451
|
Girardi V, Gularte JS, Demoliner M, da Silva MS, Filippi M, de Abreu Góes Pereira VM, Hansen AW, Rosa RB, Fleck JD, Spilki FR. Reinfection by SARS-CoV-2 by divergent Omicron sublineages, 16 days apart. Braz J Microbiol 2023; 54:1847-1851. [PMID: 37269428 PMCID: PMC10239041 DOI: 10.1007/s42770-023-01018-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: 02/22/2023] [Accepted: 05/09/2023] [Indexed: 06/05/2023] Open
Abstract
Since the beginning of the SARS-CoV-2 pandemic, studies on the variants and sublineages stand out, mainly in the cases of reinfection in a short period. In this study, we describe a case of infection by BA.1.1 sublineage in an individual from Southern Brazil. The same patient acquired reinfection with sublineage BA.2 within 16 days after the first detection. The viral extraction and RT-qPCR were performed on the samples LMM72045 (collected in May 2022) and LMM72044 (collected in June 2022). After the confirmation of SARS-CoV-2 infection, we conducted the sequencing and viral genome analysis. This case of reinfection affected a 52-year-old male patient, without comorbidities, with three doses of vaccines against COVID-19, showing symptoms on May 19. These symptoms lasted for approximately six days. The patient returned to work activities on May 30. However, on June 4, the patient felt a new round of clinical signs that lasted for approximately seven days. Analysis of the viral genomes recovered from patients' clinical samples revealed that the two COVID-19 episodes were related to two divergent VOC Omicron sublineages, namely, BA.1.1 for the first round of symptoms and BA.2 for the second infection. Based on our findings, we can say that the present case of reinfection is the shortest described so far.
Collapse
Affiliation(s)
- Viviane Girardi
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil.
| | - Juliana Schons Gularte
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Meriane Demoliner
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Mariana Soares da Silva
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Micheli Filippi
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Vyctoria Malayhka de Abreu Góes Pereira
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Alana Witt Hansen
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Raquel Borba Rosa
- Secretaria Municipal de Saúde, Porto Alegre, Rio Grande Do Sul, Brazil
| | - Juliane Deise Fleck
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| | - Fernando Rosado Spilki
- Laboratório de Microbiologia Molecular, Universidade Feevale, Rodovia ERS-239, N° 2755, Prédio Vermelho, Piso 1, Sala 103, Vila Nova, CEP, Novo Hamburgo, RS, 93525-075, Brazil
| |
Collapse
|
452
|
Tsounidi D, Angelopoulou M, Petrou P, Raptis I, Kakabakos S. Simultaneous Detection of SARS-CoV-2 Nucleoprotein and Receptor Binding Domain by a Multi-Area Reflectance Spectroscopy Sensor. BIOSENSORS 2023; 13:865. [PMID: 37754099 PMCID: PMC10526254 DOI: 10.3390/bios13090865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
The COVID-19 pandemic has emphasized the urgent need for point-of-care methods suitable for the rapid and reliable diagnosis of viral infections. To address this demand, we report the rapid, label-free simultaneous determination of two SARS-CoV-2 proteins, namely, the nucleoprotein and the receptor binding domain peptide of S1 protein, by implementing a bioanalytical device based on Multi Area Reflectance Spectroscopy. Simultaneous detection of these two proteins is achieved by using silicon chips with adjacent areas of different silicon dioxide thickness on top, each of which is modified with an antibody specific to either the nucleoprotein or the receptor binding domain of SARS-CoV-2. Both areas were illuminated by a single probe that also collected the reflected light, directing it to a spectrometer. The online conversion of the combined reflection spectra from the two silicon dioxide areas into the respective adlayer thickness enabled real-time monitoring of immunoreactions taking place on the two areas. Several antibodies have been tested to define the pair, providing the higher specific signal following a non-competitive immunoassay format. Biotinylated secondary antibodies and streptavidin were used to enhance the specific signal. Both proteins were detected in less than 12 min, with detection limits of 1.0 ng/mL. The assays demonstrated high repeatability with intra- and inter-assay coefficients of variation lower than 10%. Moreover, the recovery of both proteins from spiked samples prepared in extraction buffer from a commercial self-test kit for SARS-CoV-2 collection from nasopharyngeal swabs ranged from 90.0 to 110%. The short assay duration in combination with the excellent analytical performance and the compact instrument size render the proposed device and assay suitable for point-of-care applications.
Collapse
Affiliation(s)
- Dimitra Tsounidi
- Immunoassays-Immunosensors Lab, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, 15341 Aghia Paraskevi, Greece; (D.T.); (M.A.); (P.P.)
| | - Michailia Angelopoulou
- Immunoassays-Immunosensors Lab, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, 15341 Aghia Paraskevi, Greece; (D.T.); (M.A.); (P.P.)
| | - Panagiota Petrou
- Immunoassays-Immunosensors Lab, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, 15341 Aghia Paraskevi, Greece; (D.T.); (M.A.); (P.P.)
| | - Ioannis Raptis
- Institute of Nanoscience and Nanotechnology, NCSR “Demokritos”, 15341 Aghia Paraskevi, Greece;
| | - Sotirios Kakabakos
- Immunoassays-Immunosensors Lab, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, 15341 Aghia Paraskevi, Greece; (D.T.); (M.A.); (P.P.)
