1
|
Ali FH, Gentilcore G, Al-Jighefee HT, Taleb SA, Hssain AA, Qotba HA, Al Thani AA, Abu Raddad LJ, Nasrallah GK, Grivel JC, Yassine HM. Comprehensive analysis of human coronavirus antibody responses in ICU and non-ICU COVID-19 patients reveals IgG3 against SARS-CoV-2 spike protein as a key biomarker of disease severity. J Med Microbiol 2025; 74:002012. [PMID: 40359129 PMCID: PMC12075857 DOI: 10.1099/jmm.0.002012] [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: 07/08/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction. Pre-existing immunity to human coronaviruses (HCoVs) may shape the immune response in COVID-19 patients. Increasing evidence suggests that immune cross-reactivity between SARS-CoV-2 and other coronaviruses may determine clinical prognosis.Hypothesis. SARS-CoV-2 disease severity is influenced by pre-existing immunity to HCoVs, with distinct antibody profiles and cross-reactivity patterns.Aim. To investigate the antibody response of ICU and non-ICU SARS-CoV-2 patients against different HCoV proteins and assess the potential impact of pre-existing immunity on SARS-CoV-2 disease outcomes.Methodology. This study used a comprehensive HCoVs antigen bead array to measure antibody response to pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, SARS-CoV-2 and the four seasonal HCoVs in 70 ICU and 63 non-ICU COVID-19 patients.Results. Our analysis demonstrates an overall higher antibody response in ICU than in non-ICU COVID-19 patients. Interestingly, the anti-S1 IgG and IgA were significantly higher among ICU than in non-ICU patients. Similarly, the anti-S1 IgG against NL63 showed a lower response among ICU compared to non-ICU. Cross-reactivity was evident between SARS-CoV-2 and SARS-CoV antibodies but not with MERS-CoV and seasonal HCoVs. The subclass analysis of antibodies recognizing SARS-CoV-2 revealed that anti-S1 IgG1, IgG3, IgA1 and IgA2 were significantly higher in ICU compared to non-ICU. The predominant IgA subtype among SARS-CoV-2 patients was IgA1. We applied machine learning algorithms to subclass serological responses to build classifiers that could distinguish between ICU patients and patients with milder COVID-19. Out of 90 variables used in two different types of models, the variable of highest influence in determining the ICU status was IgG3 against SARS-CoV-2 S, and the top 8 variables of influence included the presence of IgG3 against S-trimer as well as IgA against SARS-CoV-2 S.Conclusion. Understanding the complexities of humoral immunity in various patients is critical for early medical intervention, disease management, selective vaccination and passive immunotherapy.
Collapse
Affiliation(s)
- Fatma H. Ali
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | | | - Sara Ahmad Taleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | | | - Asmaa A. Al Thani
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Laith J. Abu Raddad
- Department of Population Health Sciences, Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | - Hadi M. Yassine
- Biomedical Research Center, QH Health, Qatar University, Doha, Qatar
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Roël M, Schandl A, Jonmarker S, Hedman A, Vogel G, Joelsson-Alm E, Cronhjort M, Darlington P. Corticosteroids and long-term pulmonary function after critical illness due to COVID-19- a single-center cohort study. BMC Pulm Med 2025; 25:201. [PMID: 40287680 PMCID: PMC12032655 DOI: 10.1186/s12890-025-03659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Early in the pandemic, corticosteroids became standard treatment for patients with critical COVID-19 infections. This study aimed to investigate the possible long-term pulmonary consequences after corticosteroid treatment in patients with critical COVID-19 requiring ventilatory support. METHODS This observational single-center cohort study included patients treated for critical COVID-19 requiring ventilatory support between March 1, 2020, and August 1, 2021, with a 6-month follow-up after discharge from the intensive care unit. Corticosteroid treatment was defined according to the RECOVERY trial (6 mg dexamethasone daily or equivalent dose of another corticosteroid, initiated within eight days of hospital admittance and continued for at least one day) Pulmonary function was assessed by diffusion capacity for carbon monoxide. Health-related quality of life was measured with the questionnaire RAND-36. General linear regression was used to present mean score differences with 95% confidence intervals. RESULTS Among the 456 (69%) critically ill COVID-19 patients who survived at least 90 days after ICU discharge, 286 (63%) attended the follow-up six months later. The groups were balanced regarding invasive ventilation; 47% received invasive ventilation in both groups. Corticosteroid treatment was associated with a lower diffusion capacity for carbon monoxide (MSD - 8.3, 95% CI: -14.2 to -2.4) 6 months after ICU discharge (change > 10% were regarded as clinically significant). There were no differences in health-related quality of life between the groups. CONCLUSIONS Corticosteroids might negatively impact pulmonary function after critical COVID-19. The decrease did not seem to influence health-related quality of life. Future studies are needed to confirm the results.
Collapse
Affiliation(s)
- Mari Roël
- Department of Internal Medicine, Södersjukhuset, Stockholm, SE-118 83, Sweden.
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden.
| | - Anna Schandl
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Perioperative and intensive care, Södersjukhuset, Stockholm, SE-118 83, Sweden
| | - Sandra Jonmarker
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Perioperative and intensive care, Södersjukhuset, Stockholm, SE-118 83, Sweden
| | - Anders Hedman
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Cardiology, SE-118 83, Södersjukhuset, Stockholm, Sweden
| | - Gisela Vogel
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Perioperative and intensive care, Södersjukhuset, Stockholm, SE-118 83, Sweden
| | - Eva Joelsson-Alm
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Perioperative and intensive care, Södersjukhuset, Stockholm, SE-118 83, Sweden
| | - Maria Cronhjort
- Department of Clinical Sciences, Danderyd Hospital, Section of Anesthesiology and Intensive Care, Karolinska Institutet, Danderyds Sjukhus, Stockholm, SE-182 88, Sweden
| | - Pernilla Darlington
- Department of Internal Medicine, Södersjukhuset, Stockholm, SE-118 83, Sweden
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, SE-118 83, Sweden
| |
Collapse
|
3
|
Lupu A, Gavrilovici C, Mihai CM, Tonco DC, Nedelcu AH, Pertea L, Chisnoiu T, Baciu G, Stoicescu RM, Salaru DL, Badescu MC, Cuciureanu M, Cirstea O, Lupu VV. Multisystem inflammatory syndrome in children and Kawasaki disease. Front Immunol 2025; 16:1554787. [PMID: 40303414 PMCID: PMC12037394 DOI: 10.3389/fimmu.2025.1554787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
This narrative review aims to analyze and compare the current literature on multisystem inflammatory syndrome in children (MIS-C) and Kawasaki disease (KD), with a focus on case definitions, clinical features, diagnostic approaches, treatment strategies, and outcomes. Through a comprehensive review of relevant studies, including screening titles, abstracts, and full-text articles, key similarities and differences were identified. Both MIS-C and KD involve immune system dysregulation and share clinical manifestations such as rash, gastrointestinal symptoms, and cardiovascular involvement, with treatments often centered around immunomodulatory therapies. However, significant differences were observed, particularly in terms of age distribution, demographic prevalence, clinical presentation, and diagnostic criteria, with KD primarily affecting younger children and being associated more prominently with coronary artery abnormalities. While both diseases raise concerns about severe cardiac involvement and the need for intensive care, their pathogenic mechanisms have not been fully understood. Ongoing research is critical to elucidating these mechanisms, refining diagnostic criteria, and optimizing therapeutic approaches to improve outcomes for affected children. This comparative analysis is essential for advancing the understanding of both conditions, as accurately distinguishing between MIS-C and KD has significant implications for clinical decision-making and patient management. Given their overlapping yet distinct clinical features, precise differentiation is critical for ensuring timely diagnosis, optimizing therapeutic strategies, and improving patient outcomes. The concern among pediatric patients stems from the potential for severe complications, particularly cardiac involvement, which underscores the need for heightened awareness, early recognition, and evidence-based treatment strategies to minimize long-term morbidity and mortality.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Cristina Gavrilovici
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Denisa Claudia Tonco
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Leonard Pertea
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Ginel Baciu
- Pediatrics, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, Galati, Romania
| | | | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Magdalena Cuciureanu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Olga Cirstea
- Pediatrics, “Nicolae Testemitanu” State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Vasile Valeriu Lupu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
4
|
Ali ST, Chen D, Lau YC, Lim WW, Yeung A, Adam DC, Lau EHY, Wong JY, Xiao J, Ho F, Gao H, Wang L, Xu XK, Du Z, Wu P, Leung GM, Cowling BJ. Insights into COVID-19 epidemiology and control from temporal changes in serial interval distributions in Hong Kong. Am J Epidemiol 2025; 194:1079-1089. [PMID: 39013785 DOI: 10.1093/aje/kwae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024] Open
Abstract
The serial interval (SI) distribution of an epidemic is used to approximate the generation time distribution, an essential parameter for inferring the transmissibility (${R}_t$) of an infectious disease. However, SI distributions may change as an epidemic progresses. We examined detailed contact tracing data on laboratory-confirmed cases of COVID-19 in Hong Kong, China, during the 5 COVID-19 waves from January 2020 to July 2022. We reconstructed the transmission pairs and estimated time-varying effective SI distributions and factors associated with longer or shorter intervals. Finally, we assessed the biases in estimating transmissibility using constant SI distributions. We found clear temporal changes in mean SI estimates within each epidemic wave studied and across waves, with mean SIs ranging from 5.5 days (95% credible interval, 4.4-6.6) to 2.7 days (95% credible interval, 2.2-3.2). The mean SIs shortened or lengthened over time, which was found to be closely associated with the temporal variation in COVID-19 case profiles and public health and social measures and could lead to biases in predicting ${R}_t$. Accounting for the impact of these factors, the time-varying quantification of SI distributions could lead to improved estimation of ${R}_t$, and could provide additional insights into the impact of public health measures on transmission.
Collapse
Affiliation(s)
- Sheikh Taslim Ali
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Dongxuan Chen
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Yiu-Chung Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Wey Wen Lim
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Amy Yeung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Dillon C Adam
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Eric H Y Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Jessica Y Wong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
| | - Jingyi Xiao
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
| | - Faith Ho
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
| | - Huizhi Gao
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
| | - Lin Wang
- Department of Genetics, School of Biological Sciences, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Xiao-Ke Xu
- College of Information and Communication Engineering, Dalian Minzu University, Dalian, Liaoning Province, China 116600
| | - Zhanwei Du
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Peng Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Gabriel M Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, 999077, China
| |
Collapse
|
5
|
Costa-Pinto R, Rotherham H, O'Brien Z, Perera P, Chen V, Massarany C, Xu B, Bellomo R. Rebound Hypoxemia in Mechanically Ventilated Patients With COVID-19 Completing a Standard 10-Day Course of Corticosteroid Therapy. Respir Care 2025. [PMID: 40178917 DOI: 10.1089/respcare.12494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Background: Rebound hypoxemia may occur after cessation of corticosteroid therapy for COVID-19 pneumonitis. We aimed to determine the incidence of this phenomenon in mechanically ventilated patients with COVID-19 completing corticosteroid therapy. Methods: We conducted a retrospective observational cohort study across 2 tertiary ICUs from September to December 2021. We included all adult patients receiving invasive mechanical ventilation on completion of a 10-day course of dexamethasone for COVID pneumonitis. Our primary outcome was change in PaO2/FIO2 ratio in the 7 days following cessation of dexamethasone. Secondary outcomes included duration of ventilation, frequency of corticosteroid recommencement, and mortality. Results: We studied 88 subjects. Median age was 61 years (interquartile range [IQR] 51-67), and median duration of mechanical ventilation was 14 days (IQR 11-26). On completion of dexamethasone, 62 subjects (70%) remained off corticosteroid therapy for the subsequent 7 days. Of these, 63% (n = 20) had a stable PaO2/FIO2 ratio on day 7 (mean change 197 ± 42 mm Hg to 258 ± 83 mm Hg). Subjects who had a reduction in PaO2/FIO2 ratio >20 mm Hg by day 7 (mean change 214 ± 68 mm Hg to 152 ± 78 mm Hg) were more likely to die in hospital (P < .001). These subjects had a higher C-reactive protein level at time of steroid cessation (204 ± 87 mg/L, P = .17), which remained persistently elevated (206 ± 89 mg/L, P = .01) on day 7. The 30% of subjects who recommenced steroids required a longer duration of ventilation (13.5 vs 24.5 d, P = .002) but showed no differences in ICU mortality (19% vs 27%, P = .43). Conclusions: Rebound hypoxemia and steroid recommencement were common occurrences in mechanically ventilated subjects with COVID-19. Rebound hypoxemia was associated with higher mortality. Steroid recommencement was associated with longer duration of mechanical ventilation but no significant difference in mortality.
