1
|
Kajova M, Khawaja T, Levonen I, Pietilä JP, Virtanen J, Pakkanen SH, Välimaa H, Nousiainen A, Hepojoki J, Sironen T, Vierikko A, Ihalainen J, Vapalahti O, Kantele A. Convalescent plasma therapy for COVID-19 - Donor selection strategies and establishment of a plasma bank. New Microbes New Infect 2024; 62:101525. [PMID: 39584055 PMCID: PMC11584594 DOI: 10.1016/j.nmni.2024.101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Background Early in the COVID-19 pandemic, convalescent plasma (CP) emerged as a potentially effective treatment neutralising SARS-CoV-2. Early CP therapy with high neutralising antibody (NAb) titre may benefit COVID-19 outpatients and, in sufficient quantities even some hospitalised patients. This study details the process of setting up a CP bank, containing high- and low-titre CP for a clinical trial. Study design and methods We identified 18-65-year-old convalescents with SARS-CoV-2 NAb titres of ≥1:40 in microneutralisation test (MNT). Following eligibility pre-screening, the Finnish Red Cross Blood Service (FRCBS) determined suitability as CP donors. Results Of the 6466 COVID-19 convalescents contacted, 1481 provided serum, with 851 (57.5 %) exhibiting NAb titres ≥1:40. Participation barriers included reluctance, advanced age and, for women, insufficient body size. Of the volunteers, 125 were evaluated at FRCBS, with major exclusions for HLA antibodies (42 women), interferon antibodies (five men), and NAb titres waning below 1:20 (16 participants). Finally, 70 underwent plasmapheresis, resulting in 50 suitable CP donors (0.8 % of initial contacts and 3.4 % of those tested for NAb). Discussion The process of setting up a CP bank proved challenging. Excessive laboratory workloads during a pandemic hamper their ability to conduct MNT, underscoring the need for rapid screening tests. Only a small proportion of our convalescents exhibited high-titre CP, this fraction declining over time because of waning immunity. Strict plasmapheresis criteria further constrained donor eligibility. Establishing a plasma bank requires meticulous planning to maximize efficiency. Detailed insights from current experiences may prove critical in future pandemics before other remedies and vaccines become available.
Collapse
Affiliation(s)
- Mikael Kajova
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Tamim Khawaja
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Iris Levonen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Jenni Virtanen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sari H. Pakkanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Hanna Välimaa
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Arttu Nousiainen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, University of Helsinki, Helsinki, Finland
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Tarja Sironen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | | | | | - Olli Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Anu Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| |
Collapse
|
2
|
Cavalcanti-Dantas VDM, da Silva AF, Mendes AF, de Araújo Júnior WO, Bernardo-Menezes LC, Bresani-Salvi CC, Castellano LRC, Fernandes AIV, Lemos SG, de Magalhães JJF, Oliveira RADS, de Assis PAC, de Souza JR, de Morais CNL. Performance assessment of a new serological diagnostic test for COVID-19 with candidate peptides from spike and nucleocapsid viral proteins. Braz J Microbiol 2024; 55:2797-2803. [PMID: 39042245 PMCID: PMC11405565 DOI: 10.1007/s42770-024-01446-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/28/2024] [Indexed: 07/24/2024] Open
Abstract
Numerous commercial tests for the serological diagnosis of COVID-19 have been produced in recent years. However, it is important to note that these tests exhibit significant variability in their sensitivity, specificity, and accuracy of results. Therefore, the objective of this study was to utilize bioinformatics tools to map SARS-CoV-2 peptides, with the goal of developing a new serological diagnostic test for COVID-19. Two peptides from the S protein and one from the N protein were selected and characterized in silico, chemically synthesized, and used as a serological diagnostic tool to detect IgM, IgG, and IgA anti-SARS-CoV-2 antibodies through the ELISA technique, confirmed as positive and negative samples by RT-qPCR or serology by ELISA. The results showed a sensitivity, specificity, Positive Predictive Value and Negative Predictive Value of 100% (p < 00001, 95% CI) for the proposed test. Although preliminary, this study brings proof-of-concept results that are consistent with the high-performance rates of the ELISA test when compared to other well-established methods for diagnosing COVID-19.
Collapse
Affiliation(s)
- Vanessa de Melo Cavalcanti-Dantas
- Laboratory of Virology and Experimental Therapy, Department of Virology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Alan Frazão da Silva
- Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrei Félix Mendes
- Laboratory of Microbiology, Department of Microbiology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Waldecir Oliveira de Araújo Júnior
- Laboratory of Immunology and Hematology, Department of Physiology and Pathology, Multidisciplinary Research Group in Biotechnology and Health (GePeMBiS), Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Lucas Coêlho Bernardo-Menezes
- Laboratory of Virology and Experimental Therapy, Department of Virology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Cristiane Campello Bresani-Salvi
- Laboratory of Virology and Experimental Therapy, Department of Virology, Group of Integrated Studies in Nutrition and Health, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Instituto de Medicina Integral Prof Fernando Figueira, Recife, Pernambuco, Brazil
| | - Lúcio Roberto Cançado Castellano
- Professional and Technological Center of the Technical School of Health, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Ana Isabel Vieira Fernandes
- Health Promotion Department of the Medical Sciences Center and Division for Infectious and Parasitic Diseases, Lauro Wanderley University Hospital, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Sherlan Guimarães Lemos
- Advanced Study Group in Analytical Chemistry, Department of Chemistry, Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Jurandy Júnior Ferraz de Magalhães
- Laboratory of Virology and Experimental Therapy, Department of Virology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Renato Antônio Dos Santos Oliveira
- Laboratory of Immunology and Hematology, Department of Physiology and Pathology, Multidisciplinary Research Group in Biotechnology and Health (GePeMBiS), Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Priscilla Anne Castro de Assis
- Laboratory of Immunology and Hematology, Department of Physiology and Pathology, Multidisciplinary Research Group in Biotechnology and Health (GePeMBiS), Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Joelma Rodrigues de Souza
- Laboratory of Immunology and Hematology, Department of Physiology and Pathology, Multidisciplinary Research Group in Biotechnology and Health (GePeMBiS), Federal University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Clarice Neuenschwander Lins de Morais
- Laboratory of Virology and Experimental Therapy, Department of Virology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil.
| |
Collapse
|
3
|
Gray P, Eriksson T, Skoglund L, Lagheden C, Hellström C, Pin E, Suomenrinne-Nordvik A, Pimenoff VN, Nilsson P, Dillner J, Lehtinen M. Seroepidemiological assessment of the spread of SARS-CoV-2 among 25 and 28 year-old adult women in Finland between March 2020-June 2022. PLoS One 2024; 19:e0305285. [PMID: 38990856 PMCID: PMC11238966 DOI: 10.1371/journal.pone.0305285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 07/13/2024] Open
Abstract
INTRODUCTION Serological surveys of the prevalence of SARS-CoV-2 are instrumental to understanding the course of the COVID-19 epidemic. We evaluate the seroprevalence of SARS-CoV-2 among young adult Finnish females residing in 25 communities all over Finland from 2020 until 2022. METHODS Between 1st March 2020 and 30th June 2022, 3589 blood samples were collected from 3583 women born in 1992-95 when aged 25 or 28 years old attending the follow-up of an ongoing population-based trial of cervical screening strategies. The crude and population standardized SARS-CoV-2 seroprevalence was measured using nucleocapsid (induced by infection) and spike wild-type (WT) protein (induced both by infection and by vaccination) antigens over time and stratified by place of residence (inside or outside the Helsinki metropolitan region). RESULTS During 2020 (before vaccinations), spike-WT and nucleocapsid IgG antibodies followed each other closely, at very low levels (<5%). Spike-WT seropositivity increased rapidly concomitant with mass vaccinations in 2021 and reached 96.3% in the 2nd quartile of 2022. Antibodies to nucleocapsid IgG remained relatively infrequent throughput 2020-2021, increasing rapidly in the 1st and 2nd quartiles of 2022 (to 19.7% and 56.6% respectively). The nucleocapsid IgG seropositivity increased more profoundly in participants residing in the Helsinki metropolitan region (4.5%, 8.4% and 43.9% in 2020, 2021 and 2022 respectively) compared to those residing in communities outside the capital region (4.5%, 4.3% and 34.7%). CONCLUSIONS Low SARS-CoV-2 infection-related seroprevalence during 2020-2021 suggest a comparatively successful infection control. Antibodies to the SARS-CoV-2 WT spike protein became extremely common among young women by the end of 2021, in line with the high uptake of SARS-CoV-2 vaccination. Finally, the rapid increase of seroprevalences to the SARS-CoV-2 nucleocapsid protein during the first and second quartile of 2022, imply a high incidence of infections with SARS-CoV-2 variants able to escape vaccine-induced protection.
Collapse
Affiliation(s)
- Penelope Gray
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | | - Lovisa Skoglund
- Division of Affinity Proteomics Department of Protein Science KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Camilla Lagheden
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ceke Hellström
- Division of Affinity Proteomics Department of Protein Science KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Elisa Pin
- Division of Affinity Proteomics Department of Protein Science KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Anna Suomenrinne-Nordvik
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Tampere University Hospital, Tampere, Finland
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Ville N Pimenoff
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biobank Borealis of Northern Finland, University of Oulu, Oulu, Finland
| | - Peter Nilsson
- Division of Affinity Proteomics Department of Protein Science KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Joakim Dillner
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Matti Lehtinen
- Center for Cervical Cancer Elimination, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Budylowski P, Chau SLL, Banerjee A, Guvenc F, Samson R, Hu Q, Fiddes L, Seifried L, Chao G, Buchholz M, Estacio A, Cheatley PL, Pavenski K, Patriquin CJ, Liu Y, Sheikh-Mohamed S, Crasta K, Yue F, Pasic MD, Mossman K, Gingras AC, Gommerman JL, Ehrhardt GRA, Mubareka S, Ostrowski M. A Significant Contribution of the Classical Pathway of Complement in SARS-CoV-2 Neutralization of Convalescent and Vaccinee Sera. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1922-1931. [PMID: 38683124 DOI: 10.4049/jimmunol.2300320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/09/2024] [Indexed: 05/01/2024]
Abstract
Although high titers of neutralizing Abs in human serum are associated with protection from reinfection by SARS-CoV-2, there is considerable heterogeneity in human serum-neutralizing Abs against SARS-CoV-2 during convalescence between individuals. Standard human serum live virus neutralization assays require inactivation of serum/plasma prior to testing. In this study, we report that the SARS-CoV-2 neutralization titers of human convalescent sera were relatively consistent across all disease states except for severe COVID-19, which yielded significantly higher neutralization titers. Furthermore, we show that heat inactivation of human serum significantly lowered neutralization activity in a live virus SARS-CoV-2 neutralization assay. Heat inactivation of human convalescent serum was shown to inactivate complement proteins, and the contribution of complement in SARS-CoV-2 neutralization was often >50% of the neutralizing activity of human sera without heat inactivation and could account for neutralizing activity when standard titers were zero after heat inactivation. This effect was also observed in COVID-19 vaccinees and could be abolished in individuals who were undergoing treatment with therapeutic anti-complement Abs. Complement activity was mainly dependent on the classical pathway with little contributions from mannose-binding lectin and alternative pathways. Our study demonstrates the importance of the complement pathway in significantly increasing viral neutralization activity against SARS-CoV-2 in spike seropositive individuals.
Collapse
Affiliation(s)
- Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Serena L L Chau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arinjay Banerjee
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Furkan Guvenc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Reuben Samson
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Queenie Hu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Lindsey Fiddes
- Microscopy Imaging Lab, University of Toronto, Toronto, Ontario, Canada
| | - Laurie Seifried
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Gary Chao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Megan Buchholz
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Antonio Estacio
- Keenan Research Centre for Biomedical Science of St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Patti Lou Cheatley
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Katerina Pavenski
- Apheresis Unit, Kidney and Metabolism Program, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
- Department of Laboratory Medicine, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Christopher J Patriquin
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Yanling Liu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Kimberly Crasta
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - FengYun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Maria D Pasic
- Department of Immunology, Unity Health Toronto, Toronto, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | | | - Götz R A Ehrhardt
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science of St Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Pan Y, Li D, Zhang Z, Xu J, Sun X, Wang M, Ding J. Clinical characteristics and outcomes of maintenance hemodialysis patients with COVID-19 during the Omicron wave of the pandemic in Beijing: a single center retrospective study. BMC Nephrol 2024; 25:143. [PMID: 38649863 PMCID: PMC11036696 DOI: 10.1186/s12882-024-03575-1] [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: 09/17/2023] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The clinical manifestations and prognosis of hemodialysis patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) during the Omicron wave of the pandemic infection were still unclear. This study investigated the clinical characteristics of patients undergoing maintenance hemodialysis (MHD) infected with it. METHODS This retrospective single-center study included 151 patients undergoing MHD. Healthcare workers were selected as control group were assessed from December 1, 2022 to March 31, 2023. Clinical data, laboratory test results, treatment protocols, and prognoses were collected and analyzed. RESULTS The study population included 146 patients with MHD, 93 (63.7%) of whom were infected with SARS-CoV-2. The number of non-severe, severe, and critical cases was 84 (90.3%), 4 (4.3%), and 5 (5.3%), respectively. Six patients (6.5%) died during the study period. The main symptoms of SARS-CoV-2 infection, including fever, cough, and fatigue, were less common in patients with MHD than the controls. During SARS-CoV-2 infection, the C-reactive protein (2.9 vs. 11.8 mg/dl, p < 0.0001) and ferritin levels(257.7 vs. 537 ng/l, p < 0.0001) were elevated. The hemoglobin(113vs 111 g/L, p = 0.0001) and albumin levels(39.4 vs. 36.1 g/L, p < 0.0001) decreased. Generally, it took two months for the hemoglobin levels to recover. Positivity rate for SARS-COV-2 serum immunoglobin G (IgG) antibodies and IgG titers were lower in dialysis patients than the controls. Age was positively associated with disease severity, while age and hyponatremia were associated with death. CONCLUSIONS Patients with MHD and COVID-19 were primarily classified as non-severe. SARS-CoV-2 infection would soon lead to the increase of inflammation related acute response protein in dialysis patients, and then lead to the decrease of hemoglobin and albumin. About 9.6% in HD patients were severe cases and had poor prognosis. Advanced age and hyponatremia were associated with disease severity and prognosis.