| |
Collapse
|
453
|
Jain RK, Perumal N, Chaurasia D, Shrivastava R, Ahirwar KK, Sharma A, Kapoor G, Lalwani J. Performance Evaluation of Different RT-PCR Kits for the Direct Detection of SARS-CoV-2 in Preheated Specimens. J Lab Physicians 2023; 15:383-391. [PMID: 37564223 PMCID: PMC10411152 DOI: 10.1055/s-0043-1760752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has created high demand for molecular kits and consumables for mass screening of suspected individuals. Direct real-time polymerase chain reaction (RT-PCR) assay without nucleic acid extraction has several advantages in saving testing time and cost and helps in the rapid reporting of SARS-CoV-2. The present study evaluated the analytical performance of four SARS-CoV-2 RT-PCR for direct RT-PCR testing using preheated specimens. Methods A total of 100 clinical specimens were selected and divided into three different groups: (1) group I: 20 SARS-CoV-2 positive specimens with high viral load, viz., low Ct values (< 30 Ct), (2) group II: 50 SARS-CoV-2 positive specimens with low viral load, viz., high Ct values (> 30 Ct), and (3) group III: 30 SARS-CoV-2 negative specimens. Specimens were heat-inactivated at 70°C for 10 minutes and cooled down at 4°C and were evaluated for standard and direct RT-PCR method by using ViralDtect-II Multiplex Real-Time PCR kit, TaqPath COVID-19 Combo kit, COVIDsure Pro Multiplex RT-PCR kit, and Hi-PCR Coronavirus (COVID-19) Multiplex Probe PCR kit. Results Results showed that except ViralDtect-II kit, the other three TaqPath COVID-19 Combo kit, COVIDsure Pro kit, and Hi-PCR Coronavirus (COVID-19) RT-PCR kit were able to amplify all the SARS-CoV-2 genes in the direct RT-PCR method using preheated specimens. In group I specimens, 100% sensitivity was observed in all three RT-PCR kits. In group II specimens, COVIDsure Pro kit was found to be superior among other kits. Conclusion Direct RT-PCR method during pandemic situation is valuable and cost effective for the detection of SARS-CoV-2. All three TaqPath COVID-19 Combo kit, COVIDsure Pro kit, and Hi-PCR Coronavirus (COVID-19) RT-PCR kit can be used for direct RT-PCR method and COVIDsure Pro kit performance was found to be superior among all.
Collapse
Affiliation(s)
- Rajeev Kumar Jain
- State Virology Laboratory, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | - Nagaraj Perumal
- State Virology Laboratory, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | - Deepti Chaurasia
- Department of Microbiology, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | - Rakesh Shrivastava
- Department of Microbiology, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | | | - Archa Sharma
- Department of Microbiology, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | - Garima Kapoor
- Department of Microbiology, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| | - Jaya Lalwani
- Department of Microbiology, Gandhi Medical College, Bhopal, Madhya Pradesh, India
| |
Collapse
|
454
|
Mhlekude B, Postmus D, Stenzel S, Weiner J, Jansen J, Zapatero-Belinchón FJ, Olmer R, Richter A, Heinze J, Heinemann N, Mühlemann B, Schroeder S, Jones TC, Müller MA, Drosten C, Pich A, Thiel V, Martin U, Niemeyer D, Gerold G, Beule D, Goffinet C. Pharmacological inhibition of bromodomain and extra-terminal proteins induces an NRF-2-mediated antiviral state that is subverted by SARS-CoV-2 infection. PLoS Pathog 2023; 19:e1011657. [PMID: 37747932 PMCID: PMC10629670 DOI: 10.1371/journal.ppat.1011657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/07/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
Inhibitors of bromodomain and extra-terminal proteins (iBETs), including JQ-1, have been suggested as potential prophylactics against SARS-CoV-2 infection. However, molecular mechanisms underlying JQ-1-mediated antiviral activity and its susceptibility to viral subversion remain incompletely understood. Pretreatment of cells with iBETs inhibited infection by SARS-CoV-2 variants and SARS-CoV, but not MERS-CoV. The antiviral activity manifested itself by reduced reporter expression of recombinant viruses, and reduced viral RNA quantities and infectious titers in the culture supernatant. While we confirmed JQ-1-mediated downregulation of expression of angiotensin-converting enzyme 2 (ACE2) and interferon-stimulated genes (ISGs), multi-omics analysis addressing the chromatin accessibility, transcriptome and proteome uncovered induction of an antiviral nuclear factor erythroid 2-related factor 2 (NRF-2)-mediated cytoprotective response as an additional mechanism through which JQ-1 inhibits SARS-CoV-2 replication. Pharmacological inhibition of NRF-2, and knockdown of NRF-2 and its target genes reduced JQ-1-mediated inhibition of SARS-CoV-2 replication. Serial passaging of SARS-CoV-2 in the presence of JQ-1 resulted in predominance of ORF6-deficient variant, which exhibited resistance to JQ-1 and increased sensitivity to exogenously administered type I interferon (IFN-I), suggesting a minimised need for SARS-CoV-2 ORF6-mediated repression of IFN signalling in the presence of JQ-1. Importantly, JQ-1 exhibited a transient antiviral activity when administered prophylactically in human airway bronchial epithelial cells (hBAECs), which was gradually subverted by SARS-CoV-2, and no antiviral activity when administered therapeutically following an established infection. We propose that JQ-1 exerts pleiotropic effects that collectively induce an antiviral state in the host, which is ultimately nullified by SARS-CoV-2 infection, raising questions about the clinical suitability of the iBETs in the context of COVID-19.
Collapse
Affiliation(s)
- Baxolele Mhlekude
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Virology and Innate Immunity Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Saskia Stenzel
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - January Weiner
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Francisco J. Zapatero-Belinchón
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Clinical Microbiology, Virology & Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH—Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Anja Richter
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julian Heinze
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicolas Heinemann
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Mühlemann
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Schroeder
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Terry C. Jones
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Marcel A. Müller
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Core Facility Proteomics, Hannover, Germany
| | - Volker Thiel
- Institute of Virology and Immunology (IVI), University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH—Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gisa Gerold
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Clinical Microbiology, Virology & Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Dieter Beule
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool United Kingdom
| |
Collapse
|
455
|
Himsworth CG, Caleta JM, Coombe M, McGregor G, Dibernardo A, Lindsay R, Sekirov I, Prystajecky N. A comparison of sampling and testing approaches for the surveillance of SARS-CoV-2 in farmed American mink. J Vet Diagn Invest 2023; 35:528-534. [PMID: 37366157 PMCID: PMC10300625 DOI: 10.1177/10406387231183685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Surveillance for SARS-CoV-2 in American mink (Neovison vison) is a global priority because outbreaks on mink farms have potential consequences for animal and public health. Surveillance programs often focus on screening natural mortalities; however, significant knowledge gaps remain regarding sampling and testing approaches. Using 76 mink from 3 naturally infected farms in British Columbia, Canada, we compared the performance of 2 reverse-transcription real-time PCR (RT-rtPCR) targets (the envelope [E] and RNA-dependent RNA polymerase [RdRp] genes) as well as serology. We also compared RT-rtPCR and sequencing results from nasopharyngeal, oropharyngeal, skin, and rectal swabs, as well as nasopharyngeal samples collected using swabs and interdental brushes. We found that infected mink were generally RT-rtPCR-positive on all samples; however, Ct values differed significantly among sample types (nasopharyngeal < oropharyngeal < skin < rectal). There was no difference in the results of nasopharyngeal samples collected using swabs or interdental brushes. For most mink (89.4%), qualitative (i.e., positive vs. negative) serology and RT-rtPCR results were concordant. However, mink were positive on RT-rtPCR and negative on serology and vice versa, and there was no significant correlation between Ct values on RT-rtPCR and percent inhibition on serology. Both the E and RdRp targets were detectable in all sample types, albeit with a small difference in Ct values. Although SARS-CoV-2 RNA can be detected in multiple sample types, passive surveillance programs in mink should focus on multiple target RT-rtPCR testing of nasopharyngeal samples in combination with serology.