Collapse
Affiliation(s)
- Rahul Costa-Pinto
- Drs. Costa-Pinto, Massarany, Xu and Prof. Bellomo are affiliated with Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia
- Drs. Costa-Pinto, Rotherham and Prof. Bellomo are affiliated with Department of Critical Care, Department of Medicine, the University of Melbourne, Parkville, Victoria, Australia
| | - Hannah Rotherham
- Drs. Costa-Pinto, Rotherham and Prof. Bellomo are affiliated with Department of Critical Care, Department of Medicine, the University of Melbourne, Parkville, Victoria, Australia
- Drs. Rotherham, Perera, Chen and Prof. Bellomo are affiliated with Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Zachary O'Brien
- Dr. O'Brien is affiliated with Department of Anaesthesia, Austin Hospital, Heidelberg, Victoria, Australia
| | - Padeepa Perera
- Drs. Rotherham, Perera, Chen and Prof. Bellomo are affiliated with Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Vicky Chen
- Drs. Rotherham, Perera, Chen and Prof. Bellomo are affiliated with Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Christopher Massarany
- Drs. Costa-Pinto, Massarany, Xu and Prof. Bellomo are affiliated with Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia
| | - Brett Xu
- Drs. Costa-Pinto, Massarany, Xu and Prof. Bellomo are affiliated with Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia
| | - Rinaldo Bellomo
- Drs. Costa-Pinto, Massarany, Xu and Prof. Bellomo are affiliated with Department of Intensive Care, Austin Hospital, Heidelberg, Victoria, Australia
- Drs. Costa-Pinto, Rotherham and Prof. Bellomo are affiliated with Department of Critical Care, Department of Medicine, the University of Melbourne, Parkville, Victoria, Australia
- Drs. Rotherham, Perera, Chen and Prof. Bellomo are affiliated with Department of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Prof. Bellomo is affiliated with Australian and New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Prof. Bellomo is affiliated with Data Analytics Research and Evaluation Centre, The University of Melbourne and Austin Hospital, Melbourne, Australia
| |
Collapse
|
6
|
Fang JL, Shrestha L, Beland FA. Flow cytometric analysis of the SARS coronavirus 2 antibodies in human plasma. Sci Rep 2025; 15:10300. [PMID: 40133428 PMCID: PMC11937374 DOI: 10.1038/s41598-025-92389-8] [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: 10/09/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
COVID-19 is an infectious disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Anti-SARS-CoV-2 antibodies can provide information on patient immunity, identify asymptomatic patients, and track the spread of COVID-19. Efforts have been made to develop methods to detect anti-SARS-CoV-2 antibodies in humans. Here, we describe a flow cytometric assay for the simultaneous detection of anti-SARS-CoV-2 IgG and IgM in human plasma. To assess the antibody response against the different SARS-CoV-2 structural proteins, five viral recombinant proteins, including spike protein subunit 1 (S1), N-terminal domain of S1 (S1A), spike receptor-binding domain (RBD), spike protein subunit 2 (S2), and nucleocapsid protein (N), were generated. A comparison of the antibody profiles detected by the assay with plasma from 100 healthy blood donors collected prior to the COVID-19 pandemic and plasma from 100 virologically confirmed COVID-19 patients demonstrated a clear discrimination between the two groups. Among the COVID-19 patients, the antibody responses for the viral proteins, as determined by their prevalence, were anti-RBD IgG = anti-N IgG > anti-S1 IgG > anti-S1A IgG > anti-S2 IgG, and anti-RBD IgM > anti-S1 IgM > anti-N IgM > anti-S2 IgM. The prevalence of anti-SARS-CoV-2 IgG and IgM was not associated with sex, age, race, days after the onset of symptoms, or severity of illness, except for a higher prevalence of anti-S2 IgG being observed in men than in women. The levels of anti-RBD IgG were higher in patients 65 years and older and in patients who had severe symptoms. Similarly, patients who had severe symptoms exhibited higher levels of anti-S1 and anti-S1A IgG than patients who had mild or moderate symptoms. The levels of anti-RBD IgM tended to be higher in men but did not differ among age, race, days after the onset of symptoms, or severity of illness. Our study indicates that the flow cytometric assay, especially using RBD as target antigen, can be used to detect simultaneously anti-SARS-CoV-2 IgG and IgM antibodies in human plasma.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Leeza Shrestha
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
7
|
Kostinov M, Chuchalin A, Svitich O, Gaynitdinova V, Mashilov K, Kryukova N, Osiptsov V, Tatevosov V, Khromova E, Baranova I, Vlasenko A, Shogenova L, Mekhantseva I, Khrapunova I, Linok A, Kostinov A, Polishchuk V, Poddubikov A. Bacterial lysates in modifying sIgA levels in the upper respiratory tract in COVID-19 patients. Sci Rep 2025; 15:8325. [PMID: 40065113 PMCID: PMC11893902 DOI: 10.1038/s41598-025-92794-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
A great deal of evidence has accumulated suggesting an important role of mucosal immunity not only in preventing COVID-19 but also in the pathogenesis of this infection. The aim of the study was to evaluate the levels of secretory immunoglobulin A (sIgA) in different compartments of the upper respiratory tract in COVID-19 patients in relation to the severity of the disease and treatment with a bacteria-based immunomodulating agent (Immunovac VP4). The titers of sIgA were determined by ELISA in nasal epithelial swabs, pharyngeal swabs, and salivary gland secretions at baseline and on days 14 and 30 of treatment. The levels of nasal, pharyngeal and salivary sIgA were significantly lower in more severe patients (subgroup A) than in less severe patients (subgroup B), p < 0.01. In subgroup A, the patients who received Immunovac VP4 had higher pharyngeal sIgA levels in convalescent period than those who did not receive the therapy p < 0.05. In subgroup B patients, an increase in immunoglobulin levels was observed from baseline to day 14 of treatment whether they received the add-on therapy or not, p < 0.01. On day 30 of treatment, the sIgA levels in the standard treatment group, however, decreased, while the patients receiving the immunomodulating agent maintained high sIgA levels, p < 0.05. Oxygen saturation significantly increased by day 14 in both groups, p < 0.001. However, it was higher in the Immunovac VP4 group than in the standard treatment group, p < 0.01. Thus, addition of a bacterial lysate-based immunomodulating agent to the treatment regimen for moderate-to-severe COVID-19 induces the production of pharyngeal and salivary sIgA. SIgA production is inversely correlated to CRP levels and percentage of lung involvement on CT scan and is directly correlated to SpO2 levels.
Collapse
Affiliation(s)
- Mikhail Kostinov
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander Chuchalin
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Oksana Svitich
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
| | - Viliya Gaynitdinova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Kirill Mashilov
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
| | - Nadezhda Kryukova
- Pirogov Russian National Research Medical University, Moscow, Russian Federation.
| | - Valerij Osiptsov
- Main Military Clinical Hospital of the Troops of the National Guard of the Russian Federation, Moscow, Russian Federation
| | - Vitalij Tatevosov
- Main Military Clinical Hospital of the Troops of the National Guard of the Russian Federation, Moscow, Russian Federation
| | - Ekaterina Khromova
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
| | - Irina Baranova
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Anna Vlasenko
- Novokuznetsk State Institute for Advanced Training of Physicians, Branch Campus of the Russian Medical Academy of Continuous Professional Education, Novokuznetsk, Russian Federation
| | - Ludmila Shogenova
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | | | - Izabella Khrapunova
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
| | - Andrey Linok
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Anton Kostinov
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Valentina Polishchuk
- I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russian Federation
| | - Arseniy Poddubikov
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
8
|
Alric L, Brusq C, Migueres M, Faure S, Lebray P, Viallard JF, Chauveau D, Sailler L, Bérard E, Pugnet G, Cacoub P. Evaluation of the effects of pre-exposure treatment with hydroxychloroquine on the risk of COVID-19 infection and on the efficacy of anti-COVID-19 vaccination during lupus or Gougerot-Sjögren's disease: Prepcov multicentre trial. Lupus Sci Med 2025; 12:e001435. [PMID: 40044500 PMCID: PMC11883547 DOI: 10.1136/lupus-2024-001435] [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: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025]
Abstract
OBJECTIVES Some patients with SLE or Gougerot-Sjögren's disease (GSD) receive long-term treatment with hydroxychloroquine (HCQ), sometimes combined with immunosuppressive therapy (IS). This study sought to assess whether long-term HCQ therapy that had been initiated long before the COVID-19 pandemic had a protective or adverse effect on COVID-19 risk, severity of infection or immunity protection. METHODS This prospective multicentre study included 547 patients with SLE, GSD, autoimmune hepatitis, primary biliary cholangitis or cured viral hepatitis C divided into four groups according to HCQ (+/-) and IS (+/-) intake prior to the pandemic: HCQ+IS+ (n=112), HCQ+IS- (n=121), HCQ-IS+ (n=115) and HCQ-IS- (n=199). When COVID-19 vaccination was possible, patients were vaccinated as recommended. Vaccination efficacy was prospectively assessed on the basis of the postvaccination antibody titre. RESULTS Compared with HCQ+IS+ patients, HCQ-IS+ patients had a decreased risk of COVID-19 infection (p<0.001). Compared with HCQ+IS+ patients, HCQ-IS- patients had a decreased risk of contracting COVID-19 (p<0.001). Patients in the HCQ-IS+ or HCQ-IS- group had a lower risk of symptomatic or severe infection than HCQ+IS+ patients did (p=0.001 and p<0.001, respectively). Only patients who had two or more exposures (to vaccine and/or infection) had an increased likelihood of COVID-19 immunity after the last dose (p<0.001). CONCLUSIONS HCQ treatment that was initiated before the pandemic did not protect against COVID-19 infection. Moreover, non-exposure to HCQ treatment (combined or not with IS) was associated with decreased risk of COVID-19 infection and of developing a symptomatic or severe infection. HCQ and IS do not influence the vaccine response. Only two or more doses of vaccine result in a good vaccine response. TRIAL REGISTRATION NUMBER NCT04481633.
Collapse
Affiliation(s)
- Laurent Alric
- Toulouse III University-Paul Sabatier, Toulouse, France
| | - Clara Brusq
- Unité de Soutien Méthodologique à la Recherche (USMR), Service d'Epidémiologie Clinique et de Santé Publique, CHU de Toulouse, Toulouse III University-Paul Sabatier, Toulouse, France
| | | | - Stephanie Faure
- Hepatogastroenterology, Montpellier University, Montpellier, France
| | - Pascal Lebray
- Hepatology Unit, Hopital Universitaire Pitie-Salpetriere, Paris, France
| | | | - Dominique Chauveau
- Kidney Disease Unit, Toulouse III University-Paul Sabatier Faculty of Health, Toulouse, France
| | | | - Emilie Bérard
- Service d'Epidémiologie et Santé Publique, Toulouse III University-Paul Sabatier Faculty of Health, Toulouse, France
| | - Grégory Pugnet
- Internal Medicine Department, Toulouse III University-Paul Sabatier Faculty of Health, Toulouse, France
| | - Patrice Cacoub
- Service de Médecine Interne et Immunologie Clinique, Hopital Pitie-Salpetriere, Paris, France
| |
Collapse
|
9
|
He J, Liu X, Zhu X, Yuan HY, Chen W. Modeling the spatiotemporal transmission of COVID-19 epidemic by coupling the heterogeneous impact of detection rates: A case study in Hong Kong. Health Place 2025; 92:103422. [PMID: 39914091 DOI: 10.1016/j.healthplace.2025.103422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/22/2024] [Accepted: 01/24/2025] [Indexed: 03/24/2025]
Abstract
During the COVID-19 epidemic, many infections may have been undiagnosed in communities (hidden cases) due to low detection rates, thus exacerbating the overall prevalence of the epidemic. However, the heterogeneity of detection rates poses a challenge in simulating the proportion and spatial distribution of hidden cases. Coupling the heterogeneous impact of detection rates to extend epidemic modeling is necessary for forecasting the health burden and mitigating the inequity of testing resources. In this study, we developed an agent-based model integrated with the Susceptible-Exposed-Reported-Hidden-Removed (SERHR) model to simulate the spatiotemporal transmission of reported and hidden cases (RH-ABM). The RH-ABM was fitted with data for the fifth wave of infection in Hong Kong induced by the Omicron variant. We conducted multi-scenario simulations based on various testing strategies to assess the local variation in attack rates. The RH-ABM predicted that maintaining a constant high detection rate would reduce the average attack rate from 65.62% to 53.09%. Increasing detection rates in groups with many individuals and daily close contact can also assist in controlling the health burden of outbreaks. The variation in the attack rates is strongly associated with changes in the region-stratified detection rates. In addition, The RH-ABM estimated that allocating limited testing resources based on demographic distribution and human mobility data is effective for controlling the average attack rate.
Collapse
Affiliation(s)
- Jialyu He
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Xintao Liu
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Xiaolin Zhu
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Hsiang-Yu Yuan
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China; Centre for Applied One Health Research and Policy Advice, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Wu Chen
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China.
| |
Collapse
|
10
|
Zhang H, Osawa F, Okamoto H, Qiu Y, Liu Z, Ohshima N, Kajisa T, Sakata T, Izumi T, Sone H. Ultrasensitive Specific Detection of Anti-influenza A H1N1 Hemagglutinin Monoclonal Antibody Using Silicon Nanowire Field Effect Biosensors. ACS APPLIED BIO MATERIALS 2025; 8:1038-1049. [PMID: 39815599 DOI: 10.1021/acsabm.4c01263] [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: 01/18/2025]
Abstract
Rapid and sensitive detection of virus-related antigens and antibodies is crucial for controlling sudden seasonal epidemics and monitoring neutralizing antibody levels after vaccination. However, conventional detection methods still face challenges related to compatibility with rapid, highly sensitive, and compact detection apparatus. In this work, we developed a Si nanowire (SiNW)-based field-effect biosensor by precisely controlling the process conditions to achieve the required electrical properties via complementary metal-oxide-semiconductor (CMOS)-compatible nanofabrication processes. The SiNW surface was chemically modified with 2-aminoethylphosphonic acid, followed by a dehydration condensation reaction with influenza A H1N1 hemagglutinin (HA1), to enable specific detection of anti-HA1 immunoglobulin G (IgG). We successfully detected the anti-influenza IgG with concentrations ranging from 1 aM to 100 nM, achieving a remarkable detection limit of 6.0 aM. To demonstrate specificity, a control experiment was conducted using normal mouse IgG with concentrations of 6 aM to 600 nM. The results showed a high specificity, with the signal being 6-fold greater for the target IgG compared to the control IgG. This work demonstrates the capability of SiNW biosensors to detect anti-influenza A H1N1 hemagglutinin monoclonal antibody with enhanced detection sensitivity and specificity. This work lays the groundwork for future applications in detecting antibodies after vaccination or immunotherapy, contributing to the effective management of infectious pandemics.
Collapse
Affiliation(s)
- Hui Zhang
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| | - Fumiya Osawa
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| | - Haru Okamoto
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| | - Yawei Qiu
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| | - Zhiheng Liu
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| | - Noriyasu Ohshima
- Graduate School of Medicine, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Taira Kajisa
- SympaFit Company Limited, 16th Floor, Ark Hills South Tower 1-4-5 Roppongi, Minato-ku, Tokyo 106-0032, Japan
| | - Toshiya Sakata
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan
| | - Takashi Izumi
- Graduate School of Medicine, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
- Faculty of Health Care, Teikyo Heisei University, 2-51-4, Higashiikebukuro, Toshima-Ku, Tokyo 170-8445, Japan
| | - Hayato Sone
- Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-Cho, Kiryu, Gunma 376-8515, Japan
| |
Collapse
|
11
|
Mota Amaral GG, Sgorlon G, Batista Ferreira V, Serrano Batista F, da Silva Soares Farias L, Barbagelata LS, dos Santos MC, da Silva Bedran RL, Souza Vieira Dall'Acqua D, Benevides Matos N. SARS-CoV-2 Infection Among Children in Rondônia, Western Brazilian Amazon: SARS-CoV-2 Among Children in Rondônia. Adv Virol 2025; 2025:6655790. [PMID: 39995617 PMCID: PMC11850067 DOI: 10.1155/av/6655790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/28/2024] [Accepted: 12/19/2024] [Indexed: 02/26/2025] Open
Abstract
We analyzed 364 children symptomatic or asymptomatic for respiratory symptoms, aged 0.1 month-17 years, selected from primary healthcare units of different municipalities of Rondônia from June 2021 to September 2022. Data were collected from medical and electronic records for epidemiological characterization. The positive cohort (n = 96) was quantified using a real-time (RT) qPCR and sequenced by next-generation sequencing. Whole-genome sequences were obtained, SARS-CoV-2 strains were classified using the Pango system, and the maximum likelihood method was used for phylogenetic analyses. Among the patients, 59.34% (216/364) were male and 40.66% (148/364) were female. Children aged 10-14 years showed the highest rate of SARS-CoV-2 positivity. At the time of collection, 54.12% (197/364) of the patients were not age-eligible for immunization against COVID-19. The unvaccinated group accounted for 34.07% (124/364), with the highest proportion in the age groups of 5-9 and 10-14 years. Most patients exhibited mild symptoms. Seventy-nine high-quality genomes were obtained: Delta variant of concern (VOC) was the most prevalent (most abundant strain: AY.99.2), Omicron VOC was reported in 26 individuals (most frequent subvariant: BA.1.1), and Gamma VOC with 22 cases (12 cases of P.1 strain). The viral load showed a median of 7.26 log10 copies/mL, with a mean symptom duration of 4 days. Most of the cases were from children who were unvaccinated and age-ineligible for immunization and were associated with Delta and Omicron VOCs with an increase in subvariants during the study period.
