/2022] [Accepted: 06/28/2022] [Indexed: 11/09/2022]
Abstract
The COVID-19 pandemic caused by novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for more than 500 million cases worldwide as of April 2022. Initial estimates in 2020 found that children were less likely to become infected with SARS-CoV-2 and more likely to be asymptomatic or display mild COVID-19 symptoms. Our early understanding of COVID-19 transmission and disease in children led to a range of public health measures including school closures that have indirectly impacted child health and wellbeing. The emergence of variants of concern (particularly Delta and Omicron) has raised new issues about transmissibility in children, as preliminary data suggest that children may be at increased risk of infection, especially if unvaccinated. Global national prevalence data show that SARS-CoV-2 infection in children and adolescents is rising due to COVID-19 vaccination among adults and increased circulation of Delta and Omicron variants. To mitigate this, childhood immunisation programmes are being implemented globally to prevent direct and indirect consequences of COVID-19 including severe complications (e.g., MIS-C), debilitating long-COVID symptoms, and the indirect impacts of prolonged community and school closures on childhood education, social and behavioural development and mental health. This review explores the current state of knowledge on COVID-19 in children including COVID-19 vaccination strategies. IMPACT: Provides an up-to-date account of SARS-CoV-2 infections in children. Discusses the direct and indirect effects of COVID-19 in children. Provides the latest information on the current state of global COVID-19 vaccination in children.
Collapse
Affiliation(s)
- Jordan Nathanielsz
- grid.1021.20000 0001 0526 7079School of Medicine, Deakin University, Geelong, VIC Australia ,grid.267362.40000 0004 0432 5259Department of Medicine, Alfred Health, Melbourne, VIC Australia ,grid.1058.c0000 0000 9442 535XInfection and Immunity, Murdoch Children’s Research Institute, Melbourne, VIC Australia
| | - Zheng Quan Toh
- grid.1058.c0000 0000 9442 535XInfection and Immunity, Murdoch Children’s Research Institute, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, The University of Melbourne, Melbourne, VIC Australia
| | - Lien Anh Ha Do
- grid.1058.c0000 0000 9442 535XInfection and Immunity, Murdoch Children’s Research Institute, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, The University of Melbourne, Melbourne, VIC Australia
| | - Kim Mulholland
- grid.1058.c0000 0000 9442 535XInfection and Immunity, Murdoch Children’s Research Institute, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, The University of Melbourne, Melbourne, VIC Australia ,grid.8991.90000 0004 0425 469XDepartment of Infectious Diseases Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Paul V. Licciardi
- grid.1058.c0000 0000 9442 535XInfection and Immunity, Murdoch Children’s Research Institute, Melbourne, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Paediatrics, The University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
19
|
Kumar NP, Venkataraman A, Nancy A, Moideen K, Varadarjan P, Selladurai E, Sangaralingam T, Selvam R, Thimmaiah A, Natarajan S, Ramasamy G, Hissar S, Radayam Ranganathan U, Babu S. Enhanced Severe Acute Respiratory Syndrome Coronavirus 2 Antigen-Specific Systemic Immune Responses in Multisystem Inflammatory Syndrome in Children and Reversal After Recovery. J Infect Dis 2022; 226:1215-1223. [PMID: 35932220 PMCID: PMC9384631 DOI: 10.1093/infdis/jiac304] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 01/19/2023] Open
Abstract
Background Multisystem inflammatory syndrome in children (MIS-C) presents with inflammation and pathology of multiple organs in the pediatric population in the weeks following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Methods We characterized the SARS-CoV-2 antigen–specific cytokine and chemokine responses in children with MIS-C, coronavirus disease 2019 (COVID-19), and other infectious diseases. Results MIS-C is characterized by elevated levels of type 1 (interferon-γ, interleukin [IL] 2), type 2 (IL-4, IL-13), type 17 (IL-17), and other proinflammatory cytokines (IL-1α, IL-6, IL-12p70, IL-18, and granulocyte-macrophage colony-stimulating factor) in comparison to COVID-19 and other infectious diseases following stimulation with SARS-CoV-2–specific antigens. Similarly, upon SARS-CoV-2 antigen stimulation, CCL2, CCL3, and CXCL10 chemokines were significantly elevated in children with MIS-C in comparison to the other 2 groups. Principal component analysis based on these cytokines and chemokines could clearly distinguish MIS-C from both COVID-19 and other infections. In addition, these responses were significantly diminished and normalized 6–9 months after recovery. Conclusions Our data suggest that MIS-C is characterized by an enhanced production of cytokines and chemokines that may be associated with disease pathogenesis.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- Department of Immunology, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Aishwarya Venkataraman
- Department of Immunology, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Arul Nancy
- ICER, National Institute for Research in Tuberculosis, National Institutes of Health-International Center for Excellence in Research, Chennai, India
| | - Kadar Moideen
- ICER, National Institute for Research in Tuberculosis, National Institutes of Health-International Center for Excellence in Research, Chennai, India
| | - Poovazhagi Varadarjan
- Pediatric Intensive Care Unit, Institute of Child Health and Hospital for Children, Chennai, India
| | - Elilarasi Selladurai
- Pediatric Intensive Care Unit, Institute of Child Health and Hospital for Children, Chennai, India
| | | | - Ramya Selvam
- General Pediatrics, Dr.Mehta's Children's Hospital, Chennai, India
| | | | - Suresh Natarajan
- Pediatric Pulmonology, Rainbow Children's Hospital, Chennai, India
| | - Ganesh Ramasamy
- Pediatric Pulmonology, Rainbow Children's Hospital, Chennai, India
| | - Syed Hissar
- Department of Immunology, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Umadevi Radayam Ranganathan
- Department of Immunology, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Subash Babu
- ICER, National Institute for Research in Tuberculosis, National Institutes of Health-International Center for Excellence in Research, Chennai, India
| |
Collapse
|
20
|
Recovering or Persisting: The Immunopathological Features of SARS-CoV-2 Infection in Children. J Clin Med 2022; 11:jcm11154363. [PMID: 35955979 PMCID: PMC9369242 DOI: 10.3390/jcm11154363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 12/18/2022] Open
Abstract
Background. The profile of cellular immunological responses of children across the spectrum of COVID-19, ranging from acute SARS-CoV-2 infection to full recovery or Long COVID, has not yet been fully investigated. Methods. We examined and compared cytokines in sera and cell subsets in peripheral blood mononuclear cells (B and regulatory T lymphocytes) collected from four distinct groups of children, distributed as follows: younger than 18 years of age with either acute SARS-CoV-2 infection (n = 49); fully recovered from COVID-19 (n = 32); with persistent symptoms (Long COVID, n = 51); and healthy controls (n = 9). Results. In the later stages after SARS-CoV-2 infection, the cohorts of children, both with recovered and persistent symptoms, showed skewed T and B subsets, with remarkable differences when compared with children at the onset of the infection and with controls. The frequencies of IgD+CD27− naïve B cells, IgD+IgM+ and CD27−IgM+CD38dim B cells were higher in children with recent infection than in those with an older history of disease (p < 0.0001 for all); similarly, the total and natural Tregs compartments were more represented in children at onset when compared with Long COVID (p < 0.0001 and p = 0.0005, respectively). Despite the heterogeneity, partially due to age, sex and infection incidence, the susceptibility of certain children to develop persistent symptoms after infection appeared to be associated with the imbalance of the adaptive immune response. Following up and comparing recovered versus Long COVID patients, we analyzed the role of circulating naïve and switched B and regulatory T lymphocytes in counteracting the evolution of the symptomatology emerged, finding an interesting correlation between the amount and ability to reconstitute the natural Tregs component with the persistence of symptoms (linear regression, p = 0.0026). Conclusions. In this study, we suggest that children affected by Long COVID may have a compromised ability to switch from the innate to the adaptive immune response, as supported by our data showing a contraction of naïve and switched B cell compartment and an unstable balance of regulatory T lymphocytes occurring in these children. However, further prospective immunological studies are needed to better clarify which factors (epigenetic, diet, environment, etc.) are involved in the impairment of the immunological mechanisms in the Long COVID patients.
Collapse
|
21
|
Rowntree LC, Nguyen THO, Kedzierski L, Neeland MR, Petersen J, Crawford JC, Allen LF, Clemens EB, Chua B, McQuilten HA, Minervina AA, Pogorelyy MV, Chaurasia P, Tan HX, Wheatley AK, Jia X, Amanat F, Krammer F, Allen EK, Sonda S, Flanagan KL, Jumarang J, Pannaraj PS, Licciardi PV, Kent SJ, Bond KA, Williamson DA, Rossjohn J, Thomas PG, Tosif S, Crawford NW, van de Sandt CE, Kedzierska K. SARS-CoV-2-specific T cell memory with common TCRαβ motifs is established in unvaccinated children who seroconvert after infection. Immunity 2022; 55:1299-1315.e4. [PMID: 35750048 PMCID: PMC9174177 DOI: 10.1016/j.immuni.2022.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/31/2022] [Accepted: 06/01/2022] [Indexed: 11/05/2022]
Abstract
As the establishment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cell memory in children remains largely unexplored, we recruited convalescent COVID-19 children and adults to define their circulating memory SARS-CoV-2-specific CD4+ and CD8+ T cells prior to vaccination. We analyzed epitope-specific T cells directly ex vivo using seven HLA class I and class II tetramers presenting SARS-CoV-2 epitopes, together with Spike-specific B cells. Unvaccinated children who seroconverted had comparable Spike-specific but lower ORF1a- and N-specific memory T cell responses compared with adults. This agreed with our TCR sequencing data showing reduced clonal expansion in children. A strong stem cell memory phenotype and common T cell receptor motifs were detected within tetramer-specific T cells in seroconverted children. Conversely, children who did not seroconvert had tetramer-specific T cells of predominantly naive phenotypes and diverse TCRαβ repertoires. Our study demonstrates the generation of SARS-CoV-2-specific T cell memory with common TCRαβ motifs in unvaccinated seroconverted children after their first virus encounter.