Collapse
Affiliation(s)
- Yujing Pan
- Nephrology Department, Peking University International Hospital, No. 1 Life Park Road, Life Science Park of Zhongguancun, Changping District, 102206, Beijing, P.R. China
| | - Dan Li
- Hemodialysis Center, Peking University International Hospital, 102206, Beijing, P.R. China
| | - Zhoucang Zhang
- Nephrology Department, Peking University International Hospital, No. 1 Life Park Road, Life Science Park of Zhongguancun, Changping District, 102206, Beijing, P.R. China
| | - Jing Xu
- Nephrology Department, Peking University International Hospital, No. 1 Life Park Road, Life Science Park of Zhongguancun, Changping District, 102206, Beijing, P.R. China
| | - Xinping Sun
- Department of Clinical Laboratory, Peking University International Hospital, 102206, Beijing, P.R. China
| | - Mei Wang
- Nephrology Department, Peking University International Hospital, No. 1 Life Park Road, Life Science Park of Zhongguancun, Changping District, 102206, Beijing, P.R. China
| | - Jiaxiang Ding
- Nephrology Department, Peking University International Hospital, No. 1 Life Park Road, Life Science Park of Zhongguancun, Changping District, 102206, Beijing, P.R. China.
| |
Collapse
|
6
|
Bogacheva MS, Kuivanen S, Potdar S, Hassinen A, Huuskonen S, Pöhner I, Luck TJ, Turunen L, Feodoroff M, Szirovicza L, Savijoki K, Saarela J, Tammela P, Paavolainen L, Poso A, Varjosalo M, Kallioniemi O, Pietiäinen V, Vapalahti O. Drug repurposing platform for deciphering the druggable SARS-CoV-2 interactome. Antiviral Res 2024; 223:105813. [PMID: 38272320 DOI: 10.1016/j.antiviral.2024.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has heavily challenged the global healthcare system. Despite the vaccination programs, the new virus variants are circulating. Further research is required for understanding of the biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and for discovery of therapeutic agents against the virus. Here, we took advantage of drug repurposing to identify if existing drugs could inhibit SARS-CoV-2 infection. We established an open high throughput platform for in vitro screening of drugs against SARS-CoV-2 infection. We screened ∼1000 drugs for their ability to inhibit SARS-CoV-2-induced cell death in the African green monkey kidney cell line (Vero-E6), analyzed how the hit compounds affect the viral N (nucleocapsid) protein expression in human cell lines using high-content microscopic imaging and analysis, determined the hit drug targets in silico, and assessed their ability to cause phospholipidosis, which can interfere with the viral replication. Duvelisib was found by in silico interaction assay as a potential drug targeting virus-host protein interactions. The predicted interaction between PARP1 and S protein, affected by Duvelisib, was further validated by immunoprecipitation. Our results represent a rapidly applicable platform for drug repurposing and evaluation of the new emerging viruses' responses to the drugs. Further in silico studies help us to discover the druggable host pathways involved in the infectious cycle of SARS-CoV-2.
Collapse
Affiliation(s)
- Mariia S Bogacheva
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland.
| | - Suvi Kuivanen
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Hassinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sini Huuskonen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ina Pöhner
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tamara J Luck
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Leonora Szirovicza
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Kirsi Savijoki
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Päivi Tammela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Poso
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Department of Internal Medicine VIII, University Hospital Tubingen, Tubingen, Germany
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Science for Life Laboratory (SciLifeLab), Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland; HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
7
|
Jokiranta ST, Miettinen S, Salonen S, Kareinen L, Uusitalo R, Korhonen EM, Virtanen J, Kivistö I, Aaltonen K, Mosselhy DA, Lääveri T, Kantele A, Arstila TP, Jarva H, Vapalahti O, Heinonen S, Kekäläinen E. Stable Levels of Antibodies Against Unrelated Toxoid Vaccines After COVID-19: COVID-19 Infection Does Not Affect Toxoid Vaccine Antibody Levels. Pathog Immun 2024; 8:74-87. [PMID: 38347963 PMCID: PMC10860543 DOI: 10.20411/pai.v8i2.627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024] Open
Abstract
Background Lymphopenia is common in COVID-19. This has raised concerns that COVID-19 could affect the immune system akin to measles infection, which causes immune amnesia and a reduction in protective antibodies. Methods We recruited COVID-19 patients (n = 59) in Helsinki, Finland, and collected plasma samples on 2 to 3 occasions during and after infection. We measured IgG antibodies to diphtheria toxin, tetanus toxoid, and pertussis toxin, along with total IgG, SARS-CoV-2 spike protein IgG, and neutralizing antibodies. We also surveyed the participants for up to 17 months for long-term impaired olfaction as a proxy for prolonged post-acute COVID-19 symptoms. Results No significant differences were found in the unrelated vaccine responses while the serological response against COVID-19 was appropriate. During the acute phase of the disease, the SARSCoV-2 IgG levels were lower in outpatients when compared to inpatients. SARS-CoV-2 serology kinetics matched expectations. In the acute phase, anti-tetanus and anti-diphtheria IgG levels were lower in patients with prolonged impaired olfaction during follow up than in those without. Conclusions We could not detect significant decline in overall humoral immunity during or after COVID-19 infection. In severe COVID-19, there appears to be a temporary decline in total IgG levels.
Collapse
Affiliation(s)
- Suvi T. Jokiranta
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Simo Miettinen
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Sami Salonen
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Lauri Kareinen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Finnish Food Authority, Helsinki, Finland
| | - Ruut Uusitalo
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Essi M. Korhonen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Virtanen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilkka Kivistö
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Aaltonen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dina A. Mosselhy
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Microbiological Unit, Fish Diseases Department, Animal Health Research Institute, ARC, Dokki, Giza 12618, Egypt
| | - Tinja Lääveri
- Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Aalto University, Department of Computer Science, School of Science, Espoo, Finland
| | - Anu Kantele
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Department of Infectious Diseases, University of Helsinki, Helsinki, Finland
| | - T. Petteri Arstila
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanna Jarva
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Santtu Heinonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
8
|
Shroff S, Haapakoski M, Tapio K, Laajala M, Leppänen M, Plavec Z, Haapala A, Butcher SJ, Ihalainen JA, Toppari JJ, Marjomäki V. Antiviral action of a functionalized plastic surface against human coronaviruses. Microbiol Spectr 2024; 12:e0300823. [PMID: 38226803 PMCID: PMC10846231 DOI: 10.1128/spectrum.03008-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/16/2023] [Indexed: 01/17/2024] Open
Abstract
Viruses may persist on solid surfaces for long periods, which may contribute to indirect transmission. Thus, it is imperative to develop functionalized surfaces that will lower the infectious viral load in everyday life. Here, we have tested a plastic surface functionalized with tall oil rosin against the seasonal human coronavirus OC43 as well as severe acute respiratory syndrome coronavirus 2. All tested non-functionalized plastic surfaces showed virus persistence up to 48 h. In contrast, the functionalized plastic showed good antiviral action already within 15 min of contact and excellent efficacy after 30 min over 90% humidity. Excellent antiviral effects were also observed at lower humidities of 20% and 40%. Despite the hydrophilic nature of the functionalized plastic, viruses did not adhere strongly to it. According to helium ion microscopy, viruses appeared flatter on the rosin-functionalized surface, but after flushing away from the rosin-functionalized surface, they showed no apparent structural changes when imaged by transmission electron microscopy of cryogenic or negatively stained specimens or by atomic force microscopy. Flushed viruses were able to bind to their host cell surface and enter endosomes, suggesting that the fusion with the endosomal membrane was halted. The eluted rosin from the functionalized surface demonstrated its ability to inactivate viruses, indicating that the antiviral efficacy relied on the active leaching of the antiviral substances, which acted on the viruses coming into contact. The rosin-functionalized plastic thus serves as a promising candidate as an antiviral surface for enveloped viruses.IMPORTANCEDuring seasonal and viral outbreaks, the implementation of antiviral plastics can serve as a proactive strategy to limit the spread of viruses from contaminated surfaces, complementing existing hygiene practices. In this study, we show the efficacy of a rosin-functionalized plastic surface that kills the viral infectivity of human coronaviruses within 15 min of contact time, irrespective of the humidity levels. In contrast, non-functionalized plastic surfaces retain viral infectivity for an extended period of up to 48 h. The transient attachment on the surface or the leached active components do not cause major structural changes in the virus or prevent receptor binding; instead, they effectively block viral infection at the endosomal stage.
Collapse
Affiliation(s)
- Sailee Shroff
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Marjo Haapakoski
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kosti Tapio
- Department of Physics, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Mira Laajala
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Miika Leppänen
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Zlatka Plavec
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Antti Haapala
- Sustainable Technologies group, Department of Chemistry, University of Eastern Finland, Joensuu, Finland
- FSCN Research Centre, Mid Sweden University, Sundsvall, Sweden
| | - Sarah J. Butcher
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Janne A. Ihalainen
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - J. Jussi Toppari
- Department of Physics, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
9
|
Virtanen J, Korhonen EM, Salonen S, Vapalahti O, Sironen T, Jääskeläinen AJ. SARS-CoV-2 infections among pregnant women, 2020, Finland-Cross-testing of neutralization assays. J Med Virol 2024; 96:e29415. [PMID: 38293724 DOI: 10.1002/jmv.29415] [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: 09/08/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
We studied the development of the severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) pandemic in southern Finland in 2020 and evaluated the performance of two surrogate immunoassays for the detection of neutralizing antibodies (NAbs). The data set consisted of 12 000 retrospectively collected samples from pregnant women in their first trimester throughout 2020. All the samples were initially screened for immunoglobulin G (IgG) with SARS-CoV-2 spike antibody assay (EIM-S1, Euroimmun) followed by confirmation with nucleocapsid antibody assay (Architect SARS-CoV-2, Abbott). Samples that were reactive (positive or borderline) with both assays were subjected to testing with commercial surrogate immunoassays of NeutraLISA (EIM) and cPassTM (GenScript Biotech Corporation) by using pseudoneutralization assay (PNAbA) as a golden standard. No seropositive cases were detected between January and March. Between April and December, IgG (EIM-S1 and Abbott positive) and NAb (PNAbA positive) seroprevalences were between 0.4% and 1.4%. NeutraLISA showed 90% and cPass 55% concordant results with PNAbA among PNAbA negative samples and 49% and 92% among PNAbA positive samples giving NeutraLISA better specificity but lower sensitivity than cPass. To conclude, seroprevalence in pregnant women reflected that of the general population but the variability of the performance of serological protocols needs to be taken into account in inter-study comparison.
Collapse
Affiliation(s)
- Jenni Virtanen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Essi M Korhonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Sami Salonen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tarja Sironen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Anne J Jääskeläinen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
10
|
Saidarakath J, Joravarsinh Gadhavi B, Osman Awad M, AlSaadi MM, Chovar Kattil M, Alnuaimi AS. Anti-SARS-CoV-2 Antibody Response Among Spectators of Amir Cup 2020 With a History of Recovery From COVID-19 in Qatar: A Historic Cohort Study. Cureus 2024; 16:e54406. [PMID: 38505461 PMCID: PMC10948953 DOI: 10.7759/cureus.54406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
Aim The aim of the study is to describe the antibody response after COVID-19 infection and assess its effectiveness against reinfection. Background COVID-19 has recently emerged as a contagious infectious disease caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This infection is followed by a humoral immune antibody response, which may remain in the blood for a number of weeks. Studies have shown that antibodies protect against reinfection for at least seven months. The current study is aimed at investigating the persistence of circulating SARS-CoV-2 antibodies after COVID-19 infection and its behavior over 18 months of follow-up period, in addition to assessing the risk of reinfection of COVID-19 in unvaccinated individuals. Methodology A longitudinal historical cohort study of 3378 COVID-19 recovered individuals in connection with the Amir Cup football tournament held in Qatar, in December 2020 was analyzed. The health records of study participants were followed for a maximum of 18 months after serology testing or until the first dose of COVID-19 vaccination to detect any evidence of recurrent infection. Results The study found a statistically significant association between recurrence risk and the duration of risk exposure since the first COVID-19 episode. Compared to those with the lowest risk of exposure to reinfection (shortest duration after first infection) those beyond 299 days of at-risk exposure since the first episode, have a 51-fold higher risk of developing recurrent COVID-19. Conclusion Immunity developed after primary infection with SARS-CoV-2 may protect against reinfection from subsequent exposure to the virus in seropositive individuals up to nine months post-infection.
Collapse
|
11
|
Kantele A, Paajanen J, Pietilä JP, Vapalahti O, Pakkanen SH, Lääveri T. Long COVID-associated symptoms prevalent in both SARS-CoV-2 positive and negative individuals: A prospective follow-up study. New Microbes New Infect 2024; 56:101209. [PMID: 38174103 PMCID: PMC10761764 DOI: 10.1016/j.nmni.2023.101209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/03/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
Background Research into persistent symptoms among SARS-CoV-2-positive i.e. CoV(+) patients mostly focuses on hospitalized individuals. Our prospective follow-up study compares long COVID-associated symptoms among laboratory-confirmed CoV(+) and SARS-CoV-2 negative [CoV(-)] individuals. Methods SARS-CoV-2 RT-PCR-tested volunteers were recruited into four cohorts: 1) CoV(+) outpatients, 2) CoV(-) outpatients, 3) CoV(+) intensive care unit (ICU) inpatients, and 4) CoV(+) non-ICU inpatients. Neutralizing antibodies were assessed and questionnaires filled in at enrolment and days 90-120, 121-180, 181-270, 271-365, and 365-533. Results Of the 1326 participants, 1191 were CoV(+): 46 ICU, 123 non-ICU, and 1022 outpatients; 135 were CoV(-) outpatient controls. Both CoV(+) outpatients and CoV(-) controls showed high overall symptom rates at all time points. More prevalent among CoV(+) than CoV(-) outpatients were only impaired olfaction and taste; many others proved more frequent for CoV(-) participants. At ≥181 days, fatigue, dyspnoea, various neuropsychological symptoms and several others were recorded more often for CoV(+) inpatients than outpatients. Conclusions Long COVID-associated symptoms were more frequent among hospitalized than non-hospitalized CoV(+) participants. As for outpatients, only impaired olfaction and taste showed higher rates in the CoV(+) group; some symptoms proved even more common among those CoV(-). Besides suggesting low long COVID prevalences for outpatients, our results highlight the weight of negative controls.
Collapse
Affiliation(s)
- Anu Kantele
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Biomedicum 1, Haartmaninkatu 8, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
- Department of Infectious Diseases, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, Haartmaninkatu 4, 00014, University of Helsinki, Helsinki, Finland
| | - Juuso Paajanen
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Biomedicum 1, Haartmaninkatu 8, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, Haartmaninkatu 4, 00014, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Viral Zoonoses Research Unit, Departments of Virology and Veterinary Biosciences, Faculties of Medicine and Veterinary Medicine, 00014, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Diagnostic Center, HUSLAB, Clinical Microbiology, Topeliuksenkatu 32, 00290, Helsinki, Finland
| | - Sari H. Pakkanen
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Biomedicum 1, Haartmaninkatu 8, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, Haartmaninkatu 4, 00014, University of Helsinki, Helsinki, Finland
| | - Tinja Lääveri
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Biomedicum 1, Haartmaninkatu 8, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
- Department of Infectious Diseases, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, Haartmaninkatu 4, 00014, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Diagnostic Center, HUSLAB, Clinical Microbiology, Topeliuksenkatu 32, 00290, Helsinki, Finland
- Department of Computer Science, Aalto University, PO Box 15400, FI-00076, AALTO, Finland
| |
Collapse
|
12
|
Haapakoski M, Emelianov A, Reshamwala D, Laajala M, Tienaho J, Kilpeläinen P, Liimatainen J, Jyske T, Pettersson M, Marjomäki V. Antiviral functionalization of cellulose using tannic acid and tannin-rich extracts. Front Microbiol 2023; 14:1287167. [PMID: 38125579 PMCID: PMC10731304 DOI: 10.3389/fmicb.2023.1287167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Due to seasonally appearing viruses and several outbreaks and present pandemic, we are surrounded by viruses in our everyday life. In order to reduce viral transmission, functionalized surfaces that inactivate viruses are in large demand. Here the endeavor was to functionalize cellulose-based materials with tannic acid (TA) and tannin-rich extracts by using different binding polymers to prevent viral infectivity of both non-enveloped coxsackievirus B3 (CVB3) and enveloped human coronavirus OC43 (HCoV-OC43). Direct antiviral efficacy of TA and spruce bark extract in solution was measured: EC50 for CVB3 was 0.12 and 8.41 μg/ml and for HCoV-OC43, 78.16 and 95.49 μg/ml, respectively. TA also led to an excellent 5.8- to 7-log reduction of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infectivity. TA functionalized materials reduced infectivity already after 5-min treatment at room temperature. All the tested methods to bind TA showed efficacy on paperboard with 0.1 to 1% (w/v) TA concentrations against CVB3 whereas material hydrophobicity decreased activities. Specific signatures for TA and HCoV-OC43 were discovered by Raman spectroscopy and showed clear co-localization on the material. qPCR study suggested efficient binding of CVB3 to the TA functionalized cellulose whereas HCoV-OC43 was flushed out from the surfaces more readily. In conclusion, the produced TA-materials showed efficient and broadly acting antiviral efficacy. Additionally, the co-localization of TA and HCoV-OC43 and strong binding of CVB3 to the functionalized cellulose demonstrates an interaction with the surfaces. The produced antiviral surfaces thus show promise for future use to increase biosafety and biosecurity by reducing pathogen persistence.