Collapse
Affiliation(s)
- Gil Guibson Mota Amaral
- Department of Microorganisms Biology, Oswaldo Cruz Foundation, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Department of Postgraduate Program in Experimental Biology, Federal University of Rondônia-UNIR, Porto Velho, Rondônia, Brazil
| | - Gabriella Sgorlon
- Department of Postgraduate Program in Experimental Biology, Federal University of Rondônia-UNIR, Porto Velho, Rondônia, Brazil
- Department of Molecular Virology, Oswaldo Cruz Foundation, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Valcimar Batista Ferreira
- Department of Microorganisms Biology, Oswaldo Cruz Foundation, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Flávia Serrano Batista
- Department of Health Surveillance Agency-AGEVISA, State Coordination of Covid-19, Porto Velho, Rondônia, Brazil
| | | | | | | | | | - Deusilene Souza Vieira Dall'Acqua
- Department of Postgraduate Program in Experimental Biology, Federal University of Rondônia-UNIR, Porto Velho, Rondônia, Brazil
- Department of Molecular Virology, Oswaldo Cruz Foundation, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Division of Microbiology, Tropical Medicine Research Center-CEPEM, Porto Velho, Rondônia, Brazil
- National Institute of Epidemiology in Western Amazonia-INCT-EPIAMO, Porto Velho, Rondônia, Brazil
| | - Najla Benevides Matos
- Department of Microorganisms Biology, Oswaldo Cruz Foundation, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Department of Postgraduate Program in Experimental Biology, Federal University of Rondônia-UNIR, Porto Velho, Rondônia, Brazil
- Division of Microbiology, Tropical Medicine Research Center-CEPEM, Porto Velho, Rondônia, Brazil
- National Institute of Epidemiology in Western Amazonia-INCT-EPIAMO, Porto Velho, Rondônia, Brazil
| |
Collapse
|
12
|
Kikuchi S, Nakajima K, Kato Y, Takizawa T, Sugiyama J, Mukai T, Kakizawa Y, Kurahashi S. Quantification of droplet and contact transmission risks among elementary school students based on network analyses using video-recorded data. PLoS One 2025; 20:e0313364. [PMID: 39937726 PMCID: PMC11819611 DOI: 10.1371/journal.pone.0313364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/24/2024] [Indexed: 02/14/2025] Open
Abstract
In elementary schools, immunologically immature students come into close contact with each other and are susceptible to the spread of infectious diseases. To analyze pathogen transmission among students, it is essential to obtain behavioral data. Questionnaires and wearable sensor devices were used for communication behavior and swab sampling was employed for contact behavior. However, these methods have been insufficient in capturing information about the processes and actions of each student that contribute to pathogen transmission. Therefore, in this study, actual behavioral data were collected using video recordings to evaluate droplet and contact transmission in elementary schools. The analysis of communication behavior revealed the diverse nature of interactions among students. By calculating the droplet transmission probabilities based on conversation duration, the risk of droplet transmission was quantified. In the contact behavior, we introduced a novel approach for constructing contact networks based on contact history. According to this method, well-known items, such as students' desks, doors, and faucets, were predicted to be potential fomite. In addition, students' shirts and shared items with high contact frequency and high centrality metrics in the network, which were not evaluated in swab sampling surveys, were identified as potential fomites. The reliability of the predictions was demonstrated through micro-simulations. The micro-simulations replicated virus transmission scenarios in which virus-carrying students were present in the actual contact history. The results showed that a significant amount of virus adhered to the items predicted to be fomites. Interestingly, the micro-simulations indicated that most viral copies were transmitted through single items. The analysis of contact history, contact networks, and micro-simulations relies on video-recorded behavioral data, highlighting the importance of this method. This study contributes significantly to the prevention of infectious diseases in elementary schools by providing evidence-based information about transmission pathways and behavior-related risks.
Collapse
Affiliation(s)
- Shuta Kikuchi
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Keisuke Nakajima
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Yasuki Kato
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Takeshi Takizawa
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Junichi Sugiyama
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Taisei Mukai
- Institute of Social Simulation, Waseda University, Shinjuku, Tokyo, Japan
| | - Yasushi Kakizawa
- Advanced Analytical Science Research Laboratories, Research & Development Headquarters, Lion Corporation, Edogawa, Tokyo, Japan
| | - Setsuya Kurahashi
- Graduate School of Business Science, University of Tsukuba, Bunkyo, Tokyo, Japan
| |
Collapse
|
13
|
Trivedi R, Ashiq PL, Garg N, Jha P, Gadly T, Chakraborty B. Comparative study of alpha, beta, and omicron spike protein by computing the IR/Raman frequencies and UV-vis adsorption - A computational analysis through DFT. Microb Pathog 2025; 199:107195. [PMID: 39645095 DOI: 10.1016/j.micpath.2024.107195] [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: 03/27/2024] [Revised: 11/20/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
The outbreak of COVID-19 coronavirus disease around the end of 2019 was a pandemic. The virus has been mutated and so many strains like Alpha, Beta, and Omicron are present in different parts of the world. Hence, timely detection technique is important to overcome the diagnostic challenges. Considering the need for this pandemic situation, we used a spectroscopy test methodology to distinguish different strains of Covid-19 by computing the infrared & Raman frequencies and the optical absorption data. Optimization of spike protein of Alpha, and Omicron mutations showed the high value of dipole moment (4.44, and 4.36) Debye, and polarizability [Alpha (233.62), Omicron (228.65)] indicating more bioactivity of Alpha and Omicron instead of Beta. Molecular electrostatic potential map exhibits the presence of electrophilic and nucleophilic region suggesting charge transfer of spikes to accept/donate electrons and hence the system increased reactiveness. UV-Vis absorption analysis also shows electronic transitions (σ to π∗ and π to π∗) due to that protein probe mechanism of Alpha and Omicron becomes increasingly become unsaturated thus confirming its easy binding ability to the target human protein as compared to binding affinity of Beta spike protein.
Collapse
Affiliation(s)
- Ravi Trivedi
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Department of Physics, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India; Centre for Computational Physics, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| | - P L Ashiq
- School of Pure and Applied Physics, Mahatma Gandhi University, Priyadarsini Hills P O, Kottayam, 686560, India
| | - Nandini Garg
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Prafulla Jha
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - Trilochan Gadly
- Bio-Organic Divisions, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Brahmananda Chakraborty
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Homi Bhabha National Institute, India.
| |
Collapse
|
14
|
Avadhanula V, Creighton CJ, Ferlic-Stark L, Nagaraj D, Zhang Y, Sucgang R, Nicholson EG, Rajan A, Menon VK, Doddapaneni H, Muzny DM, Metcalf GA, Cregeen SJJ, Hoffman KL, Gibbs RA, Petrosino JF, Piedra PA. Longitudinal host transcriptional responses to SARS-CoV-2 infection in adults with extremely high viral load. PLoS One 2025; 20:e0317033. [PMID: 39820858 PMCID: PMC11737797 DOI: 10.1371/journal.pone.0317033] [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: 06/24/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Current understanding of viral dynamics of SARS-CoV-2 and host responses driving the pathogenic mechanisms in COVID-19 is rapidly evolving. Here, we conducted a longitudinal study to investigate gene expression patterns during acute SARS-CoV-2 illness. Cases included SARS-CoV-2 infected individuals with extremely high viral loads early in their illness, individuals having low SARS-CoV-2 viral loads early in their infection, and individuals testing negative for SARS-CoV-2. We could identify widespread transcriptional host responses to SARS-CoV-2 infection that were initially most strongly manifested in patients with extremely high initial viral loads, then attenuating within the patient over time as viral loads decreased. Genes correlated with SARS-CoV-2 viral load over time were similarly differentially expressed across independent datasets of SARS-CoV-2 infected lung and upper airway cells, from both in vitro systems and patient samples. We also generated expression data on the human nose organoid model during SARS-CoV-2 infection. The human nose organoid-generated host transcriptional response captured many aspects of responses observed in the above patient samples, while suggesting the existence of distinct host responses to SARS-CoV-2 depending on the cellular context, involving both epithelial and cellular immune responses. Our findings provide a catalog of SARS-CoV-2 host response genes changing over time and magnitude of these host responses were significantly correlated to viral load.
Collapse
Affiliation(s)
- Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Chad J. Creighton
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Laura Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Divya Nagaraj
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, United States of America
| | - Richard Sucgang
- Houston Methodist Research Institute, Center for Health Data Science and Analytics, Houston, Texas, United States of America
| | - Erin G. Nicholson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Vipin Kumar Menon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Harshavardhan Doddapaneni
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna Marie Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ginger A. Metcalf
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sara Joan Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Kristi Louise Hoffman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
15
|
Guo X, Yang S, Cai Z, Zhu S, Wang H, Liu Q, Zhang Z, Feng J, Chen X, Li Y, Deng J, Liu J, Li J, Tan X, Fu Z, Xu K, Zhou L, Chen Y. SARS-CoV-2 specific adaptations in N protein inhibit NF-κB activation and alter pathogenesis. J Cell Biol 2025; 224:e202404131. [PMID: 39680116 DOI: 10.1083/jcb.202404131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 12/17/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and severe acute respiratory syndrome coronavirus (SARS-CoV) exhibit differences in their inflammatory responses and pulmonary damage, yet the specific mechanisms remain unclear. Here, we discovered that the SARS-CoV-2 nucleocapsid (N) protein inhibits the activation of the nuclear factor-κB (NF-κB) pathway and downstream signal transduction by impeding the assembly of the transforming growth factor β-activated kinase1 (TAK1)-TAK1 binding protein 2/3 (TAB2/3) complex. In contrast, the SARS-CoV N protein does not impact the NF-κB pathway. By comparing the amino acid sequences of the SARS-CoV-2 and SARS-CoV N proteins, we identified Glu-290 and Gln-349 as critical residues in the C-terminal domain (CTD) of the SARS-CoV-2 N protein, essential for its antagonistic function. These findings were further validated in a SARS-CoV-2 trans-complementation system using cellular and animal models. Our results reveal the distinctions in inflammatory responses triggered by SARS-CoV-2 and SARS-CoV, highlighting the significance of specific amino acid alterations in influencing viral pathogenicity.
Collapse
Affiliation(s)
- Xiao Guo
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Shimin Yang
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zeng Cai
- Institute for Vaccine Research at Animal Bio-safety Level III Laboratory, Wuhan University School of Medicine, Wuhan, China
| | - Shunhua Zhu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Hongyun Wang
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Qianyun Liu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zhen Zhang
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jiangpeng Feng
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Xianying Chen
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Yingjian Li
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jikai Deng
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jiejie Liu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jiali Li
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Xue Tan
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zhiying Fu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Ke Xu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Li Zhou
- Institute for Vaccine Research at Animal Bio-safety Level III Laboratory, Wuhan University School of Medicine, Wuhan, China
| | - Yu Chen
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
- Institute for Vaccine Research at Animal Bio-safety Level III Laboratory, Wuhan University School of Medicine, Wuhan, China
| |
Collapse
|
16
|
Tseng CH, Lin CW, Tsai PY, Su MT. Detection of Severe Acute Respiratory Syndrome Coronavirus 2 in Pregnant Women Treated with Nirmatrelvir/Ritonavir (Paxlovid) Using Salivary Polymerase Chain Reaction: A Prospective Cohort Study. Microorganisms 2024; 12:2566. [PMID: 39770769 PMCID: PMC11677129 DOI: 10.3390/microorganisms12122566] [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: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES We aim to study the relative viral load using salivary polymerase chain reaction among pregnant women treated with Paxlovid. METHODS Pregnant women with coronavirus disease 2019 were allocated to two groups: those receiving Paxlovid and those receiving no antiviral agents. We compared the nasopharyngeal and salivary relative viral loads and their changes in saliva specimens. RESULTS Among the thirty-seven pregnant women, seventeen received Paxlovid, and twenty received no antiviral agents. The viral cycle threshold value of saliva was significantly higher than that from nasopharynx, with a median ± interquartile range of 26.44 ± 7.68 versus 17.6 ± 9.6 in the Paxlovid group (p = 0.005). Following treatment, the median salivary viral load decreased by 13.40 cycle threshold values in the Paxlovid group (from a median of [Day 0 Ct] to [Day 4/5 Ct]), compared to a change of -1.59 cycle threshold values in the no-antiviral group (from a median of [Day 0 Ct] to [Day 4/5 Ct]) (p = 0.021). The detection rate of coronavirus disease 2019 using salivary polymerase chain reaction was 83.8% (31/37). CONCLUSIONS This study showed that saliva is a useful diagnostic tool for coronavirus disease 2019 in pregnant women, and a significant decrease in the relative viral load of saliva was observed in those treated with Paxlovid.
Collapse
Affiliation(s)
| | | | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan; (C.-H.T.); (C.-W.L.)
| | - Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704, Taiwan; (C.-H.T.); (C.-W.L.)
| |
Collapse
|
17
|
Blasco A, Royuela A, García-Gómez S, Gómez-Lozano N, Sánchez-Arjona A, de la Fuente J, Anel J, Sánchez-Galarraga I, Pérez-Redondo M, González E, Silva L. Association of SARS-CoV-2 immunoserology and vaccination status with myocardial infarction severity and outcome. Vaccine 2024; 42:126305. [PMID: 39244425 DOI: 10.1016/j.vaccine.2024.126305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND The COVID-19 pandemic adversely affected the severity and prognosis of patients with acute myocardial infarction (MI) caused by atherothrombosis (type 1 MI). The effect, if any, of COVID-19 vaccination and natural SARS-CoV2 serologic immunity in these patients is unclear. Our aim was to analyze the association between the severity and outcome of patients with type 1 MI and their previous SARS-CoV2 vaccination and serostatus. METHODS A single-center retrospective cohort study conducted between March 1, 2020 and March 1, 2023. Clinical and follow-up information was collected from medical records and patients. Total antibodies (IgM, IgA, IgG) to nucleocapsid (N) antigens were measured by ECLIA (electrochemiluminescence-based immunoassay) to test the immune response to natural infection. If positive, IgM and IgG antibodies to spike (S) surface antigens were measured by CLIA to test the immune response to vaccine or natural infection. Multivariable logistic regression analysis was performed, adjusting for age, sex, hypertension, diabetes, and dyslipidemia. RESULTS Total sample of 949 patients, 656 with ST-segment elevation MI (STEMI) and 293 with non-ST-segment elevation MI (NSTEMI). Mean age was 64 (SD 13) years, 80 % men. Pre-admission vaccination status was: ≥ 1 dose, 53 % of patients; complete vaccination, 49 %; first booster dose, 25 %. The majority (84 %) of vaccines administered were mRNA-based. Six months after MI, 92 (9.7 %) patients had a major adverse cardiac event (MACE) and 50 died; 11 % of patients had severe heart failure or cardiogenic shock (Killip III-IV) after STEMI. Vaccinated patients with STEMI and positive serology (Pos/Vax group) had a higher risk of Killip III-IV on admission: OR 2.63 (1.27-5.44), p = 0.010. SARS-CoV-2 S-specific IgG titers were highest in this group (median > 2080 AU/mL, [IQR 1560- >2080] vs 91 [32-198] in the unvaccinated group). In the overall sample, a higher incidence of 6-month MACE was not demonstrated (OR 1.89 [0.98-3.61], p = 0.055). CONCLUSIONS The combination of vaccination and natural SARS-CoV2 infection was associated with the development of severe heart failure and cardiogenic shock in patients with STEMI, possibly related to an increased serological response.
