501
|
Hu J, Safir F, Chang K, Dagli S, Balch HB, Abendroth JM, Dixon J, Moradifar P, Dolia V, Sahoo MK, Pinsky BA, Jeffrey SS, Lawrence M, Dionne JA. Rapid genetic screening with high quality factor metasurfaces. Nat Commun 2023; 14:4486. [PMID: 37495593 PMCID: PMC10372074 DOI: 10.1038/s41467-023-39721-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: 10/11/2021] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
Genetic analysis methods are foundational to advancing personalized medicine, accelerating disease diagnostics, and monitoring the health of organisms and ecosystems. Current nucleic acid technologies such as polymerase chain reaction (PCR) and next-generation sequencing (NGS) rely on sample amplification and can suffer from inhibition. Here, we introduce a label-free genetic screening platform based on high quality (high-Q) factor silicon nanoantennas functionalized with nucleic acid fragments. Each high-Q nanoantenna exhibits average resonant quality factors of 2,200 in physiological buffer. We quantitatively detect two gene fragments, SARS-CoV-2 envelope (E) and open reading frame 1b (ORF1b), with high-specificity via DNA hybridization. We also demonstrate femtomolar sensitivity in buffer and nanomolar sensitivity in spiked nasopharyngeal eluates within 5 minutes. Nanoantennas are patterned at densities of 160,000 devices per cm2, enabling future work on highly-multiplexed detection. Combined with advances in complex sample processing, our work provides a foundation for rapid, compact, and amplification-free molecular assays.
Collapse
Affiliation(s)
- Jack Hu
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA.
| | - Fareeha Safir
- Department of Mechanical Engineering, Stanford University, 440 Escondido Mall, Stanford, CA, 94305, USA
| | - Kai Chang
- Department of Electrical Engineering, Stanford University, 350 Jane Stanford Way, Stanford, CA, 94305, USA
| | - Sahil Dagli
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Halleh B Balch
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - John M Abendroth
- Laboratory for Solid State Physics, ETH Zürich, CH-8093, Zürich, Switzerland
| | - Jefferson Dixon
- Department of Mechanical Engineering, Stanford University, 440 Escondido Mall, Stanford, CA, 94305, USA
| | - Parivash Moradifar
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Varun Dolia
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA
| | - Malaya K Sahoo
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA
| | - Mark Lawrence
- Department of Electrical & Systems Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO, 63130, USA.
| | - Jennifer A Dionne
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA, 94305, USA.
| |
Collapse
|
502
|
Emanuel J, Papies J, Galander C, Adler JM, Heinemann N, Eschke K, Merz S, Pischon H, Rose R, Krumbholz A, Kulić Ž, Lehner MD, Trimpert J, Müller MA. In vitro and in vivo effects of Pelargonium sidoides DC. root extract EPs ® 7630 and selected constituents against SARS-CoV-2 B.1, Delta AY.4/AY.117 and Omicron BA.2. Front Pharmacol 2023; 14:1214351. [PMID: 37564181 PMCID: PMC10410074 DOI: 10.3389/fphar.2023.1214351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
The occurrence of immune-evasive SARS-CoV-2 strains emphasizes the importance to search for broad-acting antiviral compounds. Our previous in vitro study showed that Pelargonium sidoides DC. root extract EPs® 7630 has combined antiviral and immunomodulatory properties in SARS-CoV-2-infected human lung cells. Here we assessed in vivo effects of EPs® 7630 in SARS-CoV-2-infected hamsters, and investigated properties of EPs® 7630 and its functionally relevant constituents in context of phenotypically distinct SARS-CoV-2 variants. We show that EPs® 7630 reduced viral load early in the course of infection and displayed significant immunomodulatory properties positively modulating disease progression in hamsters. In addition, we find that EPs® 7630 differentially inhibits SARS-CoV-2 variants in nasal and bronchial human airway epithelial cells. Antiviral effects were more pronounced against Omicron BA.2 compared to B.1 and Delta, the latter two preferring TMPRSS2-mediated fusion with the plasma membrane for cell entry instead of receptor-mediated low pH-dependent endocytosis. By using SARS-CoV-2 Spike VSV-based pseudo particles (VSVpp), we confirm higher EPs® 7630 activity against Omicron Spike-VSVpp, which seems independent of the serine protease TMPRSS2, suggesting that EPs® 7630 targets endosomal entry. We identify at least two molecular constituents of EPs® 7630, i.e., (-)-epigallocatechin and taxifolin with antiviral effects on SARS-CoV-2 replication and cell entry. In summary, our study shows that EPs® 7630 ameliorates disease outcome in SARS-CoV-2-infected hamsters and has enhanced activity against Omicron, apparently by limiting late endosomal SARS-CoV-2 entry.
Collapse
Affiliation(s)
- Jackson Emanuel
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Jan Papies
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Celine Galander
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Julia M. Adler
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Nicolas Heinemann
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| | - Kathrin Eschke
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | | | | | - Ruben Rose
- Institute for Infection Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andi Krumbholz
- Institute for Infection Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Labor Dr. Krause und Kollegen MVZ GmbH, Kiel, Germany
| | - Žarko Kulić
- Preclinical R&D, Dr. Willmar Schwabe GmbH and Co. KG, Karlsruhe, Germany
| | - Martin D. Lehner
- Preclinical R&D, Dr. Willmar Schwabe GmbH and Co. KG, Karlsruhe, Germany
| | - Jakob Trimpert
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Marcel A. Müller
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Partner Site Charité, Berlin, Germany
| |
Collapse
|
503
|
Generaal E, van Santen DKD, Campman SL, Booij MJ, Price D, Buster M, van Dijk C, Boyd A, Bruisten SM, van Dam AP, van der Lubben M, van Duijnhoven YTHP, Prins M. Low prevalence of current and past SARS-CoV-2 infections among visitors and staff members of homelessness services in Amsterdam at the end of the second wave of infections in the Netherlands. PLoS One 2023; 18:e0288610. [PMID: 37490469 PMCID: PMC10368265 DOI: 10.1371/journal.pone.0288610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 07/02/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND People experiencing homelessness (PEH) may be at increased risk of SARS-CoV-2 infection and severe COVID-19. The Dutch government established emergency shelters and introduced preventive measures for homelessness services. There were no major SARS-CoV-2 outbreaks noticed among PEH during the first two waves of infections. This study aimed to assess the prevalence of current and past infections among PEH and staff by conducting an on-site COVID-19 screening project at homelessness services in Amsterdam, the Netherlands. METHODS We assessed the proportion of visitors and staff members of four homelessness services at two locations in Amsterdam with positive SARS-CoV-2 qPCR and antibody results (IgG/IgM Rapid Test/Biozek) in May 2021. We also assessed sociodemographic, clinical and lifestyle characteristics, compliance with basic prevention measures and intention to vaccinate against COVID-19 among PEH and staff. RESULTS A total of 138 visitors and 53 staff members filled out a questionnaire and were tested. Among PEH, the SARS-CoV-2 positivity rate was 0% (0/133;95%CI = 0-1.9) and the antibody positivity rate was 1.6% (2/131;95%CI = 0.8-7.5) among those without prior COVID-19 vaccination. Among staff, these percentages were 3% (1/32;95%CI = 0.1-16.2) and 11% (5/53;95%CI = 3.6-23.6), respectively. Most participants were often compliant with the basic preventive measures 'not shaking hands', 'wearing a face mask' and 'washing hands', but not with 'physical distancing'. High vaccination intent was more common among staff members (55%) than among visitors (42%), while high trust in the governmental COVID-19 policies was more common among visitors (41%) than among staff (30%). CONCLUSIONS We observed a low prevalence of past and current SARS-CoV-2 infections among PEH, which may be explained by instated shelter policies, limited daily activities of PEH and compliance with prevention measures. Vaccine hesitancy and mistrust among visitors and staff could hinder vaccination uptake, suggesting that interventions towards homelessness services are needed.
Collapse
Affiliation(s)
- Ellen Generaal
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control (Cib), Bilthoven, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - D K Daniela van Santen
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Department of Disease Elimination, Burnet Institute, Melbourne, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sophie L Campman
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Marjolein J Booij
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Dylan Price
- Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Marcel Buster
- Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | | | - Anders Boyd
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Stichting HIV Monitoring, Amsterdam, the Netherlands
| | - Sylvia M Bruisten
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Alje P van Dam
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | | | - Yvonne T H P van Duijnhoven
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Public Health Service of Rotterdam, Rotterdam, the Netherlands
| | - Maria Prins
- Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| |
Collapse
|
504
|
Nicholson BD, Turner PJ, Fanshawe TR, Williams AJ, Amirthalingam G, Tonner S, Zambon M, Body R, Davies K, Perera R, de Lusignan S, Hayward GN, Hobbs FR. Evaluation of the diagnostic accuracy of two point-of-care tests for COVID-19 when used in symptomatic patients in community settings in the UK primary care COVID diagnostic accuracy platform trial (RAPTOR-C19). PLoS One 2023; 18:e0288612. [PMID: 37478103 PMCID: PMC10361479 DOI: 10.1371/journal.pone.0288612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/02/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Point-of-care lateral flow device antigen testing has been used extensively to identify individuals with active SARS-CoV-2 infection in the community. This study aimed to evaluate the diagnostic accuracy of two point-of-care tests (POCTs) for SARS-CoV-2 in routine community care. METHODS Adults and children with symptoms consistent with suspected current COVID-19 infection were prospectively recruited from 19 UK general practices and two COVID-19 testing centres between October 2020 and October 2021. Participants were tested by trained healthcare workers using at least one of two index POCTs (Roche-branded SD Biosensor Standard™ Q SARS-CoV-2 Rapid Antigen Test and/or BD Veritor™ System for Rapid Detection of SARS-CoV-2). The reference standard was laboratory triplex reverse transcription quantitative PCR (RT-PCR) using a combined nasal/oropharyngeal swab. Diagnostic accuracy parameters were estimated, with 95% confidence intervals (CIs), overall, in relation to RT-PCR cycle threshold and in pre-specified subgroups. RESULTS Of 663 participants included in the primary analysis, 39.2% (260/663, 95% CI 35.5% to 43.0%) had a positive RT-PCR result. The SD Biosensor POCT had sensitivity 84.0% (178/212, 78.3% to 88.6%) and specificity 98.5% (328/333, 96.5% to 99.5%), and the BD Veritor POCT had sensitivity 76.5% (127/166, 69.3% to 82.7%) and specificity 98.8% (249/252, 96.6% to 99.8%) compared with RT-PCR. Sensitivity of both devices dropped substantially at cycle thresholds ≥30 and in participants more than 7 days after onset of symptoms. CONCLUSIONS Both POCTs assessed exceed the Medicines and Healthcare products Regulatory Agency target product profile's minimum acceptable specificity of 95%. Confidence intervals for both tests include the minimum acceptable sensitivity of 80%. In symptomatic patients, negative results on these two POCTs do not preclude the possibility of infection. Tests should not be expected to reliably detect disease more than a week after symptom onset, when viral load may be reduced. REGISTRATION ISRCTN142269.
Collapse
Affiliation(s)
- Brian D. Nicholson
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Philip J. Turner
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Thomas R. Fanshawe
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Alice J. Williams
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Gayatri Amirthalingam
- Immunisation and Vaccine Preventable Diseases Division and Public Health Programmes, UK Health Security Agency, London, United Kingdom
| | - Sharon Tonner
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Maria Zambon
- Influenza and Respiratory Virology & Polio Reference Service, UK Health Security Agency, London, United Kingdom
- NIHR Health Protection Research Unit, Imperial College London, London, United Kingdom
| | - Richard Body
- Division of Cardiovascular Science, University of Manchester, Manchester, United Kingdom
- Emergency Department, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, & Healthcare Sciences Department, Manchester Metropolitan University, Manchester, United Kingdom
| | - Kerrie Davies
- Healthcare Associated Infections Research Group, Leeds Teaching Hospitals NHS trust and University of Leeds, Leeds, United Kingdom
- NIHR Leeds MedTech In vitro Diagnostic Co-operative, Leeds Teaching Hospitals NHS Trust and University of Leeds, Leeds, United Kingdom
| | - Rafael Perera
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Gail N. Hayward
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - F.D. Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
505
|
Abdulla SA, Elawamy HA, Mohamed NAE, Abduallah EH, Amshahar HAA, Abuzaeid NK, Eisa MAM, Osman MEM, Konozy EHE. Association of ABO blood types and clinical variables with COVID-19 infection severity in Libya. SAGE Open Med 2023; 11:20503121231187736. [PMID: 37489137 PMCID: PMC10363681 DOI: 10.1177/20503121231187736] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/25/2023] [Indexed: 07/26/2023] Open
Abstract
Objective The continuing COVID-19 pandemic is a coronavirus-related health emergency (severe acute respiratory syndrome coronavirus 2). Inadequate efforts are still being made to address the illness situation in Libya, and this must change. To address these issues, we looked into the demography and trend of the disease as well as the potential risk factors for infection. Methods This study is a retrospective case-control study conducted online among 616 COVID-19 patients. The p0.05 value, odds ratios, and 95% confidence intervals were calculated and analyzed from the drawn data. Results Males were at high risk of COVID-19 than females (odds ratio = 1.3, 95% confidence interval: 1.042-1.622; p = 0.02). Anosmia and ageusia were more prominent in females. Patients with an "AB" blood group are significantly susceptible to infection. Adults (31 and above) are highly liable to infection. The univariate logistic regression analysis revealed that smoking is a risk factor for those above 60 years (odds ratio = 2.228, 95% confidence interval: 1.145-4.336; p = 0.018). Individuals with chronic diseases such as diabetes and/or hypertension are more prone to COVID-19 (odds ratio = 10.045, 95% confidence interval: 3.078-32.794; p = 0.000 and odds ratio = 11.508, 95% confidence interval: 3.930-33.695; p = 0.000, respectively). Conclusion This study provided for the first time the demographic data and the trend of COVID-19 infection in Libya, which will assist the stakeholders and governmental bodies in planning protection strategies against the pandemic.