Collapse
Affiliation(s)
- Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Melanie R Neeland
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC 3000, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Jan Petersen
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - E Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Brendon Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Anastasia A Minervina
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mikhail V Pogorelyy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Priyanka Chaurasia
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Fatima Amanat
- Department of Microbiology, 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
| | - Florian Krammer
- Department of Microbiology, 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; Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sabrina Sonda
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS 7248, Australia
| | - Katie L Flanagan
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS 7248, Australia; Department of Immunology and Pathology, Monash University, Commercial Road, Melbourne, VIC 3004, Australia; School of Health and Biomedical Science, RMIT University, Melbourne, VIC 3000, Australia; Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Jaycee Jumarang
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Pia S Pannaraj
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Departments of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC 3000, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC 3000, Australia; Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Katherine A Bond
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Microbiology, Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Deborah A Williamson
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3000, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shidan Tosif
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC 3000, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia; Department of General Medicine, Royal Children's Hospital Melbourne, Melbourne, VIC 3000, Australia
| | - Nigel W Crawford
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC 3000, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia; Department of General Medicine, Royal Children's Hospital Melbourne, Melbourne, VIC 3000, Australia; Royal Children's Hospital Melbourne, Immunisation Service, Melbourne, VIC 3000, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| |
Collapse
|
22
|
Yu S, Zhang P, Liao M, Zhang J, Luo S, Zhai J, Zhang Y, Lin J, Yuan J, Zhang Z, Wang F, Wei L. Detection of SARS-CoV-2 Specific Antibodies in Saliva Samples. Front Immunol 2022; 13:880154. [PMID: 35898491 PMCID: PMC9309249 DOI: 10.3389/fimmu.2022.880154] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
Molecular assays on nasopharyngeal swabs act as a confirmatory test in coronavirus disease (COVID-19) diagnosis. However, the technical requirements of nasopharyngeal sampling and molecular assays limit the testing capabilities. Recent studies suggest the use of saliva for the COVID-19 diagnostic test. In this study, 44 patients diagnosed with COVID-19 in The Third People’s Hospital of Shenzhen were enrolled. Saliva and serum specimens were obtained at different time points and the immunoglobulins against SARS-CoV-2 were measured. The results showed that saliva IgA presented a higher COI value than IgG and IgM. In matched saliva and serum samples, all saliva samples presented lower IgG levels than serum samples, and only one saliva sample presented a higher IgM level. The conversion rates of saliva IgA and the detection of viral nucleic acids were analyzed in the first and second weeks after hospitalization. The positive rates increased when combining saliva IgA and viral nucleic acid detection. In conclusion, our results provide evidence that saliva IgA could serve as a useful index for the early diagnosis of COVID-19.
Collapse
Affiliation(s)
- Siyang Yu
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peiyan Zhang
- Department of Infectious Diseases, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Mingfeng Liao
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Juanjuan Zhang
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Suisui Luo
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jinglei Zhai
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yaxi Zhang
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Jingyan Lin
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Jing Yuan
- Department of Infectious Diseases, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Zheng Zhang
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, China
- *Correspondence: Lanlan Wei, ; Fuxiang Wang, ; Zheng Zhang,
| | - Fuxiang Wang
- Department of Infectious Diseases, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Lanlan Wei, ; Fuxiang Wang, ; Zheng Zhang,
| | - Lanlan Wei
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, The Third People’s Hospital of Shenzhen, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Lanlan Wei, ; Fuxiang Wang, ; Zheng Zhang,
| |
Collapse
|
23
|
Tosif S, Haycroft ER, Sarkar S, Toh ZQ, Do LAH, Donato CM, Selva KJ, Hoq M, Overmars I, Nguyen J, Lee L, Clifford V, Daley A, Mordant FL, McVernon J, Mulholland K, Marcato AJ, Smith MZ, Curtis N, McNab S, Saffery R, Kedzierska K, Subarrao K, Burgner D, Steer A, Bines JE, Sutton P, Licciardi PV, Chung AW, Neeland MR, Crawford NW. Virology and immune dynamics reveal high household transmission of ancestral SARS-CoV-2 strain. Pediatr Allergy Immunol 2022; 33:10.1111/pai.13824. [PMID: 35871459 PMCID: PMC9349415 DOI: 10.1111/pai.13824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Household studies are crucial for understanding the transmission of SARS-CoV-2 infection, which may be underestimated from PCR testing of respiratory samples alone. We aim to combine the assessment of household mitigation measures; nasopharyngeal, saliva, and stool PCR testing; along with mucosal and systemic SARS-CoV-2-specific antibodies, to comprehensively characterize SARS-CoV-2 infection and transmission in households. METHODS Between March and September 2020, we obtained samples from 92 participants in 26 households in Melbourne, Australia, in a 4-week period following the onset of infection with ancestral SARS-CoV-2 variants. RESULTS The secondary attack rate was 36% (24/66) when using nasopharyngeal swab (NPS) PCR positivity alone. However, when respiratory and nonrespiratory samples were combined with antibody responses in blood and saliva, the secondary attack rate was 76% (50/66). SARS-CoV-2 viral load of the index case and household isolation measures were key factors that determine secondary transmission. In 27% (7/26) of households, all family members tested positive by NPS for SARS-CoV-2 and were characterized by lower respiratory Ct values than low transmission families (Median 22.62 vs. 32.91; IQR 17.06-28.67 vs. 30.37-34.24). High transmission families were associated with enhanced plasma antibody responses to multiple SARS-CoV-2 antigens and the presence of neutralizing antibodies. Three distinguishing saliva SARS-CoV-2 antibody features were identified according to age (IgA1 to Spike 1, IgA1 to nucleocapsid protein (NP)), suggesting that adults and children generate distinct mucosal antibody responses during the acute phase of infection. CONCLUSION Utilizing respiratory and nonrespiratory PCR testing, along with the measurement of SARS-CoV-2-specific local and systemic antibodies, provides a more accurate assessment of infection within households and highlights some of the immunological differences in response between children and adults.
Collapse
|
24
|
Sieber J, Mayer M, Schmidthaler K, Kopanja S, Camp JV, Popovitsch A, Dwivedi V, Hoz J, Schoof A, Weseslindtner L, Szépfalusi Z, Stiasny K, Aberle JH. Long-Lived Immunity in SARS-CoV-2-Recovered Children and Its Neutralizing Capacity Against Omicron. Front Immunol 2022; 13:882456. [PMID: 35663948 PMCID: PMC9157051 DOI: 10.3389/fimmu.2022.882456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 12/26/2022] Open
Abstract
SARS-CoV-2 infection is effectively controlled by humoral and cellular immune responses. However, the durability of immunity in children as well as the ability to neutralize variants of concern are unclear. Here, we assessed T cell and antibody responses in a longitudinal cohort of children after asymptomatic or mild COVID-19 over a 12-month period. Antigen-specific CD4 T cells remained stable over time, while CD8 T cells declined. SARS-CoV-2 infection induced long-lived neutralizing antibodies against ancestral SARS-CoV-2 (D614G isolate), but with poor cross-neutralization of omicron. Importantly, recall responses to vaccination in children with pre-existing immunity yielded neutralizing antibody activities against D614G and omicron BA.1 and BA.2 variants that were 3.9-fold, 9.9-fold and 14-fold higher than primary vaccine responses in seronegative children. Together, our findings demonstrate that SARS-CoV-2 infection in children induces robust memory T cells and antibodies that persist for more than 12 months, but lack neutralizing activity against omicron. Vaccination of pre-immune children, however, substantially improves the omicron-neutralizing capacity.
Collapse
Affiliation(s)
- Justyna Sieber
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
| | - Margareta Mayer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Klara Schmidthaler
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Sonja Kopanja
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Jeremy V. Camp
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Varsha Dwivedi
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Jakub Hoz
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Anja Schoof
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | - Zsolt Szépfalusi
- Division of Pediatric Pulmonology, Allergy and Endocrinology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Lazova S, Dimitrova Y, Hristova D, Tzotcheva I, Velikova T. Cellular, Antibody and Cytokine Pathways in Children with Acute SARS-CoV-2 Infection and MIS-C-Can We Match the Puzzle? Antibodies (Basel) 2022; 11:25. [PMID: 35466278 PMCID: PMC9036295 DOI: 10.3390/antib11020025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
The newly identified strain of the Coronaviridae family called severe acute respiratory syndrome (SARS-CoV-2) recently became the most significant health threat for adults and children. Some main predictors of severe clinical course in patients with SARS-CoV-2 infection are age and concomitant health conditions. Therefore, the proper evaluation of SARS-CoV-2-specific immunity is urgently required to understand and predict the spectrum of possible clinical phenotypes and recommend vaccination options and regimens in children. Furthermore, it is critical to characterize the nature of SARS-CoV-2-specific immune responses in children following asymptomatic infection and COVID-19 and other related conditions such as multisystem inflammatory syndrome (MIS-C), para-infectious and late postinfectious consequences. Recent studies involving children revealed a variety of cytokines, T cells and antibody responses in the pathogenesis of the disease. Moreover, different clinical scenarios in children were observed-asymptomatic seroprevalence, acute SARS-CoV-2 infection, and rarely severe COVID-19 with typical cytokine storm, MIS-C, long COVID-19, etc. Therefore, to gain a better clinical view, adequate diagnostic criteria and treatment algorithms, it is essential to create a realistic picture of the immunological puzzle of SARS-CoV-2 infection in different age groups. Finally, it was demonstrated that children may exert a potent and prolonged adaptive anti-SARS-CoV-2 immune response, with significant cross-reactions against other human Corona Viruses, that might contribute to disease sparing effect in this age range. However, the immunopathology of the virus has to be elucidated first.
Collapse
Affiliation(s)
- Snezhina Lazova
- Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben”, Blvd., 1463 Sofia, Bulgaria; (Y.D.); (I.T.)
- Health Care Department, Faculty of Public Health, Medical University Sofia, Bialo More, 8 Str., 1527 Sofia, Bulgaria
| | - Yulia Dimitrova
- Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben”, Blvd., 1463 Sofia, Bulgaria; (Y.D.); (I.T.)
| | - Diana Hristova
- Department of Immunology, National Center of Infectious and Parasitic Diseases, 1504 Sofia, Bulgaria;
| | - Iren Tzotcheva
- Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben”, Blvd., 1463 Sofia, Bulgaria; (Y.D.); (I.T.)
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia University St. Kliment Ohridski, Kozyak 1 Str., 1407 Sofia, Bulgaria;
| |
Collapse
|
26
|
Fernández-Rojas MA, Luna-Ruiz Esparza MA, Campos-Romero A, Calva-Espinosa DY, Moreno-Camacho JL, Mendlovic F, Plett-Torres T, Alcántar-Fernández J. Seroconversion dynamic and SARS-CoV-2 seropositivity in unvaccinated population during the first and second outbreaks in Mexico. Sci Rep 2022; 12:5241. [PMID: 35347208 PMCID: PMC8960100 DOI: 10.1038/s41598-022-09395-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/22/2022] [Indexed: 12/23/2022] Open
Abstract
Serosurveillance helps establish reopening guidelines and determine the immunity levels in different populations to reach herd immunity. Then, there is an urgent need to estimate seroprevalence population wide. In Mexico, information about COVID-19 cases and related deaths is scarce. Also, there is no official serosurveillance, limiting our knowledge of the impact of the SARS-CoV-2 pandemic. Here, we report the prevalence of anti-SARS-CoV-2 antibodies in 522,690 unvaccinated people from July 5th to December 31st, 2020. The overall seroprevalence was 32.8% and highest in adults aged 30–39 years (38.5%) than people under 20 years (33.0%) or older (28.9%). Moreover, in a cohort of 1655 individuals confirmed COVID-19 by PCR, we found that symptomatic people (HR = 2.56) increased seroconversion than presymptomatic. Also, we identified that the most discriminative symptoms for COVID-19 that could predict seroconversion were anosmia and ageusia (HR = 1.70), fever, myalgia/arthralgia, and cough (HR = 1.75). Finally, we found that obese people had lower seroconversion (HR = 0.53) than healthy people, but the opposite happens in diabetic people (HR = 1.39). These findings reveal that around one-third of Mexican outpatients had anti-SARS-CoV-2 antibodies before vaccination. Also, some symptoms improve empirically COVID-19 diagnosis and seroconversion. This information could help fine-tune vaccination schemes and the reopening and back-to-work algorithms.