Collapse
Affiliation(s)
- Ana Blasco
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain; Research Ethics Committee, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain.
| | - Ana Royuela
- Biostatistics Unit, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain; Center for Biomedical Research in Epidemiology and Public Health Network (CIBERESP), Madrid, Spain
| | - Sergio García-Gómez
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Natalia Gómez-Lozano
- Immunology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alberto Sánchez-Arjona
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge de la Fuente
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge Anel
- Microbiology Department, Serology Section, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Marina Pérez-Redondo
- Intensive Care Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Elisa González
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Lorenzo Silva
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
18
|
Maiti AK. MDA5 Is a Major Determinant of Developing Symptoms in Critically Ill COVID-19 Patients. Clin Rev Allergy Immunol 2024; 67:58-72. [PMID: 39460899 DOI: 10.1007/s12016-024-09008-z] [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] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Apart from the skin and mucosal immune barrier, the first line of defense of the human immune system includes MDA5 (ifih1 gene) which acts as a cellular sensor protein for certain viruses including SARS-CoV-2. Upon binding with viral RNA, MDA5 activates cell-intrinsic innate immunity, humoral responses, and MAVS (mitochondrial antiviral signaling). MAVS signaling induces type I and III interferon (IFN) expressions that further induce ISGs (interferon stimulatory genes) expressions to initiate human cell-mediated immune responses and attenuate viral replication. SARS-CoV-2 counteracts by producing NSP1, NSP2, NSP3, NSP5, NSP7, NSP12, ORF3A, ORF9, N, and M protein and directs anti-MDA5 antibody production presumably to antagonize IFN signaling. Furthermore, COVID-19 resembles several diseases that carry anti-MDA5 antibodies and the current COVID-19 vaccines induced anti-MDA5 phenotypes in healthy individuals. GWAS (genome-wide association studies) identified several polymorphisms (SNPs) in the ifih1-ifn pathway genes including rs1990760 in ifih1 that are strongly associated with COVID-19, and the associated risk allele is correlated with reduced IFN production. The genetic association of SNPs in ifih1 and ifih1-ifn pathway genes reinforces the molecular findings of the critical roles of MDA5 in sensing SARS-CoV-2 and subsequently the IFN responses to inhibit viral replication and host immune evasion. Thus, MDA5 or its pathway genes could be targeted for therapeutic development of COVID-19.
Collapse
Affiliation(s)
- Amit K Maiti
- Mydnavar, Department of Genetics and Genomics, 28475 Greenfield Rd, Southfield, MI, USA.
| |
Collapse
|
19
|
Stephenson S, Eid W, Wong CH, Mercier E, D'Aoust PM, Kabir MP, Baral S, Gilbride KA, Oswald C, Straus SE, Mackenzie A, Delatolla R, Graber TE. Urban wastewater contains a functional human antibody repertoire of mucosal origin. WATER RESEARCH 2024; 267:122532. [PMID: 39369505 DOI: 10.1016/j.watres.2024.122532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
Wastewater-based surveillance of human disease offers timely insights to public health, helping to mitigate infectious disease outbreaks and decrease downstream morbidity and mortality. These systems rely on nucleic acid amplification tests for monitoring disease trends, while antibody-based seroprevalence surveys gauge community immunity. However, serological surveys are resource-intensive and subject to potentially long lead times and sampling bias. We identified and characterized a human antibody repertoire, predominantly secretory IgA, isolated from a central wastewater treatment plant and building-scale wastewater collection points. These antibodies partition to the solids fraction and retain immunoaffinity for SARS-CoV-2 and Influenza A virus antigens. This stable pool could enable real-time tracking for correlates of vaccination, infection, and immunity, aiding in establishing population-level thresholds for immune protection and assessing the efficacy of future vaccine campaigns.
Collapse
Affiliation(s)
- Sean Stephenson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Walaa Eid
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Chandler Hayyin Wong
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Elisabeth Mercier
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Patrick M D'Aoust
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Md Pervez Kabir
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Stefan Baral
- Knowledge Translation Program, Unity Health Toronto, Toronto, Ontario, Canada
| | - Kimberly A Gilbride
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Claire Oswald
- Department of Geography and Environmental Studies, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Sharon E Straus
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alex Mackenzie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Robert Delatolla
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada.
| |
Collapse
|
20
|
Eggenhöffner R, Ghisellini P, Rando C, Papa S, khakshooy A, Giacomelli L. Evaluation of rapid detection methods for H5N1 virus using biosensors: An AI-based study. Bioinformation 2024; 20:1516-1523. [PMID: 40162463 PMCID: PMC11953552 DOI: 10.6026/9732063002001516] [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: 11/01/2024] [Revised: 11/30/2024] [Accepted: 11/30/2024] [Indexed: 04/02/2025] Open
Abstract
High mortality and zoonotic potential predispose the H5N1 avian influenza virus as a critical threat. knowing that an epidemic could be occurring, quick and precise diagnostic techniques are essential for managing and containing possible epidemics. To detect H5N1 in saliva samples, this study investigates the theoretical design, simulation and evaluation of three kind of biosensors based on different technologies with potential as rapid identifications tools to diagnose quickly H5N1: Lateral Flow Tests (LFT), Field Effect transistors (FET) based electrochemical sensors and Quartz Crystal Microbalance (QCM) sensors. Through detailed AI-based simulations, we show the capabilities, sensitivities and specificities of these biosensors, highlighting their potential for applications in general biology as well as their suitability both for routine home practice and for applications by control entities in public settings. We therefore wish to pave the way to a framework for the quick creation of detection tools that can be swiftly implemented for rapid deployment in case of an outbreak of disease.
Collapse
Affiliation(s)
- Roberto Eggenhöffner
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Corso Europa 30, Genova - 16132, Italy
| | - Paola Ghisellini
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Corso Europa 30, Genova - 16132, Italy
| | - Cristina Rando
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Corso Europa 30, Genova - 16132, Italy
| | | | - Allen khakshooy
- Department of Internal Medicine, Valley Hospital Medical Center, Las Vegas, NV - 89106, USA
| | | |
Collapse
|
21
|
Vu LD, Christofferson RC, O’Neal HR, Hamer D, Phan ATQ, Vance KM, Turner EA, Kumar A, Yola IM, Lim N, Ogden B, Cormier SA. Predicting severe COVID-19 using readily available admission indicators: SpO2/FiO2 ratio, comorbidity index, and gender. Exp Biol Med (Maywood) 2024; 249:10193. [PMID: 39633683 PMCID: PMC11614601 DOI: 10.3389/ebm.2024.10193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024] Open
Abstract
The focus of this study was to identify risk factors for severe and critical COVID-19, evaluate local respiratory immune responses to SARS-CoV-2 infection, and develop a prognostic tool for COVID-19 severity using accessible early indicators. Using nasopharyngeal swab samples from hospitalized patients with COVID-19 of varying severity during the first wave of the pandemic from March to May 2020 in Louisiana, we evaluated the association between COVID-19 severity and viral load, respiratory immune mediators, and demographic/clinical factors. We found that the SpO2/FiO2 ratio at triage, total comorbidity burden (represented by Charlson Comorbidity Index), and gender were significantly associated with COVID-19 severity. Using these early significant indicators, we developed a prognostic tool for COVID-19 severity that is simple and convenient. Additionally, our study demonstrated that elevated levels of respiratory immune mediators, including IL-10, IL-6, MCP-1, and MCP-3, were significantly associated with COVID-19 severity. We also found that viral load at the time of admission was associated with disease severity. Our findings highlight the feasibility and importance of evaluating the humoral component of local mucosal immune responses and viral load at the infected site using convenient nasopharyngeal swab samples, which could be an effective method to understand the relationship between viral infection and immune responses at the early stages of infection. Our proposed prognostic tool has the potential to be useful for COVID-19 management in clinical settings, as it utilizes accessible and easy-to-collect variables at the time of admission.
Collapse
Affiliation(s)
- Luan D. Vu
- Department of Biological Sciences, Louisiana State University and Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Rebecca C. Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Hollis R. O’Neal
- Medical Director of Research, Our Lady of the Lake Regional Medical Center, Pulmonary and Critical Care Medicine, Louisiana State University Health Sciences Center, Baton Rouge, LA, United States
| | - Diana Hamer
- Office of Research Administration, Our Lady of the Lake Regional Medical Center, Baton Rouge, LA, United States
| | - Anh T. Q. Phan
- Department of Biological Sciences, Louisiana State University and Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Katie M. Vance
- Office of Research Administration, Our Lady of the Lake Regional Medical Center, Baton Rouge, LA, United States
| | - E. A. Turner
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Avinash Kumar
- Department of Biological Sciences, Louisiana State University and Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Ibrahim Musa Yola
- Office of Research Administration, Our Lady of the Lake Regional Medical Center, Baton Rouge, LA, United States
| | - Natalie Lim
- Department of Biological Sciences, Louisiana State University and Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Beverly Ogden
- Department of Research, Woman’s Hospital, Baton Rouge, LA, United States
| | - Stephania A. Cormier
- Department of Biological Sciences, Louisiana State University and Pennington Biomedical Research Center, Baton Rouge, LA, United States
| |
Collapse
|
22
|
Byrne C, Schiffer JT. Ensemble modeling of SARS-CoV-2 immune dynamics in immunologically naïve rhesus macaques predicts that potent, early innate immune responses drive viral elimination. Front Immunol 2024; 15:1426016. [PMID: 39575237 PMCID: PMC11578959 DOI: 10.3389/fimmu.2024.1426016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction An unprecedented breadth of longitudinal viral and multi-scale immunological data has been gathered during SARS-CoV-2 infection. However, due to the high complexity, non-linearity, multi-dimensionality, mixed anatomic sampling, and possible autocorrelation of available immune data, it is challenging to identify the components of the innate and adaptive immune response that drive viral elimination. Novel mathematical models and analytical approaches are required to synthesize contemporaneously gathered cytokine, transcriptomic, flow cytometry, antibody response, and viral load data into a coherent story of viral control, and ultimately to discriminate drivers of mild versus severe infection. Methods We investigated a dataset describing innate, SARS-CoV-2 specific T cell, and antibody responses in the lung during early and late stages of infection in immunologically naïve rhesus macaques. We used multi-model inference and ensemble modeling approaches from ecology and weather forecasting to compare and combine various competing models. Results and discussion Model outputs suggest that the innate immune response plays a crucial role in controlling early infection, while SARS-CoV-2 specific CD4+ T cells correspond to later viral elimination, and anti-spike IgG antibodies do not impact viral dynamics. Among the numerous genes potentially contributing to the innate response, we identified IFI27 as most closely linked to viral load decline. A 90% knockdown of the innate response from our validated model resulted in a ~10-fold increase in peak viral load during infection. Our approach provides a novel methodological framework for future analyses of similar complex, non-linear multi-component immunologic data sets.
Collapse
Affiliation(s)
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center,
Seattle, WA, United States
| |
Collapse
|
23
|
de Oliveira-Filho EF, Martínez-Vega RA, Moreira-Soto A, Beuchel C, Freyle-Román IK, Mora-Guevara E, Rincón-Orozco B, Drexler JF. Post-migration infection with SARS-CoV-2 in Venezuelan migrants: A laboratory-based epidemiological observational study. Travel Med Infect Dis 2024; 62:102772. [PMID: 39424114 DOI: 10.1016/j.tmaid.2024.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/19/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Since 2015, over 6 million Venezuelans migrated to Colombia and neighboring countries. While most people adhered to lockdown measures, migrants kept moving during the COVID-19 pandemic. METHOD To investigate the extent of migration-associated SARS-CoV-2 infections, we interviewed 1209 adult Venezuelan migrants upon arrival to Bucaramanga, Colombia, 200 km from the Colombian-Venezuelan border along the main migration route during April-September 2021, collected individual-level socio-economic and clinical data, sampled blood and saliva, and assessed SARS-CoV-2 infection by serological, molecular and phylogenetic tools. RESULTS SARS-CoV-2 RT-PCR positivity was 1.9 % (95 % Confidence Interval (CI), 1.2-2.9) without varying significantly over the study period (chi-square, p = 0.922) and significantly associated with stay in Colombia >14 days (p = 0.018; prevalence ratio 3.3, 95 % CI, 1.2-8.7). Pre-existing SARS-CoV-2-specific antibodies were neither significantly associated with preventing infection (Chi-square, p = 0.188), nor symptom development (Fisher, p = 0.246). Predominance and time of detection of SARS-CoV-2 Mu and Gamma variants in migrants in comparison to available genomic data suggested infection predominantly in Colombia. SARS-CoV-2 IgG-based seroprevalence was 34.2 % (95 % CI, 31.5-36.9). Detection of SARS-CoV-2-specific antibodies was significantly associated with previous contact with infected individuals (p = 0.002). CONCLUSIONS SARS-CoV-2 infection occurred predominantly after immigration, potentially facilitated by densely populated border camps. Improved infrastructure and health care will prevent migration-associated spread of COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Edmilson F de Oliveira-Filho
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Ruth A Martínez-Vega
- Universidad de Santander, Escuela de Medicina, Facultad de Ciencias Médicas y de la Salud Bucaramanga, Colombia
| | - Andres Moreira-Soto
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Carl Beuchel
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | | | | | | | - Jan Felix Drexler
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; German Centre for Infection Research (DZIF), associated partner site Charité, Berlin, Germany.
| |
Collapse
|
24
|
Wright AJ, Nasralla HH, Deshmukh R, Jamalzadeh M, Hannigan M, Patera A, Li Y, Manzo-Perez M, Parashar N, Huang Z, Udumulla T, Chen W, De Forni D, Weck M, de Peppo GM, Riedo E, Shahrjerdi D. Nanoscale-localized multiplexed biological activation of field effect transistors for biosensing applications. NANOSCALE 2024; 16:19620-19632. [PMID: 39324869 DOI: 10.1039/d4nr02535k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The rise in antibiotic-resistant pathogens, highly infectious viruses, and chronic diseases has prompted the search for rapid and versatile medical tests that can be performed by the patient. Field-effect transistor (FET)-based electronic biosensing platforms are particularly attractive due to their sensitivity, fast turn-around time, potential for parallel detection of multiple pathogens, and compatibility with semiconductor manufacturing. However, an unmet critical need is a scalable, site-selective multiplexed biofunctionalization method with nanoscale precision for immobilizing different types of pathogen-specific bioreceptors on individual FETs, preventing parallel detection of multiple targets. Here, we propose a paradigm shift in FET biofunctionalization using thermal scanning probe lithography (tSPL) with a thermochemically sensitive polymer. This polymer can be spin-coated on fully-fabricated FET chips, making this approach applicable to any FET sensor material and technology. Crucially, we demonstrate the spatially selective multiplexed functionalization capability of this method by immobilizing different types of bioreceptors at prescribed locations on a chip with sub-20 nm resolution, paving the way for massively parallel FET detection of multiple pathogens. Antibody- and aptamer-modified graphene FET sensors are then realized, achieving ultra-sensitive detection of a minimum measured concentrations of 3 aM of SARS-CoV-2 spike proteins and 10 human SARS-CoV-2 infectious live virus particles per ml, and selectivity against human influenza A (H1N1) live virus.