Collapse
Affiliation(s)
- Sara A Abdulla
- Department of Biochemistry, Faculty of Medicine, University of Benghazi, Benghazi, Libya
| | | | - Negia AE Mohamed
- Department of Biochemistry, Faculty of Medicine, University of Benghazi, Benghazi, Libya
| | - Enas H Abduallah
- Department of Mathematics, Faculty of Science, University of Tobruk, Tobruk, Libya
| | - Habsa AA Amshahar
- Department of Pathology, Faculty of Medicine, Sirte University, Sirte, Libya
| | - Nadir K Abuzaeid
- Department of Medical Microbiology, Faculty of Medical Laboratory of Sciences, Omdurman Islamic University, Khartoum, Sudan
| | - Mohamed AM Eisa
- Department of Physiology, Al Qunfudah Medical College, Umm Al Qura University, Al Qunfudah, Saudi Arabia
| | - Makarim Elfadil M Osman
- Laboratory of Proteomics and Glycoproteins, Biotechnology Park, Africa City of Technology, Khartoum, Sudan
| | - Emadeldin Hassan E Konozy
- Laboratory of Proteomics and Glycoproteins, Biotechnology Park, Africa City of Technology, Khartoum, Sudan
- Pharmaceutical Research and Development Centre, Faculty of Pharmacy, Karary University, Omdurman, Khartoum State, Sudan
| |
Collapse
|
506
|
Corrêa TD, Midega TD, Cordioli RL, Barbas CSV, Rabello Filho R, Silva BCD, Silva Júnior M, Nawa RK, Carvalho FRTD, Matos GFJD, Lucinio NM, Rodrigues RD, Eid RAC, Bravim BDA, Pereira AJ, Santos BFCD, Pinho JRR, Pardini A, Teich VD, Laselva CR, Cendoroglo Neto M, Klajner S, Ferraz LJR. Clinical characteristics and outcomes of patients with COVID-19 admitted to the intensive care unit during the first and second waves of the pandemic in Brazil: a single-center retrospective cohort study. EINSTEIN-SAO PAULO 2023; 21:eAO0233. [PMID: 37493832 PMCID: PMC10356126 DOI: 10.31744/einstein_journal/2023ao0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/07/2023] [Indexed: 07/27/2023] Open
Abstract
OBJECTIVE To describe and compare the clinical characteristics and outcomes of patients admitted to intensive care units during the first and second waves of the COVID-19 pandemic. METHODS In this retrospective single-center cohort study, data were retrieved from the Epimed Monitor System; all adult patients admitted to the intensive care unit between March 4, 2020, and October 1, 2021, were included in the study. We compared the clinical characteristics and outcomes of patients admitted to the intensive care unit of a quaternary private hospital in São Paulo, Brazil, during the first (May 1, 2020, to August 31, 2020) and second (March 1, 2021, to June 30, 2021) waves of the COVID-19 pandemic. RESULTS In total, 1,427 patients with COVID-19 were admitted to the intensive care unit during the first (421 patients) and second (1,006 patients) waves. Compared with the first wave group [median (IQR)], the second wave group was younger [57 (46-70) versus 67 (52-80) years; p<0.001], had a lower SAPS 3 Score [45 (42-52) versus 49 (43-57); p<0.001], lower SOFA Score on intensive care unit admission [3 (1-6) versus 4 (2-6); p=0.018], lower Charlson Comorbidity Index [0 (0-1) versus 1 (0-2); p<0.001], and were less frequently frail (10.4% versus 18.1%; p<0.001). The second wave group used more noninvasive ventilation (81.3% versus 53.4%; p<0.001) and high-flow nasal cannula (63.2% versus 23.0%; p<0.001) during their intensive care unit stay. The intensive care unit (11.3% versus 10.5%; p=0.696) and in-hospital mortality (12.3% versus 12.1%; p=0.998) rates did not differ between both waves. CONCLUSION In the first and second waves, patients with severe COVID-19 exhibited similar mortality rates and need for invasive organ support, despite the second wave group being younger and less severely ill at the time of intensive care unit admission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sidney Klajner
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | |
Collapse
|
507
|
Lee WS, Tan HX, Reynaldi A, Esterbauer R, Koutsakos M, Nguyen J, Amarasena T, Kent HE, Aggarwal A, Turville SG, Taiaroa G, Kinsella P, Liew KC, Tran T, Williamson DA, Cromer D, Davenport MP, Kent SJ, Juno JA, Khoury DS, Wheatley AK. Durable reprogramming of neutralizing antibody responses following Omicron breakthrough infection. SCIENCE ADVANCES 2023; 9:eadg5301. [PMID: 37478181 PMCID: PMC10361595 DOI: 10.1126/sciadv.adg5301] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection of vaccinated individuals is increasingly common with the circulation of highly immune evasive and transmissible Omicron variants. Here, we report the dynamics and durability of recalled spike-specific humoral immunity following Omicron BA.1 or BA.2 breakthrough infection, with longitudinal sampling up to 8 months after infection. Both BA.1 and BA.2 infections robustly boosted neutralization activity against the infecting strain while expanding breadth against BA.4, although neutralization activity was substantially reduced for the more recent XBB and BQ.1.1 strains. Cross-reactive memory B cells against both ancestral and Omicron spike were predominantly expanded by infection, with limited recruitment of de novo Omicron-specific B cells or antibodies. Modeling of neutralization titers predicts that protection from symptomatic reinfection against antigenically similar strains will be durable but is undermined by new emerging strains with further neutralization escape.
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Julie Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thakshila Amarasena
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Helen E. Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Anupriya Aggarwal
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Stuart G. Turville
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - George Taiaroa
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Paul Kinsella
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kwee Chin Liew
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thomas Tran
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Deborah A. Williamson
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
508
|
Stoto MA, Reno C, Tsolova S, Fantini MP. The European experience with testing and surveillance during the first phase of the COVID-19 pandemic. Global Health 2023; 19:51. [PMID: 37480125 PMCID: PMC10362750 DOI: 10.1186/s12992-023-00950-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/30/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND COVID-19 pandemic provides a unique opportunity to learn the challenges encountered by public health emergency preparedness systems, both in terms of problems encountered and adaptations during and after the first wave, as well as successful responses to them. RESULTS This work draws on published literature, interviews with countries and institutional documents as part of a European Centre for Disease Prevention and Control project that aims to identify the implications for preparedness measurement derived from COVID-19 pandemic experience in order to advance future preparedness efforts in European Union member states. The analysis focused on testing and surveillance themes and five countries were considered, namely Italy, Germany, Finland, Spain and Croatia. Our analysis shown that a country's ability to conduct testing at scale was critical, especially early in the pandemic, and the inability to scale up testing operations created critical issues for public health operations such as contact tracing. Countries were required to develop new strategies, approaches, and policies under pressure and to review and revise them as the pandemic evolved, also considering that public health systems operate at the national, regional, and local level with respect to testing, contact tracing, and surveillance, and involve both government agencies as well as private organizations. Therefore, communication among multiple public and private entities at all levels and coordination of the testing and surveillance activities was critical. CONCLUSION With regard to testing and surveillance, three capabilities that were essential to the COVID-19 response in the first phase, and presumably in other public health emergencies: the ability to scale-up testing, contact tracing, surveillance efforts; flexibility to develop new strategies, approaches, and policies under pressure and to review and revise them as the pandemic evolved; and the ability to coordinate and communicate in complex public health systems that operate at the national, regional, and local level with respect and involve multiple government agencies as well as private organizations.
Collapse
Affiliation(s)
- Michael A Stoto
- Department of Health Management and Policy, Georgetown University, Washington, D.C, United States of America.
- Harvard T.H. Chan School of Public Health, Boston, United States.
| | - Chiara Reno
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, Bologna, 40126, Italy
- Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Pisa, Italy
| | - Svetla Tsolova
- European Centre for Disease prevention and Control (ECDC), Solna, Sweden
| | - Maria Pia Fantini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, Bologna, 40126, Italy
| |
Collapse
|
509
|
Staples J, Dourou AM, Liampa I, Sjaarda C, Moslinger E, Wong H, Sheth PM, Arhondakis S, Prakash R. A Miniaturized System for Rapid, Isothermal Detection of SARS-CoV-2 in Human and Environmental Samples. Biomedicines 2023; 11:2038. [PMID: 37509680 PMCID: PMC10377682 DOI: 10.3390/biomedicines11072038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
We report a small-footprint cost-effective isothermal rapid DNA amplification system, with integrated microfluidics for automated sample analysis and detection of SARS-CoV-2 in human and environmental samples. Our system measures low-level fluorescent signals in real-time during amplification, while maintaining the desired assay temperature on a low power, portable system footprint. A unique soft microfluidic chip design was implemented to mitigate thermocapillary effects and facilitate optical alignment for automated image capture and signal analysis. The system-on-board prototype, coupled with the LAMP primers designed by BioCoS, was sensitive enough to detect large variations in viral loads of SARS-CoV-2 corresponding to a threshold cycle range of 16 to 39. Furthermore, tested samples consisted of a broad range of viral strains and lineages identified in Canada during 2021-2022. Clinical specimens were collected and tested at the Kingston Health Science Centre using a clinically validated PCR assay, and variants were determined using whole genome sequencing.
Collapse
Affiliation(s)
- Jake Staples
- Department of Electronics Engineering, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | | | - Calvin Sjaarda
- Kingston Health Sciences Centre, Kingston, ON K7L 2V7, Canada
| | - Emily Moslinger
- Kingston Health Sciences Centre, Kingston, ON K7L 2V7, Canada
| | - Henry Wong
- Kingston Health Sciences Centre, Kingston, ON K7L 2V7, Canada
| | - Prameet M Sheth
- Kingston Health Sciences Centre, Kingston, ON K7L 2V7, Canada
| | | | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
510
|
Kant R, Kareinen L, Ojha R, Strandin T, Saber SH, Lesnikova A, Kuivanen S, Sirnonen T, Joensuu M, Vapalahti O, Kirchhausen T, Kipar A, Balistreri G. Complete Protection from SARS-CoV-2 Lung Infection in Mice Through Combined Intranasal Delivery of PIKfyve Kinase and TMPRSS2 Protease Inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549731. [PMID: 37503261 PMCID: PMC10370096 DOI: 10.1101/2023.07.19.549731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Emerging variants of concern of SARS-CoV-2 can significantly reduce the prophylactic and therapeutic efficacy of vaccines and neutralizing antibodies due to mutations in the viral genome. Targeting cell host factors required for infection provides a complementary strategy to overcome this problem since the host genome is less susceptible to variation during the life span of infection. The enzymatic activities of the endosomal PIKfyve phosphoinositide kinase and the serine protease TMPRSS2 are essential to meditate infection in two complementary viral entry pathways. Simultaneous inhibition in cultured cells of their enzymatic activities with the small molecule inhibitors apilimod dimesylate and nafamostat mesylate synergistically prevent viral entry and infection of native SARS-CoV-2 and vesicular stomatitis virus (VSV)-SARS-CoV-2 chimeras expressing the SARS-CoV-2 surface spike (S) protein and of variants of concern. We now report prophylactic prevention of lung infection in mice intranasally infected with SARS-CoV-2 beta by combined intranasal delivery of very low doses of apilimod dimesylate and nafamostat mesylate, in a formulation that is stable for over 3 months at room temperature. Administration of these drugs up to 6 hours post infection did not inhibit infection of the lungs but substantially reduced death of infected airway epithelial cells. The efficiency and simplicity of formulation of the drug combination suggests its suitability as prophylactic or therapeutic treatment against SARS-CoV-2 infection in households, point of care facilities, and under conditions where refrigeration would not be readily available.
Collapse
Affiliation(s)
- Ravi Kant
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri Kareinen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Finnish Food Authority, Helsinki, Finland
| | - Ravi Ojha
- Medicum Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tomas Strandin
- Medicum Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Saber Hassan Saber
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Angelina Lesnikova
- Medicum Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Tarja Sirnonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Merja Joensuu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Medicum Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Anja Kipar
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Giuseppe Balistreri
- Medicum Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
511
|
de Graaf M, Langeveld J, Post J, Carrizosa C, Franz E, Izquierdo-Lara RW, Elsinga G, Heijnen L, Been F, van Beek J, Schilperoort R, Vriend R, Fanoy E, de Schepper EIT, Koopmans MPG, Medema G. Capturing the SARS-CoV-2 infection pyramid within the municipality of Rotterdam using longitudinal sewage surveillance. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 883:163599. [PMID: 37100150 PMCID: PMC10125208 DOI: 10.1016/j.scitotenv.2023.163599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/07/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
Despite high vaccination rates in the Netherlands, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to circulate. Longitudinal sewage surveillance was implemented along with the notification of cases as two parts of the surveillance pyramid to validate the use of sewage for surveillance, as an early warning tool, and to measure the effect of interventions. Sewage samples were collected from nine neighborhoods between September 2020 and November 2021. Comparative analysis and modeling were performed to understand the correlation between wastewater and case trends. Using high resolution sampling, normalization of wastewater SARS-CoV-2 concentrations, and 'normalization' of reported positive tests for testing delay and intensity, the incidence of reported positive tests could be modeled based on sewage data, and trends in both surveillance systems coincided. The high collinearity implied that high levels of viral shedding around the onset of disease largely determined SARS-CoV-2 levels in wastewater, and that the observed relationship was independent of variants of concern and vaccination levels. Sewage surveillance alongside a large-scale testing effort where 58 % of a municipality was tested, indicated a five-fold difference in the number of SARS-CoV-2-positive individuals and reported cases through standard testing. Where trends in reported positive cases were biased due to testing delay and testing behavior, wastewater surveillance can objectively display SARS-CoV-2 dynamics for both small and large locations and is sensitive enough to measure small variations in the number of infected individuals within or between neighborhoods. With the transition to a post-acute phase of the pandemic, sewage surveillance can help to keep track of re-emergence, but continued validation studies are needed to assess the predictive value of sewage surveillance with new variants. Our findings and model aid in interpreting SARS-CoV-2 surveillance data for public health decision-making and show its potential as one of the pillars of future surveillance of (re)emerging viruses.
Collapse
Affiliation(s)
- Miranda de Graaf
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Centre Rotterdam and Delft, the Netherlands.
| | - Jeroen Langeveld
- Partners4urbanwater, Nijmegen, the Netherlands; Delft University of Technology, Stevinweg 1, 2628 CN Delft, the Netherlands
| | - Johan Post
- Partners4urbanwater, Nijmegen, the Netherlands
| | - Christian Carrizosa
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Eelco Franz
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ray W Izquierdo-Lara
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Goffe Elsinga
- KWR Water Research Institute, Groningenhaven 7, 3433 PE Nieuwegein, the Netherlands
| | - Leo Heijnen
- KWR Water Research Institute, Groningenhaven 7, 3433 PE Nieuwegein, the Netherlands
| | - Frederic Been
- KWR Water Research Institute, Groningenhaven 7, 3433 PE Nieuwegein, the Netherlands
| | - Janko van Beek
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Rianne Vriend
- Regional Public Health Service Rotterdam-Rijnmond, Rotterdam, the Netherlands
| | - Ewout Fanoy
- Regional Public Health Service Rotterdam-Rijnmond, Rotterdam, the Netherlands
| | - Evelien I T de Schepper
- Department of General Practice, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands; Pandemic and Disaster Preparedness Centre Rotterdam and Delft, the Netherlands
| | - Gertjan Medema
- Pandemic and Disaster Preparedness Centre Rotterdam and Delft, the Netherlands; KWR Water Research Institute, Groningenhaven 7, 3433 PE Nieuwegein, the Netherlands; Delft University of Technology, Stevinweg 1, 2628 CN Delft, the Netherlands
| |
Collapse
|
512
|
Pinesi HT, Giugni FR, Matuck BRS, Pitta FG, Garzillo CL, Lima EG, Kalil Filho R, Serrano Junior CV. Coronavirus disease-2019 and heart: assessment of troponin and cardiovascular comorbidities as prognostic markers in patients hospitalized with coronavirus disease-2019 in a tertiary center in Brazil. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230350. [PMID: 37466607 DOI: 10.1590/1806-9282.20230350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/30/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVE Our study aimed to evaluate the correlation of cardiac troponin T levels with comorbidities and in-hospital outcomes in patients with coronavirus disease-2019 in Brazil. METHODS Data from a cohort of 3,596 patients who were admitted with suspected coronavirus disease-2019 in a Brazilian tertiary center, between March and August 2020, were reviewed. A total of 2,441 (68%) patients had cardiac troponin T determined in the first 72 h of admission and were stratified into two groups: elevated cardiac troponin T (cardiac troponin T >0.014 ng/mL) and normal cardiac troponin T. Associations between troponin, comorbidities, biomarkers, and outcomes were assessed. Regression models were built to assess the association of several variables with in-hospital mortality. RESULTS A total of 2,441 patients were embraced, of which 924 (38%) had normal cardiac troponin T and 1,517 (62%) had elevated cardiac troponin T. Patients with elevated cardiac troponin T were older and had more comorbidities, such as cardiovascular disease, hypertension, diabetes, arrhythmia, renal dysfunction, liver disease, stroke, cancer, and dementia. Patients with abnormal cardiac troponin T also had more altered laboratory parameters on admission (i.e., leukocytes, C-reactive protein, D-dimer, and B-type natriuretic peptide), as well as more need for intensive care unit, vasoactive drugs, mechanical ventilation, dialysis, and blood transfusion. All-cause mortality was markedly higher among patients with increased cardiac troponin T (42 vs. 16%, P<0.001). Multiple regression analysis demonstrated that in-hospital mortality was not independently associated with troponin elevation. CONCLUSION This study showed that cardiac troponin T elevation at admission was common and associated with several comorbidities, biomarkers, and clinical outcomes in patients hospitalized with coronavirus disease-2019, but it was not an independent marker of in-hospital mortality.