Collapse
Affiliation(s)
- Miguel A Fernández-Rojas
- Innovation and Research Department, Salud Digna A.C., Francisco Villa 113 sur, 80000, Culiacán, Sinaloa, Mexico
| | - Marco A Luna-Ruiz Esparza
- Innovation and Research Department, Salud Digna A.C., Francisco Villa 113 sur, 80000, Culiacán, Sinaloa, Mexico
| | - Abraham Campos-Romero
- Innovation and Research Department, Salud Digna A.C., Francisco Villa 113 sur, 80000, Culiacán, Sinaloa, Mexico
| | | | - José L Moreno-Camacho
- Clinical Laboratory Department, Salud Digna, 80000, Culiacán, Sinaloa, Mexico.,National Reference Center "Culiacan", Salud Digna, 80300, Culiacán, Sinaloa, Mexico
| | - Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Huixquilucan, Estado de Mexico, Mexico
| | - Tanya Plett-Torres
- Plan de Estudios Combinados en Medicina, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jonathan Alcántar-Fernández
- Innovation and Research Department, Salud Digna A.C., Francisco Villa 113 sur, 80000, Culiacán, Sinaloa, Mexico.
| |
Collapse
|
27
|
Castro Dopico X, Muschiol S, Grinberg NF, Aleman S, Sheward DJ, Hanke L, Ahl M, Vikström L, Forsell M, Coquet JM, McInerney G, Dillner J, Bogdanovic G, Murrell B, Albert J, Wallace C, Karlsson Hedestam GB. Probabilistic classification of anti-SARS-CoV-2 antibody responses improves seroprevalence estimates. Clin Transl Immunology 2022; 11:e1379. [PMID: 35284072 PMCID: PMC8891432 DOI: 10.1002/cti2.1379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/04/2022] [Accepted: 02/17/2022] [Indexed: 02/02/2023] Open
Abstract
Objectives Population-level measures of seropositivity are critical for understanding the epidemiology of an emerging pathogen, yet most antibody tests apply a strict cutoff for seropositivity that is not learnt in a data-driven manner, leading to uncertainty when classifying low-titer responses. To improve upon this, we evaluated cutoff-independent methods for their ability to assign likelihood of SARS-CoV-2 seropositivity to individual samples. Methods Using robust ELISAs based on SARS-CoV-2 spike (S) and the receptor-binding domain (RBD), we profiled antibody responses in a group of SARS-CoV-2 PCR+ individuals (n = 138). Using these data, we trained probabilistic learners to assign likelihood of seropositivity to test samples of unknown serostatus (n = 5100), identifying a support vector machines-linear discriminant analysis learner (SVM-LDA) suited for this purpose. Results In the training data from confirmed ancestral SARS-CoV-2 infections, 99% of participants had detectable anti-S and -RBD IgG in the circulation, with titers differing > 1000-fold between persons. In data of otherwise healthy individuals, 7.2% (n = 367) of samples were of uncertain serostatus, with values in the range of 3-6SD from the mean of pre-pandemic negative controls (n = 595). In contrast, SVM-LDA classified 6.4% (n = 328) of test samples as having a high likelihood (> 99% chance) of past infection, 4.5% (n = 230) to have a 50-99% likelihood, and 4.0% (n = 203) to have a 10-49% likelihood. As different probabilistic approaches were more consistent with each other than conventional SD-based methods, such tools allow for more statistically-sound seropositivity estimates in large cohorts. Conclusion Probabilistic antibody testing frameworks can improve seropositivity estimates in populations with large titer variability.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Sandra Muschiol
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden,Department of Clinical MicrobiologyKarolinska University HospitalStockholmSweden
| | - Nastasiya F Grinberg
- Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Soo Aleman
- Department of Infectious DiseasesKarolinska University HospitalHuddingeSweden
| | - Daniel J Sheward
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Marcus Ahl
- Department of Infectious DiseasesKarolinska University HospitalHuddingeSweden
| | - Linnea Vikström
- Department of Clinical MicrobiologyUmeå UniversitetUmeåSweden
| | - Mattias Forsell
- Department of Clinical MicrobiologyUmeå UniversitetUmeåSweden
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Gerald McInerney
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Joakim Dillner
- Division of PathologyDepartment of Laboratory MedicineKarolinska InstitutetHuddingeSweden
| | - Gordana Bogdanovic
- Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden,Department of Clinical MicrobiologyKarolinska University HospitalStockholmSweden
| | - Chris Wallace
- Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK,Medical Research Council Biostatistics UnitUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
28
|
Dethioux L, Dauby N, Montesinos I, Rebuffat E, Hainaut M. SARS-CoV-2 seroprevalence in children and their family members, July-October 2020, Brussels. Eur J Pediatr 2022; 181:1009-1016. [PMID: 34677664 PMCID: PMC8532097 DOI: 10.1007/s00431-021-04284-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/24/2021] [Accepted: 10/03/2021] [Indexed: 11/15/2022]
Abstract
The aim of this study was to estimate the seroprevalence of SARS-CoV-2 antibodies in a pediatric population after the first pandemic wave in Belgium. All patients requiring a blood sample between 1 July 2020 and 31 October 2020 in our institution were invited to participate. Their parents and siblings could also participate to estimate familial transmission and the congruence between serological statuses. A questionnaire was completed for each participant to identify symptoms consistent with COVID-19 in the previous months. Blood samples were tested for SARS-CoV-2-specific immunoglobulin G using ELISA. The final population included 112 children, 24 siblings of these children, and 36 adults. The seroprevalence of cases was 6.9% before 8 September, a date that corresponds to 1 week after the beginning of the second wave in Belgium and 22.5% afterwards (OR = 3.89, 95% CI (1.20; 12.58), p-value = 0.03). Twenty-five percent of children were asymptomatic, and none experienced severe disease. The symptoms associated with SARS-CoV-2-positive antibodies were diarrhoea (OR = 9.9, 95% CI [2.88; 33.87.65] p-value < 0.01), fever (OR = 3.8, 95% CI [1.44; 10.22] p-value < 0.01), rhinitis (OR = 3.9, 95% CI [1.38; 10.90] p-value = 0.01), or anosmia (OR = 31.5, 95% CI [1.45; 682.7], p-value = 0.02). A child was the first symptomatic household member in 50% of the familial clusters.Conclusion: Seroprevalence in children was comparable to that of the general population. Children could represent the source of infection in the household. What is Known: • COVID-19 infection is generally mild or asymptomatic in children and adolescents. • Belgian strategy of testing was focused on symptoms. • Adults are believed to be responsible for most of familial clusters. What is New: • Serological testing gives a more accurate view of the rate of infected children. • Based on serological results, children have been infected as frequently as adults during the first and second wave in Belgium. • Seventy-five percent of SARS-CoV-2 IgG-positive children presented a mild symptomatology, and 25% were totally asymptomatic. • Children could represent the source of infection within household.
Collapse
Affiliation(s)
- Lorraine Dethioux
- Pediatrics Department, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nicolas Dauby
- Infectious Diseases Department, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
- School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Isabel Montesinos
- Microbiology Department, Laboratoire Hospitalier Universitaire de Bruxelles (LHUB-ULB), Brussels, Belgium
| | - Elisabeth Rebuffat
- Pediatrics Department, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marc Hainaut
- Pediatrics Department, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
29
|
Toh ZQ, Anderson J, Mazarakis N, Neeland M, Higgins RA, Rautenbacher K, Dohle K, Nguyen J, Overmars I, Donato C, Sarkar S, Clifford V, Daley A, Nicholson S, Mordant FL, Subbarao K, Burgner DP, Curtis N, Bines JE, McNab S, Steer AC, Mulholland K, Tosif S, Crawford NW, Pellicci DG, Do LAH, Licciardi PV. Comparison of Seroconversion in Children and Adults With Mild COVID-19. JAMA Netw Open 2022; 5:e221313. [PMID: 35262717 PMCID: PMC8908077 DOI: 10.1001/jamanetworkopen.2022.1313] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE The immune response in children with SARS-CoV-2 infection is not well understood. OBJECTIVE To compare seroconversion in nonhospitalized children and adults with mild SARS-CoV-2 infection and identify factors that are associated with seroconversion. DESIGN, SETTING, AND PARTICIPANTS This household cohort study of SARS-CoV-2 infection collected weekly nasopharyngeal and throat swabs and blood samples during the acute (median, 7 days for children and 12 days for adults [IQR, 4-13] days) and convalescent (median, 41 [IQR, 31-49] days) periods after polymerase chain reaction (PCR) diagnosis for analysis. Participants were recruited at The Royal Children's Hospital, Melbourne, Australia, from May 10 to October 28, 2020. Participants included patients who had a SARS-CoV-2-positive nasopharyngeal or oropharyngeal swab specimen using PCR analysis. MAIN OUTCOMES AND MEASURES SARS-CoV-2 immunoglobulin G (IgG) and cellular (T cell and B cell) responses in children and adults. Seroconversion was defined by seropositivity in all 3 (an in-house enzyme-linked immunosorbent assay [ELISA] and 2 commercial assays: a SARS-CoV-2 S1/S2 IgG assay and a SARS-CoV-2 antibody ELISA) serological assays. RESULTS Among 108 participants with SARS-CoV-2-positive PCR findings, 57 were children (35 boys [61.4%]; median age, 4 [IQR, 2-10] years) and 51 were adults (28 women [54.9%]; median age, 37 [IQR, 34-45] years). Using the 3 established serological assays, a lower proportion of children had seroconversion to IgG compared with adults (20 of 54 [37.0%] vs 32 of 42 [76.2%]; P < .001). This result was not associated with viral load, which was similar in children and adults (mean [SD] cycle threshold [Ct] value, 28.58 [6.83] vs 24.14 [8.47]; P = .09). In addition, age and sex were not associated with seroconversion within children (median age, 4 [IQR, 2-14] years for both seropositive and seronegative groups; seroconversion by sex, 10 of 21 girls [47.6%] vs 10 of 33 boys [30.3%]) or adults (median ages, 37 years for seropositive and 40 years for seronegative adults [IQR, 34-39 years]; seroconversion by sex, 18 of 24 women [75.0%] vs 14 of 18 men [77.8%]) (P > .05 for all comparisons between seronegative and seropositive groups). Symptomatic adults had 3-fold higher SARS-CoV-2 IgG levels than asymptomatic adults (median, 227.5 [IQR, 133.7-521.6] vs 75.3 [IQR, 36.9-113.6] IU/mL), whereas no differences were observed in children regardless of symptoms. Moreover, differences in cellular immune responses were observed in adults compared with children with seroconversion. CONCLUSIONS AND RELEVANCE The findings of this cohort study suggest that among patients with mild COVID-19, children may be less likely to have seroconversion than adults despite similar viral loads. This finding has implications for future protection after SARS-CoV-2 infection in children and for interpretation of serosurveys that involve children. Further research to understand why seroconversion and development of symptoms are potentially less likely in children after SARS-CoV-2 infection and to compare vaccine responses may be of clinical and scientific importance.