Collapse
Affiliation(s)
- Chelsea G Himsworth
- Animal Health Centre, British Columbia Ministry of Agriculture, Abbotsford, British Columbia, Canada
| | - Jessica M Caleta
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Michelle Coombe
- Animal Health Centre, British Columbia Ministry of Agriculture, Abbotsford, British Columbia, Canada
| | - Glenna McGregor
- Animal Health Centre, British Columbia Ministry of Agriculture, Abbotsford, British Columbia, Canada
| | - Antonia Dibernardo
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Robbin Lindsay
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Inna Sekirov
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalie Prystajecky
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
456
|
Scutari R, Renica S, Cento V, Nava A, Sammartino JC, Ferrari A, Pani A, Merli M, Fanti D, Vismara C, Scaglione F, Puoti M, Bandera A, Gori A, Piralla A, Baldanti F, Perno CF, Alteri C. Quantitative SARS-CoV-2 subgenomic RNA as a surrogate marker for viral infectivity: Comparison between culture isolation and direct sgRNA quantification. PLoS One 2023; 18:e0291120. [PMID: 37656746 PMCID: PMC10473502 DOI: 10.1371/journal.pone.0291120] [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: 09/07/2022] [Accepted: 08/22/2023] [Indexed: 09/03/2023] Open
Abstract
Detection of subgenomic (sg) SARS-CoV-2 RNAs are frequently used as a correlate of viral infectiousness, but few data about correlation between sg load and viable virus are available. Here, we defined concordance between culture isolation and E and N sgRNA quantification by ddPCR assays in 51 nasopharyngeal swabs collected from SARS-CoV-2 positive hospitalized patients. Among the 51 samples, 14 were SARS-CoV-2 culture-positive and 37 were negative. According to culture results, the sensitivity and specificity of E and N sgRNA assays were 100% and 100%, and 84% and 86%, respectively. ROC analysis showed that the best E and N cut-offs to predict positive culture isolation were 32 and 161 copies/mL respectively, with an AUC (95% CI) of 0.96 (0.91-1.00) and 0.96 (0.92-1.00), and a diagnostic accuracy of 88% and 92%, respectively. Even if no significant correlations were observed between sgRNA amount and clinical presentation, a higher number of moderate/severe cases and lower number of days from symptoms onset characterized patients with sgRNA equal to or higher than sgRNA cut-offs. Overall, this study suggests that SARS-CoV-2 sgRNA quantification could be helpful to estimate the replicative activity of SARS-CoV-2 and can represent a valid surrogate marker to efficiently recognize patients with active infection. The inclusion of this assay in available SARS-CoV-2 diagnostics procedure might help in optimizing fragile patients monitoring and management.
Collapse
Affiliation(s)
- Rossana Scutari
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Multimodal Research Area, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Silvia Renica
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Valeria Cento
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCSS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alice Nava
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Alessandro Ferrari
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arianna Pani
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marco Merli
- Infectious Diseases Unit, Azienda Socio-Sanitaria Territoriale (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Diana Fanti
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Vismara
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Massimo Puoti
- Infectious Diseases Unit, Azienda Socio-Sanitaria Territoriale (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Antonio Piralla
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | | | - Claudia Alteri
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Multimodal Research Area, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| |
Collapse
|
457
|
Cui X, Ngang S, Liu DD, Cheow LF. Rapid Single-Round Pool Testing of Infectious Disease Enabled by Multicolor Digital Melting PCR. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205636. [PMID: 37209020 DOI: 10.1002/smll.202205636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/27/2023] [Indexed: 05/21/2023]
Abstract
Pooled nucleic acid amplification test is a promising strategy to reduce cost and resources for screening large populations for infectious disease. However, the benefit of pooled testing is reversed when disease prevalence is high, because of the need to retest each sample to identify infected individual when a pool is positive. Split, Amplify, and Melt analysis of Pooled Assay (SAMPA) is presented, a multicolor digital melting PCR assay in nanoliter chambers that simultaneously identify infected individuals and quantify their viral loads in a single round of pooled testing. This is achieved by early sample tagging with unique barcodes and pooling, followed by single molecule barcode identification in a digital PCR platform using a highly multiplexed melt curve analysis strategy. The feasibility is demonstrated of SAMPA for quantitative unmixing and variant identification from pools of eight synthetic DNA and RNA samples corresponding to the N1 gene, as well as from heat-inactivated SARS-CoV-2 virus. Single round pooled testing of barcoded samples with SAMPA can be a valuable tool for rapid and scalable population testing of infectious disease.