Collapse
Affiliation(s)
| | | | | | - Fabio Grusnpun Pitta
- Universidade de São Paulo, Instituto do Coracao, Faculdade de Medicina - São Paulo (SP), Brazil
| | - Cibele Larrosa Garzillo
- Universidade de São Paulo, Instituto do Coracao, Faculdade de Medicina - São Paulo (SP), Brazil
| | - Eduardo Gomes Lima
- Universidade de São Paulo, Instituto do Coracao, Faculdade de Medicina - São Paulo (SP), Brazil
| | - Roberto Kalil Filho
- Universidade de São Paulo, Instituto do Coracao, Faculdade de Medicina - São Paulo (SP), Brazil
| | | |
Collapse
|
513
|
Chen YJ, Huang JC, Yang CP, Hsu KF, Liu HF. A Comprehensive Phylogenetic Analysis of SARS-CoV-2: Utilizing a Novel and Convenient In-House RT-PCR Method for Characterization without Virus Culture and BSL-3 Facilities. Viruses 2023; 15:1562. [PMID: 37515248 PMCID: PMC10383548 DOI: 10.3390/v15071562] [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: 05/31/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
We developed a convenient method for amplifying the complete SARS-CoV-2 sequence using in-house RT-PCR without virus culture. Forty-one stored throat swabs and blood specimens were collected from eight SARS-CoV-2 infections at multiple time points. Total RNA was extracted using the QIAamp viral RNA mini kit and pooled for higher RNA levels. Only those positive specimens by commercial real-time RT-PCR (RT-qPCR) were selected and amplified by in-house RT-PCR for complete sequences, followed by sequencing. Phylogenetic trees and exploratory analyses were performed using MEGA 11 and Simplot 3.5.1 software. Swab samples had significantly higher total RNA concentrations than plasma (p < 0.01). Positive results were found mainly in swabs, but one was found in plasma. Successful gene amplification depended on Ct values (Ct < 38). A non-synonymous substitution was found in ORF1ab/Nsp3 (at NC045512.2 position 6312, C to A) and most spike protein mutations occurred in the S1 subunit (residues 14-685). The proposed method is time-saving and reliable for rapid genomic analysis. Increasing sample volume and pooling them for RNA extraction increases RNA concentration without culture. Combining nucleotide sequences from specific variable regions of the genome is more efficient than conventional methods.
Collapse
Affiliation(s)
- Yen-Ju Chen
- Research Assistant Center, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan 701033, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Jason C Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ching-Ping Yang
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli 356006, Taiwan
| | - Kuo-Feng Hsu
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Hsin-Fu Liu
- Department of Medical Research, MacKay Memorial Hospital, Taipei 25169, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 252005, Taiwan
| |
Collapse
|
514
|
Franco-Luiz APM, Fernandes NMGS, Silva TBDS, Bernardes WPDOS, Westin MR, Santos TG, Fernandes GDR, Simões TC, Silva EFE, Gava SG, Alves BM, de Carvalho Melo M, da Silva-Pereira RA, Alves PA, Fonseca CT. Longitudinal study of humoral immunity against SARS-CoV-2 of health professionals in Brazil: the impact of booster dose and reinfection on antibody dynamics. Front Immunol 2023; 14:1220600. [PMID: 37520570 PMCID: PMC10376701 DOI: 10.3389/fimmu.2023.1220600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction The pandemic caused by SARS-CoV-2 has had a major impact on health systems. Vaccines have been shown to be effective in improving the clinical outcome of COVID-19, but they are not able to fully prevent infection and reinfection, especially that caused by new variants. Methods Here, we tracked for 450 days the humoral immune response and reinfection in 52 healthcare workers from Brazil. Infection and reinfection were confirmed by RT-qPCR, while IgM and IgG antibody levels were monitored by rapid test. Results Of the 52 participants, 19 (36%) got reinfected during the follow-up period, all presenting mild symptoms. For all participants, IgM levels dropped sharply, with over 47% of them becoming seronegative by the 60th day. For IgG, 90% of the participants became seropositive within the first 30 days of follow-up. IgG antibodies also dropped after this period reaching the lowest level on day 270 (68.5 ± 72.3, p<0.0001). Booster dose and reinfection increased the levels of both antibodies, with the interaction between them resulting in an increase in IgG levels of 130.3 arbitrary units. Conclusions Overall, our data indicate that acquired humoral immunity declines over time and suggests that IgM and IgG antibody levels are not associated with the prevention of reinfection.
Collapse
Affiliation(s)
- Ana Paula Moreira Franco-Luiz
- Grupo de Pesquisa em Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Nubia Monteiro Gonçalves Soares Fernandes
- Grupo de Pesquisa em Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Thais Bárbara de Souza Silva
- Grupo de Imunologia de Doenças Virais, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | | | - Mateus Rodrigues Westin
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thais Garcia Santos
- Grupo de Pesquisa em Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriel da Rocha Fernandes
- Grupo de Pesquisa em Informática de Biossistemas, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Taynãna César Simões
- Núcleo de Estudos em Saúde Pública e Envelhecimento, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Fernandes E. Silva
- Serviço de capacitação em métodos quantitativos -SAMeQ, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Sandra Grossi Gava
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Breno Magalhães Alves
- Centro de Vigilância em Saúde e Segurança do Paciente, Hospital Metropolitano Doutor Célio de Castro, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana de Carvalho Melo
- Serviço Especializado em Segurança e Medicina do Trabalho, Hospital Metropolitano Doutor Célio de Castro, Belo Horizonte, Minas Gerais, Brazil
| | - Rosiane A. da Silva-Pereira
- Grupo de Pesquisa em Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Augusto Alves
- Grupo de Imunologia de Doenças Virais, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina Toscano Fonseca
- Grupo de Pesquisa em Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
515
|
Abdelaziz MO, Raftery MJ, Weihs J, Bielawski O, Edel R, Köppke J, Vladimirova D, Adler JM, Firsching T, Voß A, Gruber AD, Hummel LV, Fernandez Munoz I, Müller-Marquardt F, Willimsky G, Elleboudy NS, Trimpert J, Schönrich G. Early protective effect of a ("pan") coronavirus vaccine (PanCoVac) in Roborovski dwarf hamsters after single-low dose intranasal administration. Front Immunol 2023; 14:1166765. [PMID: 37520530 PMCID: PMC10372429 DOI: 10.3389/fimmu.2023.1166765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the danger posed by human coronaviruses. Rapid emergence of immunoevasive variants and waning antiviral immunity decrease the effect of the currently available vaccines, which aim at induction of neutralizing antibodies. In contrast, T cells are marginally affected by antigen evolution although they represent the major mediators of virus control and vaccine protection against virus-induced disease. Materials and methods We generated a multi-epitope vaccine (PanCoVac) that encodes the conserved T cell epitopes from all structural proteins of coronaviruses. PanCoVac contains elements that facilitate efficient processing and presentation of PanCoVac-encoded T cell epitopes and can be uploaded to any available vaccine platform. For proof of principle, we cloned PanCoVac into a non-integrating lentivirus vector (NILV-PanCoVac). We chose Roborovski dwarf hamsters for a first step in evaluating PanCoVac in vivo. Unlike mice, they are naturally susceptible to SARS-CoV-2 infection. Moreover, Roborovski dwarf hamsters develop COVID-19-like disease after infection with SARS-CoV-2 enabling us to look at pathology and clinical symptoms. Results Using HLA-A*0201-restricted reporter T cells and U251 cells expressing a tagged version of PanCoVac, we confirmed in vitro that PanCoVac is processed and presented by HLA-A*0201. As mucosal immunity in the respiratory tract is crucial for protection against respiratory viruses such as SARS-CoV-2, we tested the protective effect of single-low dose of NILV-PanCoVac administered via the intranasal (i.n.) route in the Roborovski dwarf hamster model of COVID-19. After infection with ancestral SARS-CoV-2, animals immunized with a single-low dose of NILV-PanCoVac i.n. did not show symptoms and had significantly decreased viral loads in the lung tissue. This protective effect was observed in the early phase (2 days post infection) after challenge and was not dependent on neutralizing antibodies. Conclusion PanCoVac, a multi-epitope vaccine covering conserved T cell epitopes from all structural proteins of coronaviruses, might protect from severe disease caused by SARS-CoV-2 variants and future pathogenic coronaviruses. The use of (HLA-) humanized animal models will allow for further efficacy studies of PanCoVac-based vaccines in vivo.
Collapse
Affiliation(s)
- Mohammed O. Abdelaziz
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Martin J. Raftery
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julian Weihs
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Pediatrics, Division of Gastroenterology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olivia Bielawski
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Richard Edel
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Köppke
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Julia M. Adler
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | - Theresa Firsching
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Anne Voß
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D. Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Luca V. Hummel
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivan Fernandez Munoz
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Francesca Müller-Marquardt
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Partner Site Berlin, Berlin, Germany
| | - Nooran S. Elleboudy
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Jakob Trimpert
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | - Günther Schönrich
- Institute of Virology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
516
|
Lin X, Sha Z, Trimpert J, Kunec D, Jiang C, Xiong Y, Xu B, Zhu Z, Xue W, Wu H. The NSP4 T492I mutation increases SARS-CoV-2 infectivity by altering non-structural protein cleavage. Cell Host Microbe 2023; 31:1170-1184.e7. [PMID: 37402373 DOI: 10.1016/j.chom.2023.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/13/2023] [Accepted: 06/09/2023] [Indexed: 07/06/2023]
Abstract
The historically dominant SARS-CoV-2 Delta variant and the currently dominant Omicron variants carry a T492I substitution within the non-structural protein 4 (NSP4). Based on in silico analyses, we hypothesized that the T492I mutation increases viral transmissibility and adaptability, which we confirmed with competition experiments in hamster and human airway tissue culture models. Furthermore, we showed that the T492I mutation increases the replication capacity and infectiveness of the virus and improves its ability to evade host immune responses. Mechanistically, the T492I mutation increases the cleavage efficiency of the viral main protease NSP5 by enhancing enzyme-substrate binding, which increases production of nearly all non-structural proteins processed by NSP5. Importantly, the T492I mutation suppresses viral-RNA-associated chemokine production in monocytic macrophages, which may contribute to the attenuated pathogenicity of Omicron variants. Our results highlight the importance of NSP4 adaptation in the evolutionary dynamics of SARS-CoV-2.
Collapse
Affiliation(s)
- Xiaoyuan Lin
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China; Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Straße 7, 14163 Berlin, Germany
| | - Zhou Sha
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China
| | - Jakob Trimpert
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Straße 7, 14163 Berlin, Germany
| | - Dusan Kunec
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Straße 7, 14163 Berlin, Germany
| | - Chen Jiang
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China
| | - Binbin Xu
- School of Pharmaceutical Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China
| | - Zhenglin Zhu
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China.
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, No.55 Daxuecheng South Road, Shapingba, Chongqing 401331, China.
| |
Collapse
|
517
|
Rivas-Macho A, Sorarrain A, Marimón JM, Goñi-de-Cerio F, Olabarria G. Extraction-Free Colorimetric RT-LAMP Detection of SARS-CoV-2 in Saliva. Diagnostics (Basel) 2023; 13:2344. [PMID: 37510088 PMCID: PMC10377860 DOI: 10.3390/diagnostics13142344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The pandemic situation caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need of fast, simple, and cost-effective tests for the diagnosis of emerging pathogens. RT-qPCR has been established as the reference technique for the diagnosis of SARS-CoV-2 infections. This method requires a time-consuming protocol for the extraction of the nucleic acids present in the sample. A colorimetric reverse transcription loop-mediated isothermal amplification using the calcein molecule combined with a simple extraction-free method for saliva samples (calcein RT-LAMP) has been developed. Samples are heated 95 °C for 10 min before amplification at 63 °C for 40 min. The results can be observed by fluorescence or by the naked eye with a color change from orange to green. The method was compared with commercialized available colorimetric and fluorescent RT-LAMP kits. The developed method shows better sensitivity and specificity than the colorimetric commercial RT-LAMP and the same as the fluorescent RT-LAMP, without the need of a fluorescent reader. Moreover, the calcein RT-LAMP has, compared to RT-qPCR, a sensitivity of 90% and a specificity of 100% for saliva samples with a Ct ≤ 34, without the need for expensive RT-qPCR instruments, demonstrating the potential of this method for population screening.
Collapse
Affiliation(s)
- Ane Rivas-Macho
- Gaiker, GAIKER Technology Centre, Basque Research and Technology Alliance, Parque Tecnológico, Ed. 202, 48170 Zamudio, Spain
- Molecular Biology and Biomedicine PhD Program, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Ane Sorarrain
- Biodonostia Health Research Institute, Infectious Diseases Area, Microbiology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organization, 20014 San Sebastián, Spain
| | - José M Marimón
- Biodonostia Health Research Institute, Infectious Diseases Area, Microbiology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organization, 20014 San Sebastián, Spain
| | - Felipe Goñi-de-Cerio
- Gaiker, GAIKER Technology Centre, Basque Research and Technology Alliance, Parque Tecnológico, Ed. 202, 48170 Zamudio, Spain
| | - Garbiñe Olabarria
- Gaiker, GAIKER Technology Centre, Basque Research and Technology Alliance, Parque Tecnológico, Ed. 202, 48170 Zamudio, Spain
| |
Collapse
|
518
|
Sato HI, Costa MS, Takahashi RHC, Lourenço KL, Guimarães NS, Alves CRL, Machado EL, Tupinambás U, Fonseca FGD, Teixeira SMR. RT-qPCR-based pool testing for the diagnosis of COVID-19. EINSTEIN-SAO PAULO 2023; 21:eAE0115. [PMID: 37436266 DOI: 10.31744/einstein_journal/2023ae0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 02/07/2023] [Indexed: 07/13/2023] Open
Abstract
This study proposes a strategy for large-scale testing among a large number of people for the early diagnosis of COVID-19 to elucidate the epidemiological situation. Pool testing involves the analysis of pooled samples. This study aimed to discuss a reverse transcription technique followed by quantitative real-time polymerase chain reaction using pool testing to detect SARS-CoV-2 in nasopharyngeal swab samples. The study proposes an innovative diagnostic strategy that contributes to resource optimization, cost reduction, and improved agility of feedback from results. Pool testing is simultaneously performed on multiple samples to efficiently and cost-effectively detect COVID-19. Pool testing can optimize resource utilization and expand diagnostic access, and is a viable alternative for developing countries with limited access to testing. To optimize resources, the pool size was determined by estimating COVID-19 prevalence in the study population.
Collapse
Affiliation(s)
- Hugo Itaru Sato
- Vaccine Technology Center, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Murilo Soares Costa
- Postgraduate Program in Infectious Diseases and Tropical Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Karine Lima Lourenço
- Vaccine Technology Center, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Elaine Leandro Machado
- Department of Preventive and Social Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Unaí Tupinambás
- Medical Clinic Department, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | |
Collapse
|
519
|
Zuluaga N, Martínez D, Hernández C, Ballesteros N, Castañeda S, Ramírez JD, Muñoz M. Description of pathogenic bacteria in patients with respiratory symptoms associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Colombia. Ann Clin Microbiol Antimicrob 2023; 22:56. [PMID: 37420198 DOI: 10.1186/s12941-023-00595-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/29/2023] [Indexed: 07/09/2023] Open
Abstract
Viral respiratory infections may predispose to co-infections with other pathogenic microorganisms. In this study, pathogenic respiratory bacteria were detected using commercial kit Allplex™ Respiratory Panel 4 from nasopharyngeal samples from individuals suffering respiratory symptoms with and without severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients without respiratory symptoms were included as controls. Haemophilus influenzae and Streptococcus pneumoniae were detected from 12 patients (6%) in both, patients with respiratory symptoms (including hospitalized) (n = 6) and individual without symptoms (n = 6). Pathogenic bacteria possibly proliferate due to the limited immune response of patients with SARS-CoV-2, perhaps due to dysbiosis generated by the viral infection.
Collapse
Affiliation(s)
- Nathalia Zuluaga
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - David Martínez
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Carolina Hernández
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Centro de Tecnología en Salud (CETESA), Innovaseq SAS, Bogotá, Colombia
| | - Nathalia Ballesteros
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Sergio Castañeda
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología and Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
520
|
Zhang Q, Qiao R, Niu J, Xiong X, Wang N, Zhang R, Luo S, Guo Y, Liu Z, Peng L, Zhang S, Tan G, Song K, Sun M, Xu L, Zhang R, Wu X. Evaluation of an identification method for the SARS-CoV-2 Delta variant based on the amplification-refractory mutation system. Front Cell Infect Microbiol 2023; 13:1180297. [PMID: 37475960 PMCID: PMC10354518 DOI: 10.3389/fcimb.2023.1180297] [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: 03/06/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023] Open
Abstract
The Delta variant of SARS-CoV-2 dominated the COVID-19 pandemic due to its high viral replication capacity and immune evasion, causing massive outbreaks of cases, hospitalizations, and deaths. Currently, variant identification is performed mainly by sequencing. However, the high requirements for equipment and operators as well as its high cost have limited its application in underdeveloped regions. To achieve an economical and rapid method of variant identification suitable for undeveloped areas, we applied an amplification-refractory mutation system (ARMS) based on PCR for the detection of novel coronavirus variants. The results showed that this method could be finished in 90 min and detect as few as 500 copies/mL and not react with SARS-Coronavirus, influenza A H1N1(2009), and other cross-pathogens or be influenced by fresh human blood, α- interferon, and other interfering substances. In a set of double-blind trials, tests of 262 samples obtained from patients confirmed with Delta variant infection revealed that our method was able to accurately identify the Delta variant with high sensitivity and specificity. In conclusion, the ARMS-PCR method applied in Delta variant identification is rapid, sensitive, specific, economical, and suitable for undeveloped areas. In our future study, ARMS-PCR will be further applied in the identification of other variants, such as Omicron.