Collapse
Affiliation(s)
- Zheng Quan Toh
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Jeremy Anderson
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Nadia Mazarakis
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Melanie Neeland
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Rachel A. Higgins
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Karin Rautenbacher
- Laboratory Services, The Royal Children’s Hospital, Melbourne, Australia
| | - Kate Dohle
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Jill Nguyen
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Isabella Overmars
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Celeste Donato
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Sohinee Sarkar
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Vanessa Clifford
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Andrew Daley
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Francesca L. Mordant
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO (World Health Organization) Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David P. Burgner
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Nigel Curtis
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Julie E. Bines
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of Gastroenterology, The Royal Children’s Hospital, Melbourne, Australia
| | - Sarah McNab
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Andrew C. Steer
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Kim Mulholland
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shidan Tosif
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Nigel W. Crawford
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of General Medicine, The Royal Children’s Hospital, Melbourne, Australia
| | - Daniel G. Pellicci
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lien Anh Ha Do
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Paul V. Licciardi
- Division of Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
30
|
Dobaño C, Alonso S, Vidal M, Jiménez A, Rubio R, Santano R, Barrios D, Pons Tomas G, Melé Casas M, Hernández García M, Girona-Alarcón M, Puyol L, Baro B, Millat-Martínez P, Ajanovic S, Balanza N, Arias S, Rodrigo Melero N, Carolis C, García-Miquel A, Bonet-Carné E, Claverol J, Cubells M, Fortuny C, Fumadó V, Codina A, Bassat Q, Muñoz-Almagro C, Fernández de Sevilla M, Gratacós E, Izquierdo L, García-García JJ, Aguilar R, Jordan I, Moncunill G. Multiplex Antibody Analysis of IgM, IgA and IgG to SARS-CoV-2 in Saliva and Serum From Infected Children and Their Close Contacts. Front Immunol 2022; 13:751705. [PMID: 35154094 PMCID: PMC8828491 DOI: 10.3389/fimmu.2022.751705] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19 affects children to a lesser extent than adults but they can still get infected and transmit SARS-CoV-2 to their contacts. Field deployable non-invasive sensitive diagnostic techniques are needed to evaluate the infectivity dynamics of SARS-CoV-2 in pediatric populations and guide public health interventions, particularly if this population is not fully vaccinated. We evaluated the utility of high-throughput Luminex assays to quantify saliva IgM, IgA and IgG antibodies against five SARS-CoV-2 spike (S) and nucleocapsid (N) antigens in a contacts and infectivity longitudinal study in 122 individuals (52 children and 70 adults). We compared saliva versus serum/plasma samples in infected children and adults diagnosed by weekly RT-PCR over 35 days (n=62), and those who consistently tested negative over the same follow up period (n=60), in the Summer of 2020 in Barcelona, Spain. Saliva antibody levels in SARS-CoV-2 RT-PCR positive individuals were significantly higher than in negative individuals and correlated with those measured in sera/plasmas. Asymptomatic infected individuals had higher levels of anti-S IgG than symptomatic individuals, suggesting a protective anti-disease role for antibodies. Higher anti-S IgG and IgM levels in serum/plasma and saliva, respectively, in infected children compared to infected adults could also be related to stronger clinical immunity in them. Among infected children, males had higher levels of saliva IgG to N and RBD than females. Despite overall correlation, individual clustering analysis suggested that responses that may not be detected in blood could be patent in saliva, and vice versa. In conclusion, measurement of SARS-CoV-2-specific saliva antibodies should be considered as a complementary non-invasive assay to serum/plasma to determine COVID-19 prevalence and transmission in pediatric populations before and after vaccination campaigns.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, Madrid, Spain
| | - Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Rocío Rubio
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Rebeca Santano
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Gemma Pons Tomas
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain
| | - María Melé Casas
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain
| | - María Hernández García
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain
| | - Mònica Girona-Alarcón
- Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Laura Puyol
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Barbara Baro
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | | | - Sara Ajanovic
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Núria Balanza
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Sara Arias
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Aleix García-Miquel
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Elisenda Bonet-Carné
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Universitat Politècnica de Catalunya, BarcelonaTech, Barcelona, Spain
| | - Joana Claverol
- Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Fundació Sant Joan de Déu, Barcelona, Spain
| | - Marta Cubells
- Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Fundació Sant Joan de Déu, Barcelona, Spain
| | - Claudia Fortuny
- Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Infectious Diseases Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Victoria Fumadó
- Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Infectious Diseases Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Anna Codina
- Biobank Hospital Sant Joan de Déu, Barcelona, Spain
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carmen Muñoz-Almagro
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Department of Medicine, Universitat Internacional de Catalunya, Barcelona, Spain.,Molecular Microbiology Department, Hospital Sant Joan de Déu, Esplugues, Spain
| | - Mariona Fernández de Sevilla
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Spain
| | - Eduard Gratacós
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, Madrid, Spain
| | - Juan José García-García
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Iolanda Jordan
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Spain.,Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Consorcio de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, Madrid, Spain
| |
Collapse
|
31
|
Kaaijk P, Olivo Pimentel V, Emmelot ME, Poelen MCM, Cevirgel A, Schepp RM, den Hartog G, Reukers DF, Beckers L, van Beek J, van Els CACM, Meijer A, Rots NY, de Wit J. Children and Adults With Mild COVID-19: Dynamics of the Memory T Cell Response up to 10 Months. Front Immunol 2022; 13:817876. [PMID: 35197982 PMCID: PMC8858984 DOI: 10.3389/fimmu.2022.817876] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
BackgroundSevere acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has led to considerable morbidity/mortality worldwide, but most infections, especially among children, have a mild course. However, it remains largely unknown whether infected children develop cellular immune memory.MethodsTo determine whether a memory T cell response is being developed, we performed a longitudinal assessment of the SARS-CoV-2-specific T cell response by IFN-γ ELISPOT and activation marker analyses of peripheral blood samples from unvaccinated children and adults with mild-to-moderate COVID-19.ResultsUpon stimulation of PBMCs with heat-inactivated SARS-CoV-2 or overlapping peptides of spike (S-SARS-CoV-2) and nucleocapsid proteins, we found S-SARS-CoV-2-specific IFN-γ T cell responses in infected children (83%) and adults (100%) that were absent in unexposed controls. Frequencies of SARS-CoV-2-specific T cells were higher in infected adults, especially in those with moderate symptoms, compared to infected children. The S-SARS-CoV-2 IFN-γ T cell response correlated with S1-SARS-CoV-2-specific serum antibody concentrations. Predominantly, effector memory CD4+ T cells of a Th1 phenotype were activated upon exposure to SARS-CoV-2 antigens. Frequencies of SARS-CoV-2-specific T cells were significantly reduced at 10 months after symptom onset, while S1-SARS-CoV-2-specific IgG concentrations were still detectable in 90% of all children and adults.ConclusionsOur data indicate that an antigen-specific T cell and antibody response is developed after mild SARS-CoV-2 infection in children and adults. It remains to be elucidated to what extent this SARS-CoV-2-specific response can contribute to an effective recall response after reinfection.
Collapse
Affiliation(s)
- Patricia Kaaijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- *Correspondence: Patricia Kaaijk,
| | - Verónica Olivo Pimentel
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Maarten E. Emmelot
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Martien C. M. Poelen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alper Cevirgel
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Rutger M. Schepp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Gerco den Hartog
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Daphne F.M. Reukers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Beckers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Adam Meijer
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Nynke Y. Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
32
|
Zimmermann P, Curtis N. Why Does the Severity of COVID-19 Differ With Age?: Understanding the Mechanisms Underlying the Age Gradient in Outcome Following SARS-CoV-2 Infection. Pediatr Infect Dis J 2022; 41:e36-e45. [PMID: 34966142 PMCID: PMC8740029 DOI: 10.1097/inf.0000000000003413] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 11/26/2022]
Abstract
Although there are many hypotheses for the age-related difference in the severity of COVID-19, differences in innate, adaptive and heterologous immunity, together with differences in endothelial and clotting function, are the most likely mechanisms underlying the marked age gradient. Children have a faster and stronger innate immune response to SARS-CoV-2, especially in the nasal mucosa, which rapidly controls the virus. In contrast, adults can have an overactive, dysregulated and less effective innate response that leads to uncontrolled pro-inflammatory cytokine production and tissue injury. More recent exposure to other viruses and routine vaccines in children might be associated with protective cross-reactive antibodies and T cells against SARS-CoV-2. There is less evidence to support other mechanisms that have been proposed to explain the age-related difference in outcome following SARS-CoV-2 infection, including pre-existing immunity from exposure to common circulating coronaviruses, differences in the distribution and expression of the entry receptors ACE2 and TMPRSS2, and difference in viral load.
Collapse
Affiliation(s)
- Petra Zimmermann
- From the Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital Melbourne, Parkville, Australia
| |
Collapse
|
33
|
Drozdzik A, Drozdzik M. Oral Pathology in COVID-19 and SARS-CoV-2 Infection-Molecular Aspects. Int J Mol Sci 2022; 23:1431. [PMID: 35163355 PMCID: PMC8836070 DOI: 10.3390/ijms23031431] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
This review article was designed to evaluate the existing evidence related to the molecular processes of SARS-CoV-2 infection in the oral cavity. The World Health Organization stated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission is produced by respiratory droplets and aerosols from the oral cavity of infected patients. The oral cavity structures, keratinized and non-keratinized mucosa, and salivary glands' epithelia express SARS-CoV-2 entry and transmission factors, especially angiotensin converting enzyme Type 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). Replication of the virus in cells leads to local and systemic infection spread, and cellular damage is associated with clinical signs and symptoms of the disease in the oral cavity. Saliva, both the cellular and acellular fractions, holds the virus particles and contributes to COVID-19 transmission. The review also presents information about the factors modifying SARS-CoV-2 infection potential and possible local pharmacotherapeutic interventions, which may confine SARS-CoV-2 virus entry and transmission in the oral cavity. The PubMed and Scopus databases were used to search for suitable keywords such as: SARS-CoV-2, COVID-19, oral virus infection, saliva, crevicular fluid, salivary gland, tongue, oral mucosa, periodontium, gingiva, dental pulp, ACE2, TMPRSS2, Furin, diagnosis, topical treatment, vaccine and related words in relevant publications up to 28 December 2021. Data extraction and quality evaluation of the articles were performed by two reviewers, and 63 articles were included in the final review.