Collapse
Affiliation(s)
- Marjo Haapakoski
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Aleksei Emelianov
- Department of Chemistry/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Dhanik Reshamwala
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Mira Laajala
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jenni Tienaho
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Petri Kilpeläinen
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Jaana Liimatainen
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Tuula Jyske
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Mika Pettersson
- Department of Chemistry/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
13
|
Liang Z, Wu X, Wu J, Liu S, Tong J, Li T, Yu Y, Zhang L, Zhao C, Lu Q, Qin H, Nie J, Huang W, Wang Y. Development of an automated, high-throughput SARS-CoV-2 neutralization assay based on a pseudotyped virus using a vesicular stomatitis virus (VSV) vector. Emerg Microbes Infect 2023; 12:e2261566. [PMID: 37727107 PMCID: PMC10540657 DOI: 10.1080/22221751.2023.2261566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/17/2023] [Indexed: 09/21/2023]
Abstract
ABSTRACTThe global outbreak of COVID-19 has caused a severe threat to human health; therefore, simple, high-throughput neutralization assays are desirable for developing vaccines and drugs against COVID-19. In this study, a high-titre SARS-CoV-2 pseudovirus was successfully packaged by truncating the C-terminus of the SARS-CoV-2 spike protein by 21 amino acids and infecting 293 T cells that had been stably transfected with the angiotensin-converting enzyme 2 (ACE2) receptor and furin (named AF cells), to establish a simple, high-throughput, and automated 384-well plate neutralization assay. The method was optimized for cell amount, virus inoculation, incubation time, and detection time. The automated assay showed good sensitivity, accuracy, reproducibility, Z' factor, and a good correlation with the live virus neutralization assay. The high-throughput approach would make it available for the SARS-CoV-2 neutralization test in large-scale clinical trials and seroepidemiological surveys which would aid the accelerated vaccine development and evaluation.
Collapse
Affiliation(s)
- Ziteng Liang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Xi Wu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Jiajing Wu
- Beijing Yunling Biotechnology Co., Ltd., Beijing, People’s Republic of China
| | - Shuo Liu
- Changping Laboratory, Beijing, People’s Republic of China
| | - Jincheng Tong
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Tao Li
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Yuanling Yu
- Changping Laboratory, Beijing, People’s Republic of China
| | - Li Zhang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Qiong Lu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Haiyang Qin
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People’s Republic of China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| |
Collapse
|
14
|
Reshamwala D, Shroff S, Liimatainen J, Tienaho J, Laajala M, Kilpeläinen P, Viherä-Aarnio A, Karonen M, Jyske T, Marjomäki V. Willow ( Salix spp.) bark hot water extracts inhibit both enveloped and non-enveloped viruses: study on its anti-coronavirus and anti-enterovirus activities. Front Microbiol 2023; 14:1249794. [PMID: 38029113 PMCID: PMC10663278 DOI: 10.3389/fmicb.2023.1249794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/18/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Recurring viral outbreaks have a significant negative impact on society. This creates a need to develop novel strategies to complement the existing antiviral approaches. There is a need for safe and sustainable antiviral solutions derived from nature. Objective This study aimed to investigate the antiviral potential of willow (Salix spp.) bark hot water extracts against coronaviruses and enteroviruses. Willow bark has long been recognized for its medicinal properties and has been used in traditional medicines. However, its potential as a broad-spectrum antiviral agent remains relatively unexplored. Methods Cytopathic effect inhibition assay and virucidal and qPCR-based assays were used to evaluate the antiviral potential of the bark extracts. The mechanism of action was investigated using time-of-addition assay, confocal microscopy, TEM, thermal, and binding assays. Extracts were fractionated and screened for their chemical composition using high-resolution LC-MS. Results The native Salix samples demonstrated their excellent antiviral potential against the non-enveloped enteroviruses even at room temperature and after 45 s. They were equally effective against the seasonal and pandemic coronaviruses. Confocal microscopy verified the loss of infection capacity by negligible staining of the newly synthesized capsid or spike proteins. Time-of-addition studies demonstrated that Salix bark extract had a direct effect on the virus particles but not through cellular targets. Negative stain TEM and thermal assay showed that antiviral action on enteroviruses was based on the added stability of the virions. In contrast, Salix bark extract caused visible changes in the coronavirus structure, which was demonstrated by the negative stain TEM. However, the binding to the cells was not affected, as verified by the qPCR study. Furthermore, coronavirus accumulated in the cellular endosomes and did not proceed after this stage, based on the confocal studies. None of the tested commercial reference samples, such as salicin, salicylic acid, picein, and triandrin, had any antiviral activity. Fractionation of the extract and subsequent MS analysis revealed that most of the separated fractions were very effective against enteroviruses and contained several different chemical groups such as hydroxycinnamic acid derivatives, flavonoids, and procyanidins. Conclusion Salix spp. bark extracts contain several virucidal agents that are likely to act synergistically and directly on the viruses.
Collapse
Affiliation(s)
- Dhanik Reshamwala
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Sailee Shroff
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | | | - Jenni Tienaho
- Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Mira Laajala
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | | | | | - Maarit Karonen
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Tuula Jyske
- Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
15
|
Pietiäinen V, Polso M, Migh E, Guckelsberger C, Harmati M, Diosdi A, Turunen L, Hassinen A, Potdar S, Koponen A, Sebestyen EG, Kovacs F, Kriston A, Hollandi R, Burian K, Terhes G, Visnyovszki A, Fodor E, Lacza Z, Kantele A, Kolehmainen P, Kakkola L, Strandin T, Levanov L, Kallioniemi O, Kemeny L, Julkunen I, Vapalahti O, Buzas K, Paavolainen L, Horvath P, Hepojoki J. Image-based and machine learning-guided multiplexed serology test for SARS-CoV-2. CELL REPORTS METHODS 2023; 3:100565. [PMID: 37671026 PMCID: PMC10475844 DOI: 10.1016/j.crmeth.2023.100565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023]
Abstract
We present a miniaturized immunofluorescence assay (mini-IFA) for measuring antibody response in patient blood samples. The method utilizes machine learning-guided image analysis and enables simultaneous measurement of immunoglobulin M (IgM), IgA, and IgG responses against different viral antigens in an automated and high-throughput manner. The assay relies on antigens expressed through transfection, enabling use at a low biosafety level and fast adaptation to emerging pathogens. Using severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the model pathogen, we demonstrate that this method allows differentiation between vaccine-induced and infection-induced antibody responses. Additionally, we established a dedicated web page for quantitative visualization of sample-specific results and their distribution, comparing them with controls and other samples. Our results provide a proof of concept for the approach, demonstrating fast and accurate measurement of antibody responses in a research setup with prospects for clinical diagnostics.
Collapse
Affiliation(s)
- Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Minttu Polso
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Ede Migh
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Christian Guckelsberger
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Computer Science, Aalto University, Espoo, Finland
- Finnish Center for Artificial Intelligence, Espoo, Finland
- School of Electronic Engineering and Computer Science, Queen Mary University of London, London, UK
| | - Maria Harmati
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Akos Diosdi
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Hassinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Annika Koponen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Edina Gyukity Sebestyen
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Ferenc Kovacs
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Single-Cell Technologies Ltd., Szeged, Hungary
| | - Andras Kriston
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Single-Cell Technologies Ltd., Szeged, Hungary
| | - Reka Hollandi
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Katalin Burian
- Department of Medical Microbiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gabriella Terhes
- Department of Medical Microbiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Adam Visnyovszki
- 1 Department of Internal Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Eszter Fodor
- Department of Sports Physiology, Institute of Sports and Health Sciences, University of Physical Education, Budapest, Hungary
| | - Zsombor Lacza
- Department of Sports Physiology, Institute of Sports and Health Sciences, University of Physical Education, Budapest, Hungary
| | - Anu Kantele
- Meilahti Infectious Diseases and Vaccine Research Center (MeiVac), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | | | - Laura Kakkola
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tomas Strandin
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Lev Levanov
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Lajos Kemeny
- HCEMM-USZ Skin Research Group, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Turku University Hospital, Turku, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Krisztina Buzas
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Department of Immunology, Faculty of Medicine, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Peter Horvath
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Single-Cell Technologies Ltd., Szeged, Hungary
| | - Jussi Hepojoki
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
- University of Zurich, Vetsuisse Faculty, Institute of Veterinary Pathology, Zürich, Switzerland
| |
Collapse
|
16
|
Stoto MA, Reno C, Tsolova S, Fantini MP. The European experience with testing and surveillance during the first phase of the COVID-19 pandemic. Global Health 2023; 19:51. [PMID: 37480125 PMCID: PMC10362750 DOI: 10.1186/s12992-023-00950-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/30/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND COVID-19 pandemic provides a unique opportunity to learn the challenges encountered by public health emergency preparedness systems, both in terms of problems encountered and adaptations during and after the first wave, as well as successful responses to them. RESULTS This work draws on published literature, interviews with countries and institutional documents as part of a European Centre for Disease Prevention and Control project that aims to identify the implications for preparedness measurement derived from COVID-19 pandemic experience in order to advance future preparedness efforts in European Union member states. The analysis focused on testing and surveillance themes and five countries were considered, namely Italy, Germany, Finland, Spain and Croatia. Our analysis shown that a country's ability to conduct testing at scale was critical, especially early in the pandemic, and the inability to scale up testing operations created critical issues for public health operations such as contact tracing. Countries were required to develop new strategies, approaches, and policies under pressure and to review and revise them as the pandemic evolved, also considering that public health systems operate at the national, regional, and local level with respect to testing, contact tracing, and surveillance, and involve both government agencies as well as private organizations. Therefore, communication among multiple public and private entities at all levels and coordination of the testing and surveillance activities was critical. CONCLUSION With regard to testing and surveillance, three capabilities that were essential to the COVID-19 response in the first phase, and presumably in other public health emergencies: the ability to scale-up testing, contact tracing, surveillance efforts; flexibility to develop new strategies, approaches, and policies under pressure and to review and revise them as the pandemic evolved; and the ability to coordinate and communicate in complex public health systems that operate at the national, regional, and local level with respect and involve multiple government agencies as well as private organizations.
Collapse
Affiliation(s)
- Michael A Stoto
- Department of Health Management and Policy, Georgetown University, Washington, D.C, United States of America.
- Harvard T.H. Chan School of Public Health, Boston, United States.
| | - Chiara Reno
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, Bologna, 40126, Italy
- Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| | - Svetla Tsolova
- European Centre for Disease prevention and Control (ECDC), Solna, Sweden
| | - Maria Pia Fantini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, Bologna, 40126, Italy
| |
Collapse
|
17
|
Nieminen TA, Auranen K, Kulathinal S, Härkänen T, Melin M, Palmu AA, Jokinen J. Underreporting of SARS-CoV-2 infections during the first wave of the 2020 COVID-19 epidemic in Finland-Bayesian inference based on a series of serological surveys. PLoS One 2023; 18:e0282094. [PMID: 37352274 PMCID: PMC10289354 DOI: 10.1371/journal.pone.0282094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/06/2023] [Indexed: 06/25/2023] Open
Abstract
In Finland, the first wave of the COVID-19 epidemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) took place from March to June 2020, with the majority of COVID-19 cases diagnosed in the Helsinki-Uusimaa region. The magnitude and trend in the incidence of COVID-19 is one way to monitor the course of the epidemic. The diagnosed COVID-19 cases are a subset of the infections and therefore the COVID-19 incidence underestimates the SARS-CoV-2 incidence. The likelihood that an individual with SARS-CoV-2 infection is diagnosed with COVID-19 depends on the clinical manifestation as well as the infection testing policy and capacity. These factors may fluctuate over time and the underreporting of infections changes accordingly. Quantifying the extent of underreporting allows the assessment of the true incidence of infection. To obtain information on the incidence of SARS-CoV-2 infection in Finland, a series of serological surveys was initiated in April 2020. We develop a Bayesian inference approach and apply it to data from the serological surveys, registered COVID-19 cases, and external data on antibody development, to estimate the time-dependent underreporting of SARS-Cov-2 infections during the first wave of the COVID-19 epidemic in Finland. During the entire first wave, there were 1 to 5 (95% probability) SARS-CoV-2 infections for every COVID-19 case. The underreporting was highest before April when there were 4 to 17 (95% probability) infections for every COVID-19 case. It is likely that between 0.5%-1.0% (50% probability) and no more than 1.5% (95% probability) of the adult population in the Helsinki-Uusimaa region were infected with SARS-CoV-2 by the beginning of July 2020.