Collapse
Affiliation(s)
- Qin Zhang
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Runjie Qiao
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiaojiao Niu
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xia Xiong
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Nan Wang
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ruixian Zhang
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Sha Luo
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuwan Guo
- Research and Development Department, Jiangsu Bioperfectus Technologies Company Limited, Taizhou, Jiangsu, China
| | - Zhonghua Liu
- Research and Development Department, Jiangsu Bioperfectus Technologies Company Limited, Taizhou, Jiangsu, China
| | - Li Peng
- Research and Development Department, Jiangsu Bioperfectus Technologies Company Limited, Taizhou, Jiangsu, China
| | - Shaoduo Zhang
- Research and Development Department, Jiangsu Bioperfectus Technologies Company Limited, Taizhou, Jiangsu, China
| | - Guolei Tan
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Keyu Song
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mei Sun
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lulu Xu
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Rong Zhang
- Research and Development Department, Jiangsu Bioperfectus Technologies Company Limited, Taizhou, Jiangsu, China
| | - Xuping Wu
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
521
|
Krause E, Michel J, Puyskens A, Hofmann N, Rinner T, Biere B, Dorner BG, Skiba M, Schaade L, Nitsche A. Flexible upscaling of laboratory PCR testing capacity at the Robert Koch Institute during the SARS-CoV-2 pandemic. Virol J 2023; 20:139. [PMID: 37408040 DOI: 10.1186/s12985-023-02088-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/02/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Over the course of the COVID-19 pandemic, laboratories worldwide have been facing an unprecedented increase in demand for PCR testing because of the high importance of diagnostics for prevention and control of virus spread. Moreover, testing demand has been varying considerably over time, depending on the epidemiological situation, rendering efficient resource allocation difficult. Here, we present a scalable workflow which we implemented in our laboratory to increase PCR testing capacity while maintaining high flexibility regarding the number of samples to be processed. METHODS We compared the performance of five automated extraction instruments, using dilutions of SARS-CoV-2 cell culture supernatant as well as clinical samples. To increase PCR throughput, we combined the two duplex PCR reactions of our previously published SARS-CoV-2 PCR assay into one quadruplex reaction and compared their limit of detection as well as their performance on the detection of low viral loads in clinical samples. Furthermore, we developed a sample pooling protocol with either two or four samples per pool, combined with a specifically adapted SARS-CoV-2 quadruplex PCR assay, and compared the diagnostic sensitivity of pooled testing and individual testing. RESULTS All tested automated extraction instruments yielded comparable results regarding the subsequent sensitivity of SARS-CoV-2 detection by PCR. While the limit of detection of the quadruplex SARS-CoV-2 PCR assay (E-Gene assay: 28.7 genome equivalents (ge)/reaction, orf1ab assay: 32.0 ge/reaction) was slightly higher than that of our previously published duplex PCR assays (E-Gene assay: 9.8 ge/reaction, orf1ab assay: 6.6 ge/reaction), the rate of correctly identified positive patient samples was comparable for both assays. Sample pooling with optimized downstream quadruplex PCR showed no loss in diagnostic sensitivity compared to individual testing. CONCLUSION Specific adaptation of PCR assays can help overcome the potential loss of sensitivity due to higher levels of PCR multiplexing or sample dilution in pooled testing. Combining these adapted PCR assays with different sample processing strategies provides a simple and highly adjustable workflow for resource-efficient SARS-CoV-2 diagnostics. The presented principles can easily be adopted in a variety of laboratory settings as well as be adapted to pathogens other than SARS-CoV-2, making it feasible for any laboratory that conducts PCR diagnostics.
Collapse
Affiliation(s)
- Eva Krause
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany.
| | - Janine Michel
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Andreas Puyskens
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Natalie Hofmann
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Thomas Rinner
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Barbara Biere
- Department for Infectious Diseases, Unit Influenza and Other Respiratory Viruses (FG 17), Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Brigitte G Dorner
- Centre for Biological Threats and Special Pathogens, Unit Biological Toxins (ZBS 3), WHO Collaborating Centre for Emerging Infections and Biological Threats, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Martin Skiba
- Centre for Biological Threats and Special Pathogens, Unit Biological Toxins (ZBS 3), WHO Collaborating Centre for Emerging Infections and Biological Threats, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Lars Schaade
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| | - Andreas Nitsche
- Centre for Biological Threats and Special Pathogens, Unit Highly Pathogenic Viruses (ZBS 1), WHO Collaborating Centre for Emerging Infections and Biological Threats, WHO Reference Laboratory for SARS-CoV-2, Robert Koch Institute, Seestrasse 10, 13353, Berlin, Germany
| |
Collapse
|
522
|
Cabral KMDS, Baptista RCG, Castineiras TMPP, Tanuri A, Carneiro FA, Almeida MDS, Montero-Lomeli M. Accuracy of a raw saliva-based COVID-19 RT-LAMP diagnostic assay. Braz J Infect Dis 2023; 27:102790. [PMID: 37478898 PMCID: PMC10391658 DOI: 10.1016/j.bjid.2023.102790] [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: 02/20/2023] [Revised: 06/02/2023] [Accepted: 07/04/2023] [Indexed: 07/23/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pandemic demanded rapid diagnosis to isolate new COVID-19 cases and prevent disease transmission. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) rapidly became the gold standard for diagnosis. However, due to the high cost and delay of the results, other types of diagnosis were implemented, such as COVID-19 Ag Rapid Tests and Reverse Transcription Technique followed by Loop-Mediated isothermal Amplification (RT-LAMP). In this work, we validated the use of RT-LAMP in saliva samples rather than nasopharyngeal swabs, as the collection is more comfortable. First, we selected 5 primer sets based on the limit of detection for SARS-CoV-2 RNA, then validated their sensitivity and specificity in patient samples. A total of 117 samples were analyzed by fluorometric RT-LAMP and compared with qRT-PCR results. Our results show that the use of a high-sensitive primer ORF1-a, together with a low-sensitive primer set Gene E (time to threshold of 22.9 and 36.4 minutes, respectively, using 200 copies of viral RNA), achieved sensitivity in purified RNA from saliva samples of 95.2% (95% CI 76.1‒99.8) with 90.5% specificity (95% CI 69.6‒98.8) (n = 42).As RNA purification increases the turnaround time, we tested the outcome of RT-LAMP utilizing raw saliva samples without purification. The test achieved a sensitivity of 81.8% (95% CI 59.7‒94.8) and a specificity of 90.9% (95% CI 70.8‒98.8). As a result, the accuracy of 92.9% (95% CI 80.5‒98.5) in purified RNA-saliva samples was lowered to an acceptable level of 86.4% (95% CI 72.6‒94.8) in raw saliva. Although mass vaccination has been implemented, new strains and low vaccination progress helped to spread COVID-19. This study shows that it is feasible to track new COVID-19 cases in a large population with the use of raw saliva as sample in RT-LAMP assay which yields accurate results and offers a less invasive test.
Collapse
Affiliation(s)
- Kátia Maria Dos Santos Cabral
- Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Bioquímica Médica-Leopoldo de Meis, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, Plataforma Avançada de Biomoléculas, Rio de Janeiro, RJ, Brazil
| | - Ramon Cid Gismonti Baptista
- Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Bioquímica Médica-Leopoldo de Meis, Rio de Janeiro, RJ, Brazil
| | | | - Amilcar Tanuri
- Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Biologia, Departamento de Genética, Rio de Janeiro, RJ, Brazil
| | - Fabiana Avila Carneiro
- Centro de Pesquisa de Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho, Rio de Janeiro, RJ, Brazil; Núcleo de Pesquisa (Numpex-Bio), Campus Duque de Caxias Professor Geraldo Cidade, Duque de Caxias, RJ, Brazil
| | - Marcius da Silva Almeida
- Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Bioquímica Médica-Leopoldo de Meis, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, Plataforma Avançada de Biomoléculas, Rio de Janeiro, RJ, Brazil
| | - Monica Montero-Lomeli
- Universidade Federal do Rio de Janeiro (UFRJ), Instituto de Bioquímica Médica-Leopoldo de Meis, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
523
|
Buchta C, Zeichhardt H, Aberle SW, Camp JV, Görzer I, Weseslindtner L, Puchhammer-Stöckl E, Huf W, Benka B, Allerberger F, Mielke M, Griesmacher A, Müller MM, Schellenberg I, Kammel M. Design of external quality assessment schemes and definition of the roles of their providers in future epidemics. THE LANCET. MICROBE 2023; 4:e552-e562. [PMID: 37156257 PMCID: PMC10162712 DOI: 10.1016/s2666-5247(23)00072-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 05/10/2023]
Abstract
During an epidemic, individual test results form the basis of epidemiological indicators such as case numbers or incidence. Therefore, the accuracy of measures derived from these indicators depends on the reliability of individual results. In the COVID-19 pandemic, monitoring and evaluating the performance of the unprecedented number of testing facilities in operation, and novel testing systems in use, was urgently needed. External quality assessment (EQA) schemes are unique sources of data reporting on testing performance, and their providers are recognised contacts and support for test facilities (for technical-analytical topics) and health authorities (for planning the monitoring of infection diagnostics). To identify information provided by SARS-CoV-2 genome detection EQA schemes that is relevant for public health microbiology, we reviewed the current literature published in PubMed between January, 2020, and July, 2022. We derived recommendations for EQA providers and their schemes for best practices to monitor pathogen-detection performance in future epidemics. We also showed laboratories, test facilities, and health authorities the information and benefits they can derive from EQA data, and from the non-EQA services of their providers.
Collapse
Affiliation(s)
- Christoph Buchta
- Austrian Association for Quality Assurance and Standardization of Medical and Diagnostic Tests, Vienna, Austria; European Organisation for External Quality Assurance Providers in Laboratory Medicine, Geneva, Switzerland.
| | - Heinz Zeichhardt
- INSTAND eV Society for Promoting Quality Assurance in Medical Laboratories, Düsseldorf, Germany; IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Germany; GBD Gesellschaft für Biotechnologische Diagnostik, Berlin, Germany
| | - Stephan W Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Jeremy V Camp
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Irene Görzer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | | | - Wolfgang Huf
- Karl Landsteiner Institute for Clinical Risk Management, Vienna, Austria
| | - Bernhard Benka
- Austrian Agency for Health and Food Safety, Vienna, Austria
| | | | - Martin Mielke
- Department for Infectious Diseases, Robert Koch-Institute, Berlin, Germany
| | - Andrea Griesmacher
- Austrian Association for Quality Assurance and Standardization of Medical and Diagnostic Tests, Vienna, Austria
| | - Mathias M Müller
- Austrian Association for Quality Assurance and Standardization of Medical and Diagnostic Tests, Vienna, Austria
| | - Ingo Schellenberg
- INSTAND eV Society for Promoting Quality Assurance in Medical Laboratories, Düsseldorf, Germany
| | - Martin Kammel
- INSTAND eV Society for Promoting Quality Assurance in Medical Laboratories, Düsseldorf, Germany; IQVD GmbH, Institut für Qualitätssicherung in der Virusdiagnostik, Berlin, Germany; GBD Gesellschaft für Biotechnologische Diagnostik, Berlin, Germany
| |
Collapse
|
524
|
Dusseldorp F, Bruins-van-Sonsbeek LGR, Buskermolen M, Niphuis H, Dirven M, Whelan J, Oude Munnink BB, Koopmans M, Fanoy EB, Sikkema RS, Tjon-A-Tsien A. SARS-CoV-2 in lions, gorillas and zookeepers in the Rotterdam Zoo, the Netherlands, a One Health investigation, November 2021. Euro Surveill 2023; 28:2200741. [PMID: 37440347 PMCID: PMC10347891 DOI: 10.2807/1560-7917.es.2023.28.28.2200741] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 03/07/2023] [Indexed: 07/15/2023] Open
Abstract
In November 2021, seven western lowland gorillas and four Asiatic lions were diagnosed with COVID-19 at Rotterdam Zoo. An outbreak investigation was undertaken to determine the source and extent of the outbreak and to identify possible transmission routes. Interviews were conducted with staff to identify human and animal contacts and cases, compliance with personal protective equipment (PPE) and potential transmission routes. Human and animal contacts and other animal species suspected to be susceptible to SARS-CoV-2 were tested for SARS-CoV-2 RNA. Positive samples were subjected to sequencing. All the gorillas and lions that could be tested (3/7 and 2/4, respectively) were RT-PCR positive between 12 November and 10 December 2021. No other animal species were SARS-CoV-2 RNA positive. Forty direct and indirect human contacts were identified. Two direct contacts tested RT-PCR positive 10 days after the first COVID-19 symptoms in animals. The zookeepers' viral genome sequences clustered with those of gorillas and lions. Personal protective equipment compliance was suboptimal at instances. Findings confirm transmission of SARS-CoV-2 among animals and between humans and animals but source and directionality could not be established. Zookeepers were the most likely source and should have periodic PPE training. Sick animals should promptly be tested and isolated/quarantined.
Collapse
Affiliation(s)
| | | | | | - Henk Niphuis
- Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | | | - Jane Whelan
- Public Health Services Rotterdam Rijnmond, the Netherlands
| | - Bas B Oude Munnink
- Department of Viroscience, Erasmus MC, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, the Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus MC, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, the Netherlands
| | - Ewout B Fanoy
- Public Health Services Rotterdam Rijnmond, the Netherlands
| | - Reina S Sikkema
- Department of Viroscience, Erasmus MC, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, the Netherlands
| | | |
Collapse
|
525
|
Noahsen P, Faber LL, Isidor S, Fonager J, Rasmussen M, Hansen HL. The COVID-19 pandemic in Greenland, epidemic features and impact of early strict measures, March 2020 to June 2022. Euro Surveill 2023; 28:2200767. [PMID: 37470739 PMCID: PMC10360370 DOI: 10.2807/1560-7917.es.2023.28.29.2200767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/17/2023] [Indexed: 07/21/2023] Open
Abstract
BackgroundThe COVID-19 pandemic was of major concern in Greenland. There was a high possibility of rapid transmission in settlements, and an increased risk of morbidity and mortality because of comorbidities in the population and limited access to specialised healthcare in remote areas.AimTo describe the epidemiology of the COVID-19 pandemic in Greenland and evaluate the effects of a strict COVID-19 strategy until risk groups were immunised.MethodsWe studied the epidemiology during March 2020 to June 2022. We describe the non-pharmaceutical interventions (NPIs), PCR-confirmed COVID-19 cases and vaccination coverage with data from the registries of the Greenlandic health authority.ResultsWe found 21,419 confirmed cases per 100,000 inhabitants (54% female, 46% male), 342 per 100,000 were hospitalised and 16 per 100,000 were admitted to the intensive care unit. The COVID-19 mortality rate was 39 per 100,000, all those affected were aged above 65 years. No excess overall mortality was observed. The vaccination coverage by June 2022 was 71.67 and 41% for one, two and three doses, respectively.ConclusionSARS-CoV-2 circulation in Greenland was low, given strict restrictions until all eligible inhabitants had been offered immunisation. The main impact of the pandemic was from May 2021 onwards with increasing numbers of confirmed cases. This occurred after introduction of the vaccine programme, which may have had an influence on the severity of the associated morbidity and mortality experienced. Halting community transmission of SARS-CoV-2 with NPIs until the majority of the population had been immunised was a successful strategy in Greenland.