Collapse
Affiliation(s)
- Xu Cui
- Department of Biomedical Engineering & Institute for Health Innovation and Technology, National University of Singapore, Singapore, 119077, Singapore
| | - Shaun Ngang
- Department of Biomedical Engineering & Institute for Health Innovation and Technology, National University of Singapore, Singapore, 119077, Singapore
| | - Dong Dong Liu
- Department of Biomedical Engineering & Institute for Health Innovation and Technology, National University of Singapore, Singapore, 119077, Singapore
| | - Lih Feng Cheow
- Department of Biomedical Engineering & Institute for Health Innovation and Technology, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
458
|
Subudhi PD, Rooge S, Bihari C, Thangariyal S, Goswami S, Agarwal R, Kaur S, Gupta E, Baweja S. Prolonged existence of SARS-CoV-2 RNAs in the extracellular vesicles of respiratory specimens from patients with negative reverse transcription-polymerase chain reaction. LIVER RESEARCH 2023; 7:228-236. [PMID: 39958378 PMCID: PMC11791904 DOI: 10.1016/j.livres.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/23/2023] [Accepted: 09/08/2023] [Indexed: 02/18/2025]
Abstract
Background and aim Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is primarily in the respiratory tract, particularly in patients with underlying comorbidities. This study aimed to investigate the presence of the virus inside the extracellular vesicles (EVs) in patients with and without chronic liver disease (CLD). Methods Eighty patients with positive SARS-CoV-2, including twenty-four patients with CLD and fifty-six patients without CLD, and five healthy controls with negative SARS-CoV-2 were enrolled. Nasal swab specimens were tested for the detection of SARS-CoV-2 using reverse transcription-polymerase chain reaction (RT-PCR). Patients with coronavirus disease 2019 (COVID-19) were followed up on days 7 and 14. Nasal swab, collected in viral transport media (VTM), and plasma samples were investigated at each time point. EVs were isolated from the nasal swabs (collected in VTM) and plasma using differential ultracentrifugation and estimated at each time point. The transmission or replication by the EVs was assessed in Vero E6 cells. Results In patients with baseline RT-PCR positive, SARS-CoV-2 RNAs inside the EVs were found in 68/80 (85%) patients with higher viral load in the nasal swabs than in the EVs (cycle threshold (Ct) value, 23.4 ± 5.7 vs. 30.3 ± 5.0, P < 0.001). On follow-up at day 7, of the 32 patients negative for COVID-19, 15 (46.9%) had virus persistence in the EVs (Ct value, 30.7 ± 2.7), and on day 14, of the 56 patients with negative SARS-CoV-2, 16 patients (28.6%) had positive SARS-CoV-2 RNAs in the EVs (Ct value, 31.4 ± 3.0). The mean viral load decreased on days 7 and 14 compared to baseline in the nasal swabs (P < 0.001) but not in the EVs. Additionally, SARS-CoV-2 RNAs were undetectable in the plasma, but 12.5% of patients were positive in the plasma EVs. Significantly prolonged and high viral load was found in the EVs on day 14 in COVID-19 patients combined with CLD compared with COVID-19 patients (P = 0.0004). We found significant higher levels of EV-associated with endothelial cells and hepatocytes in the COVID-19 + CLD group than COVID-19 group (P = 0.032 and P = 0.002, respectively), suggesting more endothelial cells and hepatocytes cellular injury in liver disease patients with COVID-19. Interestingly, we also found EVs could transmit SARS-CoV-2 RNAs into Vero E6 cells at 24 h post-infection. Conclusions The identification of SARS-CoV-2 RNAs in the EVs in patients with negative RT-PCR indicates the persistence of infection and likely recurrence of the infection. It is suggestive of another route of transmission as EVs harbor SARS-CoV-2 RNAs. EV-associated RNAs may determine the ongoing inflammation and clinical course of subjects with undetectable SARS-CoV-2 virus and this may have relevance to better management of patients with CLD.
Collapse
Affiliation(s)
- P. Debishree Subudhi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sheetalnath Rooge
- Department of Virology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Chhagan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Swati Thangariyal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sivang Goswami
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Reshu Agarwal
- Department of Virology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Ekta Gupta
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sukriti Baweja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
459
|
Rehan S, Tranter D, Sharp PP, Craven GB, Lowe E, Anderl JL, Muchamuel T, Abrishami V, Kuivanen S, Wenzell NA, Jennings A, Kalyanaraman C, Strandin T, Javanainen M, Vapalahti O, Jacobson MP, McMinn D, Kirk CJ, Huiskonen JT, Taunton J, Paavilainen VO. Signal peptide mimicry primes Sec61 for client-selective inhibition. Nat Chem Biol 2023; 19:1054-1062. [PMID: 37169961 PMCID: PMC10449633 DOI: 10.1038/s41589-023-01326-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/06/2023] [Indexed: 05/13/2023]
Abstract
Preventing the biogenesis of disease-relevant proteins is an attractive therapeutic strategy, but attempts to target essential protein biogenesis factors have been hampered by excessive toxicity. Here we describe KZR-8445, a cyclic depsipeptide that targets the Sec61 translocon and selectively disrupts secretory and membrane protein biogenesis in a signal peptide-dependent manner. KZR-8445 potently inhibits the secretion of pro-inflammatory cytokines in primary immune cells and is highly efficacious in a mouse model of rheumatoid arthritis. A cryogenic electron microscopy structure reveals that KZR-8445 occupies the fully opened Se61 lateral gate and blocks access to the lumenal plug domain. KZR-8445 binding stabilizes the lateral gate helices in a manner that traps select signal peptides in the Sec61 channel and prevents their movement into the lipid bilayer. Our results establish a framework for the structure-guided discovery of novel therapeutics that selectively modulate Sec61-mediated protein biogenesis.