Collapse
Affiliation(s)
- Tuomo A. Nieminen
- Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | | | | | - Tommi Härkänen
- Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | - Merit Melin
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Arto A. Palmu
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jukka Jokinen
- Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Belik M, Liedes O, Vara S, Haveri A, Pöysti S, Kolehmainen P, Maljanen S, Huttunen M, Reinholm A, Lundberg R, Skön M, Österlund P, Melin M, Hänninen A, Hurme A, Ivaska L, Tähtinen PA, Lempainen J, Kakkola L, Jalkanen P, Julkunen I. Persistent T cell-mediated immune responses against Omicron variants after the third COVID-19 mRNA vaccine dose. Front Immunol 2023; 14:1099246. [PMID: 36756112 PMCID: PMC9899862 DOI: 10.3389/fimmu.2023.1099246] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/02/2023] [Indexed: 01/24/2023] Open
Abstract
Introduction The prime-boost COVID-19 mRNA vaccination strategy has proven to be effective against severe COVID-19 disease and death. However, concerns have been raised due to decreasing neutralizing antibody levels after COVID-19 vaccination and due to the emergence of new immuno-evasive SARS-CoV-2 variants that may require additional booster vaccinations. Methods In this study, we analyzed the humoral and cell-mediated immune responses against the Omicron BA.1 and BA.2 subvariants in Finnish healthcare workers (HCWs) vaccinated with three doses of COVID-19 mRNA vaccines. We used enzyme immunoassay and microneutralization test to analyze the levels of SARS-CoV-2 specific IgG antibodies in the sera of the vaccinees and the in vitro neutralization capacity of the sera. Activation induced marker assay together with flow cytometry and extracellular cytokine analysis was used to determine responses in SARS-CoV-2 spike protein stimulated PBMCs. Results Here we show that within the HCWs, the third mRNA vaccine dose recalls both humoral and T cell-mediated immune responses and induces high levels of neutralizing antibodies against Omicron BA.1 and BA.2 variants. Three weeks after the third vaccine dose, SARS-CoV-2 wild type spike protein-specific CD4+ and CD8+ T cells are observed in 82% and 71% of HCWs, respectively, and the T cells cross-recognize both Omicron BA.1 and BA.2 spike peptides. Although the levels of neutralizing antibodies against Omicron BA.1 and BA.2 decline 2.5 to 3.8-fold three months after the third dose, memory CD4+ T cell responses are maintained for at least eight months post the second dose and three months post the third vaccine dose. Discussion We show that after the administration of the third mRNA vaccine dose the levels of both humoral and cell-mediated immune responses are effectively activated, and the levels of the spike-specific antibodies are further elevated compared to the levels after the second vaccine dose. Even though at three months after the third vaccine dose antibody levels in sera decrease at a similar rate as after the second vaccine dose, the levels of spike-specific CD4+ and CD8+ T cells remain relatively stable. Additionally, the T cells retain efficiency in cross-recognizing spike protein peptide pools derived from Omicron BA.1 and BA.2 subvariants. Altogether our results suggest durable cellmediated immunity and protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Milja Belik
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Oona Liedes
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Saimi Vara
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anu Haveri
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Sakari Pöysti
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | | | - Sari Maljanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Moona Huttunen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arttu Reinholm
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Marika Skön
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Pamela Österlund
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Merit Melin
- Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Arno Hänninen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Antti Hurme
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Infectious Diseases, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, Lapland Central Hospital, Rovaniemi, Finland
| | - Lauri Ivaska
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Paula A. Tähtinen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Johanna Lempainen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Laura Kakkola
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pinja Jalkanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| |
Collapse
|
19
|
Yilmaz A, Turan N, Kocazeybek BS, Dinc HO, Tali HE, Aydin O, Tali HB, Yilmaz SG, Konukoglu D, Borekci S, Bold D, Roman Sosa G, Gungordu N, Vardaloglu I, Gareayaghi N, Guzel M, Guner E, Sadeyen JR, Chang P, Iqbal M, Richt JA, Yilmaz H. Development of in House ELISAs to Detect Antibodies to SARS-CoV-2 in Infected and Vaccinated Humans by Using Recombinant S, S1 and RBD Proteins. Diagnostics (Basel) 2022; 12:3085. [PMID: 36553092 PMCID: PMC9777145 DOI: 10.3390/diagnostics12123085] [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: 10/19/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: The aim of this study was to produce in-house ELISAs which can be used to determine SARS-CoV-2-specific antibody levels directed against the spike protein (S), the S1 subunit of S and the receptor binding domain (RBD) of S in SARS-CoV-2 vaccinated and infected humans. (2) Methods: Three in-house ELISAs were developed by using recombinant proteins of SARS-CoV-2, namely the S, S1 and RBD proteins. Specificity and sensitivity evaluations of these tests were performed using sera from SARS-CoV-2-infected (n = 70) and SARS-CoV-2-vaccinated (n = 222; CoronaVac vaccine) humans in Istanbul, Turkey. The analyses for the presence of SARS-CoV-2-specific antibodies were performed using the in-house ELISAs, a commercial ELISA (Abbott) and a commercial surrogate virus neutralization test (sVNT). We also analyzed archival human sera (n = 50) collected before the emergence of COVID-19 cases in Turkey. (3) Results: The sensitivity of the in-house S, S1 and RBD ELISAs was found to be 88.44, 90.17 and 95.38%, while the specificity was 72.27, 89.08 and 89.92%, respectively, when compared to the commercial SARS-CoV-2 antibody test kit. The area under curve (AUC) values were 0.777 for the in-house S ELISA, 0.926 for the S1 ELISA, and 0.959 for the RBD ELISA. The kappa values were 0.62, 0.79 and 0.86 for the S, S1 and RBD ELISAs, respectively. (4) Conclusions: The in-house S1 and RBD ELISAs developed in this study have acceptable performance characteristics in terms of sensitivity, specificity, AUC and kappa values. In particular, the RBD ELISA seems viable to determine SARS-CoV-2-specific antibody levels, both in infected and vaccinated people, and help mitigate SARS-CoV-2 outbreaks and spread.
Collapse
Affiliation(s)
- Aysun Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Nuri Turan
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Bekir Sami Kocazeybek
- Department of Medical Microbiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Harika Oyku Dinc
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul 34098, Turkey
| | - Hasan Emre Tali
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Ozge Aydin
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Hamid Besim Tali
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Semaha Gul Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
| | - Dildar Konukoglu
- Department of Biocehmistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Sermin Borekci
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Dashzeveg Bold
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Gleyder Roman Sosa
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Nejdiye Gungordu
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
- Department of Occupational Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Ilgim Vardaloglu
- Department of Pulmonary Diseases, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey
| | - Nesrin Gareayaghi
- Sisli, Hamidiye Etfal Training and Research Hospiatal, Blood Center, Istanbul 34098, Turkey
| | - Mine Guzel
- Biruni Laboratories, Esentepe, Istanbul 34098, Turkey
| | - Ebru Guner
- Biruni Laboratories, Esentepe, Istanbul 34098, Turkey
| | | | - Pengxiang Chang
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Munir Iqbal
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| | - Juergen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Huseyin Yilmaz
- Department of Virology, Veterinary Faculty, Istanbul University-Cerrahpasa, Hadimkoy, Istanbul 34098, Turkey
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK
| |
Collapse
|
20
|
Ekström N, Haveri A, Solastie A, Virta C, Österlund P, Nohynek H, Nieminen T, Ivaska L, Tähtinen PA, Lempainen J, Jalkanen P, Julkunen I, Palmu AA, Melin M. Strong Neutralizing Antibody Responses to SARS-CoV-2 Variants Following a Single Vaccine Dose in Subjects With Previous SARS-CoV-2 Infection. Open Forum Infect Dis 2022; 9:ofac625. [PMID: 36519113 PMCID: PMC9745780 DOI: 10.1093/ofid/ofac625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2022] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primes the immune system; thus individuals who have recovered from infection have enhanced immune responses to subsequent vaccination (hybrid immunity). However, it remains unclear how well hybrid immunity induced by severe or mild infection can cross-neutralize emerging variants. We aimed to compare the strength and breadth of antibody responses in vaccinated recovered and uninfected subjects. METHODS We measured spike-specific immunoglobulin (Ig)G and neutralizing antibodies (NAbs) from vaccinated subjects including 320 with hybrid immunity and 20 without previous infection. From 29 subjects with a previous severe or mild infection, we also measured NAb responses against Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529/BA.1) variants following vaccination. RESULTS A single vaccine dose induced 2-fold higher anti-spike IgG concentrations and up to 4-fold higher neutralizing potency of antibodies in subjects with a previous infection compared with vaccinated subjects without a previous infection. Hybrid immunity was more enhanced after a severe than a mild infection, with sequentially decreasing NAb titers against Alpha, Beta, Delta, and Omicron variants. We found similar IgG concentrations in subjects with a previous infection after 1 or 2 vaccine doses. CONCLUSIONS Hybrid immunity induced strong IgG responses, particularly after severe infection. However, the NAb titers were low against heterologous variants, especially against Omicron.
Collapse
Affiliation(s)
- Nina Ekström
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anu Haveri
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anna Solastie
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Camilla Virta
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Pamela Österlund
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Hanna Nohynek
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tuomo Nieminen
- Data and Analytics Unit, Department of Knowledge Brokers, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Lauri Ivaska
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Paula A Tähtinen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Johanna Lempainen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pinja Jalkanen
- Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Clinical Microbiology, Turku University Hospital, Turku, Finland
- Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arto A Palmu
- Interventions Unit, Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Tampere, Finland
| | - Merit Melin
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
21
|
Bourret V, Dutra L, Alburkat H, Mäki S, Lintunen E, Wasniewski M, Kant R, Grzybek M, Venkat V, Asad H, Pradel J, Bouilloud M, Leirs H, Colombo VC, Sluydts V, Stuart P, McManus A, Eccard JA, Firozpoor J, Imholt C, Nowicka J, Goll A, Ranc N, Castel G, Charbonnel N, Sironen T. Serologic Surveillance for SARS-CoV-2 Infection among Wild Rodents, Europe. Emerg Infect Dis 2022; 28:2577-2580. [DOI: 10.3201/eid2812.221235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
22
|
Omer AAM, Hinkula J, Tran PTH, Melik W, Zattarin E, Aili D, Selegård R, Bengtsson T, Khalaf H. Plantaricin NC8 αβ rapidly and efficiently inhibits flaviviruses and SARS-CoV-2 by disrupting their envelopes. PLoS One 2022; 17:e0278419. [PMID: 36449554 PMCID: PMC9710782 DOI: 10.1371/journal.pone.0278419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Potent broad-spectrum antiviral agents are urgently needed to combat existing and emerging viral infections. This is particularly important considering that vaccine development is a costly and time consuming process and that viruses constantly mutate and render the vaccine ineffective. Antimicrobial peptides (AMP), such as bacteriocins, are attractive candidates as antiviral agents against enveloped viruses. One of these bacteriocins is PLNC8 αβ, which consists of amphipathic peptides with positive net charges that display high affinity for negatively charged pathogen membrane structures, including phosphatidylserine rich lipid membranes of viral envelopes. Due to the morphological and physiological differences between viral envelopes and host cell plasma membranes, PLNC8 αβ is thought to have high safety profile by specifically targeting viral envelopes without effecting host cell membranes. In this study, we have tested the antiviral effects of PLNC8 αβ against the flaviviruses Langat and Kunjin, coronavirus SARS-CoV-2, influenza A virus (IAV), and human immunodeficiency virus-1 (HIV-1). The concentration of PLNC8 αβ that is required to eliminate all the infective virus particles is in the range of nanomolar (nM) to micromolar (μM), which is surprisingly efficient considering the high content of cholesterol (8-35%) in their lipid envelopes. We found that viruses replicating in the endoplasmic reticulum (ER)/Golgi complex, e.g. SARS-CoV-2 and flaviviruses, are considerably more susceptible to PLNC8 αβ, compared to viruses that acquire their lipid envelope from the plasma membrane, such as IAV and HIV-1. Development of novel broad-spectrum antiviral agents can significantly benefit human health by rapidly and efficiently eliminating infectious virions and thereby limit virus dissemination and spreading between individuals. PLNC8 αβ can potentially be developed into an effective and safe antiviral agent that targets the lipid compartments of viral envelopes of extracellular virions, more or less independent of virus antigenic mutations, which faces many antiviral drugs and vaccines.
Collapse
Affiliation(s)
- Abubakr A. M. Omer
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jorma Hinkula
- Department of Biomedical and Clinical Sciences (BKV), Division of Molecular Medicine and Virology, Mucosa infection och inflammation Center (MIIC), Linköping University, Linköping, Sweden
| | - Pham-Tue-Hung Tran
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Wessam Melik
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Elisa Zattarin
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
23
|
Oksanen LAH, Virtanen J, Sanmark E, Rantanen N, Venkat V, Sofieva S, Aaltonen K, Kivistö I, Svirskaite J, Pérez AD, Kuula J, Levanov L, Hyvärinen A, Maunula L, Atanasova NS, Laitinen S, Anttila V, Lehtonen L, Lappalainen M, Geneid A, Sironen T. SARS-CoV-2 indoor environment contamination with epidemiological and experimental investigations. INDOOR AIR 2022; 32:e13118. [PMID: 36305066 PMCID: PMC9828560 DOI: 10.1111/ina.13118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/25/2022] [Accepted: 09/06/2022] [Indexed: 05/02/2023]
Abstract
SARS-CoV-2 has been detected both in air and on surfaces, but questions remain about the patient-specific and environmental factors affecting virus transmission. Additionally, more detailed information on viral sampling of the air is needed. This prospective cohort study (N = 56) presents results from 258 air and 252 surface samples from the surroundings of 23 hospitalized and eight home-treated COVID-19 index patients between July 2020 and March 2021 and compares the results between the measured environments and patient factors. Additionally, epidemiological and experimental investigations were performed. The proportions of qRT-PCR-positive air (10.7% hospital/17.6% homes) and surface samples (8.8%/12.9%) showed statistical similarity in hospital and homes. Significant SARS-CoV-2 air contamination was observed in a large (655.25 m3 ) mechanically ventilated (1.67 air changes per hour, 32.4-421 L/s/patient) patient hall even with only two patients present. All positive air samples were obtained in the absence of aerosol-generating procedures. In four cases, positive environmental samples were detected after the patients had developed a neutralizing IgG response. SARS-CoV-2 RNA was detected in the following particle sizes: 0.65-4.7 μm, 7.0-12.0 μm, >10 μm, and <100 μm. Appropriate infection control against airborne and surface transmission routes is needed in both environments, even after antibody production has begun.
Collapse
Affiliation(s)
- Lotta‐Maria A. H. Oksanen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Otorhinolaryngology and Phoniatrics – Head and Neck SurgeryHelsinki University HospitalHelsinkiFinland
| | - Jenni Virtanen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| | - Enni Sanmark
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Otorhinolaryngology and Phoniatrics – Head and Neck SurgeryHelsinki University HospitalHelsinkiFinland
| | - Noora Rantanen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Otorhinolaryngology and Phoniatrics – Head and Neck SurgeryHelsinki University HospitalHelsinkiFinland
| | - Vinaya Venkat
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| | - Svetlana Sofieva
- Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
- Finnish Meteorological InstituteHelsinkiFinland
| | - Kirsi Aaltonen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| | - Ilkka Kivistö
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| | - Julija Svirskaite
- Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | | | - Joel Kuula
- Finnish Meteorological InstituteHelsinkiFinland
| | - Lev Levanov
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - Leena Maunula
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| | - Nina S. Atanasova
- Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
- Finnish Meteorological InstituteHelsinkiFinland
| | | | - Veli‐Jukka Anttila
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- HUS Inflammation CenterHelsinki University HospitalHelsinkiFinland
| | - Lasse Lehtonen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- HUS Diagnostic Center, HUSLABHelsinki University HospitalHelsinkiFinland
| | - Maija Lappalainen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- HUS Diagnostic Center, HUSLABHelsinki University HospitalHelsinkiFinland
| | - Ahmed Geneid
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Otorhinolaryngology and Phoniatrics – Head and Neck SurgeryHelsinki University HospitalHelsinkiFinland
| | - Tarja Sironen
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Veterinary MedicineUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
24
|
Ruiz MJ, Siracusano G, Cottignies-Calamarte A, Tudor D, Real F, Zhu A, Pastori C, Capron C, Rosenberg AR, Temperton N, Cantoni D, Liao H, Ternette N, Moine P, Godement M, Geri G, Chiche JD, Annane D, Cramer Bordé E, Lopalco L, Bomsel M. Persistent but dysfunctional mucosal SARS-CoV-2-specific IgA and low lung IL-1β associate with COVID-19 fatal outcome: A cross-sectional analysis. Front Immunol 2022; 13:842468. [PMID: 36248831 PMCID: PMC9560774 DOI: 10.3389/fimmu.2022.842468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The role of the mucosal pulmonary antibody response in coronavirus disease 2019 (COVID-19) outcome remains unclear. Here, we found that in bronchoalveolar lavage (BAL) samples from 48 patients with severe COVID-19-infected with the ancestral Wuhan virus, mucosal IgG and IgA specific for S1, receptor-binding domain (RBD), S2, and nucleocapsid protein (NP) emerged in BAL containing viruses early in infection and persist after virus elimination, with more IgA than IgG for all antigens tested. Furthermore, spike-IgA and spike-IgG immune complexes were detected in BAL, especially when the lung virus has been cleared. BAL IgG and IgA recognized the four main RBD variants. BAL neutralizing titers were higher early in COVID-19 when virus replicates in the lung than later in infection after viral clearance. Patients with fatal COVID-19, in contrast to survivors, developed higher levels of mucosal spike-specific IgA than IgG but lost neutralizing activities over time and had reduced IL-1β in the lung. Altogether, mucosal spike and NP-specific IgG and S1-specific IgA persisting after lung severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance and low pulmonary IL-1β correlate with COVID-19 fatal outcome. Thus, mucosal SARS-CoV-2-specific antibodies may have adverse functions in addition to protective neutralization.