Collapse
Affiliation(s)
- Paneeraq Noahsen
- National Board of Health in Greenland, Nuuk, Greenland
- Ilisimatusarfik, University of Greenland, Nuuk, Greenland
- Aalborg University, Aalborg, Denmark
- Arctic Health Research Centre, Aalborg University Hospital, Aalborg, Denmark
| | | | - Silvia Isidor
- Ilisimatusarfik, University of Greenland, Nuuk, Greenland
| | - Jannik Fonager
- Virus Research and Development section, Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Morten Rasmussen
- Virus Research and Development section, Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | |
Collapse
|
526
|
Razzaq M, Han JH, Ravichandran S, Kim J, Bae JY, Park MS, Kannappan S, Chung WC, Ahn JH, Song MJ, Kim KK. Stabilization of RNA G-quadruplexes in the SARS-CoV-2 genome inhibits viral infection via translational suppression. Arch Pharm Res 2023; 46:598-615. [PMID: 37563335 DOI: 10.1007/s12272-023-01458-x] [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: 01/13/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
The G-quadruplex (G4) formed in single-stranded DNAs or RNAs plays a key role in diverse biological processes and is considered as a potential antiviral target. In the genome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), 25 putative G4-forming sequences are predicted; however, the effects of G4-binding ligands on SARS-CoV-2 replication have not been studied in the context of viral infection. In this study, we investigated whether G4-ligands suppressed SARS-CoV-2 replication and whether their antiviral activity involved stabilization of viral RNA G4s and suppression of viral gene expression. We found that pyridostatin (PDS) suppressed viral gene expression and genome replication as effectively as the RNA polymerase inhibitor remdesivir. Biophysical analyses revealed that the 25 predicted G4s in the SARS-CoV-2 genome formed a parallel G4 structure. In particular, G4-644 and G4-3467 located in the 5' region of ORF1a, formed a G4 structure that could be effectively stabilized by PDS. We also showed that PDS significantly suppressed translation of the reporter genes containing these G4s. Taken together, our results demonstrate that stabilization of RNA G4s by PDS in the SARS-CoV-2 genome inhibits viral infection via translational suppression, highlighting the therapeutic potential of G4-ligands in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Maria Razzaq
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Ji Ho Han
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Subramaniyam Ravichandran
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
- Department of Biology, Stanford University, Stanford, United States of America
| | - Jaehyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Biosafety Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Shrute Kannappan
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Woo-Chang Chung
- Department of Microbiology, Graduate School of Basic Medical Science (GSBMS), Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Jin-Hyun Ahn
- Department of Microbiology, Graduate School of Basic Medical Science (GSBMS), Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Moon Jung Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
527
|
Giang TV, Hoa LNM, Hien TT, Cuong QD, Cap NT, Lam Vuong N, Thach PN. Traditional Vietnamese Medicine Containing Garlic Extract for Patients With Non-severe COVID-19: A Phase-II, Double-Blind, Randomized Controlled Trial. Cureus 2023; 15:e42484. [PMID: 37637574 PMCID: PMC10452047 DOI: 10.7759/cureus.42484] [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] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is still ongoing with the omicron variant. Low-cost, effective treatments are still needed, particularly in low-to-middle-income countries. This study assessed the safety and efficacy of TD0068, an herbal medicine developed from mainly garlic, for patients with non-severe COVID-19. Methods This is a phase-II, double-blind, randomized controlled trial to compare oral capsule TD0068 and placebo in adults aged 18-65 years with non-severe COVID-19 between September and October 2021. The efficacy outcomes measured included daily cycle threshold (Ct) value from the time of the initial reverse transcription-polymerase chain reaction (RT-PCR) test, time to viral clearance, daily symptom severity score from 15 symptoms of interest, time to symptom resolution, and progression to severe/critical COVID-19. Safety outcomes included adverse events (AEs) and serious adverse events (SAEs). Results Sixty patients were randomized (31 received TD0068, and 29 received a placebo). The two groups were balanced in baseline characteristics: mean age was 39 years, and female was predominant (66%). Daily Ct value (median on days 3, 5, 7, and 9 was 25.7, 30.8, 35.4, and 37.6 in the TD0068 group, and 26.4, 31.2, 36.0, and 37.4 in the placebo group, respectively) and time to viral clearance (median: 10 vs. 11 days in TD0068 and placebo groups) were similar between groups. Daily symptom severity score (median on days 3, 5, 7, and 9 was 2, 2, 1, and 0 in the TD0068 group, and 3, 2, 1, and 1 in the placebo group), and time to symptom resolution (median: seven vs. nine days, respectively) were also comparable between groups. No SAE occurred in the study. Conclusions TD0068 is safe but does not show an effect for non-severe COVID-19 patients. Further research is needed to explore the potential benefits of garlic in other forms or dosages for the treatment of COVID-19.
Collapse
Affiliation(s)
- Tran Van Giang
- Department of Viral and Parasitic Diseases, National Hospital for Tropical Diseases, Hanoi, VNM
- Department of Infectious Diseases, Hanoi Medical University, Hanoi, VNM
| | - Le Nguyen Minh Hoa
- Department of Microbiology and Molecular Biology, National Hospital for Tropical Diseases, Hanoi, VNM
| | - Tran Thi Hien
- Department of Viral and Parasitic Diseases, National Hospital for Tropical Diseases, Hanoi, VNM
| | - Quach Duy Cuong
- Department of Viral and Parasitic Diseases, National Hospital for Tropical Diseases, Hanoi, VNM
| | - Nguyen Trung Cap
- Emergency Department, National Hospital for Tropical Diseases, Hanoi, VNM
| | - Nguyen Lam Vuong
- Department of Medical Statistics and Informatics, Faculty of Public Health, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, VNM
| | - Pham Ngoc Thach
- Emergency Department, National Hospital for Tropical Diseases, Hanoi, VNM
| |
Collapse
|
528
|
Faucheux L, Bassolli de Oliveira Alves L, Chevret S, Rocha V. Comparison of characteristics and laboratory tests of COVID-19 hematological patients from France and Brazil during the pre-vaccination period: identification of prognostic profiles for survival. Hematol Transfus Cell Ther 2023; 45:306-316. [PMID: 35673599 PMCID: PMC9159977 DOI: 10.1016/j.htct.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/29/2022] [Accepted: 05/04/2022] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION COVID-19 disease presentation is heterogeneous, from asymptomatic up to severe life-threatening forms. Getting further insights into patients with specific diseases is of particular interest. We aimed to identify profiles of hematology patients hospitalized with COVID-19 that would be associated with survival and to assess the differences between cohorts METHODS: A binational cohort of 263 patients with COVID-19 and hematological disease was studied in Paris, France and São Paulo, Brazil. Patient profiles were based on age, comorbidities, biological measurements, COVID-19 symptoms and hematological disease characteristics. A semi-supervised learning method with a survival endpoint was first used, following which, a classifier was identified to allow the classification of patients using only baseline information MAIN RESULTS: Two profiles of patients were identified, one being young patients with few comorbidities and low C-reactive protein (CRP), D-dimers, lactate dehydrogenase (LDH) and creatinine levels, and the other, older patients, with several comorbidities and high levels of the 4 biology markers. The profiles were strongly associated with survival (p < 0.0001), even after adjusting for age (p = 0.0002). The 30-day survival rate was 77.1% in the first profiles, versus 46.7% in the second. The Brazilian analysis emphasized the importance of age, while the French focused on the comorbidities CONCLUSION: This analysis showed the importance of CRP, LHD and creatinine in the COVID-19 presentation and prognosis, whatever the geographic origin of the patients.
Collapse
Affiliation(s)
- Lilith Faucheux
- Hôpital Saint Louis, Université de Paris, Paris, France; Université de Paris, INSERM U976, Paris, France.
| | | | | | - Vanderson Rocha
- Hospital das Clinicas, Faculty of Medicine, Universidade de São Paulo (HCFM-USP), São Paulo, SP, Brazil; Churchill Hospital, Oxford University, Oxford, UK
| |
Collapse
|
529
|
Devignot S, Sha TW, Burkard TR, Schmerer P, Hagelkruys A, Mirazimi A, Elling U, Penninger JM, Weber F. Low-density lipoprotein receptor-related protein 1 (LRP1) as an auxiliary host factor for RNA viruses. Life Sci Alliance 2023; 6:e202302005. [PMID: 37072184 PMCID: PMC10114362 DOI: 10.26508/lsa.202302005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
Viruses with an RNA genome are often the cause of zoonotic infections. In order to identify novel pro-viral host cell factors, we screened a haploid insertion-mutagenized mouse embryonic cell library for clones that are resistant to Rift Valley fever virus (RVFV). This screen returned the low-density lipoprotein receptor-related protein 1 (LRP1) as a top hit, a plasma membrane protein involved in a wide variety of cell activities. Inactivation of LRP1 in human cells reduced RVFV RNA levels already at the attachment and entry stages of infection. Moreover, the role of LRP1 in promoting RVFV infection was dependent on physiological levels of cholesterol and on endocytosis. In the human cell line HuH-7, LRP1 also promoted early infection stages of sandfly fever Sicilian virus and La Crosse virus, but had a minor effect on late infection by vesicular stomatitis virus, whereas encephalomyocarditis virus was entirely LRP1-independent. Moreover, siRNA experiments in human Calu-3 cells demonstrated that also SARS-CoV-2 infection benefitted from LRP1. Thus, we identified LRP1 as a host factor that supports infection by a spectrum of RNA viruses.
Collapse
Affiliation(s)
- Stephanie Devignot
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Tim Wai Sha
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Thomas R Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Patrick Schmerer
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Astrid Hagelkruys
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Ali Mirazimi
- Public Health Agency of Sweden, Solna, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Solna, Sweden
| | - Ulrich Elling
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
- German Centre for Infection Research (DZIF), Partner Site Giessen, Giessen, Germany
| |
Collapse
|
530
|
Huang W, Zhang Z, Lin D, Deng Y, Chen X, Huang J. RT-nestRPA is a new technology for the rapid and sensitive detection of nucleic acid detection of pathogens used for a variety of medical application scenarios. Anal Chim Acta 2023; 1262:341263. [PMID: 37179064 PMCID: PMC10123537 DOI: 10.1016/j.aca.2023.341263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND The effective detection of pathogens is of great importance for the diagnosis and treatment of infectious diseases. We have proposed the novel RT-nestRPA technique for SARS-CoV-2 detection, which is a rapid RNA detection technique with ultra-high sensitivity. RESULTS The RT-nestRPA technology has a sensitivity of 0.5 copies/uL of synthetic RNA targeting the ORF7a/7b/8 gene or 1 copy/uL synthetic RNA targeting the N gene of SARS-CoV-2. The entire detection process of RT-nestRPA only takes only 20 min, which is significantly shorter than RT-qPCR (nearly 100 min). Additionally, RT-nestRPA is capable of detecting dual genes of SARS-CoV-2 and human RPP30 simultaneously in one reaction tube. The excellent specificity of RT-nestRPA was verified by analyzing twenty-two SARS-CoV-2 unrelated pathogens. Furthermore, RT-nestRPA had great performance in detecting samples treated with cell lysis buffer without RNA extraction. The innovative double-layer reaction tube for RT-nestRPA can prevent aerosol contamination and simplify the reaction operation. Moreover, the ROC analysis revealed that RT-nestRPA had high diagnostic value (AUC = 0.98), while the AUC of RT-qPCR was 0.75. SIGNIFICANCE Our current findings suggested that RT-nestRPA could serve as a novel technology for nucleic acid detection of pathogens with rapid and ultrahigh sensitive features used in various medical application scenarios.
Collapse
Affiliation(s)
- Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Zhaoqi Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dachuan Lin
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
531
|
Cabrera A, Al Mutawah F, Kadour M, Schofield S, Conkey B, Fuller J, Payne M, Elsayed S, Delport J. Increasing SARS-CoV-2 testing capacity through specimen pooling: An acute care center experience. PLoS One 2023; 18:e0267137. [PMID: 37379564 DOI: 10.1371/journal.pone.0267137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/01/2022] [Indexed: 06/30/2023] Open
Abstract
Innovation in laboratory testing algorithms to address seemingly uncontrollable global supply chain shortages in plastics and other consumables during emergencies such as the current COVID-19 pandemic have been urgently needed. We report our experience with specimen pooling on SARS-CoV-2 testing in an acute care hospital microbiology laboratory during a high testing demand period that exceeded available processing capacity. A fully automated four-in-one pooling algorithm was designed and validated. Correlation and agreement were calculated. A custom Microsoft Excel tool was designed for use by the technologists to aid interpretation, verification and result entry. Cost-per-test impact for pooling was measured in reference to the consumable cost and was denoted as the percentage reduction of cost versus the baseline cost-per-test of testing specimens individually. Validation showed a strong correlation between the signals observed when testing specimens individually versus those that were pooled. Average crossing point difference was 1.352 cycles (95% confidence interval of -0.235 and 2.940). Overall agreement observed between individually and pooled tested specimens was 96.8%. Stratified agreement showed an expected decreased performance of pooling for weakly positive specimens dropping below 60% after a crossing point of 35. Post-implementation data showed the consumable cost-savings achieved through this algorithm was 85.5% after 8 months, creating both testing and resource capacity. Pooling is an effective method to be used for SARS-CoV-2 testing during the current pandemic to address resource shortages and provide quick turnaround times for high test volumes without compromising performance.
Collapse
Affiliation(s)
- Ana Cabrera
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Microbiology and Immunology Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Fatimah Al Mutawah
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Mike Kadour
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Shannon Schofield
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
| | - Beverley Conkey
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
| | - Jeffrey Fuller
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Michael Payne
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sameer Elsayed
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Johan Delport
- Pathology and Laboratory Medicine Department, London Health Sciences Centre, London, Ontario, Canada
- Pathology and Laboratory Medicine Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Microbiology and Immunology Department, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
532
|
Jonker L, Linde KJ, de Boer AR, Ding E, Zhang D, de Hoog MLA, Herfst S, Heederik DJJ, Fraaij PLA, Bluyssen PM, Wouters IM, Bruijning-Verhagen PCJL. SARS-CoV-2 incidence in secondary schools; the role of national and school-initiated COVID-19 measures. BMC Public Health 2023; 23:1243. [PMID: 37370045 DOI: 10.1186/s12889-023-16146-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION Our aim was to gain insight into the effect of COVID-19 measures on SARS-CoV-2 incidence in secondary schools and the association with classroom CO2 concentration and airborne contamination. METHODS Between October 2020-June 2021, 18 schools weekly reported SARS-CoV-2 incidence and completed surveys on school-initiated COVID-19 measures (e.g. improving hygiene or minimizing contacts). CO2 was measured in occupied classrooms twice, and SARS-CoV-2 air contamination longitudinally using electrostatic dust collectors (EDC) and analyzed using RT-qPCR. National COVID-19 policy measures varied during pre-lockdown, lockdown and post-lockdown periods. During the entire study, schools were recommended to improve ventilation. SARS-CoV-2 incidence rate ratios (IRR) were estimated by Generalized Estimating Equation (GEE) models. RESULTS During 18 weeks follow-up (range: 10-22) SARS-CoV-2 school-incidence decreased during national lockdown (adjusted IRR: 0.41, 95%CI: 0.21-0.80) and post-lockdown (IRR: 0.60, 0.39-0.93) compared to pre-lockdown. School-initiated COVID-19 measures had no additional effect. Pre-lockdown, IRRs per 10% increase in time CO2 exceeded 400, 550 and 800 ppm above outdoor level respectively, were 1.08 (1.00-1.16), 1.10 (1.02-1.19), and 1.08 (0.95-1.22). Post-lockdown, CO2-concentrations were considerably lower and not associated with SARS-CoV-2 incidence. No SARS-CoV-2 RNA was detected in any of the EDC samples. CONCLUSION During a period with low SARS-CoV-2 population immunity and increased attention to ventilation, with CO2 levels most of the time below acceptable thresholds, only the national policy during and post-lockdown of reduced class-occupancy, stringent quarantine, and contact testing reduced SARS-CoV-2 incidence in Dutch secondary schools. Widespread SARS-CoV-2 air contamination could not be demonstrated in schools under the prevailing conditions during the study.