Collapse
Affiliation(s)
- Agnieszka Drozdzik
- Department of Integrated Dentistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp 72, 70-111 Szczecin, Poland;
| | - Marek Drozdzik
- Department of Pharmacology, Pomeranian Medical University in Szczecin, Powstancow Wlkp 72, 70-111 Szczecin, Poland
| |
Collapse
|
34
|
Lee J, Cho K, Kook H, Kang S, Lee Y, Lee J. The Different Immune Responses by Age Are due to the Ability of the Fetal Immune System to Secrete Primal Immunoglobulins Responding to Unexperienced Antigens. Int J Biol Sci 2022; 18:617-636. [PMID: 35002513 PMCID: PMC8741860 DOI: 10.7150/ijbs.67203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
Among numerous studies on coronavirus 2019 (COVID-19), we noted that the infection and mortality rates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) increased with age and that fetuses known to be particularly susceptible to infection were better protected despite various mutations. Hence, we established the hypothesis that a new immune system exists that forms before birth and decreases with aging. Methods: To prove this hypothesis, we established new ex-vivo culture conditions simulating the critical environmental factors of fetal stem cells (FSCs) in early pregnancy. Then, we analyzed the components from FSCs cultivated newly developed ex-vivo culture conditions and compared them from FSCs cultured in a normal condition. Results: We demonstrated that immunoglobulin M (IgM), a natural antibody (NAb) produced only in early B-1 cells, immunoglobulins (Igs) including IgG3, which has a wide range of antigen-binding capacity and affinity, complement proteins, and antiviral proteins are induced in FSCs only cultured in newly developed ex-vivo culture conditions. Particularly we confirmed that their extracellular vesicles (EVs) contained NAbs, Igs, various complement proteins, and antiviral proteins, as well as human leukocyte antigen G (HLA-G), responsible for immune tolerance. Conclusion: Our results suggest that FSCs in early pregnancy can form an independent immune system responding to unlearned antigens as a self-defense mechanism before establishing mature immune systems. Moreover, we propose the possibility of new solutions to cope with various infectious diseases based on the factors in NAbs-containing EVs, especially not causing unnecessary immune reaction due to HLA-G.
Collapse
Affiliation(s)
- Jangho Lee
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| | - Kyoungshik Cho
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| | - Hyejin Kook
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| | - Suman Kang
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| | - Yunsung Lee
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| | - Jiwon Lee
- R&D Center of Stemmedicare Ltd, Seoul, 06095, Republic of Korea
| |
Collapse
|
35
|
Abstract
Adaptive immune responses play critical roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional is the rapid characterization of the immune response to the virus performed by researchers during the first 20 months of the pandemic. This has given us a more detailed understanding of SARS-CoV-2 compared to many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
Collapse
Affiliation(s)
- Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
36
|
Howard‐Jones AR, Burgner DP, Crawford NW, Goeman E, Gray PE, Hsu P, Kuek S, McMullan BJ, Tosif S, Wurzel D, Bowen AC, Danchin M, Koirala A, Sharma K, Yeoh DK, Britton PN. COVID-19 in children. II: Pathogenesis, disease spectrum and management. J Paediatr Child Health 2022; 58:46-53. [PMID: 34694037 PMCID: PMC8662268 DOI: 10.1111/jpc.15811] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022]
Abstract
The global disruption of the COVID-19 pandemic has impacted the life of every child either directly or indirectly. This review explores the pathophysiology, immune response, clinical presentation and treatment of COVID-19 in children, summarising the most up-to-date data including recent developments regarding variants of concern. The acute infection with SARS-CoV-2 is generally mild in children, whilst the post-infectious manifestations, including paediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2 (PIMS-TS) and 'long COVID' in children, are more complex. Given that most research on COVID-19 has focused on adult cohorts and that clinical manifestations, treatment availability and impacts differ markedly in children, research that specifically examines COVID-19 in children needs to be prioritised.
Collapse
Affiliation(s)
- Annaleise R Howard‐Jones
- Discipline of Child and Adolescent HealthUniversity of Sydney, The Children's Hospital at WestmeadSydneyNew South WalesAustralia
- NSW Health Pathology‐NepeanNepean HospitalSydneyNew South WalesAustralia
| | - David P Burgner
- Infectious Diseases UnitDepartment of General Medicine, Royal Children's HospitalMelbourneVictoriaAustralia
- Infection and Immunity ThemeMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
| | - Nigel W Crawford
- Infection and Immunity ThemeMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
- Department of General MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Emma Goeman
- Department of Infectious Diseases and MicrobiologyRoyal Prince Alfred HospitalSydneyNew South WalesAustralia
| | - Paul E Gray
- Department of Immunology and Infectious DiseasesSydney Children's HospitalSydneyNew South WalesAustralia
- School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Peter Hsu
- Discipline of Child and Adolescent HealthUniversity of Sydney, The Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Department of ImmunologyThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Stephanie Kuek
- Department of Respiratory and Sleep MedicineThe Royal Children's HospitalMelbourneVictoriaAustralia
| | - Brendan J McMullan
- Department of Immunology and Infectious DiseasesSydney Children's HospitalSydneyNew South WalesAustralia
- School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Shidan Tosif
- Infection and Immunity ThemeMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
- Department of General MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Danielle Wurzel
- Infection and Immunity ThemeMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of Respiratory and Sleep MedicineThe Royal Children's HospitalMelbourneVictoriaAustralia
- School of Population and Global HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Asha C Bowen
- Department of Infectious DiseasesPerth Children's HospitalPerthWestern AustraliaAustralia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids InstituteUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Margie Danchin
- Infection and Immunity ThemeMurdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
- Department of General MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Archana Koirala
- Discipline of Child and Adolescent HealthUniversity of Sydney, The Children's Hospital at WestmeadSydneyNew South WalesAustralia
- National Centre for Immunisation Research and SurveillanceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Department of Infectious DiseasesNepean HospitalPenrithNew South WalesAustralia
| | - Ketaki Sharma
- Discipline of Child and Adolescent HealthUniversity of Sydney, The Children's Hospital at WestmeadSydneyNew South WalesAustralia
- National Centre for Immunisation Research and SurveillanceThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Daniel K Yeoh
- Department of Infectious DiseasesPerth Children's HospitalPerthWestern AustraliaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Philip N Britton
- Discipline of Child and Adolescent HealthUniversity of Sydney, The Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Department of Infectious Diseases and MicrobiologyThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| |
Collapse
|
37
|
Dowell AC, Butler MS, Jinks E, Tut G, Lancaster T, Sylla P, Begum J, Bruton R, Pearce H, Verma K, Logan N, Tyson G, Spalkova E, Margielewska-Davies S, Taylor GS, Syrimi E, Baawuah F, Beckmann J, Okike IO, Ahmad S, Garstang J, Brent AJ, Brent B, Ireland G, Aiano F, Amin-Chowdhury Z, Jones S, Borrow R, Linley E, Wright J, Azad R, Waiblinger D, Davis C, Thomson EC, Palmarini M, Willett BJ, Barclay WS, Poh J, Amirthalingam G, Brown KE, Ramsay ME, Zuo J, Moss P, Ladhani S. Children develop robust and sustained cross-reactive spike-specific immune responses to SARS-CoV-2 infection. Nat Immunol 2022; 23:40-49. [PMID: 34937928 PMCID: PMC8709786 DOI: 10.1038/s41590-021-01089-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022]
Abstract
SARS-CoV-2 infection is generally mild or asymptomatic in children but a biological basis for this outcome is unclear. Here we compare antibody and cellular immunity in children (aged 3-11 years) and adults. Antibody responses against spike protein were high in children and seroconversion boosted responses against seasonal Beta-coronaviruses through cross-recognition of the S2 domain. Neutralization of viral variants was comparable between children and adults. Spike-specific T cell responses were more than twice as high in children and were also detected in many seronegative children, indicating pre-existing cross-reactive responses to seasonal coronaviruses. Importantly, children retained antibody and cellular responses 6 months after infection, whereas relative waning occurred in adults. Spike-specific responses were also broadly stable beyond 12 months. Therefore, children generate robust, cross-reactive and sustained immune responses to SARS-CoV-2 with focused specificity for the spike protein. These findings provide insight into the relative clinical protection that occurs in most children and might help to guide the design of pediatric vaccination regimens.