Collapse
Affiliation(s)
- Maria Julia Ruiz
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Gabriel Siracusano
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Andréa Cottignies-Calamarte
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Daniela Tudor
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Fernando Real
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Aiwei Zhu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Claudia Pastori
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Claude Capron
- AP-HP, Hôpital Ambroise Paré, Service d'Hématologie, Boulogne-Billancourt, France
| | - Arielle R. Rosenberg
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Hanqing Liao
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola Ternette
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pierre Moine
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Mathieu Godement
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Guillaume Geri
- AP-HP, Hôpital Ambroise Paré, Service de Réanimation, Boulogne-Billancourt, France
- Université de Versailles-St Quentin en Yvelines, Versailles, France
| | | | - Djillali Annane
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | | | - Lucia Lopalco
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- *Correspondence: Morgane Bomsel,
| |
Collapse
|
25
|
Obach D, Solastie A, Liedes O, Vara S, Krzyżewska‐Dudek E, Brinkmann L, Haveri A, Hammer CC, Dub T, Meri S, Freitag TL, Lyytikäinen O, Melin M. Impaired immunity and high attack rates caused by SARS-CoV-2 variants among vaccinated long-term care facility residents. Immun Inflamm Dis 2022; 10:e679. [PMID: 36039644 PMCID: PMC9382858 DOI: 10.1002/iid3.679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Long-term care facilities (LTCF) residents are at high risk for severe coronavirus disease 2019 (COVID-19), and therefore, COVID-19 vaccinations were prioritized for residents and personnel in Finland at the beginning of 2021. METHODS We investigated COVID-19 outbreaks in two LTCFs, where residents were once or twice vaccinated. After the outbreaks we measured immunoglobulin G (IgG) antibodies to severe acute respiratory syndrome coronavirus 2 spike glycoprotein, neutralizing antibody (NAb) titers, and cell-mediated immunity markers from residents and healthcare workers (HCWs). RESULTS In LTFC-1, the outbreak was caused by an Alpha variant (B.1.1.7) and the attack rate (AR) among once vaccinated residents was 23%. In LTCF-2 the outbreak was caused by a Beta variant (B.1.351). Its AR was 47% although all residents had received their second dose 1 month before the outbreak. We observed that vaccination had induced lower IgG concentrations, NAb titers and cell-mediated immune responses in residents compared to HCWs. Only 1/8 residents had NAb to the Beta variant after two vaccine doses. CONCLUSIONS The vaccinated elderly remain susceptible to breakthrough infections caused by Alpha and Beta variants. The weaker vaccine response in the elderly needs to be addressed in vaccination protocols, while new variants capable of evading vaccine-induced immunity continue to emerge.
Collapse
Affiliation(s)
- Dorothée Obach
- Department of Health Security, Infectious Disease Control and Vaccinations UnitFinnish Institute for Health and WelfareHelsinkiFinland
- European Program for Intervention Epidemiology Training (EPIET)European Centre for Disease Prevention and Control, (ECDC)StockholmSweden
| | - Anna Solastie
- Department of Health Security, Expert Microbiology UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Oona Liedes
- Department of Health Security, Expert Microbiology UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Saimi Vara
- Department of Health Security, Expert Microbiology UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Eva Krzyżewska‐Dudek
- Department of Bacteriology and Immunology, Translational Immunology Research ProgramUniversity of HelsinkiFinland
- Hirszfeld Institute of Immunology and Experimental Therapy, Department of Immunology of Infectious DiseasesPolish Academy of SciencesWroclawPoland
| | - Luise Brinkmann
- Department of Bacteriology and Immunology, Translational Immunology Research ProgramUniversity of HelsinkiFinland
| | - Anu Haveri
- Department of Health Security, Expert Microbiology UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Charlotte C. Hammer
- Department of Health Security, Infectious Disease Control and Vaccinations UnitFinnish Institute for Health and WelfareHelsinkiFinland
- European Program for Intervention Epidemiology Training (EPIET)European Centre for Disease Prevention and Control, (ECDC)StockholmSweden
| | - Timothée Dub
- Department of Health Security, Infectious Disease Control and Vaccinations UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Seppo Meri
- Department of Bacteriology and Immunology, Translational Immunology Research ProgramUniversity of HelsinkiFinland
| | - Tobias L. Freitag
- Department of Bacteriology and Immunology, Translational Immunology Research ProgramUniversity of HelsinkiFinland
| | - Outi Lyytikäinen
- Department of Health Security, Infectious Disease Control and Vaccinations UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - Merit Melin
- Department of Health Security, Expert Microbiology UnitFinnish Institute for Health and WelfareHelsinkiFinland
| |
Collapse
|
26
|
Srivastava AK, Gupta A, Chauhan D, Meena RC, Sugadev R, Eslavath MR, Gupta H, Karuna, Singh S, Singh Y, Tiwari RP, Kohli V, Varshney R, Ganju L. Development of robust, indigenous ELISA for detection of IgG antibodies against CoV-2 N and S proteins: mass screening. Appl Microbiol Biotechnol 2022; 106:6225-6238. [PMID: 35976427 PMCID: PMC9382608 DOI: 10.1007/s00253-022-12113-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022]
Abstract
Abstract The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic has adversely affected humankind and caused millions of deaths globally since January 2020. Robust and quick serological tests such as antibody detection assays for SARS-CoV-2 provide relevant information and aid in the process of vaccine development and diagnostics, as well as in sero-epidemiological monitoring of antibody response to the virus. The receptor-binding domain (RBD) of spike and nucleocapsid protein are specific targets for detecting SARS-CoV-2 antibodies. Here, we present the development of a stable spike (S) and nucleocapsid (N) protein-based ELISA antibody detection test “CoroSuchak,” with 99% sensitivity, 98% specificity, cost-effective, and detection in a minimum time for serodiagnosis and mass screening of the population for antibodies against SARS-CoV-2. Blood samples were analyzed from 374 SARS-CoV-2 reverse transcription-polymerase chain reaction (RT-PCR) positive, 772 negative and asymptomatic, and 874 random groups of subjects. We found that the antibody titer was significantly higher (p < 0.0001) in infected and vaccinated group compared to the only vaccinated and only infected group. Using enzyme-linked immunosorbent assay (ELISA), we detected SARS-CoV-2 immunoglobulin G (IgG) antibodies in 118/123 (96%) infected individuals, 570/653 (87%) non-infected but vaccinated individuals, 231/237 (97%) individuals who were both infected and vaccinated, and 499/874 (57%) from randomly selected individuals from the first and second waves of the pandemic. Similarly in the third wave, 14/14 (100%) infected and 16/20 (80%) RT-PCR-negative but symptomatic subjects were detected. Thus, the highly sensitive and specific in-house developed ELISA antibody detection kit “CoroSuchak” is extremely useful to determine the seroprevalence of SARS-CoV-2 antibodies in the coronavirus-exposed population. Key points •Indigenous kit using a combination of spike and nucleocapsid proteins and peptide sequences. •High sensitivity and specificity to detect variants. •Highly sensitive for mass screening. Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12113-8.
Collapse
Affiliation(s)
- Ashish Kumar Srivastava
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Avinash Gupta
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Deepika Chauhan
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Ramesh Chand Meena
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Ragumani Sugadev
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Malleswara Rao Eslavath
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Harshita Gupta
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Karuna
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sayar Singh
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Yamini Singh
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - R P Tiwari
- Vanguard Diagnostics Private Limited, Okhla Industrial Area, New Delhi, 110020, India
| | - Veena Kohli
- Vanguard Diagnostics Private Limited, Okhla Industrial Area, New Delhi, 110020, India
| | - Rajeev Varshney
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Lilly Ganju
- Defence Institute of Physiology and Allied Sciences (DIPAS), Ministry of Defence, DRDO, Govt. of India, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
27
|
Mishra S. Computational Structural and Functional Analyses of ORF10 in Novel Coronavirus SARS-CoV-2 Variants to Understand Evolutionary Dynamics. Evol Bioinform Online 2022; 18:11769343221108218. [PMID: 35909986 PMCID: PMC9336178 DOI: 10.1177/11769343221108218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction: In an effort to combat SARS-CoV-2 through multi-subunit vaccine design,
during studies using whole genome and immunome, ORF10, located at the 3′ end
of the genome, displayed unique features. It showed no homology to any known
protein in other organisms, including SARS-CoV. It was observed that its
nucleotide sequence is 100% identical in the SARS-CoV-2 genomes sourced
worldwide, even in the recent-most VoCs and VoIs of B.1.1.529 (Omicron),
B.1.617 (Delta), B.1.1.7 (Alpha), B.1.351 (Beta), and P.1 (Gamma) lineages,
implicating its constant nature throughout the evolution of deadly
variants. Aim: The structure and function of SARS-CoV-2 ORF10 and the role it may play in
the viral evolution is yet to be understood clearly. The aim of this study
is to predict its structure, function, and understand evolutionary dynamics
on the basis of mutations and likely heightened immune responses in the
immunopathogenesis of this deadly virus. Methods: Sequence analysis, ab-initio structure modeling and an understanding of the
impact of likely substitutions in key regions of protein was carried out.
Analyses of viral T cell epitopes and primary anchor residue mutations was
done to understand the role it may play in the evolution as a molecule with
likely enhanced immune response and consequent immunopathogenesis. Results: Few amino acid substitution mutations are observed, most probably due to the
ribosomal frameshifting, and these mutations may not be detrimental to its
functioning. As ORF10 is observed to be an expressed protein, ab-initio
structure modeling shows that it comprises mainly an α-helical region and
maybe an ER-targeted membrane mini-protein. Analyzing the whole proteome, it
is observed that ORF10 presents amongst the highest number of likely
promiscuous and immunogenic CTL epitopes, specifically 11 out of 30
promiscuous ones and 9 out of these 11, immunogenic CTL epitopes. Reactive T
cells to these epitopes have been uncovered in independent studies. Majority
of these epitopes are located on the α-helix region of its structure, and
the substitution mutations of primary anchor residues in these epitopes do
not affect immunogenicity. Its conserved nucleotide sequence throughout the
evolution and diversification of virus into several variants is a puzzle yet
to be solved. Conclusions: On the basis of its sequence, structure, and epitope mapping, it is concluded
that it may function like those mini-proteins used to boost immune responses
in medical applications. Due to the complete nucleotide sequence
conservation even a few years after SARS-CoV-2 genome was first sequenced,
it poses a unique puzzle to be solved, in view of the evolutionary dynamics
of variants emerging in the populations worldwide.
Collapse
Affiliation(s)
- Seema Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| |
Collapse
|
28
|
Dub T, Solastie A, Hagberg L, Liedes O, Nohynek H, Haveri A, Virta C, Vara S, Lasander M, Ekström N, Österlund P, Lind K, Valtonen H, Hemmilä H, Ikonen N, Lukkarinen T, Palmu AA, Melin M. High secondary attack rate and persistence of SARS-CoV-2 antibodies in household transmission study participants, Finland 2020–2021. Front Med (Lausanne) 2022; 9:876532. [PMID: 35966873 PMCID: PMC9366099 DOI: 10.3389/fmed.2022.876532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Household transmission studies offer the opportunity to assess both secondary attack rate (SAR) and persistence of SARS-CoV-2 antibodies over time. Methods In Spring 2020, we invited confirmed COVID-19 cases and their household members to four visits, where we collected nasopharyngeal and serum samples over 28 days after index case onset. We calculated SAR based on the presence of SARS-CoV-2 neutralizing antibodies (NAb) and assessed the persistence of NAb and IgG antibodies (Ab) against SARS-CoV-2 spike glycoprotein and nucleoprotein. Results SAR was 45% (39/87), including 35 symptomatic secondary cases. During the initial 28-day follow-up, 62% (80/129) of participants developed NAb. Of those that seroconverted, 90% (63/70), 85% (63/74), and 78% (45/58) still had NAb to early B-lineage SARS-CoV-2 3, 6, and 12 months after the onset of the index case. Anti-spike IgG Ab persisted in 100% (69/69), 97% (72/74), and 93% (55/59) of seroconverted participants after 3, 6, and 12 months, while anti-nucleoprotein IgG Ab levels waned faster, persisting in 99% (68/69), 78% (58/74), and 55% (39/71) of participants, respectively. Conclusion Following detection of a COVID-19 case in a household, other members had a high risk of becoming infected. NAb to early B-lineage SARS-CoV-2 persisted for at least a year in most cases.
Collapse
Affiliation(s)
- Timothée Dub
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Anna Solastie
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- *Correspondence: Anna Solastie,
| | - Lotta Hagberg
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Oona Liedes
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Hanna Nohynek
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Anu Haveri
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Camilla Virta
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Saimi Vara
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Mervi Lasander
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Nina Ekström
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Pamela Österlund
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Katja Lind
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- Centre for Military Medicine, Finnish Defence Forces, Helsinki, Finland
| | - Hanna Valtonen
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Heidi Hemmilä
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- Centre for Military Medicine, Finnish Defence Forces, Helsinki, Finland
| | - Niina Ikonen
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Timo Lukkarinen
- Health Stations and Internal Medicine Clinic, Social and Health Care Sector, Helsinki, Finland
| | - Arto A. Palmu
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Tampere, Finland
| | - Merit Melin
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|
29
|
Alzahrani OR, Alanazi AD, Kareinen L, Hawsawi YM, Alhadrami HA, Khogeer AA, Alatwi HE, Alharbi AA, Sironen T, Vapalahti O, Hepojoki J, Zakham F. Clinical and Serological Findings of COVID-19 Participants in the Region of Makkah, Saudi Arabia. Diagnostics (Basel) 2022; 12:1725. [PMID: 35885629 PMCID: PMC9318194 DOI: 10.3390/diagnostics12071725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Makkah in Saudi Arabia hosts the largest annual religious event in the world. Despite the many strict rules enacted, including Hajj cancellation, city lockdowns, and social distancing, the region has the second highest number of new COVID-19 cases in Saudi Arabia. Public health interventions that identify, isolate, and manage new cases could slow the infection rate. While RT-PCR is the current gold standard in SARS-CoV-2 identification, it yields false positive and negative results, which mandates the use of complementary serological tests. Here, we report the utility of serological assays during the acute phase of individuals with moderate and severe clinical manifestations of SARS-CoV-2 (COVID19). Fifty participants with positive RT-PCR results for SARS-CoV-2 were enrolled in this study. Following RT-PCR diagnosis, serum samples from the same participants were analyzed using in-house ELISA (IgM, IgA, and IgG) and microneutralization test (MNT) for the presence of antibodies. Of the 50 individuals analyzed, 43 (86%) showed a neutralizing antibody titer of ≥20. Univariate analysis with neutralizing antibodies as a dependent variable and the degree of disease severity and underlying medical conditions as fixed factors revealed that patients with no previous history of non-communicable diseases and moderate clinical manifestation had the strongest neutralizing antibody response "Mean: 561.11". Participants with severe symptoms and other underlying disorders, including deceased individuals, demonstrated the lowest neutralizing antibody response. Anti-spike protein antibody responses, as measured by ELISA, showed a statistically significant correlation with neutralizing antibodies. This reinforces the speculation that serological assays complement molecular testing for diagnostics; however, patients' previous medical history (anamnesis) should be considered in interpreting serological results.
Collapse
Affiliation(s)
- Othman R. Alzahrani
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (O.R.A.); (H.E.A.)