Collapse
Affiliation(s)
- L Jonker
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - K J Linde
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | - A R de Boer
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| | - E Ding
- Faculty of Architecture and the Built Environment, Delft University of Technology, Julianalaan 134, 2628 BL, Delft, the Netherlands
| | - D Zhang
- Faculty of Architecture and the Built Environment, Delft University of Technology, Julianalaan 134, 2628 BL, Delft, the Netherlands
| | - M L A de Hoog
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - S Herfst
- Department of Viroscience, Erasmus MC, Dr. Molewaterplein 50, 3015 GE, 3000 CA, Rotterdam, Netherlands
| | - D J J Heederik
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | - P L A Fraaij
- Department of Viroscience, Erasmus MC, Dr. Molewaterplein 50, 3015 GE, 3000 CA, Rotterdam, Netherlands
- Department of Pediatrics, Erasmus MC, Dr. Molewaterplein 50, 3015 GE, 3000 CA, Rotterdam, Netherlands
| | - P M Bluyssen
- Faculty of Architecture and the Built Environment, Delft University of Technology, Julianalaan 134, 2628 BL, Delft, the Netherlands
| | - I M Wouters
- Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | - P C J L Bruijning-Verhagen
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
533
|
Solís-Huerta F, Martinez-Guerra BA, Roman-Montes CM, Tamez-Torres KM, Rajme-Lopez S, Ortíz-Conchi N, López-García NI, Villalobos-Zapata GY, Rangel-Cordero A, Santiago-Cruz J, Xancal-Salvador LF, Méndez-Ramos S, Ochoa-Hein E, Galindo-Fraga A, Ponce-de-Leon A, Gonzalez-Lara MF, Sifuentes-Osornio J. Risk Factors Associated with the Development of Hospital-Acquired Infections in Hospitalized Patients with Severe COVID-19. Antibiotics (Basel) 2023; 12:1108. [PMID: 37508204 PMCID: PMC10376785 DOI: 10.3390/antibiotics12071108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Recognition of risk factors for hospital-acquired infections (HAI) in patients with COVID-19 is warranted. We aimed to describe factors associated with the development of HAI in patients with severe COVID-19. We conducted a retrospective cohort study including all adult patients admitted with severe COVID-19 between March 2020 and November 2020. The primary outcome was HAI development. Bivariate and multiple logistic regression models were constructed. Among 1540 patients, HAI occurred in 221 (14%). A total of 299 episodes of HAI were registered. The most common HAI were hospital-acquired/ventilation-associated pneumonia (173 episodes) and primary bloodstream infection (66 episodes). Death occurred in 387 (35%) patients and was more frequent in patients with HAI (38% vs. 23%, p < 0.01). Early mechanical ventilation (aOR 18.78, 95% CI 12.56-28.07), chronic kidney disease (aOR 3.41, 95% CI 1.4-8.27), use of corticosteroids (aOR 2.95, 95% CI 1.92-4.53) and tocilizumab (aOR 2.68, 95% CI 1.38-5.22), age ≥ 60 years (aOR 1.91, 95% CI 1.27-2.88), male sex (aOR 1.52, 95% CI 1.03-2.24), and obesity (aOR 1.49, 95% CI 1.03-2.15) were associated with HAI. In patients with severe COVID-19, mechanical ventilation within the first 24 h upon admission, chronic kidney disease, use of corticosteroids, use of tocilizumab, age ≥ 60 years, male sex, and obesity were associated with a higher risk of HAI.
Collapse
Affiliation(s)
- Fernando Solís-Huerta
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Medicine, Mexico City 14080, Mexico;
| | - Bernardo Alfonso Martinez-Guerra
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (C.M.R.-M.); (K.M.T.-T.); (S.R.-L.); (A.P.-d.-L.)
| | - Carla Marina Roman-Montes
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (C.M.R.-M.); (K.M.T.-T.); (S.R.-L.); (A.P.-d.-L.)
| | - Karla Maria Tamez-Torres
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (C.M.R.-M.); (K.M.T.-T.); (S.R.-L.); (A.P.-d.-L.)
| | - Sandra Rajme-Lopez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (C.M.R.-M.); (K.M.T.-T.); (S.R.-L.); (A.P.-d.-L.)
| | - Narciso Ortíz-Conchi
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Norma Irene López-García
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Guadalupe Yvonne Villalobos-Zapata
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Andrea Rangel-Cordero
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Janet Santiago-Cruz
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Luis Fernando Xancal-Salvador
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Steven Méndez-Ramos
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Eric Ochoa-Hein
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Hospital Epidemiology Department, Mexico City 14080, Mexico; (E.O.-H.); (A.G.-F.)
| | - Arturo Galindo-Fraga
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Hospital Epidemiology Department, Mexico City 14080, Mexico; (E.O.-H.); (A.G.-F.)
| | - Alfredo Ponce-de-Leon
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (C.M.R.-M.); (K.M.T.-T.); (S.R.-L.); (A.P.-d.-L.)
| | - Maria Fernanda Gonzalez-Lara
- Clinical Microbiology Laboratory, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Department of Infectious Diseases, Mexico City 14080, Mexico; (N.O.-C.); (N.I.L.-G.); (G.Y.V.-Z.); (A.R.-C.); (J.S.-C.); (L.F.X.-S.); (S.M.-R.); (M.F.G.-L.)
| | - Jose Sifuentes-Osornio
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, General Direction, Mexico City 14080, Mexico
| |
Collapse
|
534
|
Prebensen C, Lefol Y, Myhre PL, Lüders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023; 13:10368. [PMID: 37365222 PMCID: PMC10293211 DOI: 10.1038/s41598-023-37606-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
A maladaptive inflammatory response has been implicated in the pathogenesis of severe COVID-19. This study aimed to characterize the temporal dynamics of this response and investigate whether severe disease is associated with distinct gene expression patterns. We performed microarray analysis of serial whole blood RNA samples from 17 patients with severe COVID-19, 15 patients with moderate disease and 11 healthy controls. All study subjects were unvaccinated. We assessed whole blood gene expression patterns by differential gene expression analysis, gene set enrichment, two clustering methods and estimated relative leukocyte abundance using CIBERSORT. Neutrophils, platelets, cytokine signaling, and the coagulation system were activated in COVID-19, and this broad immune activation was more pronounced in severe vs. moderate disease. We observed two different trajectories of neutrophil-associated genes, indicating the emergence of a more immature neutrophil phenotype over time. Interferon-associated genes were strongly enriched in early COVID-19 before falling markedly, with modest severity-associated differences in trajectory. In conclusion, COVID-19 necessitating hospitalization is associated with a broad inflammatory response, which is more pronounced in severe disease. Our data suggest a progressively more immature circulating neutrophil phenotype over time. Interferon signaling is enriched in COVID-19 but does not seem to drive severe disease.
Collapse
Affiliation(s)
- Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Kirkeveien 166, 0450, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yohan Lefol
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Blomfeldt
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan-Erik Berdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
535
|
Colado Simão AN, Perugini Stadtlober N, Stinghen Garcia Lonni AA, Venâncio LM, Lerner Trigo G, de Souza Cassela PLC, Mastellini Sanches Silva T, De Fátima Oliveira Hirth Ruiz M, Batisti Lozovoy MA, Tano ZN, da Fonseca Orcina B, Vieira Vilhena F, da Silva Santos PS. Effect of phthalocyanine oral and nasal antiseptic solutions on the infectivity of SARS-CoV-2 in patients with COVID-19: a randomized controlled trial. GERMAN MEDICAL SCIENCE : GMS E-JOURNAL 2023; 21:Doc07. [PMID: 37426884 PMCID: PMC10326526 DOI: 10.3205/000321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 11/16/2022] [Indexed: 07/11/2023]
Abstract
Background In individuals with coronavirus disease (COVID-19), the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral load (VL) plays an important role in infectivity. Objectives This study aimed to evaluate the reduction in the VL and infectivity induced by phthalocyanine mouthwash and nasal spray in patients with COVID-19. Methods Patients with mild COVID-19 were recruited to participate in a triple-blinded randomized controlled trial. Participants were assigned to one of three groups: Group 1, non-active mouthwash and saline nasal spray (SNS); Group 2, phthalocyanine mouthwash and SNS; and Group 3 phthalocyanine mouthwash and phthalocyanine nasal spray. VL was assessed in nasopharyngeal and oropharyngeal swabs collected at the time of clinical diagnosis at baseline as well as 24 and 72 hours after starting the rinsing protocols. Findings Forty-six participants were included in the analysis: 15, 16, and 15 in Groups 1, 2, and 3, respectively. After 72 hours, the reduction in VL was significantly higher in Group 3 (mean cycle threshold (Ct) decrease: 11.21) than in Group 1 (mean Ct decrease: 5.53). Additionally, only the mean VL in Group 3 was reduced to a non-contagious level after 72 hours. Main conclusions Use of phthalocyanine mouthwash and nasal spray is effective at reducing SARS-CoV-2 infectivity.
Collapse
Affiliation(s)
| | | | | | - Luiza Mara Venâncio
- Research Laboratory in Applied Immunology, State University of Londrina, Brazil
| | | | | | | | | | | | - Zuleica Naomi Tano
- Research Laboratory in Applied Immunology, State University of Londrina, Brazil
| | - Bernardo da Fonseca Orcina
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| |
Collapse
|
536
|
Lista F, Peragallo MS, Biselli R, De Santis R, Mariotti S, Nisini R, D'Amelio R. Have Diagnostics, Therapies, and Vaccines Made the Difference in the Pandemic Evolution of COVID-19 in Comparison with "Spanish Flu"? Pathogens 2023; 12:868. [PMID: 37513715 PMCID: PMC10384375 DOI: 10.3390/pathogens12070868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
In 1918 many countries, but not Spain, were fighting World War I. Spanish press could report about the diffusion and severity of a new infection without censorship for the first-time, so that this pandemic is commonly defined as "Spanish flu", even though Spain was not its place of origin. "Spanish flu" was one of the deadliest pandemics in history and has been frequently compared with the coronavirus disease (COVID)-19 pandemic. These pandemics share similarities, being both caused by highly variable and transmissible respiratory RNA viruses, and diversity, represented by diagnostics, therapies, and especially vaccines, which were made rapidly available for COVID-19, but not for "Spanish flu". Most comparison studies have been carried out in the first period of COVID-19, when these resources were either not yet available or their use had not long started. Conversely, we wanted to analyze the role that the advanced diagnostics, anti-viral agents, including monoclonal antibodies, and innovative COVID-19 vaccines, may have had in the pandemic containment. Early diagnosis, therapies, and anti-COVID-19 vaccines have markedly reduced the pandemic severity and mortality, thus preventing the collapse of the public health services. However, their influence on the reduction of infections and re-infections, thus on the transition from pandemic to endemic condition, appears to be of minor relevance. The high viral variability of influenza and coronavirus may probably be contained by the development of universal vaccines, which are not easy to be obtained. The only effective weapon still remains the disease prevention, to be achieved with the reduction of promiscuity between the animal reservoirs of these zoonotic diseases and humans.
Collapse
Affiliation(s)
- Florigio Lista
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Mario Stefano Peragallo
- Centro Studi e Ricerche di Sanità e Veterinaria, Comando Logistico dell'Esercito, 00184 Roma, Italy
| | - Roberto Biselli
- Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Riccardo De Santis
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza, Università di Roma, 00161 Roma, Italy
| | - Sabrina Mariotti
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Raffaele D'Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza, Università di Roma, 00198 Roma, Italy
| |
Collapse
|
537
|
de Moor WRJ, Williamson AL, Schäfer G, Douglass N, Gers S, Sutherland AD, Blumenthal MJ, Margolin E, Shaw ML, Preiser W, Chapman R. LSDV-Vectored SARS-CoV-2 S and N Vaccine Protects against Severe Clinical Disease in Hamsters. Viruses 2023; 15:1409. [PMID: 37515096 PMCID: PMC10383203 DOI: 10.3390/v15071409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 07/30/2023] Open
Abstract
The SARS-CoV-2 pandemic demonstrated the need for potent and broad-spectrum vaccines. This study reports the development and testing of a lumpy skin disease virus (LSDV)-vectored vaccine against SARS-CoV-2, utilizing stabilized spike and conserved nucleocapsid proteins as antigens to develop robust immunogenicity. Construction of the vaccine (LSDV-SARS2-S,N) was confirmed by polymerase chain reaction (PCR) amplification and sequencing. In vitro characterization confirmed that cells infected with LSDV-SARS2-S,N expressed SARS-CoV-2 spike and nucleocapsid protein. In BALB/c mice, the vaccine elicited high magnitude IFN-γ ELISpot responses (spike: 2808 SFU/106 splenocytes) and neutralizing antibodies (ID50 = 6552). Testing in hamsters, which emulate human COVID-19 disease progression, showed the development of high titers of neutralizing antibodies against the Wuhan and Delta SARS-CoV-2 variants (Wuhan ID50 = 2905; Delta ID50 = 4648). Additionally, hamsters vaccinated with LSDV-SARS2-S,N displayed significantly less weight loss, lung damage, and reduced viral RNA copies following SARS-CoV-2 infection with the Delta variant as compared to controls, demonstrating protection against disease. These data demonstrate that LSDV-vectored vaccines display promise as an effective SARS-CoV-2 vaccine and as a potential vaccine platform for communicable diseases in humans and animals. Further efficacy testing and immune response analysis, particularly in non-human primates, are warranted.
Collapse
Affiliation(s)
- Warren R J de Moor
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Anna-Lise Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Georgia Schäfer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town 7925, South Africa
- Wellcome Trust Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town 7925, South Africa
| | - Nicola Douglass
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | | | - Andrew D Sutherland
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
| | - Melissa J Blumenthal
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town 7925, South Africa
| | - Emmanuel Margolin
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Trust Centre for Infectious Disease Research in Africa, University of Cape Town, Cape Town 7925, South Africa
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Cape Town 7701, South Africa
| | - Megan L Shaw
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Bellville, Cape Town 7535, South Africa
| | - Wolfgang Preiser
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Cape Town 7505, South Africa
| | - Rosamund Chapman
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
538
|
Wang J, Xie Q, Song H, Chen X, Zhang X, Zhao X, Hao Y, Zhang Y, Li H, Li N, Fan K, Wang X. Utilizing nanozymes for combating COVID-19: advancements in diagnostics, treatments, and preventative measures. J Nanobiotechnology 2023; 21:200. [PMID: 37344839 PMCID: PMC10283317 DOI: 10.1186/s12951-023-01945-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/29/2023] [Indexed: 06/23/2023] Open
Abstract
The emergence of human severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses significant challenges to global public health. Despite the extensive efforts of researchers worldwide, there remains considerable opportunities for improvement in timely diagnosis, specific treatment, and effective vaccines for SARS-CoV-2. This is due, in part, to the large number of asymptomatic carriers, rapid virus mutations, inconsistent confinement policies, untimely diagnosis and limited clear treatment plans. The emerging of nanozymes offers a promising approach for combating SARS-CoV-2 due to their stable physicochemical properties and high surface areas, which enable easier and multiple nano-bio interactions in vivo. Nanozymes inspire the development of sensitive and economic nanosensors for rapid detection, facilitate the development of specific medicines with minimal side effects for targeted therapy, trigger defensive mechanisms in the form of vaccines, and eliminate SARS-CoV-2 in the environment for prevention. In this review, we briefly present the limitations of existing countermeasures against coronavirus disease 2019 (COVID-19). We then reviewed the applications of nanozyme-based platforms in the fields of diagnostics, therapeutics and the prevention in COVID-19. Finally, we propose opportunities and challenges for the further development of nanozyme-based platforms for COVID-19. We expect that our review will provide valuable insights into the new emerging and re-emerging infectious pandemic from the perspective of nanozymes.
Collapse
Affiliation(s)
- Jia Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Qingpeng Xie
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Haoyue Song
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Xiaohang Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Xiaoxuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Xiangyu Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Yujia Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Yuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Huifei Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Na Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001 China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 China
| |
Collapse
|
539
|
Martins F, Gonçalves FT, Imamura M, Barboza DS, Matheus D, Pereira MFB, Marques HHS, Correa-Silva S, Montenegro MM, Fink TT, Lindoso L, Bain V, Ferreira JCOA, Astley C, Matsuo OM, Suguita P, Trindade V, Paula CSY, Litvinov N, Palmeira P, Gualano B, Delgado AF, Carneiro-Sampaio M, Forsait S, Odone-Filho V, Antonangelo L, Battistella LR, Silva CA. Health-related quality of life and functionality in primary caregiver of surviving pediatric COVID-19. Front Public Health 2023; 11:1117854. [PMID: 37408752 PMCID: PMC10319046 DOI: 10.3389/fpubh.2023.1117854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
Objectives To prospectively assess health-related quality of life (HRQoL), global functionality, and disability in primary caregivers of surviving children and adolescents after COVID-19. Methods A longitudinal observational study was carried out on primary caregivers of surviving pediatric post-COVID-19 patients (n = 51) and subjects without COVID-19 (n = 60). EuroQol five-dimension five-level questionnaire (EQ-5D-5L) and 12-question WHO Disability Assessment Schedule 2.0 (WHODAS 2.0) were answered for both groups. The univariate regression analysis was carried out using SPSS (v 20) and significance was established at 5%. Results The median duration between COVID-19 diagnosis in children and adolescents and longitudinal follow-up visits was 4.4 months (0.8-10.7). The median age of children and adolescents caregivers with laboratory-confirmed COVID-19 was similar to primary caregivers of subjects without laboratory-confirmed COVID-19 [43.2 (31.6-60.9) vs. 41.5 (21.6-54.8) years, p = 0.08], as well as similar female sex (p = 1.00), level of schooling (p = 0.11), social assistance program (p = 0.28), family income/month U$ (p = 0.25) and the number of household's members in the residence (p = 0.68). The frequency of slight to extreme problems (level ≥ 2) of the pain/discomfort domain according to EQ-5D-5L score was significantly higher in the former group [74% vs. 52.5%, p = 0.03, OR = 2.57 (1.14-5.96)]. The frequency of disability according to WHODAS 2.0 total score was similar to those without disability and unknown (p = 0.79); however, with a very high disability in both groups (72.5% and 78.3%). Further analysis of primary caregivers of children and adolescents with post-COVID-19 condition (PCC) [n = 12/51 (23%)] compared to those without PCC [n = 39/51(77%)] revealed no differences between demographic data, EQ-5D-5L and WHODAS 2.0 scores in both groups (p > 0.05). Conclusion We longitudinally demonstrated that pain/discomfort were predominantly reported in approximately 75% of primary caregiver of COVID-19 patients, with high disability in approximately three-quarters of both caregiver groups. These data emphasized the prospective and systematic caregiver burden evaluation relevance of pediatric COVID-19.