Collapse
Affiliation(s)
- Shahid Rehan
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Dale Tranter
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Phillip P Sharp
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Gregory B Craven
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Eric Lowe
- Kezar Life Sciences, South San Francisco, CA, USA
| | | | | | - Vahid Abrishami
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Suvi Kuivanen
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicole A Wenzell
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | | | - Chakrapani Kalyanaraman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of California, San Francisco, CA, USA
| | - Tomas Strandin
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Javanainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of California, San Francisco, CA, USA
| | | | | | - Juha T Huiskonen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| | - Ville O Paavilainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
460
|
Qin X, Paul R, Zhou Y, Wu Y, Cheng X, Liu Y. Multiplex solid-phase RPA coupled CRISPR-based visual detection of SARS-CoV-2. BIOSENSORS & BIOELECTRONICS: X 2023; 14:100381. [PMID: 38293281 PMCID: PMC10827331 DOI: 10.1016/j.biosx.2023.100381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The COVID-19 pandemic has presented a significant challenge to the world's public health and led to over 6.9 million deaths reported to date. A rapid, sensitive, and cost-effective point-of-care virus detection device is essential for the control and surveillance of the contagious severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic. The study presented here aimed to demonstrate a solid-phase isothermal recombinase polymerase amplification coupled CRISPR-based (spRPA-CRISPR) assay for on-chip multiplexed, sensitive and visual COVID-19 DNA detection. The assay targets the SARS-CoV-2 structure protein encoded genomes and can simultaneously detect two specific genes without cross-interaction. The amplified target sequences were immobilized on the one-pot device surface and detected using the mixed Cas12a-crRNA collateral cleavage of reporter-released fluorescent signal when specific genes were recognized. The endpoint signal can be directly visualized for rapid detection of COVID-19. The system was tested with samples of a broad range of concentrations (20 to 2 × 104 copies) and showed analytical sensitivity down to 20 copies per microliter. Furthermore, a low-cost blue LED flashlight (~$12) was used to provide a visible SARS-CoV-2 detection signal of the spRPA-CRISPR assay which could be purchased online easily. Thus, our platform provides a sensitive and easy-to-read multiplexed gene detection method that can specifically identify low concentration genes.
Collapse
Affiliation(s)
- Xiaochen Qin
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Ratul Paul
- Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, PA, 18015, USA
| | - Yuyuan Zhou
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Yue Wu
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Xuanhong Cheng
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Yaling Liu
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
- Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, PA, 18015, USA
| |
Collapse
|
461
|
McBride DS, Garushyants SK, Franks J, Magee AF, Overend SH, Huey D, Williams AM, Faith SA, Kandeil A, Trifkovic S, Miller L, Jeevan T, Patel A, Nolting JM, Tonkovich MJ, Genders JT, Montoney AJ, Kasnyik K, Linder TJ, Bevins SN, Lenoch JB, Chandler JC, DeLiberto TJ, Koonin EV, Suchard MA, Lemey P, Webby RJ, Nelson MI, Bowman AS. Accelerated evolution of SARS-CoV-2 in free-ranging white-tailed deer. Nat Commun 2023; 14:5105. [PMID: 37640694 PMCID: PMC10462754 DOI: 10.1038/s41467-023-40706-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
The zoonotic origin of the COVID-19 pandemic virus highlights the need to fill the vast gaps in our knowledge of SARS-CoV-2 ecology and evolution in non-human hosts. Here, we detected that SARS-CoV-2 was introduced from humans into white-tailed deer more than 30 times in Ohio, USA during November 2021-March 2022. Subsequently, deer-to-deer transmission persisted for 2-8 months, disseminating across hundreds of kilometers. Newly developed Bayesian phylogenetic methods quantified how SARS-CoV-2 evolution is not only three-times faster in white-tailed deer compared to the rate observed in humans but also driven by different mutational biases and selection pressures. The long-term effect of this accelerated evolutionary rate remains to be seen as no critical phenotypic changes were observed in our animal models using white-tailed deer origin viruses. Still, SARS-CoV-2 has transmitted in white-tailed deer populations for a relatively short duration, and the risk of future changes may have serious consequences for humans and livestock.
Collapse
Affiliation(s)
- Dillon S McBride
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Sofya K Garushyants
- Division of Intramural Research, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - John Franks
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrew F Magee
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Steven H Overend
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Devra Huey
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Amanda M Williams
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| | - Seth A Faith
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| | - Ahmed Kandeil
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Sanja Trifkovic
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lance Miller
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Trushar Jeevan
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jacqueline M Nolting
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | | | - J Tyler Genders
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Columbus, OH, USA
| | - Andrew J Montoney
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Columbus, OH, USA
| | - Kevin Kasnyik
- Columbus and Franklin County Metro Parks, Westerville, OH, USA
| | - Timothy J Linder
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Disease Program, Fort Collins, CO, USA
| | - Sarah N Bevins
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Disease Program, Fort Collins, CO, USA
| | - Julianna B Lenoch
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Disease Program, Fort Collins, CO, USA
| | - Jeffrey C Chandler
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Wildlife Disease Diagnostic Laboratory, Fort Collins, CO, USA
| | - Thomas J DeLiberto
- U.S. Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Fort Collins, CO, USA
| | - Eugene V Koonin
- Division of Intramural Research, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Marc A Suchard
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biomathematics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Philippe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Richard J Webby
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Martha I Nelson
- Division of Intramural Research, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA.
| | - Andrew S Bowman
- Department of Veterinary Preventive Medicine, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA.
| |
Collapse
|
462
|
Zarouri A, Barnes AMT, Aboubakr H, Novi VT, Dong Q, Nelson A, Goyal S, Abbas A. A High-Performance Polymer Composite Column for Coronavirus Nucleic Acid Purification. RESEARCH SQUARE 2023:rs.3.rs-3261727. [PMID: 37674719 PMCID: PMC10479450 DOI: 10.21203/rs.3.rs-3261727/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Here, we report the development of a novel polymer composite (PC) purification column and kit. The performance of the PC columns was compared to conventional silica gel (SG) columns for the purification of nucleic acids from coronaviruses, including SARS-CoV-2, in 82 clinical samples. The results shows that PC-based purification outperforms silica gel (SG)-based purification by enabling a higher sensitivity (94%), accuracy (97%), and by eliminating false positives (100% selectivity). The high selectivity is critical for efficient patient triage and resource management during pandemics. Furthermore, PC-based purification exhibits three times higher analytical precision than a commonly used SG-based nucleic acid purification thereby enabling a more accurate quantification of viral loads and higher reproducibility.