- Genome and Biotechnology Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Abdullah D. Alanazi
- Department of Biological Sciences, Faculty of Science and Humanities, Shaqra University, P.O. Box 1040, Ad-Dawadimi 11911, Saudi Arabia;
| | - Lauri Kareinen
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (L.K.); (T.S.); (O.V.); (J.H.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Yousef M. Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 40047, Jeddah 21499, Saudi Arabia;
- College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Hani A. Alhadrami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia;
- Special Infectious Agent Unit, King Fahd Medical Research Centre, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia
| | - Asim A. Khogeer
- Plan and Research Department, General Directorate of Health Affairs Makkah Region, MOH, Mecca 24321, Saudi Arabia;
| | - Hanan E. Alatwi
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (O.R.A.); (H.E.A.)
- Genome and Biotechnology Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Amnah A. Alharbi
- Genome and Biotechnology Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Biochemistry, Faculty of Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Tarja Sironen
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (L.K.); (T.S.); (O.V.); (J.H.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (L.K.); (T.S.); (O.V.); (J.H.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (L.K.); (T.S.); (O.V.); (J.H.)
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, 8057 Zürich, Switzerland
| | - Fathiah Zakham
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (L.K.); (T.S.); (O.V.); (J.H.)
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
30
|
Truong Nguyen P, Kant R, Van den Broeck F, Suvanto MT, Alburkat H, Virtanen J, Ahvenainen E, Castren R, Hong SL, Baele G, Ahava MJ, Jarva H, Jokiranta ST, Kallio-Kokko H, Kekäläinen E, Kirjavainen V, Kortela E, Kurkela S, Lappalainen M, Liimatainen H, Suchard MA, Hannula S, Ellonen P, Sironen T, Lemey P, Vapalahti O, Smura T. The phylodynamics of SARS-CoV-2 during 2020 in Finland. COMMUNICATIONS MEDICINE 2022; 2:65. [PMID: 35698660 PMCID: PMC9187640 DOI: 10.1038/s43856-022-00130-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused millions of infections and fatalities globally since its emergence in late 2019. The virus was first detected in Finland in January 2020, after which it rapidly spread among the populace in spring. However, compared to other European nations, Finland has had a low incidence of SARS-CoV-2. To gain insight into the origins and turnover of SARS-CoV-2 lineages circulating in Finland in 2020, we investigated the phylogeographic and -dynamic history of the virus. Methods The origins of SARS-CoV-2 introductions were inferred via Travel-aware Bayesian time-measured phylogeographic analyses. Sequences for the analyses included virus genomes belonging to the B.1 lineage and with the D614G mutation from countries of likely origin, which were determined utilizing Google mobility data. We collected all available sequences from spring and fall peaks to study lineage dynamics. Results We observed rapid turnover among Finnish lineages during this period. Clade 20C became the most prevalent among sequenced cases and was replaced by other strains in fall 2020. Bayesian phylogeographic reconstructions suggested 42 independent introductions into Finland during spring 2020, mainly from Italy, Austria, and Spain. Conclusions A single introduction from Spain might have seeded one-third of cases in Finland during spring in 2020. The investigations of the original introductions of SARS-CoV-2 to Finland during the early stages of the pandemic and of the subsequent lineage dynamics could be utilized to assess the role of transboundary movements and the effects of early intervention and public health measures.
Collapse
Affiliation(s)
- Phuoc Truong Nguyen
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ravi Kant
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Frederik Van den Broeck
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Maija T. Suvanto
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Hussein Alburkat
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Virtanen
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Ella Ahvenainen
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Robert Castren
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Samuel L. Hong
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Guy Baele
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Maarit J. Ahava
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Jarva
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Suvi Tuulia Jokiranta
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Hannimari Kallio-Kokko
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Vesa Kirjavainen
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Elisa Kortela
- Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu Kurkela
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Maija Lappalainen
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Liimatainen
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marc A. Suchard
- Departments of Biomathematics, Biostatistics and Human Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA USA
| | - Sari Hannula
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Tarja Sironen
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Philippe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
31
|
Siljander M, Uusitalo R, Pellikka P, Isosomppi S, Vapalahti O. Spatiotemporal clustering patterns and sociodemographic determinants of COVID-19 (SARS-CoV-2) infections in Helsinki, Finland. Spat Spatiotemporal Epidemiol 2022; 41:100493. [PMID: 35691637 PMCID: PMC8817446 DOI: 10.1016/j.sste.2022.100493] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/21/2022] [Accepted: 02/04/2022] [Indexed: 12/22/2022]
Abstract
This study aims to elucidate the variations in spatiotemporal patterns and sociodemographic determinants of SARS-CoV-2 infections in Helsinki, Finland. Global and local spatial autocorrelation were inspected with Moran's I and LISA statistics, and Getis-Ord Gi* statistics was used to identify the hot spot areas. Space-time statistics were used to detect clusters of high relative risk and regression models were implemented to explain sociodemographic determinants for the clusters. The findings revealed the presence of spatial autocorrelation and clustering of COVID-19 cases. High-high clusters and high relative risk areas emerged primarily in Helsinki's eastern neighborhoods, which are socioeconomically vulnerable, with a few exceptions revealing local outbreaks in other areas. The variation in COVID-19 rates was largely explained by median income and the number of foreign citizens in the population. Furthermore, the use of multiple spatiotemporal analysis methods are recommended to gain deeper insights into the complex spatiotemporal clustering patterns and sociodemographic determinants of the COVID-19 cases.
Collapse
Affiliation(s)
- Mika Siljander
- Earth Change Observation Laboratory, Department of Geosciences and Geography, P.O. Box 64, FI-00014 University of Helsinki, Helsinki, Finland; Department of Virology, Haartmaninkatu 3, P.O. Box 21, FI-00014 University of Helsinki, Helsinki, Finland.
| | - Ruut Uusitalo
- Earth Change Observation Laboratory, Department of Geosciences and Geography, P.O. Box 64, FI-00014 University of Helsinki, Helsinki, Finland; Department of Virology, Haartmaninkatu 3, P.O. Box 21, FI-00014 University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, Agnes Sjöberginkatu 2, P.O. Box 66, FI-00014 University of Helsinki, Helsinki, Finland
| | - Petri Pellikka
- Earth Change Observation Laboratory, Department of Geosciences and Geography, P.O. Box 64, FI-00014 University of Helsinki, Helsinki, Finland; Helsinki Institute of Sustainability Science, University of Helsinki, Helsinki, Finland; Institute for Atmospheric and Earth System Research, University of Helsinki, Helsinki, Finland; State Key Laboratory of Information Engineering in Surveying, Mapping and Remote Sensing, Wuhan University, Wuhan 430000, China
| | - Sanna Isosomppi
- Epidemiological Operations Unit, P.O. Box 8650, 00099 City of Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Haartmaninkatu 3, P.O. Box 21, FI-00014 University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, Agnes Sjöberginkatu 2, P.O. Box 66, FI-00014 University of Helsinki, Helsinki, Finland; Virology and Immunology, Diagnostic Center, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
32
|
Haveri A, Solastie A, Ekström N, Österlund P, Nohynek H, Nieminen T, Palmu AA, Melin M. Neutralizing antibodies to SARS-CoV-2 Omicron variant after third mRNA vaccination in health care workers and elderly subjects. Eur J Immunol 2022; 52:816-824. [PMID: 35312186 PMCID: PMC9087434 DOI: 10.1002/eji.202149785] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 02/28/2022] [Indexed: 01/05/2023]
Abstract
The emergence of SARS-CoV-2 Omicron variant (B.1.1.529) with major spike protein mutations has raised concern over potential neutralization escape and breakthrough infections among vaccinated and previously SARS-CoV-2-infected subjects. We measured cross-protective antibodies against variants in health care workers (HCW, n = 20) and nursing home residents (n = 9) from samples collected at 1-2 months, following the booster (3rd) dose. We also assessed the antibody responses in subjects infected before the Omicron era (n = 38) with subsequent administration of a single mRNA vaccine dose. Following booster vaccination, HCWs had high IgG antibody concentrations to the spike protein and neutralizing antibodies (NAb) were detectable against all variants. IgG concentrations among the elderly remained lower, and some lacked NAbs against the Beta and Omicron variants. NAb titers were significantly reduced against Delta, Beta, and Omicron compared to WT virus regardless of age. Vaccination induced high IgG concentrations and variable titers of cross-reactive NAbs in previously infected subjects, whereas NAb titers against Omicron were barely detectable 1 month postinfection. High IgG concentrations with cross-protective neutralizing activity were detected after three Coronavirus Disease 2019 (COVID-19) vaccine doses in HCWs. However, lower NAb titers seen in the frail elderly suggest inadequate protection against Omicron breakthrough infections, yet protection against severe COVID-19 is expected.
Collapse
Affiliation(s)
- Anu Haveri
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Anna Solastie
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Nina Ekström
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Pamela Österlund
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Hanna Nohynek
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| | - Tuomo Nieminen
- Department of Information ServicesFinnish Institute for Health and WelfareHelsinkiFinland
| | - Arto A. Palmu
- Department of Public Health and WelfareFinnish Institute for Health and WelfareTampereFinland
| | - Merit Melin
- Department of Health SecurityFinnish Institute for Health and WelfareHelsinkiFinland
| |
Collapse
|
33
|
Jalkanen P, Kolehmainen P, Haveri A, Huttunen M, Laine L, Österlund P, Tähtinen PA, Ivaska L, Maljanen S, Reinholm A, Belik M, Smura T, Häkkinen HK, Ortamo E, Kantele A, Julkunen I, Lempainen J, Kakkola L. Vaccine-Induced Antibody Responses against SARS-CoV-2 Variants-Of-Concern Six Months after the BNT162b2 COVID-19 mRNA Vaccination. Microbiol Spectr 2022; 10:e0225221. [PMID: 35262410 PMCID: PMC9045126 DOI: 10.1128/spectrum.02252-21] [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] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/06/2022] [Indexed: 01/11/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has raised concern about increased transmissibility, infectivity, and immune evasion from a vaccine and infection-induced immune responses. Although COVID-19 mRNA vaccines have proven to be highly effective against severe COVID-19 disease, the decrease in vaccine efficacy against emerged Beta and Delta variants emphasizes the need for constant monitoring of new virus lineages and studies on the persistence of vaccine-induced neutralizing antibodies. To analyze the dynamics of COVID-19 mRNA vaccine-induced antibody responses, we followed 52 health care workers in Finland for 6 months after receiving two doses of BNT162b2 vaccine with a 3-week interval. We demonstrate that, although anti-S1 antibody levels decrease 2.3-fold compared to peak antibody levels, anti-SARS-CoV-2 antibodies persist for months after BNT162b2 vaccination. Variants D614G, Alpha, and Eta are neutralized by sera of 100% of vaccinees, whereas neutralization of Delta is 3.8-fold reduced and neutralization of Beta is 5.8-fold reduced compared to D614G. Despite this reduction, 85% of sera collected 6 months postvaccination neutralizes Delta variant. IMPORTANCE A decrease in vaccine efficacy against emerging SARS-CoV-2 variants has increased the importance of assessing the persistence of SARS-CoV-2 spike protein-specific antibodies and neutralizing antibodies. Our data show that after 6 months post two doses of BNT162b2 vaccine, antibody levels decrease yet remain detectable and capable of neutralizing emerging variants. By monitoring the vaccine-induced antibody responses, vaccination strategies and administration of booster doses can be optimized.
Collapse
Affiliation(s)
- Pinja Jalkanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Anu Haveri
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Moona Huttunen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Larissa Laine
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | | | - Paula A. Tähtinen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Lauri Ivaska
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Sari Maljanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arttu Reinholm
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milja Belik
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Teemu Smura
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Hanni K. Häkkinen
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Department of Infectious Diseases, University Hospital and University of Helsinki, Helsinki, Finland
| | - Eeva Ortamo
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Department of Infectious Diseases, University Hospital and University of Helsinki, Helsinki, Finland
| | - Anu Kantele
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Department of Infectious Diseases, University Hospital and University of Helsinki, Helsinki, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Johanna Lempainen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Kakkola
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
34
|
Paju S, Tallgren M, Kivimäki A, Lahdentausta L, Salminen A, Oksanen L, Sanmark E, Geneid A, Pussinen P, Pietiäinen M. Effect of RNA quality to SARS-CoV-2 RT-qPCR detection from saliva. J Med Microbiol 2022; 71. [PMID: 35417320 DOI: 10.1099/jmm.0.001507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Saliva is an alternative sample material to nasopharyngeal swab in SARS-CoV-2 diagnostics. We investigated possible aspects to improve the reliability of SARS-CoV-2 detection from saliva. Saliva was collected from asymptomatic healthy subjects (n=133) and COVID-19 patients (n=9). SARS-CoV-2 detection was performed with quantitative reverse-transcriptase PCR (RT-qPCR) with two viral and one host target serving as an internal control. The use of internal control revealed that in the first RT-qPCR run 25-30 % of assays failed. The failure is associated with poor RNA quality. When the amount of RNA was cut down to half from the original amount, the performance of RT-qPCR was greatly enhanced (95 % of the assays succeeded). The quality of RNA was not affected by the use of different nucleic acid stabilizing buffers. Our study showed that saliva is suitable material for RT-qPCR based SARS-CoV-2 diagnostics, but the use of internal control is essential to distinguish the true negative samples from failed assays.
Collapse
Affiliation(s)
- Susanna Paju
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
| | | | - Anne Kivimäki
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
- Medical Nutrition Physiology, Pharmacology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Laura Lahdentausta
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
| | - Aino Salminen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
| | - Lotta Oksanen
- Department of Otorhinolaryngology and Phoniatrics - Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
| | - Enni Sanmark
- Department of Otorhinolaryngology and Phoniatrics - Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
| | - Ahmed Geneid
- Department of Otorhinolaryngology and Phoniatrics - Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
| | - Pirkko Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
| | - Milla Pietiäinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland
| |
Collapse
|
35
|
Intranasal Coronavirus SARS-CoV-2 Immunization with Lipid Adjuvants Provides Systemic and Mucosal Immune Response against SARS-CoV-2 S1 Spike and Nucleocapsid Protein. Vaccines (Basel) 2022; 10:vaccines10040504. [PMID: 35455253 PMCID: PMC9029453 DOI: 10.3390/vaccines10040504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/16/2022] [Indexed: 01/14/2023] Open
Abstract
In this preclinical two-dose mucosal immunization study, using a combination of S1 spike and nucleocapsid proteins with cationic (N3)/or anionic (L3) lipids were investigated using an intranasal delivery route. The study showed that nasal administration of low amounts of antigens/adjuvants induced a primary and secondary immune response in systemic IgG, mIL-5, and IFN-gamma secreting T lymphocytes, as well as humoral IgA in nasal and intestinal mucosal compartments. It is believed that recipients will benefit from receiving a combination of viral antigens in promoting a border immune response against present and evolving contagious viruses. Lipid adjuvants demonstrated an enhanced response in the vaccine effect. This was seen in the significant immunogenicity effect when using the cationic lipid N3. Unlike L3, which showed a recognizable effect when administrated at a slightly higher concentration. Moreover, the findings of the study proved the efficiency of an intranasally mucosal immunization strategy, which can be less painful and more effective in enhancing the respiratory tract immunity against respiratory infectious diseases.