Collapse
Affiliation(s)
- Alexander C Dowell
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Megan S Butler
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Elizabeth Jinks
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gokhan Tut
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Tara Lancaster
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Panagiota Sylla
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jusnara Begum
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel Bruton
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Kriti Verma
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nicola Logan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Grace Tyson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Eliska Spalkova
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sandra Margielewska-Davies
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Graham S Taylor
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eleni Syrimi
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | | | - Ifeanyichukwu O Okike
- Public Health England, 61 Colindale Avenue, London, UK
- University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Shazaad Ahmad
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Joanna Garstang
- Birmingham Community Healthcare NHS Trust, Aston, UK
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew J Brent
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- University of Oxford, Wellington Square, Oxford, UK
| | | | | | | | | | - Samuel Jones
- Public Health England, 61 Colindale Avenue, London, UK
| | - Ray Borrow
- Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - Ezra Linley
- Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Rafaq Azad
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Dagmar Waiblinger
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Chris Davis
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Emma C Thomson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Wendy S Barclay
- Department of Infectious Disease, Imperial College, London, UK
| | - John Poh
- Public Health England, 61 Colindale Avenue, London, UK
| | | | - Kevin E Brown
- Public Health England, 61 Colindale Avenue, London, UK
| | - Mary E Ramsay
- Public Health England, 61 Colindale Avenue, London, UK
| | - Jianmin Zuo
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paul Moss
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Shamez Ladhani
- Public Health England, 61 Colindale Avenue, London, UK
- Paediatric Infectious Diseases Research Group, St. George's University of London, London, UK
| |
Collapse
|
38
|
Dobaño C, Alonso S, Fernández de Sevilla M, Vidal M, Jiménez A, Pons Tomas G, Jairoce C, Melé Casas M, Rubio R, Hernández García M, Ruiz-Olalla G, Girona-Alarcón M, Barrios D, Santano R, Mitchell RA, Puyol L, Mayer L, Chi J, Rodrigo Melero N, Carolis C, Garcia-Miquel A, Bonet-Carne E, Claverol J, Cubells M, Fortuny C, Fumadó V, Jou C, Muñoz-Almagro C, Izquierdo L, Bassat Q, Gratacós E, Aguilar R, García-García JJ, Moncunill G, Jordan I. Antibody conversion rates to SARS-CoV-2 in saliva from children attending summer schools in Barcelona, Spain. BMC Med 2021; 19:309. [PMID: 34809617 PMCID: PMC8608564 DOI: 10.1186/s12916-021-02184-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Surveillance tools to estimate viral transmission dynamics in young populations are essential to guide recommendations for school opening and management during viral epidemics. Ideally, sensitive techniques are required to detect low viral load exposures among asymptomatic children. We aimed to estimate SARS-CoV-2 infection rates in children and adult populations in a school-like environment during the initial COVID-19 pandemic waves using an antibody-based field-deployable and non-invasive approach. METHODS Saliva antibody conversion defined as ≥ 4-fold increase in IgM, IgA, and/or IgG levels to five SARS-CoV-2 antigens including spike and nucleocapsid constructs was evaluated in 1509 children and 396 adults by high-throughput Luminex assays in samples collected weekly in 22 summer schools and 2 pre-schools in 27 venues in Barcelona, Spain, from June 29th to July 31st, 2020. RESULTS Saliva antibody conversion between two visits over a 5-week period was 3.22% (49/1518) or 2.36% if accounting for potentially cross-reactive antibodies, six times higher than the cumulative infection rate (0.53%) assessed by weekly saliva RT-PCR screening. IgG conversion was higher in adults (2.94%, 11/374) than children (1.31%, 15/1144) (p=0.035), IgG and IgA levels moderately increased with age, and antibodies were higher in females. Most antibody converters increased both IgG and IgA antibodies but some augmented either IgG or IgA, with a faster decay over time for IgA than IgG. Nucleocapsid rather than spike was the main antigen target. Anti-spike antibodies were significantly higher in individuals not reporting symptoms than symptomatic individuals, suggesting a protective role against COVID-19. CONCLUSION Saliva antibody profiling including three isotypes and multiplexing antigens is a useful and user-friendlier tool for screening pediatric populations to detect low viral load exposures among children, particularly while they are not vaccinated and vulnerable to highly contagious variants, and to recommend public health policies during pandemics.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain. .,CIBER de Enfermedades Infecciosas, Madrid, Spain. .,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| | - Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Mariona Fernández de Sevilla
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Gemma Pons Tomas
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Chenjerai Jairoce
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - María Melé Casas
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Rocío Rubio
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - María Hernández García
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Mònica Girona-Alarcón
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Diana Barrios
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Rebeca Santano
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Robert A Mitchell
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Laura Puyol
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Leonie Mayer
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Chi
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Aleix Garcia-Miquel
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Elisenda Bonet-Carne
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Universitat Politècnica de Catalunya, BarcelonaTech, Barcelona, Spain
| | - Joana Claverol
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Fundació Sant Joan de Déu, Barcelona, Spain
| | - Marta Cubells
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Fundació Sant Joan de Déu, Barcelona, Spain
| | - Claudia Fortuny
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Infectious Diseases Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Victoria Fumadó
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Infectious Diseases Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cristina Jou
- Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Department of Pathology and Biobank Hospital Sant Joan de Déu, Barcelona, Spain.,CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Carmen Muñoz-Almagro
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Department of Medicine, Universitat Internacional de Catalunya, Barcelona, Spain.,Molecular Microbiology Department, Hospital Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain.,Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| | - Eduard Gratacós
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Juan José García-García
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain.,Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues, Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Iolanda Jordan
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain. .,Institut de Recerca Sant Joan de Déu, Esplugues, Barcelona, Spain. .,Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
39
|
Tsukinoki K, Yamamoto T, Handa K, Iwamiya M, Saruta J, Ino S, Sakurai T. Detection of cross-reactive immunoglobulin A against the severe acute respiratory syndrome-coronavirus-2 spike 1 subunit in saliva. PLoS One 2021; 16:e0249979. [PMID: 34813596 PMCID: PMC8610234 DOI: 10.1371/journal.pone.0249979] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 10/06/2021] [Indexed: 11/20/2022] Open
Abstract
Abundant secretory immunoglobulin A (SIgA) in the mucus, breast milk, and saliva provides immunity against infection of mucosal surfaces. Pre-pandemic breast milk samples containing SIgA have been reported to cross-react with SARS-CoV-2; however, it remains unknown whether SIgA showing the cross-reaction with SARS-CoV-2 exists in saliva. We aimed to clarify whether SIgA in saliva cross-reacts with SARS-CoV-2 spike 1 subunit in individuals who have not been infected with this virus. The study involved 137 (men, n = 101; women, n = 36; mean age, 38.7; age range, 24-65 years) dentists and doctors from Kanagawa Dental University Hospital. Saliva and blood samples were analyzed by polymerase chain reaction (PCR) and immunochromatography for IgG and IgM, respectively. We then identified patients with saliva samples that were confirmed to be PCR-negative and IgM-negative for SARS-CoV-2. The cross-reactivity of IgA-positive saliva samples with SARS-CoV-2 was determined by enzyme-linked immunosorbent assay using a biotin-labeled spike recombinant protein (S1-mFc) covering the receptor-binding domain of SARS-CoV-2. The proportion of SARS-CoV-2 cross-reactive IgA-positive individuals was 46.7%, which correlated negatively with age (r = -0.218, p = 0.01). The proportion of IgA-positive individuals aged ≥50 years was significantly lower than that of patients aged ≤49 years (p = 0.008). SIgA was purified from the saliva of patients, which could partially suppress the binding of SARS-CoV-2 spike protein to the angiotensin converting enzyme-2 receptor. This study demonstrates the presence of SARS-CoV-2 cross-reactive SIgA in the saliva of individuals who had never been infected with the virus, suggesting that SIgA may help prevent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Keiichi Tsukinoki
- Department of Environmental Pathology, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| | - Tatsuo Yamamoto
- Department of Dental Sociology, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| | - Keisuke Handa
- Department of Oral Biochemistry, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| | - Mariko Iwamiya
- Department of Clinical Laboratory, Kanagawa Dental University Hospital, Yokosuka, Kanagawa, Japan
| | - Juri Saruta
- Department of Environmental Pathology, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| | - Satoshi Ino
- Department of Minimal Intervention Prosthodontics, Graduate School of Dentistry, Kanagawa Dental University, Yokohama, Kanagawa, Japan
| | - Takashi Sakurai
- Department of Maxillofacial Radiology, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Kanagawa, Japan
| |
Collapse
|
40
|
Ehara H. Detailed Analysis of Immune Tolerance Mechanisms to SARS-CoV-2 in Children Is Needed. Front Pediatr 2021; 9:652838. [PMID: 34796148 PMCID: PMC8594572 DOI: 10.3389/fped.2021.652838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Hiroshi Ehara
- Pediatrics and Primary Care, Ehara Clinic, Tokyo, Japan
| |
Collapse
|
41
|
Neeland MR, Bannister S, Clifford V, Nguyen J, Dohle K, Overmars I, Toh ZQ, Anderson J, Donato CM, Sarkar S, Do LAH, McCafferty C, Licciardi PV, Ignjatovic V, Monagle P, Bines JE, Mulholland K, Curtis N, McNab S, Steer AC, Burgner DP, Saffery R, Tosif S, Crawford NW. Children and Adults in a Household Cohort Study Have Robust Longitudinal Immune Responses Following SARS-CoV-2 Infection or Exposure. Front Immunol 2021; 12:741639. [PMID: 34721408 PMCID: PMC8548628 DOI: 10.3389/fimmu.2021.741639] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Children have reduced severity of COVID-19 compared to adults and typically have mild or asymptomatic disease. The immunological mechanisms underlying these age-related differences in clinical outcomes remain unexplained. Here, we quantify 23 immune cell populations in 141 samples from children and adults with mild COVID-19 and their PCR-negative close household contacts at acute and convalescent time points. Children with COVID-19 displayed marked reductions in myeloid cells during infection, most prominent in children under the age of five. Recovery from infection in both children and adults was characterised by the generation of CD8 TCM and CD4 TCM up to 9 weeks post infection. SARS-CoV-2-exposed close contacts also had immunological changes over time despite no evidence of confirmed SARS-CoV-2 infection on PCR testing. This included an increase in low-density neutrophils during convalescence in both exposed children and adults, as well as increases in CD8 TCM and CD4 TCM in exposed adults. In comparison to children with other common respiratory viral infections, those with COVID-19 had a greater change in innate and T cell-mediated immune responses over time. These findings provide new mechanistic insights into the immune response during and after recovery from COVID-19 in both children and adults.
Collapse
Affiliation(s)
- Melanie R Neeland
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Samantha Bannister
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Vanessa Clifford
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia.,Laboratory Services, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Jill Nguyen
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Kate Dohle
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Isabella Overmars
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Zheng Quan Toh
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Jeremy Anderson
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Celeste M Donato
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Sohinee Sarkar
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Lien Anh Ha Do
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Conor McCafferty
- Clinical Sciences Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Paul V Licciardi
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Vera Ignjatovic
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Clinical Sciences Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Paul Monagle
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Clinical Sciences Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Clinical Haematology, The Royal Children's Hospital, Parkville, VIC, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Julie E Bines
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Parkville, VIC, Australia
| | - Kim Mulholland
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Epidemiology Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nigel Curtis
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Sarah McNab
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Parkville, VIC, Australia
| | - Andrew C Steer
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - David P Burgner
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Richard Saffery
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Shidan Tosif
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of General Medicine, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Nigel W Crawford
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of General Medicine, The Royal Children's Hospital, Parkville, VIC, Australia
| |
Collapse
|
42
|
Alonso S, Alvarez-Lacalle E, Català M, López D, Jordan I, García-García JJ, Soriano-Arandes A, Lazcano U, Sallés P, Masats M, Urrutia J, Gatell A, Capdevila R, Soler-Palacin P, Bassat Q, Prats C. Age-dependency of the Propagation Rate of Coronavirus Disease 2019 Inside School Bubble Groups in Catalonia, Spain. Pediatr Infect Dis J 2021; 40:955-961. [PMID: 34321438 PMCID: PMC8505138 DOI: 10.1097/inf.0000000000003279] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/03/2021] [Indexed: 12/05/2022]
Abstract
BACKGROUND We analyzed contagions of coronavirus disease 2019 inside school bubble groups in Catalonia, Spain, in the presence of strong nonpharmaceutical interventions from September to December 2020. More than 1 million students were organized in bubble groups and monitored and analyzed by the Health and the Educational departments. METHODS We had access to 2 data sources, and both were employed for the analysis, one is the Catalan school surveillance system and the other of the educational department. As soon as a positive index case is detected by the health system, isolation is required for all members of the bubble group, in addition to a mandatory proactive systematic screening of each individual. All infected cases are reported. It permits the calculation of the average reproductive number (R*), corresponding to the average number of infected individuals per index case. RESULTS We found that propagation inside of the bubble group was small. Among 75% index cases, there was no transmission to other members in the classroom, with an average R* across all ages inside the bubble of R* = 0.4. We found a significant age trend in the secondary attack rates, with the R* going from 0.2 in preschool to 0.6 in high school youth. CONCLUSIONS The secondary attack rate depends on the school level and therefore on the age. Super-spreading events (outbreaks of 5 cases or more) in childhood were rare, only occurring in 2.5% of all infections triggered from a pediatric index case.