Collapse
|
463
|
Clancy CS, Meade-White K, Shaia C, Saturday G, Feldmann H, Rosenke K. Histopathologic Characterization of Experimental Peracute SARS-CoV-2 Infection in the Syrian Hamster. Vet Sci 2023; 10:536. [PMID: 37756057 PMCID: PMC10536131 DOI: 10.3390/vetsci10090536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
Coronavirus Infectious Disease 2019 (COVID-19) initiated a global pandemic that thus far has resulted in the death of over 6.5 million people internationally. Understanding the viral tropism during the initial, subclinical phase of infection is critical to develop targeted vaccines and therapeutics. With the continued emergence of variants of concern, particularly those that appear to have a tropism for the upper respiratory tract, understanding the complete pathogenesis is critical to develop more effective interventions. Thus far, the Syrian hamster has served as the most consistent small animal model of SARS-CoV-2 infection for mild to moderate respiratory disease. Herein, we utilize histopathology and immunohistochemistry to characterize the peracute phase of disease initiating at 6-h-post-inoculation in the intranasal inoculation route Syrian hamster model. Inflammation and viral replication initiates in the respiratory epithelium of nasal turbinates as early as 12-h-post-inoculation and moves caudally through the nasal cavity by 36-h-post inoculation. Lower respiratory involvement can be detected as early as 12-h-post inoculation in the intranasal inoculated hamster model. These data highlight the importance of rostral nasal cavity sampling at early timepoints for detection of SARS-CoV-2 in the Syrian hamster model.
Collapse
Affiliation(s)
- Chad S Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kyle Rosenke
- Laboratory of Virology, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT 59840, USA
| |
Collapse
|
464
|
Alpuche-Lazcano SP, Stuible M, Akache B, Tran A, Kelly J, Hrapovic S, Robotham A, Haqqani A, Star A, Renner TM, Blouin J, Maltais JS, Cass B, Cui K, Cho JY, Wang X, Zoubchenok D, Dudani R, Duque D, McCluskie MJ, Durocher Y. Preclinical evaluation of manufacturable SARS-CoV-2 spike virus-like particles produced in Chinese Hamster Ovary cells. COMMUNICATIONS MEDICINE 2023; 3:116. [PMID: 37612423 PMCID: PMC10447459 DOI: 10.1038/s43856-023-00340-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND As the COVID-19 pandemic continues to evolve, novel vaccines need to be developed that are readily manufacturable and provide clinical efficacy against emerging SARS-CoV-2 variants. Virus-like particles (VLPs) presenting the spike antigen at their surface offer remarkable benefits over other vaccine antigen formats; however, current SARS-CoV-2 VLP vaccines candidates in clinical development suffer from challenges including low volumetric productivity, poor spike antigen density, expression platform-driven divergent protein glycosylation and complex upstream/downstream processing requirements. Despite their extensive use for therapeutic protein manufacturing and proven ability to produce enveloped VLPs, Chinese Hamster Ovary (CHO) cells are rarely used for the commercial production of VLP-based vaccines. METHODS Using CHO cells, we aimed to produce VLPs displaying the full-length SARS-CoV-2 spike. Affinity chromatography was used to capture VLPs released in the culture medium from engineered CHO cells expressing spike. The structure, protein content, and glycosylation of spikes in VLPs were characterized by several biochemical and biophysical methods. In vivo, the generation of neutralizing antibodies and protection against SARS-CoV-2 infection was tested in mouse and hamster models. RESULTS We demonstrate that spike overexpression in CHO cells is sufficient by itself to generate high VLP titers. These VLPs are evocative of the native virus but with at least three-fold higher spike density. In vivo, purified VLPs elicit strong humoral and cellular immunity at nanogram dose levels which grant protection against SARS-CoV-2 infection. CONCLUSIONS Our results show that CHO cells are amenable to efficient manufacturing of high titers of a potently immunogenic spike protein-based VLP vaccine antigen.
Collapse
Affiliation(s)
- Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Bassel Akache
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Anh Tran
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - John Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resources Development Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Arsalan Haqqani
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Alexandra Star
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Dr, Ottawa, ON, K1A 0R6, Canada
| | - Tyler M Renner
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Julie Blouin
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Jean-Sébastien Maltais
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Brian Cass
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Kai Cui
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Jae-Young Cho
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Xinyu Wang
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB, T6G 2M9, Canada
| | - Daria Zoubchenok
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Renu Dudani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Diana Duque
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada.
| |
Collapse
|
465
|
Lombardi A, Voli A, Mancusi A, Girardi S, Proroga YTR, Pierri B, Olivares R, Cossentino L, Suffredini E, La Rosa G, Fusco G, Pizzolante A, Porta A, Campiglia P, Torre I, Pennino F, Tosco A. SARS-CoV-2 RNA in Wastewater and Bivalve Mollusk Samples of Campania, Southern Italy. Viruses 2023; 15:1777. [PMID: 37632119 PMCID: PMC10459311 DOI: 10.3390/v15081777] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
SARS-CoV-2 can be detected in the feces of infected people, consequently in wastewater, and in bivalve mollusks, that are able to accumulate viruses due to their ability to filter large amounts of water. This study aimed to monitor SARS-CoV-2 RNA presence in 168 raw wastewater samples collected from six wastewater treatment plants (WWTPs) and 57 mollusk samples obtained from eight harvesting sites in Campania, Italy. The monitoring period spanned from October 2021 to April 2022, and the results were compared and correlated with the epidemiological situation. In sewage, the ORF1b region of SARS-CoV-2 was detected using RT-qPCR, while in mollusks, three targets-RdRp, ORF1b, and E-were identified via RT-dPCR. Results showed a 92.3% rate of positive wastewater samples with increased genomic copies (g.c.)/(day*inhabitant) in December-January and March-April 2022. In the entire observation period, 54.4% of mollusks tested positive for at least one SARS-CoV-2 target, and the rate of positive samples showed a trend similar to that of the wastewater samples. The lower SARS-CoV-2 positivity rate in bivalve mollusks compared to sewages is a direct consequence of the seawater dilution effect. Our data confirm that both sample types can be used as sentinels to detect SARS-CoV-2 in the environment and suggest their potential use in obtaining complementary information on SARS-CoV-2.