Collapse
|
36
|
Ochola L, Ogongo P, Mungai S, Gitaka J, Suliman S. Performance Evaluation of Lateral Flow Assays for Coronavirus Disease-19 Serology. Clin Lab Med 2022; 42:31-56. [PMID: 35153047 PMCID: PMC8563367 DOI: 10.1016/j.cll.2021.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The coronavirus disease of 2019 (COVID-19) pandemic, caused by infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has undoubtedly resulted in significant morbidities, mortalities, and economic disruptions across the globe. Affordable and scalable tools to monitor the transmission dynamics of the SARS-CoV-2 virus and the longevity of induced antibodies will be paramount to monitor and control the pandemic as multiple waves continue to rage in many countries. Serologic assays detect humoral responses to the virus, to determine seroprevalence in target populations, or induction of antibodies at the individual level following either natural infection or vaccination. With multiple vaccines rolling out globally, serologic assays to detect anti-SARS-CoV-2 antibodies will be important tools to monitor the development of herd immunity. To address this need, serologic lateral flow assays (LFAs), which can be easily implemented for both population surveillance and home use, will be vital to monitor the evolution of the pandemic and inform containment measures. Such assays are particularly important for monitoring the transmission dynamics and durability of immunity generated by natural infections and vaccination, particularly in resource-limited settings. In this review, we discuss considerations for evaluating the accuracy of these LFAs, their suitability for different use cases, and implementation opportunities.
Collapse
Affiliation(s)
- Lucy Ochola
- Department of Tropical and Infectious Diseases, Institute of Primate Research, National Museums of Kenya, PO Box 24481, Nairobi 00502, Kenya
| | - Paul Ogongo
- Department of Tropical and Infectious Diseases, Institute of Primate Research, National Museums of Kenya, PO Box 24481, Nairobi 00502, Kenya; Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Samuel Mungai
- Directorate of Research and Innovation, Mount Kenya University, PO Box 342-01000, Thika, Kenya
| | - Jesse Gitaka
- Directorate of Research and Innovation, Mount Kenya University, PO Box 342-01000, Thika, Kenya
| | - Sara Suliman
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Characterization of the First SARS-CoV-2 Isolates from Aotearoa New Zealand as Part of a Rapid Response to the COVID-19 Pandemic. Viruses 2022; 14:v14020366. [PMID: 35215963 PMCID: PMC8877023 DOI: 10.3390/v14020366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has wreaked havoc across the globe for the last two years. More than 300 million cases and over 5 million deaths later, we continue battling the first real pandemic of the 21st century. SARS-CoV-2 spread quickly, reaching most countries within the first half of 2020, and New Zealand was not an exception. Here, we describe the first isolation and characterization of SARS-CoV-2 variants during the initial virus outbreak in New Zealand. Patient-derived nasopharyngeal samples were used to inoculate Vero cells and, three to four days later, a cytopathic effect was observed in seven viral cultures. Viral growth kinetics was characterized using Vero and VeroE6/TMPRSS2 cells. The identity of the viruses was verified by RT-qPCR, Western blot, indirect immunofluorescence assays, and electron microscopy. Whole-genome sequences were analyzed using two different yet complementary deep sequencing platforms (MiSeq/Illumina and Ion PGM™/Ion Torrent™), classifying the viruses as SARS-CoV-2 B.55, B.31, B.1, or B.1.369 based on the Pango Lineage nomenclature. All seven SARS-CoV-2 isolates were susceptible to remdesivir (EC50 values from 0.83 to 2.42 µM) and β-D-N4-hydroxycytidine (molnupiravir, EC50 values from 0.96 to 1.15 µM) but not to favipiravir (>10 µM). Interestingly, four SARS-CoV-2 isolates, carrying the D614G substitution originally associated with increased transmissibility, were more susceptible (2.4-fold) to a commercial monoclonal antibody targeting the spike glycoprotein than the wild-type viruses. Altogether, this seminal work allowed for early access to SARS-CoV-2 isolates in New Zealand, paving the way for numerous clinical and scientific research projects in the country, including the development and validation of diagnostic assays, antiviral strategies, and a national COVID-19 vaccine development program.
Collapse
|
38
|
Salmi H, Heinonen S, Hästbacka J, Lääperi M, Rautiainen P, Miettinen PJ, Vapalahti O, Hepojoki J, Knip M. New-onset type 1 diabetes in Finnish children during the COVID-19 pandemic. Arch Dis Child 2022; 107:180-185. [PMID: 34045208 PMCID: PMC8172262 DOI: 10.1136/archdischild-2020-321220] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/13/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Viral infections may trigger type 1 diabetes (T1D), and recent reports suggest an increased incidence of paediatric T1D and/or diabetic ketoacidosis (DKA) during the COVID-19 pandemic. OBJECTIVE To study whether the number of children admitted to the paediatric intensive care unit (PICU) for DKA due to new-onset T1D increased during the COVID-19 pandemic, and whether SARS-CoV-2 infection plays a role. METHODS This retrospective cohort study comprises two datasets: (1) children admitted to PICU due to new-onset T1D and (2) children diagnosed with new-onset T1D and registered to the Finnish Pediatric Diabetes Registry in the Helsinki University Hospital from 1 April to 31 October in 2016-2020. We compared the incidence, number and characteristics of children with newly diagnosed T1D between the prepandemic and pandemic periods. RESULTS The number of children admitted to PICU due to new-onset T1D increased from an average of 6.25 admissions in 2016-2019 to 20 admissions in 2020 (incidence rate ratio [IRR] 3.24 [95% CI 1.80 to 5.83]; p=0.0001). On average, 57.75 children were registered to the FPDR in 2016-2019, as compared with 84 in 2020 (IRR 1.45; 95% CI 1.13 to 1.86; p=0.004). 33 of the children diagnosed in 2020 were analysed for SARS-CoV-2 antibodies, and all were negative. CONCLUSIONS More children with T1D had severe DKA at diagnosis during the pandemic. This was not a consequence of SARS-CoV-2 infection. Instead, it probably stems from delays in diagnosis following changes in parental behaviour and healthcare accessibility.
Collapse
Affiliation(s)
- Heli Salmi
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Anesthesia and Intensive Care, New Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Santtu Heinonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna Hästbacka
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Anesthesia and Intensive Care, New Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mitja Lääperi
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Paula Rautiainen
- Department of Anesthesia and Intensive Care, New Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi J Miettinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUSLAB, Helsinki University Hospital, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Mikael Knip
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Stocking C, de Miguel L, Suteu G, Dressel A, Soricelli A, Roskos M, Valor S, Mutschmann C, März W. Evaluation of five widely used serologic assays for antibodies to SARS-CoV-2. Diagn Microbiol Infect Dis 2022; 102:115587. [PMID: 34826767 PMCID: PMC8554041 DOI: 10.1016/j.diagmicrobio.2021.115587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/26/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022]
Abstract
Reliable diagnostic technologies are pivotal to the fight against COVID-19. While real-time reverse transcription-polymerase chain reaction (rRT-PCR) remains the gold standard, commercial assays for antibodies against (SARS-CoV-2) have emerged. We sought to examine 5 widely used commercial methods. We measured antibodies against SARS-CoV-2 with assays, Abbott-IgG, Roche-IgT (total antibodies, isotype-unspecific), EUROIMMUN-IgG, EUROIMMUN-IgA, DiaSorin-IgG, in 191 serum samples from patients with rRT-PCR proven COVID-19 between days 0 and 47 after the onset of clinical symptoms and in biobank samples collected in 2018. The assays were calibrated using the manufacturers' instructions; results are in multiples of the assay specific cut-offs (Abbott, Roche, EUROIMMUN) or in arbitrary units (AU/mL, DiaSorin). The assays for IgG and IgT have approximately the same sensitivity and specificity for detecting seroconversion which starts at approximately day 3 after symptom onset, sensitivity reached 93% on day 16 and was 100% for each assay on day 20. The assay for IgA antibodies was superior in sensitivity and had a lower specificity than the others. Bivariate non-parametric correlation coefficients ranged between 0.738 and 0.991. Commercial assays for IgG or total antibodies against SARS-CoV-2 are largely equivalent for establishing seroconversion but differ at high antibody titres. Increased sensitivity to detect seroconversion is afforded by including IgA antibodies. Further international efforts to harmonise assays for antibodies against SARS-CoV-2 are urgently needed.
Collapse
Affiliation(s)
| | | | - Gabriela Suteu
- SYNLAB Medical Care Center Leverkusen GmbH, Leverkusen, Germany
| | | | | | | | | | | | - Winfried März
- SYNLAB Academy, Mannheim, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz; Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
40
|
Kajova M, Kekäläinen E, Anttila VJ, Paajanen J. Successful treatment with a short course of remdesivir in a case of prolonged COVID-19 in a lymphoma patient. Infect Dis (Lond) 2022; 54:455-459. [PMID: 35086417 DOI: 10.1080/23744235.2022.2028896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Patients with haematological malignancies have an increased susceptibility for COVID-19 and higher mortality. They may also have prolonged symptoms and viral shedding. Clinical trials have not specifically addressed the management of this patient group. We present a lymphoma patient with COVID-19 who was treated with remdesivir, and a literature review of similar cases. METHODS SARS-CoV-2 RT-PCR, virus culture and whole-genome sequencing were performed from nasopharyngeal swabs and antibody testing from serum. In addition, SARS-CoV-2 nucleocapsid antigen was tested from serum. Medline was searched for reported cases of lymphoma and COVID-19 treated with remdesivir. RESULTS The patient was undergoing lymphoma treatment including chemotherapy, rituximab and prednisolone. After diagnosis of COVID-19, broad-spectrum antibiotics were administered due to neutropenia and fever. After 20 d of fever with no signs of co-infection, remdesivir was initiated with rapid response. The treatment was continued for 4 d. Serum SARS-CoV-2 antibody tests were negative 20, 30 and 66 d from symptom onset. Before starting remdesivir, the SARS-CoV-2 PCR and virus culture from the nasopharynx and serum antigen test were positive. From earlier reports, we identified a total of eleven cases of lymphoma and COVID-19 treated with remdesivir accompanied by other antivirals and anti-inflammatory agents. CONCLUSIONS As shown in this and earlier reports on lymphoma patients, the clinical course of COVID-19 may be protracted and a humoral immune response may remain absent. In addition, optimal management remains undecided. The presented patient responded well to a short course of remdesivir.
Collapse
Affiliation(s)
- Mikael Kajova
- Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eliisa Kekäläinen
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Translational Immunology Research Program and Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Veli-Jukka Anttila
- Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Juuso Paajanen
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
41
|
Aptamers-Diagnostic and Therapeutic Solution in SARS-CoV-2. Int J Mol Sci 2022; 23:ijms23031412. [PMID: 35163338 PMCID: PMC8836149 DOI: 10.3390/ijms23031412] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
The SARS-CoV-2 virus is currently the most serious challenge to global public health. Its emergence has severely disrupted the functioning of health services and the economic and social situation worldwide. Therefore, new diagnostic and therapeutic tools are urgently needed to allow for the early detection of the SARS-CoV-2 virus and appropriate treatment, which is crucial for the effective control of the COVID-19 disease. The ideal solution seems to be the use of aptamers—short fragments of nucleic acids, DNA or RNA—that can bind selected proteins with high specificity and affinity. They can be used in methods that base the reading of the test result on fluorescence phenomena, chemiluminescence, and electrochemical changes. Exploiting the properties of aptamers will enable the introduction of rapid, sensitive, specific, and low-cost tests for the routine diagnosis of SARS-CoV-2. Aptamers are excellent candidates for the development of point-of-care diagnostic devices and are potential therapeutic tools for the treatment of COVID-19. They can effectively block coronavirus activity in multiple fields by binding viral proteins and acting as carriers of therapeutic substances. In this review, we present recent developments in the design of various types of aptasensors to detect and treat the SARS-CoV-2 infection.
Collapse
|
42
|
Liu W, Zhou Z, Zhang L, Li L, Wang L, Song L, Qiu S, Zhang L, Xu D, Tian X, Li X, Yang Y, Liang J, Liu Y, Li X, Zhou R. Establishment and evaluation of a 30-minute detection method for SARS-CoV-2 nucleic acid using a novel ultra-fast real-time PCR instrument. J Thorac Dis 2022; 13:6866-6875. [PMID: 35070371 PMCID: PMC8743416 DOI: 10.21037/jtd-21-1288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023]
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic is still raging worldwide. Efficient, fast and low-cost severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleic acid detection methods are urgently needed. Methods A rapid PCR temperature change mode was explored by moving the reaction tube between the independent temperature modules with large temperature differences and a portable ultra-fast real-time PCR instrument were developed. We established a rapid SARS-CoV-2 test method using the ultra-fast real-time PCR instrument, a China Food and Drug Administration-certified SARS-CoV-2 reagent and optimized reaction condition. The analytical and clinical performances of the rapid tests were evaluated by comparing with the standard SARS-CoV-2 tests. Results The new temperature change mode can effectively shorten the amplification reaction time and be successfully used in the development of the ultra-fast real-time PCR instrument. The rapid SARS-CoV-2 test method was established and the time to yield results were greatly shortened from 81 min of the standard test to 31 min. Specificity of the rapid test was assessed and no non-specific amplification (0/63) was observed. The limits of detection of the rapid and standard tests were similar. Clinical performance was evaluated using 184 respiratory specimens from patients with suspected SARS-CoV-2 infection. The positive agreement between the rapid and standard tests was 100% (67/67), the negative agreement was 97.4% (114/117), and the kappa statistic was 0.965 (P<0.001). No significant differences in the Ct values for each target gene were observed between the rapid test and the standard test (P>0.05). Conclusions We had developed a 30-minute detection method for SARS-CoV-2 nucleic acid using a novel ultra-fast real-time PCR instrument. The rapid test method may impact on patient management.
Collapse
Affiliation(s)
- Wenkuan Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Zhichao Zhou
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Lu Zhang
- Technology Center, Guangzhou Custom, Guangzhou, China
| | - Lei Li
- GIRM Biosafety (Guangzhou) Co., Ltd., Guangzhou, China
| | - Lin Wang
- Kingmed Virology Diagnostic & Translational Center, Guangzhou Kingmed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Linxiu Song
- Kingmed Virology Diagnostic & Translational Center, Guangzhou Kingmed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Shuyan Qiu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Duo Xu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.,Technology Center, Guangzhou Custom, Guangzhou, China.,GIRM Biosafety (Guangzhou) Co., Ltd., Guangzhou, China.,Kingmed Virology Diagnostic & Translational Center, Guangzhou Kingmed Center for Clinical Laboratory Co., Ltd., Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xingui Tian
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Xiao Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Yujie Yang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Jiaxin Liang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China
| | - Yong Liu
- Kingmed Virology Diagnostic & Translational Center, Guangzhou Kingmed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Xiaobo Li
- Technology Center, Guangzhou Custom, Guangzhou, China
| | - Rong Zhou
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
43
|
Carnicelli A, Fiori B, Ricci R, Piano A, Bonadia N, Taddei E, Fantoni M, Murri R, Cingolani A, Barillaro C, Cutuli SL, Marchesini D, Della Polla DA, Forte E, Fuorlo M, Di Maurizio L, Amorini P, Cattani P, Franceschi F, Sanguinetti M. Characteristic of IgA and IgG antibody response to SARS-CoV-2 infection in an Italian referral COVID-19 Hospital. Intern Emerg Med 2022; 17:53-64. [PMID: 33970428 PMCID: PMC8107418 DOI: 10.1007/s11739-021-02750-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/17/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Antibody response plays a fundamental role in the natural history of infectious disease. A better understanding of the immune response in patients with SARS-CoV-2 infection could be important for identifying patients at greater risk of developing a more severe form of disease and with a worse prognosis. METHODS We performed a cross-sectional analysis to determine the presence and the levels of both anti-SARS-CoV-2 IgG and IgA in a cohort of hospitalized patients with confirmed infection at different times in the natural history of the disease. Patients enrolled when admitted at the emergency department were prospectively followed up during hospital stay. RESULTS Overall, 131 patients were considered with a total of 237 samples processed. Cross-sectional analysis showed that seroconversion for IgA seems to occur between days 6 and 15, while IgG response seems to occur slightly later, peaking at day 20 after symptoms onset. Both IgA and IgG were maintained beyond 2 months. Severe patients showed a higher IgA response compared with mild patients when analyzing optical density (8.3 versus 5.6, p < 0.001). Prospective analysis conducted on 55 patients confirmed that IgA appear slightly earlier than IgG. After stratifying for the severity of disease, both the IgA and IgG responses were more vigorous in severe cases. Moreover, while IgG tended to stabilize, there was a relevant decline after the first month of IgA levels in mild cases. CONCLUSION IgA and IgG antibody response is closely related, although seroconversion for IgA occurs earlier. Both IgA and IgG are maintained beyond 2 months. Severe patients showed a more vigorous IgA and IgG response. IgA levels seem to decline after 1 month since the onset of symptoms in mild cases. Our results should be interpreted with cautions due to several limitations in our study, mainly the small number of cases, lack of data on viral load and clinical setting.