Collapse
Affiliation(s)
- Sergio Alonso
- From the Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Enric Alvarez-Lacalle
- From the Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Martí Català
- From the Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Daniel López
- From the Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Iolanda Jordan
- Paediatric Intensive Care Unit, Hospital Sant Joan de Déu, University of Barcelona
- Institut de Recerca Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Juan José García-García
- Institut de Recerca Sant Joan de Déu, University of Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Pediatrics, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues
| | - Antoni Soriano-Arandes
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d’Hebron
| | - Uxue Lazcano
- Agència de Qualitat i Avaluació Sanitàries de Catalunya, AQuAS, Generalitat de Catalunya, Barcelona, Catalonia, Spain
| | | | | | | | - Anna Gatell
- Equip Pediatria Territorial Alt Penedès-Garraf, Barcelona, Catalonia, Spain
| | | | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d’Hebron
| | - Quique Bassat
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Pediatrics, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ICREA, Barcelona, Spain
| | - Clara Prats
- From the Department of Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Catalonia, Spain
| |
Collapse
|
43
|
Conti MG, Terreri S, Piano Mortari E, Albano C, Natale F, Boscarino G, Zacco G, Palomba P, Cascioli S, Corrente F, Capponi C, Mirabella M, Salinas AF, Marciano A, De Luca F, Pangallo I, Quaranta C, Alteri C, Russo C, Galoppi P, Brunelli R, Perno CF, Terrin G, Carsetti R. Immune Response of Neonates Born to Mothers Infected With SARS-CoV-2. JAMA Netw Open 2021; 4:e2132563. [PMID: 34730817 PMCID: PMC8567114 DOI: 10.1001/jamanetworkopen.2021.32563] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
IMPORTANCE Although several studies have provided information on short-term clinical outcomes in children with perinatal exposure to SARS-CoV-2, data on the immune response in the first months of life among newborns exposed to the virus in utero are lacking. OBJECTIVE To characterize systemic and mucosal antibody production during the first 2 months of life among infants who were born to mothers infected with SARS-CoV-2. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study enrolled 28 pregnant women who tested positive for SARS-CoV-2 infection and who gave birth at Policlinico Umberto I in Rome, Italy, from November 2020 to May 2021, and their newborns. Maternal and neonatal systemic immune responses were investigated by detecting spike-specific antibodies in serum, and the mucosal immune response was assessed by measuring specific antibodies in maternal breastmilk and infant saliva 48 hours after delivery and 2 months later. EXPOSURES Maternal infection with SARS-CoV-2 in late pregnancy. MAIN OUTCOMES AND MEASURES The systemic immune response was evaluated by the detection of SARS-CoV-2 IgG and IgA antibodies and receptor binding domain-specific IgM antibodies in maternal and neonatal serum. The mucosal immune response was assessed by measuring spike-specific antibodies in breastmilk and in infant saliva, and the presence of antigen-antibody spike IgA immune complexes was investigated in breastmilk samples. All antibodies were detected using an enzyme-linked immunosorbent assay. RESULTS In total, 28 mother-infant dyads (mean [SD] maternal age, 31.8 [6.4] years; mean [SD] gestational age, 38.1 [2.3] weeks; 18 [60%] male infants) were enrolled at delivery, and 21 dyads completed the study at 2 months' follow-up. Because maternal infection was recent in all cases, transplacental transfer of virus spike-specific IgG antibodies occurred in only 1 infant. One case of potential vertical transmission and 1 case of horizontal infection were observed. Virus spike protein-specific salivary IgA antibodies were significantly increased (P = .01) in infants fed breastmilk (0.99 arbitrary units [AU]; IQR, 0.39-1.68 AU) vs infants fed an exclusive formula diet (0.16 AU; IQR, 0.02-0.83 AU). Maternal milk contained IgA spike immune complexes at 48 hours (0.53 AU; IQR, 0.25-0.39 AU) and at 2 months (0.09 AU; IQR, 0.03-0.17 AU) and may have functioned as specific stimuli for the infant mucosal immune response. CONCLUSIONS AND RELEVANCE In this cohort study, SARS-CoV-2 spike-specific IgA antibodies were detected in infant saliva, which may partly explain why newborns are resistant to SARS-CoV-2 infection. Mothers infected in the peripartum period appear to not only passively protect the newborn via breastmilk secretory IgA but also actively stimulate and train the neonatal immune system via breastmilk immune complexes.
Collapse
Affiliation(s)
- Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Eva Piano Mortari
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Christian Albano
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabio Natale
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giovanni Boscarino
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giulia Zacco
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Patrizia Palomba
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Italy
| | - Simona Cascioli
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesco Corrente
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Italy
| | - Claudia Capponi
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Italy
| | - Mattia Mirabella
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Italy
| | - Ane Fernandez Salinas
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandra Marciano
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Francesca De Luca
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ida Pangallo
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Cecilia Quaranta
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Claudia Alteri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Cristina Russo
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Galoppi
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Roberto Brunelli
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Carlo Federico Perno
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
44
|
BNT162b2 mRNA Vaccination Leads to Long-Term Protection from COVID-19 Disease. Vaccines (Basel) 2021; 9:vaccines9101164. [PMID: 34696272 PMCID: PMC8538967 DOI: 10.3390/vaccines9101164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 11/18/2022] Open
Abstract
The efficacy of SARS-CoV-2 mRNA-based vaccines in preventing COVID-19 disease has been extensively demonstrated; however, it is of uttermost importance to acquire knowledge on the persistence of immune-protection both in terms of levels of neutralizing antibodies and specialized memory cells. This can provide important scientific basis for decisions on the need of additional vaccine doses and on when these should be administered thus resulting in an improvement in vaccination schedules. Here, we briefly report the changes in antibody levels and cellular immunity following BNT162b2 administration. We show an important fall in anti S1-Spike antibodies in BNT162b2 vaccinated subjects overtime, paralleled by a contextual consolidation of specific spike (S) T-cells, mainly of the CD8+ compartment. Contrariwise, CD4+ S-specific response shows a considerable interindividual variability. These data suggest that the well-known antibody drop in vaccinated subjects is replaced by memory cell consolidation that can protect from severe adverse effects of SARS-CoV-2 infection.
Collapse
|
45
|
Chauhan N, Soni S, Jain U. Optimizing testing regimes for the detection of COVID-19 in children and older adults. Expert Rev Mol Diagn 2021; 21:999-1016. [PMID: 34324823 PMCID: PMC8425447 DOI: 10.1080/14737159.2021.1962708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is a major pandemic and continuously emerging due to unclear prognosis and unavailability of reliable detection tools. Older adults are more susceptible to COVID-19 than children showing mature Angiotensin-Converting Enzyme 2 (ACE2), low concentration of immune targets, and comorbid conditions. Several detection platforms have been commercialized to date and more are in pipeline, however, the rate of false-positive results and rapid mutation of SARS-CoV-2 is increasing. Additionally, physiological, and geographical variations of affected individuals are also calling for diagnostic methods optimization.Areas Covered: Extensive information related to the optimization and usefulness of SARS-CoV-2 diagnostic methods based on sensitivity and specificity as definitive and feasible investigative tools is discussed. Moreover, an option of combining laboratory diagnostic methods to improve diagnostic strategies is also proposed and discussed in the comparative section of optimization studies.Expert Opinion: The review article explains the importance of optimization strategies for SARS-CoV-2 detection in children and older adults. There are advancements in COVID-19 detection including CRISPR-based, electrochemical, and optical-based sensing systems. However, the lack of sufficient studies on a comparative evaluation of standardized SARS-CoV-2 diagnostic methods among children and older adults, limit the authentication of commercialized kits.
Collapse
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Shringika Soni
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| |
Collapse
|
46
|
Lindsay L, Secrest MH, Rizzo S, Keebler DS, Yang F, Tsai L. Factors associated with COVID-19 viral and antibody test positivity and assessment of test concordance: a retrospective cohort study using electronic health records from the USA. BMJ Open 2021; 11:e051707. [PMID: 34598988 PMCID: PMC8488284 DOI: 10.1136/bmjopen-2021-051707] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To identify factors associated with COVID-19 test positivity and assess viral and antibody test concordance. DESIGN Observational retrospective cohort study. SETTING Optum de-identified electronic health records including over 700 hospitals and 7000 clinics in the USA. PARTICIPANTS There were 891 754 patients who had a COVID-19 test identified in their electronic health record between 20 February 2020 and 10 July 2020. PRIMARY AND SECONDARY OUTCOME MEASURES Per cent of viral and antibody tests positive for COVID-19 ('positivity rate'); adjusted ORs for factors associated with COVID-19 viral and antibody test positivity; and per cent concordance between positive viral and subsequent antibody test results. RESULTS Overall positivity rate was 9% (70 472 of 771 278) and 12% (11 094 of 91 741) for viral and antibody tests, respectively. Positivity rate was inversely associated with the number of individuals tested and decreased over time across regions and race/ethnicities. Antibody test concordance among patients with an initial positive viral test was 91% (71%-95% depending on time between tests). Among tests separated by at least 2 weeks, discordant results occurred in 7% of patients and 9% of immunocompromised patients. Factors associated with increased odds of viral and antibody positivity in multivariable models included: male sex, Hispanic or non-Hispanic black or Asian race/ethnicity, uninsured or Medicaid insurance and Northeast residence. We identified a negative dose effect between the number of comorbidities and viral and antibody test positivity. Paediatric patients had reduced odds (OR=0.60, 95% CI 0.57 to 0.64) of a positive viral test but increased odds (OR=1.90, 95% CI 1.62 to 2.23) of a positive antibody test compared with those aged 18-34 years old. CONCLUSIONS This study identified sociodemographic and clinical factors associated with COVID-19 test positivity and provided real-world evidence demonstrating high antibody test concordance among viral-positive patients.