Collapse
Affiliation(s)
- Annalisa Lombardi
- Department of Public Health, University “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (A.L.)
| | - Antonia Voli
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (A.V.); (A.P.); (P.C.)
| | - Andrea Mancusi
- Department of Food Security Coordination, Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (A.M.); (S.G.); (Y.T.R.P.); (B.P.)
| | - Santa Girardi
- Department of Food Security Coordination, Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (A.M.); (S.G.); (Y.T.R.P.); (B.P.)
| | - Yolande Thérèse Rose Proroga
- Department of Food Security Coordination, Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (A.M.); (S.G.); (Y.T.R.P.); (B.P.)
| | - Biancamaria Pierri
- Department of Food Security Coordination, Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (A.M.); (S.G.); (Y.T.R.P.); (B.P.)
| | - Renato Olivares
- Campania Regional Environmental Protection Agency (ARPAC), Via Vicinale Santa Maria del Pianto, 80143 Naples, Italy; (R.O.); (L.C.)
| | - Luigi Cossentino
- Campania Regional Environmental Protection Agency (ARPAC), Via Vicinale Santa Maria del Pianto, 80143 Naples, Italy; (R.O.); (L.C.)
| | - Elisabetta Suffredini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Giuseppina La Rosa
- Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Giovanna Fusco
- Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (G.F.); (A.P.)
| | - Antonio Pizzolante
- Zooprophylactic Institute of Southern Italy, Via Salute 2, 80055 Portici, Italy; (G.F.); (A.P.)
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (A.V.); (A.P.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (A.V.); (A.P.); (P.C.)
| | - Ida Torre
- Department of Public Health, University “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (A.L.)
| | - Francesca Pennino
- Department of Public Health, University “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (A.L.)
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (A.V.); (A.P.); (P.C.)
| |
Collapse
|
466
|
Breulmann M, Kallies R, Bernhard K, Gasch A, Müller RA, Harms H, Chatzinotas A, van Afferden M. A long-term passive sampling approach for wastewater-based monitoring of SARS-CoV-2 in Leipzig, Germany. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 887:164143. [PMID: 37182773 PMCID: PMC10181866 DOI: 10.1016/j.scitotenv.2023.164143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/16/2023]
Abstract
Wastewater-based monitoring of SARS-CoV-2 has become a promising and useful tool in tracking the potential spread or dynamics of the virus. Its recording can be used to predict how the potential number of infections in a population will develop. Recent studies have shown that the use of passive samplers is also suitable for the detection of SARS-CoV-2 genome copies (GC) in wastewater. They can be used at any site, provide timely data and may collect SARS-CoV-2 GC missed by traditional sampling methods. Therefore, the aim of this study was to evaluate the suitability of passive samplers for the detection of SARS-CoV-2 GC in wastewater in the long-term at two different scales. Polyethylene-based plastic passive samplers were deployed at the city-scale level of Leipzig at 13 different locations, with samples being taken from March 2021 to August 2022. At the smaller city district level, three types of passive samplers (cotton-cloth, unravelled polypropylene plastic rope and polyethylene-based plastic strips) were used and sampled on a weekly basis from March to August 2022. The results are discussed in relation to wastewater samples taken at the individual passive sampling point. Our results show that passive samplers can indicate at a city-scale level an accurate level of positive infections in the population (positive-rate: 86 %). On a small-scale level, the use of passive samplers was also feasible and effective to detect SARS-CoV-2 GC easily and cost-effectively, mirroring a similar trend to that at a city-scale level. Thus, this study demonstrated that passive samplers provide reproducible SARS-CoV-2 GC signals from wastewater and a time-integrated measurement of the sampled matrix with greater sensitivity compared to wastewater. We thus recommend the use of passive samplers as an alternative method for wastewater-based epidemiology. Passive samplers can in particular be considered for a better estimation of infections compared to incidence levels.
Collapse
Affiliation(s)
- Marc Breulmann
- Centre for Environmental Biotechnology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany.
| | - René Kallies
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Katy Bernhard
- Centre for Environmental Biotechnology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Andrea Gasch
- Wastewater Monitoring Department, Kommunale Wasserwerke Leipzig GmbH, Johannisgasse 7-9, 04103 Leipzig, Germany
| | - Roland Arno Müller
- Centre for Environmental Biotechnology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Hauke Harms
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Antonis Chatzinotas
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany; Institute of Biology, Leipzig University, 04103 Leipzig, Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103 Leipzig, Germany
| | - Manfred van Afferden
- Centre for Environmental Biotechnology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| |
Collapse
|
467
|
Galperine T, Choi Y, Pagani JL, Kritikos A, Papadimitriou-Olivgeris M, Méan M, Scherz V, Opota O, Greub G, Guery B, Bertelli C. Temporal changes in fecal microbiota of patients infected with COVID-19: a longitudinal cohort. BMC Infect Dis 2023; 23:537. [PMID: 37596518 PMCID: PMC10436399 DOI: 10.1186/s12879-023-08511-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a multifaceted disease potentially responsible for various clinical manifestations including gastro-intestinal symptoms. Several evidences suggest that the intestine is a critical site of immune cell development, gut microbiota could therefore play a key role in lung immune response. We designed a monocentric longitudinal observational study to describe the gut microbiota profile in COVID-19 patients and compare it to a pre-existing cohort of ventilated non-COVID-19 patients. METHODS From March to December 2020, we included patients admitted for COVID-19 in medicine (43 not ventilated) or intensive care unit (ICU) (14 ventilated) with a positive SARS-CoV-2 RT-PCR assay in a respiratory tract sample. 16S metagenomics was performed on rectal swabs from these 57 COVID-19 patients, 35 with one and 22 with multiple stool collections. Nineteen non-COVID-19 ICU controls were also enrolled, among which 14 developed ventilator-associated pneumonia (pneumonia group) and five remained without infection (control group). SARS-CoV-2 viral loads in fecal samples were measured by qPCR. RESULTS Although similar at inclusion, Shannon alpha diversity appeared significantly lower in COVID-19 and pneumonia groups than in the control group at day 7. Furthermore, the microbiota composition became distinct between COVID-19 and non-COVID-19 groups. The fecal microbiota of COVID-19 patients was characterized by increased Bacteroides and the pneumonia group by Prevotella. In a distance-based redundancy analysis, only COVID-19 presented significant effects on the microbiota composition. Moreover, patients in ICU harbored increased Campylobacter and decreased butyrate-producing bacteria, such as Lachnospiraceae, Roseburia and Faecalibacterium as compared to patients in medicine. Both the stay in ICU and patient were significant factors affecting the microbiota composition. SARS-CoV-2 viral loads were higher in ICU than in non-ICU patients. CONCLUSIONS Overall, we identified distinct characteristics of the gut microbiota in COVID-19 patients compared to control groups. COVID-19 patients were primarily characterized by increased Bacteroides and decreased Prevotella. Moreover, disease severity showed a negative correlation with butyrate-producing bacteria. These features could offer valuable insights into potential targets for modulating the host response through the microbiota and contribute to a better understanding of the disease's pathophysiology. TRIAL REGISTRATION CER-VD 2020-00755 (05.05.2020) & 2017-01820 (08.06.2018).