Collapse
Affiliation(s)
- Annamaria Carnicelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Barbara Fiori
- Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rosalba Ricci
- Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alfonso Piano
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nicola Bonadia
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eleonora Taddei
- Division of Infectious Diseases, Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimo Fantoni
- Division of Infectious Diseases, Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rita Murri
- Division of Infectious Diseases, Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonella Cingolani
- Division of Infectious Diseases, Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Christian Barillaro
- Department of Geriatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Salvatore Lucio Cutuli
- Institute of Anesthesia and Resuscitation, Department of Emergency Medicine, Anesthesiology and Resuscitation, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Debora Marchesini
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Evelina Forte
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mariella Fuorlo
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Di Maurizio
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Amorini
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Cattani
- Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maurizio Sanguinetti
- Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
44
|
Rasool G, Riaz M, Abbas M, Fatima H, Qamar MM, Zafar F, Mahmood Z. COVID-19: Clinical laboratory diagnosis and monitoring of novel coronavirus infected patients using molecular, serological and biochemical markers: A review. Int J Immunopathol Pharmacol 2022; 36:3946320221115316. [PMID: 35840546 PMCID: PMC9289644 DOI: 10.1177/03946320221115316] [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: 02/15/2022] [Accepted: 07/06/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19, a novel coronavirus disease, has provoked a variety of health and safety concerns, and socioeconomic challenges around the globe. The laboratory diagnosis of SARS-CoV-2 was quickly established utilizing nucleic acid amplification techniques (NAAT) after the disease causing virus has been identified, and its genetic sequence has been determined. In addition to NAAT, serological tests based on antibodies testing against SARS-CoV-2 were introduced for diagnostic and epidemiologic studies. Other biochemical investigations include monitoring of peripheral blood cells count, platelets/lymphocyte ratio, coagulation profile, cardiac, and inflammatory markers such as cytokines storm are also crucial in combating COVID-19 pandemic. Further, accurate and reliable laboratory results for SARS-CoV-2 play very important role in the initiation of early treatment and timely management of COVID-19 patients, provide support in clinical decision-making process to control infection, and detection of asymptomatic cases. The Task Force on Coronavirus-19 constituted by International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) has recognized informational framework for epidemiology, pathogenesis, and recommended the PCR-based analysis, serological and biochemical assays for analysis, monitoring, and management of disease. This literature review provides an overview of the currently used diagnostic techniques in clinical laboratories for the diagnosis, treatment monitoring, and management of COVID-19 patients. We concluded that each assays differ in their performance characteristics and the utilization of multiple techniques is necessary for the accurate diagnosis and management of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ghulam Rasool
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Riaz
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Mazhar Abbas
- Department of Biochemistry, University of Veterinary and Animal Sciences, Lahore, (Jhang Campus) Pakistan
| | - Hina Fatima
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | | | - Farzana Zafar
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Zahed Mahmood
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
45
|
Ali Al Shehri S, Al-Sulaiman AM, Azmi S, Alshehri SS. Bio-safety and bio-security: A major global concern for ongoing COVID-19 pandemic. Saudi J Biol Sci 2022; 29:132-139. [PMID: 34483699 PMCID: PMC8404373 DOI: 10.1016/j.sjbs.2021.08.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Besides its impacts on governance, economics, human culture, geostrategic partnership and environment, globalization greatly exerted control over science and security policies. Biosecurity is the critical job of efforts, policy and preparation to protect health of human, animal and environmental against any biological threats. With the transition into a global village, the possibility of biosecurity breaches has significantly increased. The COVID-19 pandemic is an example of an infringement on biosecurity that has posed a serious threat to the world. Since the first report on the recognition of COVID-19, a number of governments have taken preventive measures, like; lockdown, screening and early detection of suspected and implementing the required response to protect the loss of life and economy. Unfortunately, some of these measures have only recently been taken in some countries, which have contributed significantly to an increased morbidity and loss of life on a daily basis. In this article, the biological risks affecting human, animal and environmental conditions, biosafety violations and preventive measures have been discussed in order to reduce the outbreak and impacts of a pandemic like COVID-19.
Collapse
Affiliation(s)
| | - AM Al-Sulaiman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sarfuddin Azmi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Sultan S. Alshehri
- Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Science, Riyadh, Saudi Arabia
| |
Collapse
|
46
|
Muzammil I, Aqib AI, Tanveer Q, Muzmmal S, Naseer MA, Tahir M. COVID-19 diagnosis—myths and protocols. DATA SCIENCE FOR COVID-19 2022. [PMCID: PMC8988925 DOI: 10.1016/b978-0-323-90769-9.00027-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) belonging to betacoronaviruses, on the basis of sequence analysis, mainly infects the lower respiratory tract in humans while symptoms remain milder than to those of severe acute respiratory syndrome and Middle East respiratory syndrome. The outbreak of coronavirus disease 2019 (COVID-19) has surprised the world with its rapid spread and potential virulence by compromising personal safety and economic perspectives. Its clinical diagnosis is mainly based on epidemiologic history, clinical manifestations, and auxiliary examinations including nucleic acid detection, computed tomographic scan, and immune identification technology. However, atypical signs and symptoms in patients and discrepancies in the identification techniques have also become the reason for the spread of the virus. Genetic mutations by the virus or sensitivity/specificity of diagnostic tests are becoming a major issue to report COVID-19. This chapter thus details the available diagnostic tests and their mechanisms and limitations, and finally, the approaches to identify COVID-19 with valid precision are discussed.
Collapse
|
47
|
Veenhuis RT, Zeiss CJ. Animal Models of COVID-19 II. Comparative Immunology. ILAR J 2021; 62:17-34. [PMID: 33914873 PMCID: PMC8135340 DOI: 10.1093/ilar/ilab010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022] Open
Abstract
Developing strong animal models is essential for furthering our understanding of how the immune system functions in response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. The alarming speed at which SARS-CoV-2 has spread, and the high mortality rate of severe Coronavirus Disease 2019 (COVID-19), has required both basic science and clinical research to move at an unprecedented pace. Models previously developed to study the immune response against SARS-CoV have been rapidly deployed to now study SARS-CoV-2. To date, both small and large animal models are remarkably consistent when infected with SARS-CoV-2; however, certain models have proven more useful when answering specific immunological questions than others. Small animal models, such as Syrian hamsters, ferrets, and mice carrying the hACE2 transgene, appear to reliably recapitulate the initial cytokine surge seen in COVID-19 as well as show significant innate and adaptive cell infiltration in to the lung early in infection. Additionally, these models develop strong antibody responses to the virus, are protected from reinfection, and genetically modified versions exist that can be used to ask specific immunological questions. Large animal models such as rhesus and cynomologus macaques and African green monkeys are critical to understanding how the immune system responds to SARS-CoV-2 infection because they are considered to be the most similar to humans. These models are considered the gold standard for assessing vaccine efficacy and protection, and recapitulate the initial cytokine surge, immune cell infiltration into the lung, certain aspects of thrombosis, and the antibody and T-cell response to the virus. In this review, we discuss both small and large animal model studies previously used in SARS-CoV-2 research that may be useful in elucidating the immunological contributions to hallmark syndromes observed with COVID-19.
Collapse
Affiliation(s)
- Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
48
|
Wang Z, Chen J, Khan SA, Li F, Shen J, Duan Q, Liu X, Zhu J. Plasmonic Metasurfaces for Medical Diagnosis Applications: A Review. SENSORS (BASEL, SWITZERLAND) 2021; 22:133. [PMID: 35009676 PMCID: PMC8747222 DOI: 10.3390/s22010133] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 05/25/2023]
Abstract
Plasmonic metasurfaces have been widely used in biosensing to improve the interaction between light and biomolecules through the effects of near-field confinement. When paired with biofunctionalization, plasmonic metasurface sensing is considered as a viable strategy for improving biomarker detection technologies. In this review, we enumerate the fundamental mechanism of plasmonic metasurfaces sensing and present their detection in human tumors and COVID-19. The advantages of rapid sampling, streamlined processes, high sensitivity, and easy accessibility are highlighted compared with traditional detection techniques. This review is looking forward to assisting scientists in advancing research and developing a new generation of multifunctional biosensors.
Collapse
Affiliation(s)
- Zhenbiao Wang
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Science, Xiamen University, Xiamen 361003, China;
| | - Sayed Ali Khan
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
| | - Fajun Li
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
| | - Jiaqing Shen
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
| | - Qilin Duan
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
| | - Xueying Liu
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
| | - Jinfeng Zhu
- Key Laboratory of Electromagnetic Wave Science and Detection Technology, Institute of Electromagnetics and Acoustics, Xiamen University, Xiamen 361005, China; (Z.W.); (S.A.K.); (F.L.); (J.S.); (Q.D.); (X.L.)
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, China
| |
Collapse
|
49
|
Solastie A, Virta C, Haveri A, Ekström N, Kantele A, Miettinen S, Lempainen J, Jalkanen P, Kakkola L, Dub T, Julkunen I, Melin M. A Highly Sensitive and Specific SARS-CoV-2 Spike- and Nucleoprotein-Based Fluorescent Multiplex Immunoassay (FMIA) to Measure IgG, IgA, and IgM Class Antibodies. Microbiol Spectr 2021; 9:e0113121. [PMID: 34787485 PMCID: PMC8597651 DOI: 10.1128/spectrum.01131-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022] Open
Abstract
Validation and standardization of accurate serological assays are crucial for the surveillance of the coronavirus disease 2019 (COVID-19) pandemic and population immunity. We describe the analytical and clinical performance of an in-house fluorescent multiplex immunoassay (FMIA) for simultaneous quantification of antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleoprotein and spike glycoprotein. Furthermore, we calibrated IgG-FMIA against World Health Organization (WHO) International Standard and compared FMIA results to an in-house enzyme immunoassay (EIA) and a microneutralization test (MNT). We also compared the MNT results of two laboratories. IgG-FMIA displayed 100% specificity and sensitivity for samples collected 13 to 150 days post-onset of symptoms (DPO). For IgA- and IgM-FMIA, 100% specificity and sensitivity were obtained for a shorter time window (13 to 36 and 13 to 28 DPO for IgA- and IgM-FMIA, respectively). FMIA and EIA results displayed moderate to strong correlation, but FMIA was overall more specific and sensitive. IgG-FMIA identified 100% of samples with neutralizing antibodies (NAbs). Anti-spike IgG concentrations correlated strongly (ρ = 0.77 to 0.84, P < 2.2 × 10-16) with NAb titers, and the two laboratories' NAb titers displayed a very strong correlation (ρ = 0.95, P < 2.2 × 10-16). Our results indicate good correlation and concordance of antibody concentrations measured with different types of in-house SARS-CoV-2 antibody assays. Calibration against the WHO international standard did not, however, improve the comparability of FMIA and EIA results. IMPORTANCE SARS-CoV-2 serological assays with excellent clinical performance are essential for reliable estimation of the persistence of immunity after infection or vaccination. In this paper we present a thoroughly validated SARS-CoV-2 serological assay with excellent clinical performance and good comparability to neutralizing antibody titers. Neutralization tests are still considered the gold standard for SARS-CoV-2 serological assays, but our assay can identify samples with neutralizing antibodies with 100% sensitivity and 96% specificity without the need for laborious and slow biosafety level 3 (BSL-3) facility-requiring analyses.
Collapse
Affiliation(s)
- Anna Solastie
- Department of Health Security, Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Camilla Virta
- Department of Health Security, Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anu Haveri
- Department of Health Security, Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nina Ekström
- Department of Health Security, Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Anu Kantele
- Meilahti Infectious Diseases and Vaccination Research Center, MeiVac, Department of Infectious Diseases, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Simo Miettinen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Johanna Lempainen
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pinja Jalkanen
- Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Kakkola
- Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Timothée Dub
- Department of Health Security, Infectious Disease Control and Vaccinations Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Ilkka Julkunen
- Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Merit Melin
- Department of Health Security, Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
50
|
Tian Y, Parsons LM, Jankowska E, Cipollo JF. Site-Specific Glycosylation Patterns of the SARS-CoV-2 Spike Protein Derived From Recombinant Protein and Viral WA1 and D614G Strains. Front Chem 2021; 9:767448. [PMID: 34869209 PMCID: PMC8640487 DOI: 10.3389/fchem.2021.767448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
The SARS-CoV-2 spike protein is heavily glycosylated, having 22 predicted N-glycosylation sites per monomer. It is also O-glycosylated, although the number of O-glycosites is less defined. Recent studies show that spike protein glycans play critical roles in viral entry and infection. The spike monomer has two subdomains, S1 and S2, and a receptor-binding domain (RBD) within the S1 domain. In this study, we have characterized the site-specific glycosylation patterns of the HEK293 recombinant spike RBD and S1 domains as well as the intact spike derived from the whole virus produced in Vero cells. The Vero cell-derived spike from the WA1 strain and a D614G variant was analyzed. All spike proteins, S1, and RBDs were analyzed using hydrophilic interaction chromatography (HILIC) and LC-MS/MS on an Orbitrap Eclipse Tribrid mass spectrometer. N-glycans identified in HEK293-derived S1 were structurally diverse. Those found in the HEK293-derived RBD were highly similar to those in HEK293 S1 where N-glycosites were shared. Comparison of the whole cell-derived WA1 and D614G spike proteins revealed that N-glycosites local to the mutation site appeared to be more readily detected, hinting that these sites are more exposed to glycosylation machinery. Moreover, recombinant HEK293-derived S1 was occupied almost completely with complex glycan, while both WA1 and D614G derived from the Vero E6 cell whole virus were predominantly high-mannose glycans. This stands in stark contrast to glycosylation patterns seen in both CHO- and HEK cell-derived recombinant S1, S2, and the whole spike previously reported. Concerning O-glycosylation, our analyses revealed that HEK293 recombinant proteins possessed a range of O-glycosites with compositions consistent with Core type 1 and 2 glycans. The O-glycosites shared between the S1 and RBD constructs, sites T323 and T523, were occupied by a similar range of Core 1 and 2 type O-glycans. Overall, this study reveals that the sample nature and cell substrate used for production of these proteins can have a dramatic impact on the glycosylation profile. SARS-CoV-2 spike glycans are associated with host ACE2 receptor interaction efficiency. Therefore, understanding such differences will serve to better understand these host–pathogen interactions and inform the choice of cell substrates to suite downstream investigations.
Collapse
Affiliation(s)
- Yuan Tian
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Bacterial, Parasitic and Allergenic Products, Silver Spring, MD, United States
| | - Lisa M Parsons
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Bacterial, Parasitic and Allergenic Products, Silver Spring, MD, United States
| | - Ewa Jankowska
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Bacterial, Parasitic and Allergenic Products, Silver Spring, MD, United States
| | - John F Cipollo
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Bacterial, Parasitic and Allergenic Products, Silver Spring, MD, United States
| |
Collapse
|