Collapse
Affiliation(s)
- Alexander James Wright
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Hashem Hassan Nasralla
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Rahul Deshmukh
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Moeid Jamalzadeh
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Matthew Hannigan
- Department of Chemistry, New York University, New York, NY, 10003, USA
| | - Andrew Patera
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Mirimus, Inc, 760 Parkside Ave, Brooklyn, NY, 11226, USA
| | - Yanxiao Li
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Miguel Manzo-Perez
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Nitika Parashar
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Zhujun Huang
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | | | - Weiqiang Chen
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Davide De Forni
- ViroStatics S.r.l., Viale Umberto I, 46, 07100 Sassari, Italy
| | - Marcus Weck
- Department of Chemistry, New York University, New York, NY, 10003, USA
| | | | - Elisa Riedo
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| | - Davood Shahrjerdi
- Department of Electrical and Computer Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA.
| |
Collapse
|
25
|
Feng W, Chen Y, Han Y, Diao Z, Zhao Z, Zhang Y, Huang T, Ma Y, Li Z, Jiang J, Li J, Li J, Zhang R. Key performance evaluation of commercialized multiplex rRT-PCR kits for respiratory viruses: implications for application and optimization. Microbiol Spectr 2024; 12:e0164124. [PMID: 39470276 PMCID: PMC11619282 DOI: 10.1128/spectrum.01641-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/30/2024] Open
Abstract
Respiratory tract infections (RTIs) caused by viruses are prevalent and significant conditions in clinical settings. Accurate and effective detection is of paramount importance in the diagnosis, treatment, and prevention of viral RTIs. With technological advancements, multiplex real-time reverse transcription polymerase chain reaction (rRT-PCR) assays have been developed and extensively adopted for the diagnosis of viral RTIs. Given the potential challenges in the detection performance of multiplex assays, this study evaluated the analytical sensitivity and competitive interference of the six most commonly used multiplex rRT-PCR kits for detection of respiratory viruses in China. The results revealed that the limits of detection were variable across the viruses and kits. Most of the evaluated multiplex kits demonstrated comparable or enhanced analytical sensitivity compared with singleplex kits for clinically significant viruses, including human adenovirus (HAdV)-3, HAdV-7, Omicron BA.5, H1N1pdm09, H3N2, B/Victoria, respiratory syncytial virus subtype A, and respiratory syncytial virus subtype B, whereas multiplex kits showed relatively less analytical sensitivity for human rhinovirus-B72, human metapneumovirus-A2, parainfluenza virus (PIV)-1, and PIV-3. In addition, most multiplex kits successfully identified co-infections when one analyte was present at a low concentration and another analyte was present at a high concentration. IMPORTANCE The complexity and severity of viral respiratory tract infections (RTIs) emphasize the pivotal role of precise diagnosis for viral RTIs in guiding effective public health responses and ensuring appropriate medical interventions, given the substantial population at risk. This study highlights the necessity and importance of evaluating the analytical validity of multiplex real-time reverse transcription polymerase chain reaction assays, offering valuable insights into their optimization and application.
Collapse
Affiliation(s)
- Wanyu Feng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Yuqing Chen
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Yanxi Han
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Zhenli Diao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Zihong Zhao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Yuanfeng Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Tao Huang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Yu Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Ziqiang Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jian Jiang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jing Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jinming Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Rui Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| |
Collapse
|
26
|
Nakamoto T, Iwamoto N, Oshiro Y, Inamura N, Nemoto T, Ide S, Nakamura K, Nomoto H, Akiyama Y, Suzuki T, Miyazato Y, Suzuki M, Suzuki K, Kimura M, Saito S, Kutsuna S, Ohmagari N. COVID-19 severity and corticosteroid treatment have minimal effect on specific antibody production. BMC Infect Dis 2024; 24:1197. [PMID: 39443853 PMCID: PMC11515524 DOI: 10.1186/s12879-024-10090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Dexamethasone is currently administered for Coronavirus disease 2019(COVID-19); however, there are concerns about its effect on specific antibodies' production. The aim of this study was to evaluate whether specific antibodies were affected by COVID-19 severity and corticosteroid treatment. METHODS Of 251 confirmed COVID-19 patients admitted to our hospital between January 26 and August 10, 2020, the early period of the pandemic, 75 patients with sera within 1 month of onset and 1 month or longer were included in the research. A total of 253 serum samples from these patients were collected. The levels of specific antibodies for severe acute respiratory syndrome coronavirus 2(SARS-CoV-2), immunoglobulin G (IgG) and M (IgM), were measured retrospectively. The results were compared separately of each COVID-19 severity, and with or without corticosteroid treatment. RESULTS Among the 75 patients, 47, 18, and 10 had mild, moderate, and severe disease, respectively. The median age was 53.0 years and 22 (29%) were women. The most common comorbidities were hypertension and dyslipidemia. Corticosteroids were administered to 20 (27%) and 10 (53%), patients with moderate and severe disease, respectively. The positivity rates IgM increased first, and IgG was almost always positive after day 16, regardless of the severity of COVID-19. On days 6-10, both IgG and IgM positivity rates were higher in patients with moderate disease than in those with mild or severe disease. In patients with moderate disease, IgG positivity was similar over time, regardless of corticosteroid treatment. CONCLUSIONS In COVID-19 patients, specific IgG is positive and maintained for a long period of time, even after corticosteroid treatment. The effect of corticosteroid treatment in a COVID-19 epidemiological study using specific IgG antibodies was considered minor. COVID-19 patients were more likely to receive oxygen if IgM was positive 1 week after onset, but not mechanical ventilation. IgM measurement 1 week after onset may predict COVID-19 severity.
Collapse
Affiliation(s)
- Takato Nakamoto
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan.
| | - Noriko Iwamoto
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Yusuke Oshiro
- Clinical Laboratory, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Natsumi Inamura
- Clinical Laboratory, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Takashi Nemoto
- Clinical Laboratory, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Satohi Ide
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Keiji Nakamura
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
- Center for the Study fo Global IInfection, Kushu University Hospital, Fukuoka, Japan
| | - Hidetoshi Nomoto
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Yutaro Akiyama
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Tetsuya Suzuki
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Yusuke Miyazato
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Michiyo Suzuki
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Kumiko Suzuki
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Moto Kimura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Tokyo, 162-8655, Japan
| | - Sho Saito
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Satoshi Kutsuna
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
- Department of Infection Control, Graduate School of Medicine / Faculty of Medicine, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center/Travel Clinic, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| |
Collapse
|
27
|
Drori P, Mouhadeb O, Moya Muñoz GG, Razvag Y, Alcalay R, Klocke P, Cordes T, Zahavy E, Lerner E. Rapid and specific detection of nanoparticles and viruses one at a time using microfluidic laminar flow and confocal fluorescence microscopy. iScience 2024; 27:110982. [PMID: 39391727 PMCID: PMC11466642 DOI: 10.1016/j.isci.2024.110982] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/24/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Mainstream virus detection relies on the specific amplification of nucleic acids via polymerase chain reaction, a process that is slow and requires extensive laboratory expertise and equipment. Other modalities, such as antigen-based tests, allow much faster virus detection but have reduced sensitivity. In this study, we introduce an approach for rapid and specific detection of single nanoparticles using a confocal-based flow virometer. The combination of laminar flow in a microfluidic channel and correlated fluorescence signals emerging from both free dyes and fluorescently labeled primary antibodies provide insights into nanoparticle volumes and specificities. We evaluate and validate the assay using fluorescent beads and viruses, including SARS-CoV-2 with fluorescently labeled primary antibodies. Additionally, we demonstrate how hydrodynamic focusing enhances the assay sensitivity for detecting viruses at relevant loads. Based on our results, we envision the future use of this technology for clinically relevant bio-nanoparticles, supported by the implementation of the assay in a portable and user-friendly setup.
Collapse
Affiliation(s)
- Paz Drori
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Faculty of Mathematics & Science, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Odelia Mouhadeb
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Gabriel G. Moya Muñoz
- Physical and Synthetic Biology. Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
- Biophysical Chemistry, Department of Chemistry and Chemical Biology, Technische Universität Dortmund, Dortmund, Germany
| | - Yair Razvag
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Faculty of Mathematics & Science, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ron Alcalay
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Philipp Klocke
- Physical and Synthetic Biology. Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
| | - Thorben Cordes
- Physical and Synthetic Biology. Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
- Biophysical Chemistry, Department of Chemistry and Chemical Biology, Technische Universität Dortmund, Dortmund, Germany
| | - Eran Zahavy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Eitan Lerner
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Faculty of Mathematics & Science, The Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
28
|
Yousef A, Solomon I, Cheng G, Makani S, Boys J, Weissbrod PA. COVID-19 Related Tracheal Stenosis Requiring Tracheal Resection: A Case Series. Ann Otol Rhinol Laryngol 2024; 133:879-885. [PMID: 39127881 PMCID: PMC12064082 DOI: 10.1177/00034894241266489] [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] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To characterize the preoperative and intraoperative findings of symptomatic tracheal stenosis associated with COVID-19 related respiratory failure requiring tracheal resection. METHOD We performed a retrospective review identifying all patients with a history of tracheal stenosis secondary to COVID-19 related respiratory failure who subsequently received a tracheal resection at our institution between January 2020 and June 2023. Clinical, radiological, pathological, and surgical characteristics were recorded to describe and characterize pre-operative and intraoperative findings associated with tracheal stenosis in the setting of a previous COVID-19 infection. RESULTS We retrospectively reviewed 11 patients with COVID-19 related tracheal stenosis that required open tracheal or cricotracheal resection. The mean age was 54.1. Patients were hospitalized for a mean of 49.5 days related to COVID-19 complications. Tracheotomy was completed in 10 patients (90.9%) during their initial hospitalization with COVID-19 related respiratory failure. Patients were intubated a mean of 18.6 days prior to tracheotomy completion. Ten patients (90.9%) underwent endoscopic operative interventions for their tracheal stenosis prior to open resection. Intraoperatively, the mean stenosis length was 3.33 cm. The mean tracheal resection length was 3.96 cm. Patients were hospitalized for a mean of 8.27 days post operatively with no significant post operative complications. CONCLUSIONS Symptomatic tracheal stenosis in the setting of prolonged intubation due to COVID-19 is an under-described etiology. This is one of the largest single institution retrospective reviews that identifies 11 patients with prolonged intubation who developed symptomatic tracheal stenosis refractory to conservative management and ultimately requiring tracheal resection.
Collapse
Affiliation(s)
- Andrew Yousef
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| | - Isaac Solomon
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| | - George Cheng
- Division of Pulmonology, Critical Care, Sleep Medicine & Physiology, University of California, San Diego
| | - Samir Makani
- Department of Pulmonology and Critical Care Medicine, Scripps Hospital Encinitas, Encinitas, California
| | - Joshua Boys
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego
| | - Philip A. Weissbrod
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| |
Collapse
|
29
|
Lozano C, Pible O, Eschlimann M, Giraud M, Debroas S, Gaillard JC, Bellanger L, Taysse L, Armengaud J. Universal Identification of Pathogenic Viruses by Liquid Chromatography Coupled with Tandem Mass Spectrometry Proteotyping. Mol Cell Proteomics 2024; 23:100822. [PMID: 39084562 PMCID: PMC11795680 DOI: 10.1016/j.mcpro.2024.100822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024] Open
Abstract
Accurate and rapid identification of viruses is crucial for an effective medical diagnosis when dealing with infections. Conventional methods, including DNA amplification techniques or lateral-flow assays, are constrained to a specific set of targets to search for. In this study, we introduce a novel tandem mass spectrometry proteotyping-based method that offers a universal approach for the identification of pathogenic viruses and other components, eliminating the need for a priori knowledge of the sample composition. Our protocol relies on a time and cost-efficient peptide sample preparation, followed by an analysis with liquid chromatography coupled to high-resolution tandem mass spectrometry. As a proof of concept, we first assessed our method on publicly available shotgun proteomics datasets obtained from virus preparations and fecal samples of infected individuals. Successful virus identification was achieved with 53 public datasets, spanning 23 distinct viral species. Furthermore, we illustrated the method's capability to discriminate closely related viruses within the same sample, using alphaviruses as an example. The clinical applicability of our method was demonstrated by the accurate detection of the vaccinia virus in spiked saliva, a matrix of paramount clinical significance due to its non-invasive and easily obtainable nature. This innovative approach represents a significant advancement in pathogen detection and paves the way for enhanced diagnostic capabilities.
Collapse
Affiliation(s)
- Clément Lozano
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France.
| | - Olivier Pible
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Marine Eschlimann
- Direction Générale de l'Armement Maîtrise NRBC, Vert-le-Petit, France
| | - Mathieu Giraud
- Direction Générale de l'Armement Maîtrise NRBC, Vert-le-Petit, France
| | - Stéphanie Debroas
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Jean-Charles Gaillard
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Laurent Bellanger
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Laurent Taysse
- Direction Générale de l'Armement Maîtrise NRBC, Vert-le-Petit, France
| | - Jean Armengaud
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France.
| |
Collapse
|
30
|
Au MT, Ni J, Tang K, Wang W, Zhang L, Wang H, Zhao F, Li Z, Luo P, Lau LCM, Chan PK, Luo C, Zhou B, Zhu L, Zhang CY, Jiang T, Lauwers M, Chan JFW, Yuan S, Wen C. Blockade of endothelin receptors mitigates SARS-CoV-2-induced osteoarthritis. Nat Microbiol 2024; 9:2538-2552. [PMID: 39261580 DOI: 10.1038/s41564-024-01802-x] [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: 06/19/2023] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
Joint pain and osteoarthritis can occur as coronavirus disease 2019 (COVID-19) sequelae after infection. However, little is known about the damage to articular cartilage. Here we illustrate knee joint damage after wild-type, Delta and Omicron variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in vivo. Rapid joint injury with cystic lesions at the osteochondral junction was observed in two patients with post-COVID osteoarthritis and recapitulated in a golden Syrian hamster model. SARS-CoV-2-activated endothelin-1 signalling increased vascular permeability and caused viral spike proteins leakage into the subchondral bone. Osteoclast activation, chondrocyte dropout and cyst formation were confirmed histologically. The US Food and Drug Administration-approved endothelin receptor antagonist, macitentan, mitigated cystic lesions and preserved chondrocyte number in the acute phase of viral infection in hamsters. Delayed macitentan treatment at post-acute infection phase alleviated chondrocyte senescence and restored subchondral bone loss. It is worth noting that it could also attenuate viral spike-induced joint pain. Our work suggests endothelin receptor blockade as a novel therapeutic strategy for post-COVID arthritis.
Collapse
Affiliation(s)
- Man Ting Au
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Junguo Ni
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Kaiming Tang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wei Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Lanlan Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Hantang Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Fangyi Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Zhan Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Peng Luo
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Lawrence Chun-Man Lau
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ping-Keung Chan
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Cuiting Luo
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Bo Zhou
- Department of Biology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong Special Administrative Region, China
| | - Charlie Yuli Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Tianshu Jiang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Marianne Lauwers
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.
| | - Chunyi Wen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China.
- Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China.
| |
Collapse
|
31
|
Porat Ben Amy D, Rohana H, Azrad M, V Joachim M, Bar O, Abu El-Naaj I, Peretz A. Exploring the relationship between cycle threshold values and oral manifestations in COVID-19: a comprehensive overview. Acta Odontol Scand 2024; 83:516-521. [PMID: 39311869 PMCID: PMC11443029 DOI: 10.2340/aos.v83.41390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 07/28/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE This cross-sectional study aimed to compare oral manifestations between severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-positive and SARS-CoV-2-negative patients and to examine associations between oral symptoms, Ct values of E and N SARS-CoV-2 viral genes, and the implications of low Ct values indicating a high viral load, which is a predictive factor for the outcome of COVID-19. METHODS A total of 353 participating patients were aged ≥18 years with clinical manifestations of COVID-19 infection and tested for SARS-CoV-2 carriage at the medical center, by reverse transcriptase polymerase chain reaction (RT-PCR). All patients filled out an anonymous digital questionnaire regarding oral and general symptoms and overall medical health. Results: A significant association was found between SARS-CoV-2 carriage and dry mouth, unpleasant taste and changes in taste (p < 0.001); for example, 37.4% of the 147 SARS-CoV-2- positive participants had a dry mouth, compared to 18.9% of the 206 SARS-CoV-2- negative participants. Oral blisters were experienced by patients with an E gene Ct value of 10-20 (50%) or 21-30 (50%) (p = 0.041). Bad breath, dry mouth, unpleasant taste and changes in taste were mostly present in participants whose Ct values of both E and N genes were between 21 and 30. Conclusions: This study found significant associations between low Ct values of E and N SARS-CoV-2 viral genes and high viral load, indicating that Ct values can serve as predictive factors for COVID-19 outcomes. The findings suggest that while oral symptoms are present, the Ct values and associated high viral loads are more critical indicators of disease severity and prognosis.
Collapse
Affiliation(s)
- Dalit Porat Ben Amy
- Unit of Oral Medicine, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Hanan Rohana
- Clinical Microbiology Laboratory, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel
| | - Maya Azrad
- Clinical Microbiology Laboratory, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel
| | - Michael V Joachim
- Unit of Oral and Maxillofacial Surgery, Shamir (Assaf ha-Rofeh) Medical Center, Tzrifin, Israel; Department of Oral and Maxillofacial Surgery, the Maurice and Gabriela Goldschleger School of Dentistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ori Bar
- Department of Oral and Cranio-Maxillofacial Surgery, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel
| | - Imad Abu El-Naaj
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; Department of Oral and Cranio-Maxillofacial Surgery, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel
| | - Avi Peretz
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; Clinical Microbiology Laboratory, the Baruch Padeh "Tzafon" Medical Center, Poriya, Israel
| |
Collapse
|
32
|
Farrell ML, Bryksin AV, Ryan E, Lin J, Djeddar N, Khunteev G, Holton B, Paca M, Speller N, Merrill JT, Ross TM, Hogan RJ, Gibson G, García AJ, Shannon MP. Validation of Saliva as the Clinical Specimen Type for a University-Wide COVID-19 Surveillance Program. Viruses 2024; 16:1494. [PMID: 39339970 PMCID: PMC11437455 DOI: 10.3390/v16091494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
At the beginning of the COVID-19 pandemic, the Georgia Institute of Technology made the decision to keep the university doors open for on-campus attendance. To manage COVID-19 infection rates, internal resources were applied to develop and implement a mass asymptomatic surveillance program. The objective was to identify infections early for proper follow-on verification testing, contact tracing, and quarantine/isolation as needed. Program success depended on frequent and voluntary sample collection from over 40,000 students, faculty, and staff personnel. At that time, the nasopharyngeal (NP) swab, not saliva, was the main accepted sample type for COVID-19 testing. However, due to collection discomfort and the inability to be self-collected, the NP swab was not feasible for voluntary and frequent self-collection. Therefore, saliva was selected as the clinical sample type and validated. A saliva collection kit and a sample processing and analysis workflow were developed. The results of a clinical sample-type comparison study between co-collected and matched NP swabs and saliva samples showed 96.7% positive agreement and 100% negative agreement. During the Fall 2020 and Spring 2021 semesters, 319,988 samples were collected and tested. The program resulted in maintaining a low overall mean positivity rate of 0.78% and 0.54% for the Fall 2020 and Spring 2021 semesters, respectively. For this high-throughput asymptomatic COVID-19 screening application, saliva was an exceptionally good sample type.
Collapse
Affiliation(s)
- Michael L Farrell
- Advanced Concepts Lab, Georgia Tech Research Institute, Atlanta, GA 30318, USA
| | - Anton V Bryksin
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Emily Ryan
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Jessica Lin
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Naima Djeddar
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - German Khunteev
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Benjamin Holton
- Stamps Student Health Services, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Miles Paca
- Advanced Concepts Lab, Georgia Tech Research Institute, Atlanta, GA 30318, USA
| | - Nicholas Speller
- Advanced Concepts Lab, Georgia Tech Research Institute, Atlanta, GA 30318, USA
| | - James T Merrill
- Advanced Concepts Lab, Georgia Tech Research Institute, Atlanta, GA 30318, USA
| | - Ted M Ross
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Robert J Hogan
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Greg Gibson
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Michael P Shannon
- Advanced Concepts Lab, Georgia Tech Research Institute, Atlanta, GA 30318, USA
- Office of the President, University of North Georgia, Dahlonega, GA 30597, USA
| |
Collapse
|
33
|
Niu C, Liang T, Chen Y, Zhu S, Zhou L, Chen N, Qian L, Wang Y, Li M, Zhou X, Cui J. SARS-CoV-2 spike protein induces the cytokine release syndrome by stimulating T cells to produce more IL-2. Front Immunol 2024; 15:1444643. [PMID: 39359733 PMCID: PMC11445618 DOI: 10.3389/fimmu.2024.1444643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Cytokine release syndrome (CRS) is one of the leading causes of mortality in patients with COVID-19 caused by the SARS-CoV-2 coronavirus. However, the mechanism of CRS induced by SARS-CoV-2 is vague. Methods Using spike protein combined with IL-2, IFN-γ, and TNF-α to stimulate human peripheral blood mononuclear cells (PBMCs) to secrete CRS-related cytokines, the content of cytokines in the supernatant was detected, and the effects of NK, T, and monocytes were analyzed. Results This study shows that dendritic cells loaded with spike protein of SARS-CoV-2 stimulate T cells to release much more interleukin-2 (IL-2,) which subsequently cooperates with spike protein to facilitate PBMCs to release IL-1β, IL-6, and IL-8. These effects are achieved via IL-2 stimulation of NK cells to release tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), as well as T cells to release IFN-γ Mechanistically, IFN-γ and TNF-α enhance the transcription of CD40, and the interaction of CD40 and its ligand stabilizes the membrane expression of toll-like receptor 4 (TLR4) that serves as a receptor of spike protein on the surface of monocytes. As a result, there is a constant interaction between spike protein and TLR4, leading to continuous activation of nuclear factor-κ-gene binding (NF-κB). Furthermore, TNF-α also activates NF-κB signaling in monocytes, which further cooperates with IFN-γ and spike protein to modulate NF-κB-dependent transcription of CRS-related inflammatory cytokines. Discussion Targeting TNF-α/IFN-γ in combination with TLR4 may represent a promising therapeutic approach for alleviating CRS in individuals with COVID-19.
Collapse
Affiliation(s)
- Chao Niu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yongchong Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Qian
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yufeng Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Min Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xin Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Seaman WT, Keener O, Nanayakkara D, Mollan KR, Premkumar L, Cuadra EC, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. Sci Rep 2024; 14:21788. [PMID: 39294156 PMCID: PMC11411107 DOI: 10.1038/s41598-024-67504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 07/11/2024] [Indexed: 09/20/2024] Open
Abstract
Oral fluids provide ready detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to evaluate relationships between oral virus, oral and systemic anti-SARS-CoV-2-specific antibodies, and symptoms. Oral fluids (saliva/throat wash (saliva/TW)) and serum were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+ human participants (n = 45). SARS-CoV-2 RT-qPCR and N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR for subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA and ELISA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. At time of enrollment (baseline, BL), LFA-detected N-antigen in 86% of TW and was immunoblot-confirmed. However, only 3/17 were saliva/TW qPCR+ . Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three anti-spike sero-negative participants suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At enrollment, symptomatic participants demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (63%/54%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral and serum IgG correlated 100% with NP+ PCR status. Cough and fatigue severity (p = 0.010 and 0.018 respectively), and presence of weakness, nausea, and composite upper respiratory symptoms (p = 0.037, 0.005, and 0.017, respectively) were negatively associated with saliva IgM but not TW or serum IgM. Throat wash IgM levels were higher in women compared to men, although the association did not reach statistical significance (median: 290 (female) versus 0.697, p = 0.056). Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms and early oral IgM responses during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Olive Keener
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- North Carolina School of Math and Science, Durham, NC, USA
| | - Dinelka Nanayakkara
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katie R Mollan
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edwing Centeno Cuadra
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corbin D Jones
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey Pettifor
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie M Bowman
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
| | | | - Jennifer Webster-Cyriaque
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA.
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
35
|
Jamil K, Abdulrazack N, Fakhraldeen S, Kamal H, Al-Mutairi A, Al-Feili B, Ahmed I, Kumar V. Detection of SARS-CoV-2 on the environmental surfaces and its implications for pandemic preparedness. Front Public Health 2024; 12:1396334. [PMID: 39319298 PMCID: PMC11420012 DOI: 10.3389/fpubh.2024.1396334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Even though death due to COVID-19 is no longer a public health emergency, less virulent but highly transmissible forms of SARS-CoV-2 continue to spread in many countries leading to outbreaks and rise in hospitalizations in the affected regions. Lessons learned during the pandemic must be put into action to protect the world's population from another catastrophe like COVID-19. Novel approaches that were developed for tracking the spread of SARS-CoV-2 included analysis of wastewater, air samples, and various environmental surfaces. We conducted a study in Kuwait during the peak of COVID-19 pandemic to examine if SARS-CoV-2 could be detected in swabs taken from frequently touched environmental surfaces. We selected 12 Cooperative Society Stores-two from each governorate of Kuwait-for collection of surface samples. The Cooperative Society Stores are widely distributed across the whole country and cater to daily household needs including groceries and other essential items. These stores operated even during the "lockdown" imposed at the height of the pandemic. We collected swabs from high-touch surfaces including the handles of the shopping carts and freezers, the elevators, the keypads of the point-of-service terminals of cash counters, and the automated teller machines. All the surfaces tested showed a variable presence of SARS-CoV-2 by reverse transcriptase quantitative PCR, showing the validity of the proof-of-concept study. Monitoring of the presence of SARS-CoV-2 by surface sampling thus offers a cheap but effective means of environmental surveillance for coronaviruses. We therefore strongly recommend the addition of surface environmental sampling as a strategy for pandemic preparedness everywhere.
Collapse
Affiliation(s)
- Kazi Jamil
- Environment and Life Sciences Research Center, Kuwait Institute for Scientific Research, Safat, Kuwait
| | | | | | | | | | | | | | - Vinod Kumar
- Environment and Life Sciences Research Center, Kuwait Institute for Scientific Research, Safat, Kuwait
| |
Collapse
|
36
|
Braeye T, Proesmans K, Van Cauteren D, Brondeel R, Hens N, Vermeiren E, Hammami N, Rosas A, Taame A, André E, Cuypers L. Personal characteristics and transmission dynamics associated with SARS-CoV-2 semi-quantitative PCR test results: an observational study from Belgium, 2021-2022. Front Public Health 2024; 12:1429021. [PMID: 39319296 PMCID: PMC11420023 DOI: 10.3389/fpubh.2024.1429021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Following harmonization efforts by the Belgian National Reference Center for SARS-CoV-2, semi-quantitative PCR test (SQ-PCR) results, used as a proxy for viral load, were routinely collected after performing RT-qPCR tests. Methods We investigated both the personal characteristics associated with SQ-PCR results and the transmission dynamics involving these results. We used person-level laboratory test data and contact tracing data collected in Belgium from March 2021 to February 2022. Personal characteristics (age, sex, vaccination, and laboratory-confirmed prior infection) and disease stage by date of symptom onset were analyzed in relation to SQ-PCR results using logistic regression. Vaccine effectiveness (VE) against a high viral load (≥107 copies/mL) was estimated from the adjusted probabilities. Contact tracing involves the mandatory testing of high-risk exposure contacts (HREC) after contact with an index case. Odds ratios for test positivity and high viral load in HREC were calculated based on the SQ-PCR result of the index case using logistic regression models adjusted for age, sex, immunity status (vaccination, laboratory-confirmed prior infection), variant (Alpha, Delta, Omicron), calendar time, and contact tracing covariates. Results We included 909,157 SQ-PCR results of COVID-19 cases, 379,640 PCR results from index cases, and 72,052 SQ-PCR results of HREC. High viral load was observed more frequently among recent cases, symptomatic cases, cases over 25 years of age, and those not recently vaccinated (>90 days). The vaccine effectiveness (VE) of the primary schedule in the first 30 days after vaccination was estimated at 47.3% (95%CI 40.8-53.2) during the Delta variant period. A high viral load in index cases was associated with an increased test positivity in HREC (OR 2.7, 95%CI 2.62-2.79) and, among those testing positive, an increased likelihood of a high viral load (OR 2.84, 95%CI 2.53-3.19).
Collapse
Affiliation(s)
- Toon Braeye
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
- I-BioStat, Data Science Institute, Hasselt University, Hasselt, Belgium
| | - Kristiaan Proesmans
- Faculty of Pharmaceutical Sciences, Department of Bio-analysis, Ghent University, Ghent, Belgium
| | | | - Ruben Brondeel
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
| | - Niel Hens
- I-BioStat, Data Science Institute, Hasselt University, Hasselt, Belgium
- Centre for Health Economic Research and Modelling Infectious Diseases, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Elias Vermeiren
- Epidemiology of Infectious Diseases, Sciensano, Brussels, Belgium
| | - Naïma Hammami
- Department of Care, Infection Prevention and Control, Flemish Community, Brussels, Belgium
| | - Angel Rosas
- Direction Surveillance des Maladies Infectieuses, Agence out une Vie de Qualité (AVIQ), Charleroi, Belgium
| | - Adrae Taame
- Cellule de médecine préventive- Direction santé et aide aux personnes – Vivalis/Cocom, Brussels, Belgium
| | - Emmanuel André
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Lize Cuypers
- Department of Laboratory Medicine, National Reference Centre for Respiratory Pathogens, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Kowitdamrong E, Anoma S, Loykaew T, Hansasuta P, Bhattarakosol P. ƩS COVID-19 is a rapid high throughput and sensitive one-step quadruplex real-time RT-PCR assay. Sci Rep 2024; 14:20590. [PMID: 39232060 PMCID: PMC11374890 DOI: 10.1038/s41598-024-71705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Real-time reverse transcription polymerase chain reaction (RT-PCR), a standard method recommended for the diagnosis of coronavirus disease 2019 (COVID-19) requires 2-4 h to get the result. Although antigen test kit (ATK) is used for COVID-19 screening within 15-30 min, the drawback is its limited sensitivity. Hence, a rapid one-step quadruplex real-time RT-PCR assay: termed ƩS COVID-19 targeting ORF1ab, ORF3a, and N genes of SARS-CoV-2; and Avocado sunblotch viroid (ASBVd) as an internal control was developed. Based on strategies including designing high melting temperature primers with short amplicons, applying a fast ramp rate, minimizing hold time, and reducing the range between denaturation and annealing/extension temperatures; the assay could be accomplished within 25 min. The limit of detection of ORF1ab, ORF3a, and N genes were 1.835, 1.310, and 1 copy/reaction, respectively. Validation was performed in 205 combined nasopharyngeal and oropharyngeal swabs. The sensitivity, specificity, positive predictive value, and negative predictive value were 92.8%, 100%, 100%, and 97.1%, respectively with 96.7% accuracy. Cohen's Kappa was 0.93. The newly developed rapid real-time RT-PCR assay was highly sensitive, specific, and fast, making it suitable for use as an alternative method to support laboratory diagnosis of COVID-19 in outpatient and emergency departments.