Collapse
Affiliation(s)
- Fernanda Martins
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
- Instituto de Medicina Física e Reabilitação, Hospital das Clínicas HCFMUSP, São Paulo, SP, Brazil
| | - Fernanda T. Gonçalves
- Laboratorio de Imunohematologia e Hematologia Forense (LIM-40), Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marta Imamura
- Instituto de Medicina Física e Reabilitação, Hospital das Clínicas HCFMUSP, São Paulo, SP, Brazil
- Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Daniela S. Barboza
- Instituto de Medicina Física e Reabilitação, Hospital das Clínicas HCFMUSP, São Paulo, SP, Brazil
| | - Denise Matheus
- Instituto de Medicina Física e Reabilitação, Hospital das Clínicas HCFMUSP, São Paulo, SP, Brazil
| | - Maria Fernanda B. Pereira
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Heloisa H. S. Marques
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Simone Correa-Silva
- Laboratório de Pediatria Clínica (LIM-36), Departamento de Pediatria, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marilia M. Montenegro
- Laboratório de Pediatria Clínica (LIM-36), Departamento de Pediatria, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Thais T. Fink
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Livia Lindoso
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vera Bain
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Juliana C. O. A. Ferreira
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Camilla Astley
- Faculdade de Medicina, Divisão de Reumatologia, Hospital das Clínicas, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Olivia M. Matsuo
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Priscila Suguita
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vitor Trindade
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Camila S. Y. Paula
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Nadia Litvinov
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Patricia Palmeira
- Laboratório de Pediatria Clínica (LIM-36), Departamento de Pediatria, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bruno Gualano
- Faculdade de Medicina, Divisão de Reumatologia, Hospital das Clínicas, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Artur F. Delgado
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Magda Carneiro-Sampaio
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvana Forsait
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vicente Odone-Filho
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Leila Antonangelo
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Linamara R. Battistella
- Instituto de Medicina Física e Reabilitação, Hospital das Clínicas HCFMUSP, São Paulo, SP, Brazil
- Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Clovis A. Silva
- Faculdade de Medicina, Instituto da Criança e do Adolescente, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
540
|
Ciannella S, González-Fernández C, Gomez-Pastora J. Recent progress on wastewater-based epidemiology for COVID-19 surveillance: A systematic review of analytical procedures and epidemiological modeling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:162953. [PMID: 36948304 PMCID: PMC10028212 DOI: 10.1016/j.scitotenv.2023.162953] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 05/13/2023]
Abstract
On March 11, 2020, the World Health Organization declared the coronavirus disease 2019 (COVID-19), whose causative agent is the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a pandemic. This virus is predominantly transmitted via respiratory droplets and shed via sputum, saliva, urine, and stool. Wastewater-based epidemiology (WBE) has been able to monitor the circulation of viral pathogens in the population. This tool demands both in-lab and computational work to be meaningful for, among other purposes, the prediction of outbreaks. In this context, we present a systematic review that organizes and discusses laboratory procedures for SARS-CoV-2 RNA quantification from a wastewater matrix, along with modeling techniques applied to the development of WBE for COVID-19 surveillance. The goal of this review is to present the current panorama of WBE operational aspects as well as to identify current challenges related to it. Our review was conducted in a reproducible manner by following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews. We identified a lack of standardization in wastewater analytical procedures. Regardless, the reverse transcription-quantitative polymerase chain reaction (RT-qPCR) approach was the most reported technique employed to detect and quantify viral RNA in wastewater samples. As a more convenient sample matrix, we suggest the solid portion of wastewater to be considered in future investigations due to its higher viral load compared to the liquid fraction. Regarding the epidemiological modeling, the data-driven approach was consistently used for the prediction of variables associated with outbreaks. Future efforts should also be directed toward the development of rapid, more economical, portable, and accurate detection devices.
Collapse
Affiliation(s)
- Stefano Ciannella
- Department of Chemical Engineering, Texas Tech University, Lubbock 79409, TX, USA.
| | - Cristina González-Fernández
- Department of Chemical Engineering, Texas Tech University, Lubbock 79409, TX, USA; Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Avda. Los Castros, s/n, 39005 Santander, Spain.
| | | |
Collapse
|
541
|
Fujisawa M, Adachi Y, Onodera T, Shiwa-Sudo N, Iwata-Yoshikawa N, Nagata N, Suzuki T, Takeoka S, Takahashi Y. High-throughput isolation of SARS-CoV-2 nucleocapsid antibodies for improved antigen detection. Biochem Biophys Res Commun 2023; 673:114-120. [PMID: 37379800 PMCID: PMC10279465 DOI: 10.1016/j.bbrc.2023.06.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
SARS-CoV-2 nucleocapsid protein (NP) is the main target for COVID-19-diagnostic PCR and antigen rapid diagnostic tests (Ag-RDTs). Ag-RDTs are more convenient than PCR tests for point-of-care testing or self-testing to identify the SARS-CoV-2 antigen. The sensitivity and specificity of this method depends mainly on the affinity and specificity of NP-binding antibodies; therefore, antigen-antibody binding is key elements for the Ag-RDTs. Here, we applied the high-throughput antibody isolation platform that has been utilized to isolate therapeutic antibodies against rare epitopes. Two NP antibodies were identified to recognize non-overlapping epitopes with high affinity. One antibody specifically binds to SARS-CoV-2 NP, and the other rapidly and tightly binds to SARS-CoV-2 NP with cross-reactivity to SARS-CoV NP. Furthermore, these antibodies were compatible with a sandwich enzyme-linked immunosorbent assay that exhibited enhanced sensitivity for NP detection compared to the previously isolated NP antibodies. Thus, the NP antibody pair is applicable to more sensitive and specific Ag-RDTs, highlighting the utility of a high-throughput antibody isolation platform for diagnostics development.
Collapse
Affiliation(s)
- Mizuki Fujisawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Nozomi Shiwa-Sudo
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Research Institute for Science and Engineering, Waseda University, 3-4-1, Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
542
|
Ebraham L, Xu C, Wang A, Hernandez C, Siclari N, Rajah D, Walter L, Marras SAE, Tyagi S, Fine DH, Daep CA, Chang TL. Oral Epithelial Cells Expressing Low or Undetectable Levels of Human Angiotensin-Converting Enzyme 2 Are Susceptible to SARS-CoV-2 Virus Infection In Vitro. Pathogens 2023; 12:843. [PMID: 37375533 DOI: 10.3390/pathogens12060843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The oral cavity is thought to be one of the portals for SARS-CoV-2 entry, although there is limited evidence of active oral infection by SARS-CoV-2 viruses. We assessed the capacity of SARS-CoV-2 to infect and replicate in oral epithelial cells. Oral gingival epithelial cells (hTERT TIGKs), salivary gland epithelial cells (A-253), and oral buccal epithelial cells (TR146), which occupy different regions of the oral cavity, were challenged with replication-competent SARS-CoV-2 viruses and with pseudo-typed viruses expressing SARS-CoV-2 spike proteins. All oral epithelial cells expressing undetectable or low levels of human angiotensin-converting enzyme 2 (hACE2) but high levels of the alternative receptor CD147 were susceptible to SARS-CoV-2 infection. Distinct viral dynamics were seen in hTERT TIGKs compared to A-253 and TR146 cells. For example, levels of viral transcripts were sustained in hTERT TIGKs but were significantly decreased in A-253 and TR146 cells on day 3 after infection. Analysis of oral epithelial cells infected by replication-competent SARS-CoV-2 viruses expressing GFP showed that the GFP signal and SARS-CoV-2 mRNAs were not evenly distributed. Furthermore, we found cumulative SARS-CoV-2 RNAs from released viruses in the media from oral epithelial cells on day 1 and day 2 after infection, indicating productive viral infection. Taken together, our results demonstrated that oral epithelial cells were susceptible to SARS-CoV-2 viruses despite low or undetectable levels of hACE2, suggesting that alternative receptors contribute to SARS-CoV-2 infection and may be considered for the development of future vaccines and therapeutics.
Collapse
Affiliation(s)
- Laith Ebraham
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Chuan Xu
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Annie Wang
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Cyril Hernandez
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Nicholas Siclari
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Divino Rajah
- Global Technology Center, Colgate-Palmolive Company, Piscataway, NJ 08855, USA
| | - Lewins Walter
- Global Technology Center, Colgate-Palmolive Company, Piscataway, NJ 08855, USA
| | - Salvatore A E Marras
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Daniel H Fine
- Department of Oral Biology, School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Carlo Amorin Daep
- Global Technology Center, Colgate-Palmolive Company, Piscataway, NJ 08855, USA
| | - Theresa L Chang
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
543
|
Perdiguero B, Marcos-Villar L, López-Bravo M, Sánchez-Cordón PJ, Zamora C, Valverde JR, Sorzano CÓS, Sin L, Álvarez E, Ramos M, Del Val M, Esteban M, Gómez CE. Immunogenicity and efficacy of a novel multi-patch SARS-CoV-2/COVID-19 vaccine candidate. Front Immunol 2023; 14:1160065. [PMID: 37404819 PMCID: PMC10316789 DOI: 10.3389/fimmu.2023.1160065] [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: 02/06/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction While there has been considerable progress in the development of vaccines against SARS-CoV-2, largely based on the S (spike) protein of the virus, less progress has been made with vaccines delivering different viral antigens with cross-reactive potential. Methods In an effort to develop an immunogen with the capacity to induce broad antigen presentation, we have designed a multi-patch synthetic candidate containing dominant and persistent B cell epitopes from conserved regions of SARS-CoV-2 structural proteins associated with long-term immunity, termed CoV2-BMEP. Here we describe the characterization, immunogenicity and efficacy of CoV2-BMEP using two delivery platforms: nucleic acid DNA and attenuated modified vaccinia virus Ankara (MVA). Results In cultured cells, both vectors produced a main protein of about 37 kDa as well as heterogeneous proteins with size ranging between 25-37 kDa. In C57BL/6 mice, both homologous and heterologous prime/boost combination of vectors induced the activation of SARS-CoV-2-specific CD4 and CD8 T cell responses, with a more balanced CD8+ T cell response detected in lungs. The homologous MVA/MVA immunization regimen elicited the highest specific CD8+ T cell responses in spleen and detectable binding antibodies (bAbs) to S and N antigens of SARS-CoV-2. In SARS-CoV-2 susceptible k18-hACE2 Tg mice, two doses of MVA-CoV2-BMEP elicited S- and N-specific bAbs as well as cross-neutralizing antibodies against different variants of concern (VoC). After SARS-CoV-2 challenge, all animals in the control unvaccinated group succumbed to the infection while vaccinated animals with high titers of neutralizing antibodies were fully protected against mortality, correlating with a reduction of virus infection in the lungs and inhibition of the cytokine storm. Discussion These findings revealed a novel immunogen with the capacity to control SARS-CoV-2 infection, using a broader antigen presentation mechanism than the approved vaccines based solely on the S antigen.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - María López-Bravo
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Veterinary Pathology Department, Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - José Ramón Valverde
- Scientific Computing, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit and Computational Genomics, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Laura Sin
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Enrique Álvarez
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Manuel Ramos
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Margarita Del Val
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
544
|
Caffry J, Selby M, Barr K, Morgan G, McGurk D, Scully P, Park C, Caridis AM, Southworth E, Morrison J, Clark DJ, Davies BMO, Eckersley NM, Groppelli E, Kirwan DE, Monahan I, Augustin Y, Toombs C, Planche T, Staines HM, Krishna S. The QuantuMDx Q-POC SARS-CoV-2 RT-PCR assay for rapid detection of COVID-19 at point-of-care: preliminary evaluation of a novel technology. Sci Rep 2023; 13:9827. [PMID: 37330592 PMCID: PMC10276817 DOI: 10.1038/s41598-023-35479-9] [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: 01/11/2023] [Accepted: 05/18/2023] [Indexed: 06/19/2023] Open
Abstract
Accurate and rapid point-of-care (PoC) diagnostics are critical to the control of the COVID-19 pandemic. The current standard for accurate diagnosis of SARS-CoV-2 is laboratory-based reverse transcription polymerase chain reaction (RT-PCR) assays. Here, a preliminary prospective performance evaluation of the QuantuMDx Q-POC SARS-CoV-2 RT-PCR assay is reported. Between November 2020 and March 2021, 49 longitudinal combined nose/throat (NT) swabs from 29 individuals hospitalised with RT-PCR confirmed COVID-19 were obtained at St George's Hospital, London. In addition, 101 mid-nasal (MN) swabs were obtained from healthy volunteers in June 2021. These samples were used to evaluate the Q-POC SARS-CoV-2 RT-PCR assay. The primary analysis was to compare the sensitivity and specificity of the Q-POC test against a reference laboratory-based RT-PCR assay. The overall sensitivity of the Q-POC test compared with the reference test was 96.88% (83.78- 99.92% CI) for a cycle threshold (Ct) cut-off value for the reference test of 35 and 80.00% (64.35-90.95% CI) without altering the reference test's Ct cut-off value of 40. The Q-POC test is a sensitive, specific and rapid PoC test for SARS-CoV-2 at a reference Ct cut-off value of 35. The Q-POC test provides an accurate option for RT-PCR at PoC without the need for sample pre-processing and laboratory handling, enabling rapid diagnosis and clinical triage in acute care and other settings.
Collapse
Affiliation(s)
- Jessica Caffry
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Matthew Selby
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Katie Barr
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - George Morgan
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - David McGurk
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Philip Scully
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Catherine Park
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | | | - Emily Southworth
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Jack Morrison
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - David J Clark
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Benedict M O Davies
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Nicholas M Eckersley
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Elisabetta Groppelli
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Daniela E Kirwan
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Irene Monahan
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Yolanda Augustin
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
| | - Colin Toombs
- QuantuMDx, Lugano Building, 57 Melbourne St, Newcastle Upon Tyne, UK
| | - Tim Planche
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Henry M Staines
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK.
| | - Sanjeev Krishna
- Clinical Academic Group in Institute for Infection and Immunity, St George's University of London, London, UK.
- St George's University Hospitals NHS Foundation Trust, London, UK.
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
| |
Collapse
|
545
|
Dorta-Gorrín A, Navas-Méndez J, Gozalo-Margüello M, Miralles L, García-Hevia L. Detection of SARS-CoV-2 Based on Nucleic Acid Amplification Tests (NAATs) and Its Integration into Nanomedicine and Microfluidic Devices as Point-of-Care Testing (POCT). Int J Mol Sci 2023; 24:10233. [PMID: 37373381 DOI: 10.3390/ijms241210233] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The coronavirus SARS-CoV-2 has highlighted the criticality of an accurate and rapid diagnosis in order to contain the spread of the virus. Knowledge of the viral structure and its genome is essential for diagnosis development. The virus is still quickly evolving and the global scenario could easily change. Thus, a greater range of diagnostic options is essential to face this threat to public health. In response to the global demand, there has been a rapid advancement in the understanding of current diagnostic methods. In fact, innovative approaches have emerged, leveraging the benefits of nanomedicine and microfluidic technologies. Although this development has been incredibly fast, several key areas require further investigation and optimization, such as sample collection and preparation, assay optimization and sensitivity, cost effectiveness, scalability device miniaturization, and portability and integration with smartphones. Addressing these gaps in the knowledge and these technological challenges will contribute to the development of reliable, sensitive, and user-friendly NAAT-based POCTs for the diagnosis of SARS-CoV-2 and other infectious diseases, facilitating rapid and effective patient management. This review aims to provide an overview of current SARS-CoV-2 detection methods based on nucleic acid detection tests (NAATs). Additionally, it explores promising approaches that combine nanomedicine and microfluidic devices with high sensitivity and relatively fast 'time to answer' for integration into point-of-care testing (POCT).