Collapse
Affiliation(s)
- Lisa Lindsay
- Personalized Healthcare, Data Science, Genentech, South San Francisco, California, USA
| | - Matthew H Secrest
- Personalized Healthcare, Data Science, Genentech, South San Francisco, California, USA
| | - Shemra Rizzo
- Personalized Healthcare, Data Science, Genentech, South San Francisco, California, USA
| | - Daniel S Keebler
- Personalized Healthcare, Data Science, Genentech, South San Francisco, California, USA
| | - Fei Yang
- Roche Information Solutions, Roche Diagnostics International, Rotkreuz, Switzerland
| | - Larry Tsai
- Product Development, Genentech, South San Francisco, California, USA
| |
Collapse
|
47
|
Piano Mortari E, Russo C, Vinci MR, Terreri S, Fernandez Salinas A, Piccioni L, Alteri C, Colagrossi L, Coltella L, Ranno S, Linardos G, Agosta M, Albano C, Agrati C, Castilletti C, Meschi S, Romania P, Roscilli G, Pavoni E, Camisa V, Santoro A, Brugaletta R, Magnavita N, Ruggiero A, Cotugno N, Amodio D, Ciofi Degli Atti ML, Giorgio D, Russo N, Salvatori G, Corsetti T, Locatelli F, Perno CF, Zaffina S, Carsetti R. Highly Specific Memory B Cells Generation after the 2nd Dose of BNT162b2 Vaccine Compensate for the Decline of Serum Antibodies and Absence of Mucosal IgA. Cells 2021; 10:cells10102541. [PMID: 34685521 PMCID: PMC8533837 DOI: 10.3390/cells10102541] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Specific memory B cells and antibodies are a reliable read-out of vaccine efficacy. We analysed these biomarkers after one and two doses of BNT162b2 vaccine. The second dose significantly increases the level of highly specific memory B cells and antibodies. Two months after the second dose, specific antibody levels decline, but highly specific memory B cells continue to increase, thus predicting a sustained protection from COVID-19. We show that although mucosal IgA is not induced by the vaccination, memory B cells migrate in response to inflammation and secrete IgA at mucosal sites. We show that the first vaccine dose may lead to an insufficient number of highly specific memory B cells and low concentration of serum antibodies, thus leaving vaccinees without the immune robustness needed to ensure viral elimination and herd immunity. We also clarify that the reduction of serum antibodies does not diminish the force and duration of the immune protection induced by vaccination. The vaccine does not induce sterilizing immunity. Infection after vaccination may be caused by the lack of local preventive immunity because of the absence of mucosal IgA.
Collapse
Affiliation(s)
- Eva Piano Mortari
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Cristina Russo
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Maria Rosaria Vinci
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Ane Fernandez Salinas
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Molecular Medicine, Sapienza University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Livia Piccioni
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Claudia Alteri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via festa del Perdono, 7, 20122 Milan, Italy
| | - Luna Colagrossi
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Luana Coltella
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Stefania Ranno
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Giulia Linardos
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Marilena Agosta
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Christian Albano
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Chiara Agrati
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Concetta Castilletti
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Silvia Meschi
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Paolo Romania
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Molecular Medicine, Sapienza University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Giuseppe Roscilli
- Takis s.r.l., Via di Castel Romano, 100, 00128 Rome, Italy; (G.R.); (E.P.)
| | - Emiliano Pavoni
- Takis s.r.l., Via di Castel Romano, 100, 00128 Rome, Italy; (G.R.); (E.P.)
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Annapaola Santoro
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Rita Brugaletta
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Nicola Magnavita
- Section of Occupational Medicine and Labor Law, Post-Graduate School of Occupational Health, University Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy;
- Department of Woman, Child & Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Via della Pineta Sacchetti, 217, 00168 Rome, Italy
| | - Alessandra Ruggiero
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Via San Francesco, 22, 37129 Verona, Italy
| | - Nicola Cotugno
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
| | - Donato Amodio
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
| | - Marta Luisa Ciofi Degli Atti
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
| | - Daniela Giorgio
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Nicoletta Russo
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Tiziana Corsetti
- Hospital Pharmacy Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
| | - Franco Locatelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
- Department of Pediatrics, Sapienza, University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Carlo Federico Perno
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
- Correspondence:
| |
Collapse
|
48
|
Fröberg J, Gillard J, Philipsen R, Lanke K, Rust J, van Tuijl D, Teelen K, Bousema T, Simonetti E, van der Gaast-de Jongh CE, Bos M, van Kuppeveld FJ, Bosch BJ, Nabuurs-Franssen M, van der Geest-Blankert N, van Daal C, Huynen MA, de Jonge MI, Diavatopoulos DA. SARS-CoV-2 mucosal antibody development and persistence and their relation to viral load and COVID-19 symptoms. Nat Commun 2021; 12:5621. [PMID: 34556667 PMCID: PMC8460778 DOI: 10.1038/s41467-021-25949-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
Although serological studies have shown that antibodies against SARS-CoV-2 play an important role in protection against (re)infection, the dynamics of mucosal antibodies during primary infection and their potential impact on viral load and the resolution of disease symptoms remain unclear. During the first pandemic wave, we assessed the longitudinal nasal antibody response in index cases with mild COVID-19 and their household contacts. Nasal and serum antibody responses were analysed for up to nine months. Higher nasal receptor binding domain and spike protein-specific antibody levels at study inclusion were associated with lower viral load. Older age was correlated with more frequent COVID-19 related symptoms. Receptor binding domain and spike protein-specific mucosal antibodies were associated with the resolution of systemic, but not respiratory symptoms. Finally, receptor binding domain and spike protein-specific mucosal antibodies remained elevated up to nine months after symptom onset.
Collapse
Affiliation(s)
- Janeri Fröberg
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Joshua Gillard
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- Centre for molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Ria Philipsen
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Kjerstin Lanke
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Joyce Rust
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Diana van Tuijl
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
- RTC CS Radboud Technology Center Clinical Studies, Radboudumc, Nijmegen, The Netherlands
| | - Karina Teelen
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Teun Bousema
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Elles Simonetti
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Christa E van der Gaast-de Jongh
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Mariska Bos
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Frank J van Kuppeveld
- Utrecht University, Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Utrecht, The Netherlands
| | - Berend-Jan Bosch
- Utrecht University, Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Utrecht, The Netherlands
| | - Marrigje Nabuurs-Franssen
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | | | - Charlotte van Daal
- Department of Occupational Health, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Centre for molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Marien I de Jonge
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Laboratory of Medical Immunology, Section Paediatric Infectious Diseases, 6525 GA, Nijmegen, The Netherlands.
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
49
|
Lemke MM, McLean MR, Lee CY, Lopez E, Bozich ER, Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kratochvil S, Wines BD, Hogarth PM, Kent SJ, Chung AW, Arnold KB. A systems approach to elucidate personalized mechanistic complexities of antibody-Fc receptor activation post-vaccination. CELL REPORTS MEDICINE 2021; 2:100386. [PMID: 34622227 PMCID: PMC8484512 DOI: 10.1016/j.xcrm.2021.100386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/16/2021] [Accepted: 08/11/2021] [Indexed: 11/25/2022]
Abstract
Immunoglobulin G (IgG) antibodies that activate Fc-mediated immune functions have been correlated with vaccine efficacy, but it is difficult to unravel the relative roles of multiple IgG and Fc receptor (FcR) features that have the capacity to influence IgG-FcR complex formation but vary on a personalized basis. Here, we develop an ordinary differential-equation model to determine how personalized variability in IgG subclass concentrations and binding affinities influence IgG-FcγRIIIa complex formation and validate it with samples from the HIV RV144 vaccine trial. The model identifies individuals who are sensitive, insensitive, or negatively affected by increases in HIV-specific IgG1, which is validated with the addition of HIV-specific IgG1 monoclonal antibodies to vaccine samples. IgG1 affinity to FcγRIIIa is also prioritized as the most influential parameter for dictating activation broadly across a population. Overall, this work presents a quantitative tool for evaluating personalized differences underlying FcR activation, which is relevant to ongoing efforts to improve vaccine efficacy. Fc-mediated immune functions have been correlated with protection in HIV vaccine trials A model reveals personalized mechanisms that drive variation in FcγR activation The model predicts individuals who are sensitive to changes in IgG1 concentration IgG1 affinity to FcγR best dictates activation across a heterogeneous population
Collapse
Affiliation(s)
- Melissa M Lemke
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Milla R McLean
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Christina Y Lee
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ester Lopez
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Emily R Bozich
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Punnee Pitisuttithum
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Sven Kratochvil
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Bruce D Wines
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - P Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, VIC, Australia.,Melbourne Sexual Health Centre, Alfred Hospital, Monash University Central Clinical School, Carlton, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
50
|
Blanchard-Rohner G, Didierlaurent A, Tilmanne A, Smeesters P, Marchant A. Pediatric COVID-19: Immunopathogenesis, Transmission and Prevention. Vaccines (Basel) 2021; 9:1002. [PMID: 34579240 PMCID: PMC8473426 DOI: 10.3390/vaccines9091002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022] Open
Abstract
Children are unique in the context of the COVID-19 pandemic. Overall, SARS-CoV-2 has a lower medical impact in children as compared to adults. A higher proportion of children than adults remain asymptomatic following SARS-CoV-2 infection and severe disease and death are also less common. This relative resistance contrasts with the high susceptibility of children to other respiratory tract infections. The mechanisms involved remain incompletely understood but could include the rapid development of a robust innate immune response. On the other hand, children develop a unique and severe complication, named multisystem inflammatory syndrome in children, several weeks after the onset of symptoms. Although children play an important role in the transmission of many pathogens, their contribution to the transmission of SARS-CoV-2 appears lower than that of adults. These unique aspects of COVID-19 in children must be considered in the benefit-risk analysis of vaccination. Several COVID-19 vaccines have been authorized for emergency use in adolescents and clinical studies are ongoing in children. As the vaccination of adolescents is rolled out in several countries, we shall learn about the impact of this strategy on the health of children and on transmission within communities.
Collapse
Affiliation(s)
- Geraldine Blanchard-Rohner
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Pediatric Immunology and Vaccinology Unit, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, University of Geneva, 1205 Geneva, Switzerland;
- Children’s Hospital of Geneva, 6, Rue Willy-Donzé, 1211 Geneva, Switzerland
| | - Arnaud Didierlaurent
- Pediatric Immunology and Vaccinology Unit, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, University of Geneva, 1205 Geneva, Switzerland;
| | - Anne Tilmanne
- Children’s Hospital Queen Fabiola, Université libre de Bruxelles, 1020 Brussels, Belgium; (A.T.); (P.S.)
| | - Pierre Smeesters
- Children’s Hospital Queen Fabiola, Université libre de Bruxelles, 1020 Brussels, Belgium; (A.T.); (P.S.)
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, 1050 Charleroi, Belgium;
| |
Collapse
|