Collapse
Affiliation(s)
- Ekasit Kowitdamrong
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Applied Medical Virology, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Sasiprapa Anoma
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Applied Medical Virology, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thitiya Loykaew
- Department of Microbiology, King Chulalongkorn Memorial Hospital, Thai Red Cross, Bangkok, 10330, Thailand
| | - Pokrath Hansasuta
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Parvapan Bhattarakosol
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Applied Medical Virology, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
38
|
McBride JA, DeMuri G, Nelson C, Cranley S, Alfaro E, Eickhoff J, Lim A, O'Connor S, Wald ER. Performance characteristics of "lollipop" swabs for the diagnosis of infection with SARS-CoV-2. Diagn Microbiol Infect Dis 2024; 110:116404. [PMID: 38896889 DOI: 10.1016/j.diagmicrobio.2024.116404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Common biologic samples used to diagnose COVID-19 include nasopharyngeal, nasal, or oropharyngeal swabs, and salivary samples. The performance characteristics of a sucked "lollipop" swab to detect SARS-CoV-2 virus is assessed in four small sub-studies. METHODS In each sub-study, a flocked swab was sucked for 20 s and submitted for PCR detection of SARS-CoV-2 virus. RESULTS Across all studies, 52 of 69 (75.4%) COVID-19 positive participants had positive "lollipop" swabs. Twelve of the 17 COVID-19 positive participants with negative "lollipop" swabs had known corresponding cycle threshold values of >37 from their nasal/nasopharyngeal swabs, an indication of low viral load at time of sampling. In a paired samples sub-study, the sensitivity and specificity of the "lollipop" swabs were 100% and 98%. CONCLUSIONS "Lollipop" swabs performed satisfactorily especially in individuals with acute infection of COVID-19. "Lollipop" swabs are a simple method of sample collection for detecting SARS-CoV-2 virus and warrants additional consideration.
Collapse
Affiliation(s)
- Joseph A McBride
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States; Univeristy of Wisconsin School of Medicine and Public Health, Department of Medicine, Madison, WI, United States.
| | - Gregory DeMuri
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| | - Cassandra Nelson
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| | - Suzanne Cranley
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| | - Elena Alfaro
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| | - Jens Eickhoff
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| | - Ailam Lim
- University of Wisconsin Veterinary Diagnostic Laboratory Madison, WI, United States
| | - Shelby O'Connor
- University of Wisconsin School of Medicine and Public Health Department of Pathology and Laboratory Medicine Madison WI, United States
| | - Ellen R Wald
- University of Wisconsin School of Medicine and Public Health Department of Pediatrics Madison, WI, United States
| |
Collapse
|
39
|
Yang TH, Syu GD, Chen CS, Chen GR, Jhong SE, Lin PH, Lin PC, Wang YC, Shah P, Tseng YY, Wu WS. BAPCP: A comprehensive and user-friendly web tool for identifying biomarkers from protein microarray technologies. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 254:108260. [PMID: 38878357 DOI: 10.1016/j.cmpb.2024.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/06/2024] [Accepted: 05/29/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Proteome microarrays are one of the popular high-throughput screening methods for large-scale investigation of protein interactions in cells. These interactions can be measured on protein chips when coupled with fluorescence-labeled probes, helping indicate potential biomarkers or discover drugs. Several computational tools were developed to help analyze the protein chip results. However, existing tools fail to provide a user-friendly interface for biologists and present only one or two data analysis methods suitable for limited experimental designs, restricting the use cases. METHODS In order to facilitate the biomarker examination using protein chips, we implemented a user-friendly and comprehensive web tool called BAPCP (Biomarker Analysis tool for Protein Chip Platforms) in this research to deal with diverse chip data distributions. RESULTS BAPCP is well integrated with standard chip result files and includes 7 data normalization methods and 7 custom-designed quality control/differential analysis filters for biomarker extraction among experiment groups. Moreover, it can handle cost-efficient chip designs that repeat several blocks/samples within one single slide. Using experiments of the human coronavirus (HCoV) protein microarray and the E. coli proteome chip that helps study the immune response of Kawasaki disease as examples, we demonstrated that BAPCP can accelerate the time-consuming week-long manual biomarker identification process to merely 3 min. CONCLUSIONS The developed BAPCP tool provides substantial analysis support for protein interaction studies and conforms to the necessity of expanding computer usage and exchanging information in bioscience and medicine. The web service of BAPCP is available at https://cosbi.ee.ncku.edu.tw/BAPCP/.
Collapse
Affiliation(s)
- Tzu-Hsien Yang
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan.
| | - Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, No. 1, University Road, Tainan 701, Taiwan.
| | - Chien-Sheng Chen
- Department of Food Safety/Hygiene and Risk Management, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Guan-Ru Chen
- Department of Electrical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Song-En Jhong
- Department of Electrical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Po-Heng Lin
- Department of Electrical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Pei-Chun Lin
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Yun-Cih Wang
- Department of Electrical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| | - Pramod Shah
- Institute of Systems Biology and Bioinformatics, Department of Biomedical Sciences and Engineering, College of Health Sciences and Technology, National Central University, No. 300, Zhongda Rd., Zhongli District, 320317 Taoyuan, Taiwan.
| | - Yan-Yuan Tseng
- Center for Molecular Medicine and Genetics, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| | - Wei-Sheng Wu
- Department of Electrical Engineering, National Cheng Kung University, No. 1, University Road, 701 Tainan, Taiwan.
| |
Collapse
|
40
|
Ip JD, Chu WM, Chan WM, Chu AWH, Leung RCY, Peng Q, Tam AR, Chan BPC, Cai JP, Yuen KY, Kok KH, Shi Y, Hung IFN, To KKW. The significance of recurrent de novo amino acid substitutions that emerged during chronic SARS-CoV-2 infection: an observational study. EBioMedicine 2024; 107:105273. [PMID: 39146693 PMCID: PMC11379563 DOI: 10.1016/j.ebiom.2024.105273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND De novo amino acid substitutions (DNS) frequently emerge among immunocompromised patients with chronic SARS-CoV-2 infection. While previous studies have reported these DNS, their significance has not been systematically studied. METHODS We performed a review of DNS that emerged during chronic SARS-CoV-2 infection. We searched PubMed until June 2023 using the keywords "(SARS-CoV-2 or COVID-19) and (mutation or sequencing) and ((prolonged infection) or (chronic infection) or (long term))". We included patients with chronic SARS-CoV-2 infection who had SARS-CoV-2 sequencing performed for at least 3 time points over at least 60 days. We also included 4 additional SARS-CoV-2 patients with chronic infection of our hospital not reported previously. We determined recurrent DNS that has appeared in multiple patients and determined the significance of these mutations among epidemiologically-significant variants. FINDINGS A total of 34 cases were analyzed, including 30 that were published previously and 4 from our hospital. Twenty two DNS appeared in ≥3 patients, with 14 (64%) belonging to lineage-defining mutations (LDMs) of epidemiologically-significant variants and 10 (45%) emerging among chronically-infected patients before the appearance of the corresponding variant. Notably, nsp9-T35I substitution (Orf1a T4175I) emerged in all three patients with BA.2.2 infection in 2022 before the appearance of Variants of Interest that carry nsp9-T35I as LDM (EG.5 and BA.2.86/JN.1). Structural analysis suggests that nsp9-T35I substitution may affect nsp9-nsp12 interaction, which could be critical for the function of the replication and transcription complex. INTERPRETATION DNS that emerges recurrently in different chronically-infected patients may be used as a marker for potential epidemiologically-significant variants. FUNDING Theme-Based Research Scheme [T11/709/21-N] of the Research Grants Council (See acknowledgements for full list).
Collapse
Affiliation(s)
- Jonathan Daniel Ip
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wing-Ming Chu
- Division of Infectious Diseases, Department of Medicine, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Wan-Mui Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Allen Wing-Ho Chu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Rhoda Cheuk-Ying Leung
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Qi Peng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Anthony Raymond Tam
- Division of Infectious Diseases, Department of Medicine, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Brian Pui-Chun Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China; Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kin-Hang Kok
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Yi Shi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Ivan Fan-Ngai Hung
- Division of Infectious Diseases, Department of Medicine, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Infectious Diseases Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China; Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
41
|
Shahbazi E, Moradi A, Mollasalehi H, Mohebbi SR. Unravelling the diagnostic methodologies for SARS-CoV-2; the Indispensable need for developing point-of-care testing. Talanta 2024; 275:126139. [PMID: 38696900 DOI: 10.1016/j.talanta.2024.126139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/13/2024] [Accepted: 04/20/2024] [Indexed: 05/04/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused COVID-19 pandemic that continues to be a global menace and since its emergence in the late 2019, SARS-CoV-2 has been vigorously spreading throughout the globe putting the whole world into a multidimensional calamity. The suitable diagnosis strategies are on the front line of the battle against preventing the spread of infections. Since the clinical manifestation of COVID-19 is shared between various diseases, detection of the unique impacts of the pathogen on the host along with the diagnosis of the virus itself should be addressed. Employing the most suitable approaches to specifically, sensitively and effectively recognize the infected cases may be a real game changer in controlling the outbreak and the crisis management. In that matter, point-of-care assays (POC) appears to be the potential option, due to sensitivity, specificity, affordable, and availability. Here we brief the most recent findings about the virus, its variants, and the conventional methods that have been used for its detection, along with the POC strategies that have been applied to the virus diagnosis and the developing technologies which can accelerate the diagnosis procedure yet maintain its efficiency.
Collapse
Affiliation(s)
- Erfan Shahbazi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Asma Moradi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Hamidreza Mollasalehi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Roohi A, Gharagozlou S. Vitamin D supplementation and calcium: Many-faced gods or nobody in fighting against Corona Virus Disease 2019. Clin Nutr ESPEN 2024; 62:172-184. [PMID: 38901939 DOI: 10.1016/j.clnesp.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
In December 2019, Corona Virus Disease 2019 (COVID-19) was first identified and designated as a pandemic in March 2020 due to rapid spread of the virus globally. At the beginning of the pandemic, only a few treatment options, mainly focused on supportive care and repurposing medications, were available. Due to its effects on immune system, vitamin D was a topic of interest during the pandemic, and researchers investigated its potential impact on COVID-19 outcomes. However, the results of studies about the impact of vitamin D on the disease are inconclusive. In the present narrative review, different roles of vitamin D regarding the COVID-19 have been discussed to show that vitamin D supplementation should be recommended carefully.
Collapse
Affiliation(s)
- Azam Roohi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
43
|
Sharma S, Caputi M, Asghar W. Development of a Diagnostic Microfluidic Chip for SARS-CoV-2 Detection in Saliva and Nasopharyngeal Samples. Viruses 2024; 16:1190. [PMID: 39205164 PMCID: PMC11360425 DOI: 10.3390/v16081190] [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: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
The novel coronavirus SARS-CoV-2 was first isolated in late 2019; it has spread to all continents, infected over 700 million people, and caused over 7 million deaths worldwide to date. The high transmissibility of the virus and the emergence of novel strains with altered pathogenicity and potential resistance to therapeutics and vaccines are major challenges in the study and treatment of the virus. Ongoing screening efforts aim to identify new cases to monitor the spread of the virus and help determine the danger connected to the emergence of new variants. Given its sensitivity and specificity, nucleic acid amplification tests (NAATs) such as RT-qPCR are the gold standard for SARS-CoV-2 detection. However, due to high costs, complexity, and unavailability in low-resource and point-of-care (POC) settings, the available RT-qPCR assays cannot match global testing demands. An alternative NAAT, RT-LAMP-based SARS-CoV-2 detection offers scalable, low-cost, and rapid testing capabilities. We have developed an automated RT-LAMP-based microfluidic chip that combines the RNA isolation, purification, and amplification steps on the same device and enables the visual detection of SARS-CoV-2 within 40 min from saliva and nasopharyngeal samples. The entire assay is executed inside a uniquely designed, inexpensive disposable microfluidic chip, where assay components and reagents have been optimized to provide precise and qualitative results and can be effectively deployed in POC settings. Furthermore, this technology could be easily adapted for other novel emerging viruses.
Collapse
Affiliation(s)
- Sandhya Sharma
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
| | - Massimo Caputi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Waseem Asghar
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
- Department of Biological Sciences (Courtesy Appointment), Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
44
|
Bayraktar C, Kayabolen A, Odabas A, Durgun A, Kok I, Sevinc K, Supramaniam A, Idris A, Bagci-Onder T. ACE2-Decorated Virus-Like Particles Effectively Block SARS-CoV-2 Infection. Int J Nanomedicine 2024; 19:6931-6943. [PMID: 39005960 PMCID: PMC11246629 DOI: 10.2147/ijn.s446093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/02/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Over the past three years, extensive research has been dedicated to understanding and combating COVID-19. Targeting the interaction between the SARS-CoV-2 Spike protein and the ACE2 receptor has emerged as a promising therapeutic strategy against SARS-CoV-2. This study aimed to develop ACE2-coated virus-like particles (ACE2-VLPs), which can be utilized to prevent viral entry into host cells and efficiently neutralize the virus. Methods Virus-like particles were generated through the utilization of a packaging plasmid in conjunction with a plasmid containing the ACE2 envelope sequence. Subsequently, ACE2-VLPs and ACE2-EVs were purified via ultracentrifugation. The quantification of VLPs was validated through multiple methods, including Nanosight 3000, TEM imaging, and Western blot analysis. Various packaging systems were explored to optimize the ACE2-VLP configuration for enhanced neutralization capabilities. The evaluation of neutralization effectiveness was conducted using pseudoviruses bearing different spike protein variants. Furthermore, the study assessed the neutralization potential against the Omicron BA.1 variant in Vero E6 cells. Results ACE2-VLPs showed a high neutralization capacity even at low doses and demonstrated superior efficacy in in vitro pseudoviral assays compared to extracellular vesicles carrying ACE2. ACE2-VLPs remained stable under various environmental temperatures and effectively blocked all tested variants of concern in vitro. Notably, they exhibited significant neutralization against Omicron BA.1 variant in Vero E6 cells. Given their superior efficacy compared to extracellular vesicles and proven success against live virus, ACE2-VLPs stand out as crucial candidates for treating SARS-CoV-2 infections. Conclusion This novel therapeutic approach of coating VLPs with receptor particles provides a proof-of-concept for designing effective neutralization strategies for other viral diseases in the future.