Collapse
Affiliation(s)
- Alexis Dorta-Gorrín
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria (UC), 39011 Santander, Spain
- Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
- Environmental Genetics Department, Ecohydros S.L., 39600 Maliaño, Spain
| | - Jesús Navas-Méndez
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria (UC), 39011 Santander, Spain
- Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Mónica Gozalo-Margüello
- Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
- Microbiology Service of University Hospital Marqués de Valdecilla (HUMV), 39008 Santander, Spain
- CIBER de Enfermedades Infecciosas-CIBERINFEC (CB21/13/00068), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Miralles
- Environmental Genetics Department, Ecohydros S.L., 39600 Maliaño, Spain
- Department of Functional Biology, Area of Genetics, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Lorena García-Hevia
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria (UC), 39011 Santander, Spain
- Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
546
|
Gerhards NM, Gonzales JL, Vreman S, Ravesloot L, van den Brand JMA, Doekes HP, Egberink HF, Stegeman A, Oreshkova N, van der Poel WHM, de Jong MCM. Efficient Direct and Limited Environmental Transmission of SARS-CoV-2 Lineage B.1.22 in Domestic Cats. Microbiol Spectr 2023; 11:e0255322. [PMID: 37222603 PMCID: PMC10269887 DOI: 10.1128/spectrum.02553-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 05/04/2023] [Indexed: 05/25/2023] Open
Abstract
The susceptibility of domestic cats to infection with SARS-CoV-2 has been demonstrated by several experimental studies and field observations. We performed an extensive study to further characterize the transmission of SARS-CoV-2 between cats, through both direct and indirect contact. To that end, we estimated the transmission rate parameter and the decay parameter for infectivity in the environment. Using four groups of pair-transmission experiment, all donor (inoculated) cats became infected, shed virus, and seroconverted, while three out of four direct contact cats got infected, shed virus, and two of those seroconverted. One out of eight cats exposed to a SARS-CoV-2-contaminated environment became infected but did not seroconvert. Statistical analysis of the transmission data gives a reproduction number R0 of 2.18 (95% CI = 0.92 to 4.08), a transmission rate parameter β of 0.23 day-1 (95% CI = 0.06 to 0.54), and a virus decay rate parameter μ of 2.73 day-1 (95% CI = 0.77 to 15.82). These data indicate that transmission between cats is efficient and can be sustained (R0 > 1), however, the infectiousness of a contaminated environment decays rapidly (mean duration of infectiousness 1/2.73 days). Despite this, infections of cats via exposure to a SARS-CoV-2-contaminated environment cannot be discounted if cats are exposed shortly after contamination. IMPORTANCE This article provides additional insight into the risk of infection that could arise from cats infected with SARS-CoV-2 by using epidemiological models to determine transmission parameters. Considering that transmission parameters are not always provided in the literature describing transmission experiments in animals, we demonstrate that mathematical analysis of experimental data is crucial to estimate the likelihood of transmission. This article is also relevant to animal health professionals and authorities involved in risk assessments for zoonotic spill-overs of SARS-CoV-2. Last but not least, the mathematical models to calculate transmission parameters are applicable to analyze the experimental transmission of other pathogens between animals.
Collapse
Affiliation(s)
- Nora M. Gerhards
- Department of Bioinformatics, Epidemiology and Animal Models, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | - Jose L. Gonzales
- Department of Bioinformatics, Epidemiology and Animal Models, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | - Sandra Vreman
- Department of Bacteriology, Host-Pathogen Interactions and Diagnostic Development, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | - Lars Ravesloot
- Department of Bacteriology, Host-Pathogen Interactions and Diagnostic Development, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | | | - Harmen P. Doekes
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, the Netherlands
| | - Herman F. Egberink
- Division Infectious Diseases and Immunology, Section Virology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Arjan Stegeman
- Department of Population Health Sciences, Veterinary Epidemiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Nadia Oreshkova
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | - Wim H. M. van der Poel
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, the Netherlands
| | - Mart C. M. de Jong
- Quantitative Veterinary Epidemiology, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
547
|
Fontenele RS, Yang Y, Driver EM, Magge A, Kraberger S, Custer JM, Dufault-Thompson K, Cox E, Newell ME, Varsani A, Halden RU, Scotch M, Jiang X. Wastewater surveillance uncovers regional diversity and dynamics of SARS-CoV-2 variants across nine states in the USA. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 877:162862. [PMID: 36933724 PMCID: PMC10017378 DOI: 10.1016/j.scitotenv.2023.162862] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 05/06/2023]
Abstract
Wastewater-based epidemiology (WBE) is a non-invasive and cost-effective approach for monitoring the spread of a pathogen within a community. WBE has been adopted as one of the methods to monitor the spread and population dynamics of the SARS-CoV-2 virus, but significant challenges remain in the bioinformatic analysis of WBE-derived data. Here, we have developed a new distance metric, CoVdist, and an associated analysis tool that facilitates the application of ordination analysis to WBE data and the identification of viral population changes based on nucleotide variants. We applied these new approaches to a large-scale dataset from 18 cities in nine states of the USA using wastewater collected from July 2021 to June 2022. We found that the trends in the shift between the Delta and Omicron SARS-CoV-2 lineages were largely consistent with what was seen in clinical data, but that wastewater analysis offered the added benefit of revealing significant differences in viral population dynamics at the state, city, and even neighborhood scales. We also were able to observe the early spread of variants of concern and the presence of recombinant lineages during the transitions between variants, both of which are challenging to analyze based on clinically-derived viral genomes. The methods outlined here will be beneficial for future applications of WBE to monitor SARS-CoV-2, particularly as clinical monitoring becomes less prevalent. Additionally, these approaches are generalizable, allowing them to be applied for the monitoring and analysis of future viral outbreaks.
Collapse
Affiliation(s)
- Rafaela S Fontenele
- National Library of Medicine, National Institute of Health, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Yiyan Yang
- National Library of Medicine, National Institute of Health, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Erin M Driver
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Arjun Magge
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Simona Kraberger
- The Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA
| | - Joy M Custer
- The Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA
| | - Keith Dufault-Thompson
- National Library of Medicine, National Institute of Health, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Erin Cox
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Melanie Engstrom Newell
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Arvind Varsani
- The Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA; School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; Center of Evolution and Medicine, Arizona State University, Tempe, AZ 85287, USA
| | - Rolf U Halden
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85281, USA; OneWaterOneHealth, Nonprofit Project of the Arizona State University Foundation, Tempe, AZ 85287, USA
| | - Matthew Scotch
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Xiaofang Jiang
- National Library of Medicine, National Institute of Health, 8600 Rockville Pike, Bethesda, MD 20894, USA.
| |
Collapse
|
548
|
Edimiris P, Doehmen C, Müller L, Andrée M, Baston-Buest DM, Buest S, Adams O, Krüssel JS, Bielfeld AP. Mild COVID-19 has no detrimental effect on semen quality. Basic Clin Androl 2023; 33:15. [PMID: 37316770 DOI: 10.1186/s12610-023-00190-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/06/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND As of today, the effect of coronavirus disease 2019 (COVID-19) on male fertility remains unclear. Studies published so far have partly contradictory results, likely due to very small sample sizes and heterogeneous populations. To gain a deeper understanding of the impact of COVID-19 on male fertility, we performed a prospective case-control study, in which we examined the ejaculate of 37 subjects, including 25 subjects in the acute phase of mild COVID-19 and 12 subjects who did not suffer from COVID-19. Determination of semen parameters, severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) qPCR, and infectivity analysis were performed in the acute phase of the disease and in series. RESULTS Semen parameter values did not differ significantly between subjects with mild COVID-19 and the control group. The serial examination of semen parameters revealed no significant changes between 4, 18, and 82 days after the onset of symptoms. SARS-CoV-2 RNA or infectious particles could not be detected in any ejaculate. CONCLUSION Mild COVID-19 seems to have no detrimental effect on semen parameter values.
Collapse
Affiliation(s)
- Philippos Edimiris
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Cornelius Doehmen
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
- Kinderwunschzentrum Niederrhein, Madrider Str. 6, 41069, Moenchengladbach, Germany
| | - Lisa Müller
- Institute of Virology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Marcel Andrée
- Institute of Virology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Dunja Maria Baston-Buest
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Sebastian Buest
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Ortwin Adams
- Institute of Virology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jan-Steffen Krüssel
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Alexandra Petra Bielfeld
- Department of OB/GYN and REI (UniKiD), Medical Center, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| |
Collapse
|
549
|
Thompson CR, Torres PM, Kontogianni K, Byrne RL, Noguera SV, Luna-Muschi A, Marchi AP, Andrade PS, dos Santos Barboza A, Nishikawara M, Body R, de Vos M, Escadafal C, Adams E, Figueiredo Costa S, Cubas-Atienzar AI. Multicenter Diagnostic Evaluation of OnSite COVID-19 Rapid Test (CTK Biotech) among Symptomatic Individuals in Brazil and the United Kingdom. Microbiol Spectr 2023; 11:e0504422. [PMID: 37212699 PMCID: PMC10269675 DOI: 10.1128/spectrum.05044-22] [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: 12/07/2022] [Accepted: 04/17/2023] [Indexed: 05/23/2023] Open
Abstract
The COVID-19 pandemic has given rise to numerous commercially available antigen rapid diagnostic tests (Ag-RDTs). To generate and to share accurate and independent data with the global community requires multisite prospective diagnostic evaluations of Ag-RDTs. This report describes the clinical evaluation of the OnSite COVID-19 rapid test (CTK Biotech, CA, USA) in Brazil and the United Kingdom. A total of 496 paired nasopharyngeal (NP) swabs were collected from symptomatic health care workers at Hospital das Clínicas in São Paulo, Brazil, and 211 NP swabs were collected from symptomatic participants at a COVID-19 drive-through testing site in Liverpool, United Kingdom. Swabs were analyzed by Ag-RDT, and results were compared to quantitative reverse transcriptase PCR (RT-qPCR). The clinical sensitivity of the OnSite COVID-19 rapid test in Brazil was 90.3% (95% confidence interval [CI], 75.1 to 96.7%) and in the United Kingdom was 75.3% (95% CI, 64.6 to 83.6%). The clinical specificity in Brazil was 99.4% (95% CI, 98.1 to 99.8%) and in the United Kingdom was 95.5% (95% CI, 90.6 to 97.9%). Concurrently, analytical evaluation of the Ag-RDT was assessed using direct culture supernatant of SARS-CoV-2 strains from wild-type (WT), Alpha, Delta, Gamma, and Omicron lineages. This study provides comparative performance of an Ag-RDT across two different settings, geographical areas, and populations. Overall, the OnSite Ag-RDT demonstrated a lower clinical sensitivity than claimed by the manufacturer. The sensitivity and specificity from the Brazil study fulfilled the performance criteria determined by the World Health Organization, but the performance obtained from the UK study failed to do. Further evaluation of Ag-RDTs should include harmonized protocols between laboratories to facilitate comparison between settings. IMPORTANCE Evaluating rapid diagnostic tests in diverse populations is essential to improving diagnostic responses as it gives an indication of the accuracy in real-world scenarios. In the case of rapid diagnostic testing within this pandemic, lateral flow tests that meet the minimum requirements for sensitivity and specificity can play a key role in increasing testing capacity, allowing timely clinical management of those infected, and protecting health care systems. This is particularly valuable in settings where access to the test gold standard is often restricted.
Collapse
Affiliation(s)
- Caitlin R. Thompson
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
| | - Pablo Muñoz Torres
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Konstantina Kontogianni
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
| | - Rachel L. Byrne
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
| | - LSTM Diagnostic group
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Department of Epidemiology, School of Public Health of University of São Paulo, São Paulo, Brazil
- Centro de atendimento ao colaborador, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Divisão de Laboratório Central, Hospital das Clinicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- FIND, Geneva, Switzerland
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Global Access Diagnostics, Thurleigh, Bedfordshire, United Kingdom
| | - Saidy Vásconez Noguera
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Alessandra Luna-Muschi
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Marchi
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Pâmela S. Andrade
- Department of Epidemiology, School of Public Health of University of São Paulo, São Paulo, Brazil
| | - Antonio dos Santos Barboza
- Centro de atendimento ao colaborador, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marli Nishikawara
- Centro de atendimento ao colaborador, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - CONDOR steering group
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Department of Epidemiology, School of Public Health of University of São Paulo, São Paulo, Brazil
- Centro de atendimento ao colaborador, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Divisão de Laboratório Central, Hospital das Clinicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- FIND, Geneva, Switzerland
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Global Access Diagnostics, Thurleigh, Bedfordshire, United Kingdom
| | - Richard Body
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | | | - Camille Escadafal
- Divisão de Laboratório Central, Hospital das Clinicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Emily Adams
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
- Global Access Diagnostics, Thurleigh, Bedfordshire, United Kingdom
| | - Silvia Figueiredo Costa
- LIM-49, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana I. Cubas-Atienzar
- Liverpool School of Tropical Medicine, Centre for Drugs and Diagnostics, Liverpool, United Kingdom
| |
Collapse
|
550
|
Ahti J, Österback R, Keskitalo A, Mokkala K, Vidbäck S, Veikkolainen V, Vuorinen T, Peltola V, Hakanen AJ, Waris M, Laine M. Diagnostic Performance and Tolerability of Saliva and Nasopharyngeal Swab Specimens in the Detection of SARS-CoV-2 by RT-PCR. Microbiol Spectr 2023; 11:e0532422. [PMID: 37093085 PMCID: PMC10269602 DOI: 10.1128/spectrum.05324-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
Saliva is a promising alternative for a nasopharyngeal swab (NPS) in specimen collection to detect SARS-CoV-2. We compared the diagnostic performance and tolerability of saliva collection versus NPS in a clinical setting. Paired NPS and saliva specimens were collected sequentially from participants (n = 250) at the Turku University Hospital drive-in coronavirus testing station in the spring of 2022, with Omicron BA.2 as the dominant SARS-CoV-2 variant. Discomfort and preference for the sampling method were assessed. The specimens were analyzed for SARS-CoV-2 using real-time multiplex reverse transcriptase PCR (RT-PCR) with a laboratory-developed test (LDT) and two commercial kits (PerkinElmer SARS-CoV-2 and PerkinElmer SARS-CoV-2 Plus) for several target genes. Among the 250 participants, 246 had respiratory symptoms. With LDT, SARS-CoV-2 was detected in 135 and 134 participants from NPS and saliva, respectively. Of the 250 specimens, 11 gave a discordant outcome, resulting in excellent agreement between the specimen types (Cohen's kappa coefficient of 0.911; P = 0.763). The cycle threshold (CT) values of LDT and commercial kit target genes were significantly lower from NPS than from saliva. A total of 172 (69%) participants assessed saliva sampling as more tolerable than NPS (P < 0.0001). Our findings present saliva as an applicable alternative for SARS-CoV-2 diagnostics. However, the lower CT values obtained from NPS indicate that NPS may be a slightly more sensitive specimen type. Participants preferred saliva sampling, although delivering an adequate volume of saliva was challenging for some participants. IMPORTANCE The extensive testing of SARS-CoV-2 is vital in controlling the spread of COVID-19. The reference standard for specimen collection is a nasopharyngeal swab (NPS). However, the discomfort of NPS sampling, the risk of nosocomial infections, and global material shortages have accelerated the development of alternative testing methods. Our study demonstrates that patients tolerate saliva sampling better than NPS. Of importance, although the RT-PCR qualitative test results seem to correspond between NPS and saliva, we show significantly lower CT values for NPS, compared to saliva, thus contradicting the suggested superiority of the saliva specimen over NPS in the detection of the Omicron variants of SARS-CoV-2. Future research is still required to enable individual planning for specimen collection and to determine the effects of different SARS-CoV-2 variants on the sensitivity of the saliva matrix.
Collapse
Affiliation(s)
- Jaakko Ahti
- Department of Paediatrics and Adolescent Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Riikka Österback
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anniina Keskitalo
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | | | - Tytti Vuorinen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Antti J. Hakanen
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Waris
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Miia Laine
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|