Collapse
Affiliation(s)
- Canan Bayraktar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Alisan Kayabolen
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Arda Odabas
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Aysegul Durgun
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Ipek Kok
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Kenan Sevinc
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Aroon Supramaniam
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, Brisbane, QLD, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, Brisbane, QLD, Australia
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Tugba Bagci-Onder
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| |
Collapse
|
45
|
Ko JH, Kim CM, Bang MS, Lee DY, Kim DY, Seo JW, Yun NR, Yang JY, Peck KR, Lee KW, Jung SH, Bang HJ, Bae WK, Kim TJ, Byeon KH, Kim SH, Kim DM. Risk Factors for Impaired Cellular or Humoral Immunity after Three Doses of SARS-CoV-2 Vaccine in Healthy and Immunocompromised Individuals. Vaccines (Basel) 2024; 12:752. [PMID: 39066390 PMCID: PMC11281526 DOI: 10.3390/vaccines12070752] [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: 05/21/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Background: We aimed to identify the risk factors for impaired cellular and humoral immunity after three doses of the SARS-CoV-2 vaccine. Methods: Six months after the third vaccine dose, T-cell immunity was evaluated using interferon-gamma release assays (IGRAs) in 60 healthy and 139 immunocompromised (IC) individuals, including patients with hematologic malignancy (HM), solid malignancy (SM), rheumatic disease (RD), and kidney transplantation (KT). Neutralizing antibody titers were measured using the plaque reduction neutralization test (PRNT) and surrogate virus neutralization test (sVNT). Results: T-cell immunity results showed that the percentages of IGRA-positive results using wild-type/alpha spike protein (SP) and beta/gamma SP were 85% (51/60) and 75% (45/60), respectively, in healthy individuals and 45.6% (62/136) and 40.4% (55/136), respectively, in IC individuals. IC with SM or KT showed a high percentage of IGRA-negative results. The underlying disease poses a risk for impaired cellular immune response to wild-type SP. The risk was low when all doses were administered as mRNA vaccines. The risk factors for an impaired cellular immune response to beta/gamma SP were underlying disease and monocyte%. In the sVNT using wild-type SP, 12 of 191 (6.3%) individuals tested negative. In the PRNT of 46 random samples, 6 (13%) individuals tested negative for the wild-type virus, and 19 (41.3%) tested negative with omicrons. KT poses a risk for an impaired humoral immune response. Conclusions: Underlying disease poses a risk for impaired cellular immune response after the third dose of the SARS-CoV-2 vaccine; KT poses a risk for impaired humoral immune response, emphasizing the requirement of precautions in patients.
Collapse
Affiliation(s)
- Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Choon-Mee Kim
- Department of Premedical Science, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea;
| | - Mi-Seon Bang
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Da-Yeon Lee
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Da-Young Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Jun-Won Seo
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Na-Ra Yun
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Jin-Young Yang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Kyong-Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Kyo-Won Lee
- Division of Transplantation, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Sung-Hoon Jung
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Hyun-Jin Bang
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Woo-Kyun Bae
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea;
| | - Kyeong-Hwan Byeon
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea;
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Dong-Min Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| |
Collapse
|
46
|
Liu P, Cao K, Dai G, Chen T, Zhao Y, Xu H, Xu X, Cao Q, Zhan Y, Zuo X. Omicron variant and pulmonary involvements: a chest imaging analysis in asymptomatic and mild COVID-19. Front Public Health 2024; 12:1325474. [PMID: 39035180 PMCID: PMC11258674 DOI: 10.3389/fpubh.2024.1325474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Objectives To identify clinical characteristics and risk factors for pulmonary involvements in asymptomatic and mildly symptomatic patients infected with SARS-CoV-2 Omicron variant by chest imaging analysis. Methods Detailed data and chest computed tomography (CT) imaging features were retrospectively analyzed from asymptomatic and mildly symptomatic patients infected with Omicron between 24 April and 10 May 2022. We scored chest CT imaging features and categorized the patients into obvious pulmonary involvements (OPI) (score > 2) and not obvious pulmonary involvements (NOPI) (score ≤ 2) groups based on the median score. The risk factors for OPI were identified with analysis results visualized by nomogram. Results In total, 339 patients were included (145 were male and 194 were female), and the most frequent clinical symptoms were cough (75.5%); chest CT imaging features were mostly linear opacities (42.8%). Pulmonary involvements were more likely to be found in the left lower lung lobe, with a significant difference in the lung total severity score of the individual lung lobes (p < 0.001). Logistic regression analysis revealed age stratification [odds ratio (OR) = 1.92, 95% confidence interval (CI) (1.548-2.383); p < 0.001], prolonged nucleic acid negative conversion time (NCT) (NCT > 8d) [OR = 1.842, 95% CI (1.104-3.073); p = 0.019], and pulmonary diseases [OR = 4.698, 95% CI (1.159-19.048); p = 0.03] as independent OPI risk factors. Conclusion Asymptomatic and mildly symptomatic patients infected with Omicron had pulmonary involvements which were not uncommon. Potential risk factors for age stratification, prolonged NCT, and pulmonary diseases can help clinicians to identify OPI in asymptomatic and mildly symptomatic patients infected with Omicron.
Collapse
Affiliation(s)
- Peiben Liu
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kejun Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guanqun Dai
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingzhen Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yifan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hai Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoquan Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quan Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiyang Zhan
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiangrong Zuo
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
47
|
Kudryavtsev DS, Mozhaeva VA, Ivanov IA, Siniavin AE, Kalmykov AS, Gritchenko AS, Khlebtsov BN, Wang SP, Kang B, Tsetlin VI, Balykin VI, Melentiev PN. Optical detection of infectious SARS-CoV-2 virions by counting spikes. NANOSCALE 2024; 16:12424-12430. [PMID: 38887059 DOI: 10.1039/d4nr01236d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Existing methods for the mass detection of viruses are limited to the registration of small amounts of a viral genome or specific protein markers. In spite of high sensitivity, the applied methods cannot distinguish between virulent viral particles and non-infectious viral particle debris. We report an approach to solve this long-standing challenge using the SARS-CoV-2 virus as an example. We show that wide-field optical microscopy with the state-of-the-art mesoscopic fluorescent labels, formed by a core-shell plasmonic nanoparticle with fluorescent dye molecules in the core-shell that are strongly coupled to the plasmonic nanoparticle, not only rapidly, i.e. in less than 20 minutes after sampling, detects SARS-CoV-2 virions directly in a patient sample without a pre-concentration step, but can also distinguish between infectious and non-infectious virus strains by counting the spikes on the lipid envelope of individual viral particles.
Collapse
Affiliation(s)
- Denis S Kudryavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the RAS, Moscow 117997, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Vera A Mozhaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the RAS, Moscow 117997, Russia
| | - Igor A Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the RAS, Moscow 117997, Russia
| | - Andrey E Siniavin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the RAS, Moscow 117997, Russia
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ivanovsky Institute of Virology, Ministry of Health, Moscow, 123098, Russia
| | | | | | - Boris N Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences, Saratov, 410049, Russia
| | - Shao-Peng Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Bin Kang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the RAS, Moscow 117997, Russia
| | | | - Pavel N Melentiev
- Institute of Spectroscopy RAS, Moscow, Troitsk 108840, Russia.
- Higher School of Economics, National Research University, Moscow, 101000, Russia
| |
Collapse
|
48
|
Asplin P, Mancy R, Finnie T, Cumming F, Keeling MJ, Hill EM. Symptom propagation in respiratory pathogens of public health concern: a review of the evidence. J R Soc Interface 2024; 21:20240009. [PMID: 39045688 PMCID: PMC11267474 DOI: 10.1098/rsif.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024] Open
Abstract
Symptom propagation occurs when the symptom set an individual experiences is correlated with the symptom set of the individual who infected them. Symptom propagation may dramatically affect epidemiological outcomes, potentially causing clusters of severe disease. Conversely, it could result in chains of mild infection, generating widespread immunity with minimal cost to public health. Despite accumulating evidence that symptom propagation occurs for many respiratory pathogens, the underlying mechanisms are not well understood. Here, we conducted a scoping literature review for 14 respiratory pathogens to ascertain the extent of evidence for symptom propagation by two mechanisms: dose-severity relationships and route-severity relationships. We identify considerable heterogeneity between pathogens in the relative importance of the two mechanisms, highlighting the importance of pathogen-specific investigations. For almost all pathogens, including influenza and SARS-CoV-2, we found support for at least one of the two mechanisms. For some pathogens, including influenza, we found convincing evidence that both mechanisms contribute to symptom propagation. Furthermore, infectious disease models traditionally do not include symptom propagation. We summarize the present state of modelling advancements to address the methodological gap. We then investigate a simplified disease outbreak scenario, finding that under strong symptom propagation, isolating mildly infected individuals can have negative epidemiological implications.
Collapse
Affiliation(s)
- Phoebe Asplin
- EPSRC & MRC Centre for Doctoral Training in Mathematics for Real-World Systems, University of Warwick, Coventry, UK
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
| | - Rebecca Mancy
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- MRC/CSO Social and Public Health Sciences Unit, University of Glasgow, Glasgow, UK
| | - Thomas Finnie
- Data, Analytics and Surveillance, UK Health Security Agency, London, UK
| | - Fergus Cumming
- Foreign, Commonwealth and Development Office, London, UK
| | - Matt J. Keeling
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Edward M. Hill
- Mathematics Institute, University of Warwick, Coventry, UK
- The Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, UK
| |
Collapse
|
49
|
Sasso-Cerri E, Martinelli VD, de Oliveira SA, da Silva AAS, de Moraes JCG, Cerri PS. Submandibular Gland Pathogenesis Following SARS-CoV-2 Infection and Implications for Xerostomia. Int J Mol Sci 2024; 25:6820. [PMID: 38999930 PMCID: PMC11241347 DOI: 10.3390/ijms25136820] [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: 05/09/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 07/14/2024] Open
Abstract
Although SARS-CoV-2 induces mucin hypersecretion in the respiratory tract, hyposalivation/xerostomia has been reported by COVID-19 patients. We evaluate the submandibular gland (SMGs) pathogenesis in SARS-CoV-2-infected K18-hACE2 mice, focusing on the impact of infection on the mucin production and structural integrity of acini, ductal system, myoepithelial cells (MECs) and telocytes. The spike protein, the nucleocapsid protein, hACE2, actin, EGF, TNF-α and IL-1β were detected by immunofluorescence, and the Egfr and Muc5b expression was evaluated. In the infected animals, significant acinar hypertrophy was observed in contrast to ductal atrophy. Nucleocapsid proteins and/or viral particles were detected in the SMG cells, mainly in the nuclear membrane-derived vesicles, confirming the nuclear role in the viral formation. The acinar cells showed intense TNF-α and IL-1β immunoexpression, and the EGF-EGFR signaling increased, together with Muc5b upregulation. This finding explains mucin hypersecretion and acinar hypertrophy, which compress the ducts. Dying MECs and actin reduction were also observed, indicating failure of contraction and acinar support, favoring acinar hypertrophy. Viral assembly was found in the dying telocytes, pointing to these intercommunicating cells as viral transmitters in SMGs. Therefore, EGF-EGFR-induced mucin hypersecretion was triggered by SARS-CoV-2 in acinar cells, likely mediated by cytokines. The damage to telocytes and MECs may have favored the acinar hypertrophy, leading to ductal obstruction, explaining xerostomia in COVID-19 patients. Thus, acinar cells, telocytes and MECs may be viral targets, which favor replication and cell-to-cell viral transmission in the SMG, corroborating the high viral load in saliva of infected individuals.
Collapse
Affiliation(s)
- Estela Sasso-Cerri
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Dental School–São Paulo State University (UNESP), Araraquara 14801-903, Brazil; (V.D.M.); (J.C.G.d.M.)
| | - Vitor Dallacqua Martinelli
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Dental School–São Paulo State University (UNESP), Araraquara 14801-903, Brazil; (V.D.M.); (J.C.G.d.M.)
| | - Salmo Azambuja de Oliveira
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo 04023-900, Brazil; (S.A.d.O.); (A.A.S.d.S.)
| | - André Acácio Souza da Silva
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo 04023-900, Brazil; (S.A.d.O.); (A.A.S.d.S.)
| | - Juliana Cerini Grassi de Moraes
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Dental School–São Paulo State University (UNESP), Araraquara 14801-903, Brazil; (V.D.M.); (J.C.G.d.M.)
| | - Paulo Sérgio Cerri
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Dental School–São Paulo State University (UNESP), Araraquara 14801-903, Brazil; (V.D.M.); (J.C.G.d.M.)
| |
Collapse
|
50
|
Claus J, ten Doesschate T, Taks E, Debisarun PA, Smits G, van Binnendijk R, van der Klis F, Verhagen LM, de Jonge MI, Bonten MJM, Netea MG, van de Wijgert JHHM. Determinants of Systemic SARS-CoV-2-Specific Antibody Responses to Infection and to Vaccination: A Secondary Analysis of Randomised Controlled Trial Data. Vaccines (Basel) 2024; 12:691. [PMID: 38932420 PMCID: PMC11209274 DOI: 10.3390/vaccines12060691] [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: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
SARS-CoV-2 infections elicit antibodies against the viral spike (S) and nucleocapsid (N) proteins; COVID-19 vaccines against the S-protein only. The BCG-Corona trial, initiated in March 2020 in SARS-CoV-2-naïve Dutch healthcare workers, captured several epidemic peaks and the introduction of COVID-19 vaccines during the one-year follow-up. We assessed determinants of systemic anti-S1 and anti-N immunoglobulin type G (IgG) responses using trial data. Participants were randomised to BCG or placebo vaccination, reported daily symptoms, SARS-CoV-2 test results, and COVID-19 vaccinations, and donated blood for SARS-CoV-2 serology at two time points. In the 970 participants, anti-S1 geometric mean antibody concentrations (GMCs) were much higher than anti-N GMCs. Anti-S1 GMCs significantly increased with increasing number of immune events (SARS-CoV-2 infection or COVID-19 vaccination): 104.7 international units (IU)/mL, 955.0 IU/mL, and 2290.9 IU/mL for one, two, and three immune events, respectively (p < 0.001). In adjusted multivariable linear regression models, anti-S1 and anti-N log10 concentrations were significantly associated with infection severity, and anti-S1 log10 concentration with COVID-19 vaccine type/dose. In univariable models, anti-N log10 concentration was also significantly associated with acute infection duration, and severity and duration of individual symptoms. Antibody concentrations were not associated with long COVID or long-term loss of smell/taste.
Collapse
Affiliation(s)
- Juana Claus
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Thijs ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
- Department of Internal Medicine, Jeroen Bosch Ziekenhuis, 5223 GZ Hertogenbosch, The Netherlands
| | - Esther Taks
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Priya A. Debisarun
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
| | - Gaby Smits
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Rob van Binnendijk
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Fiona van der Klis
- National Institute of Public Health and the Environment, 3720 BA Bilthoven, The Netherlands; (G.S.); (R.v.B.); (F.v.d.K.)
| | - Lilly M. Verhagen
- Department of Paediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marien I. de Jonge
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marc J. M. Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| | - Mihai G. Netea
- Department of Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (E.T.); (P.A.D.); (M.G.N.)
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53113 Bonn, Germany
| | - Janneke H. H. M. van de Wijgert
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (J.C.); (T.t.D.); (J.H.H.M.v.d.W.)
| |
Collapse
|