Collapse
Affiliation(s)
- Tatiana Galperine
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, BH10-553, 1011, Lausanne, Switzerland
| | - Yangji Choi
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jean-Luc Pagani
- Service of Intensive Care, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Antonios Kritikos
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, BH10-553, 1011, Lausanne, Switzerland
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthaios Papadimitriou-Olivgeris
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, BH10-553, 1011, Lausanne, Switzerland
| | - Marie Méan
- Division of Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Valentin Scherz
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Onya Opota
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gilbert Greub
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Benoit Guery
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, BH10-553, 1011, Lausanne, Switzerland.
| | - Claire Bertelli
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
468
|
Oyesola OO, Hilligan KL, Namasivayam S, Howard N, Clancy CS, Zhao M, Oland SD, Kiwanuka KN, Garza NL, Lafont BAP, Johnson RF, Mayer-Barber KD, Sher A, Loke P. Exposure to lung-migrating helminth protects against murine SARS-CoV-2 infection through macrophage-dependent T cell activation. Sci Immunol 2023; 8:eadf8161. [PMID: 37566678 DOI: 10.1126/sciimmunol.adf8161] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/19/2023] [Indexed: 08/13/2023]
Abstract
Helminth endemic regions report lower COVID-19 morbidity and mortality. Here, we show that lung remodeling from a prior infection with a lung-migrating helminth, Nippostrongylus brasiliensis, enhances viral clearance and survival of human-ACE2 transgenic mice challenged with SARS-CoV-2 (SCV2). This protection is associated with a lymphocytic infiltrate, including increased accumulation of pulmonary SCV2-specific CD8+ T cells, and anti-CD8 antibody depletion abrogated the N. brasiliensis-mediated reduction in viral loads. Pulmonary macrophages with a type 2 transcriptional and epigenetic signature persist in the lungs of N. brasiliensis-exposed mice after clearance of the parasite and establish a primed environment for increased CD8+ T cell recruitment and activation. Accordingly, depletion of macrophages ablated the augmented viral clearance and accumulation of CD8+ T cells driven by prior N. brasiliensis infection. Together, these findings support the concept that lung-migrating helminths can limit disease severity during SCV2 infection through macrophage-dependent enhancement of antiviral CD8+ T cell responses.
Collapse
Affiliation(s)
- Oyebola O Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kerry L Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nina Howard
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chad S Clancy
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Mingming Zhao
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra D Oland
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kasalina N Kiwanuka
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole L Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bernard A P Lafont
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - P'ng Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
469
|
Deckert A, Anders S, Morales I, De Allegri M, Nguyen HT, Souares A, McMahon S, Meurer M, Burk R, Lou D, Brugnara L, Sand M, Koeppel L, Maier-Hein L, Ross T, Adler TJ, Brenner S, Dyer C, Herbst K, Ovchinnikova S, Marx M, Schnitzler P, Knop M, Bärnighausen T, Denkinger CM. Comparison of Four Active SARS-CoV-2 Surveillance Strategies in Representative Population Sample Points: Two-Factor Factorial Randomized Controlled Trial. JMIR Public Health Surveill 2023; 9:e44204. [PMID: 37235704 PMCID: PMC10437130 DOI: 10.2196/44204] [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: 11/14/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic is characterized by rapid increases in infection burden owing to the emergence of new variants with higher transmissibility and immune escape. To date, monitoring the COVID-19 pandemic has mainly relied on passive surveillance, yielding biased epidemiological measures owing to the disproportionate number of undetected asymptomatic cases. Active surveillance could provide accurate estimates of the true prevalence to forecast the evolution of the pandemic, enabling evidence-based decision-making. OBJECTIVE This study compared 4 different approaches of active SARS-CoV-2 surveillance focusing on feasibility and epidemiological outcomes. METHODS A 2-factor factorial randomized controlled trial was conducted in 2020 in a German district with 700,000 inhabitants. The epidemiological outcome comprised SARS-CoV-2 prevalence and its precision. The 4 study arms combined 2 factors: individuals versus households and direct testing versus testing conditioned on symptom prescreening. Individuals aged ≥7 years were eligible. Altogether, 27,908 addresses from 51 municipalities were randomly allocated to the arms and 15 consecutive recruitment weekdays. Data collection and logistics were highly digitized, and a website in 5 languages enabled low-barrier registration and tracking of results. Gargle sample collection kits were sent by post. Participants collected a gargle sample at home and mailed it to the laboratory. Samples were analyzed with reverse transcription loop-mediated isothermal amplification (RT-LAMP); positive and weak results were confirmed with real-time reverse transcription-polymerase chain reaction (RT-PCR). RESULTS Recruitment was conducted between November 18 and December 11, 2020. The response rates in the 4 arms varied between 34.31% (2340/6821) and 41.17% (2043/4962). The prescreening classified 16.61% (1207/7266) of the patients as COVID-19 symptomatic. Altogether, 4232 persons without prescreening and 7623 participating in the prescreening provided 5351 gargle samples, of which 5319 (99.4%) could be analyzed. This yielded 17 confirmed SARS-CoV-2 infections and a combined prevalence of 0.36% (95% CI 0.14%-0.59%) in the arms without prescreening and 0.05% (95% CI 0.00%-0.108%) in the arms with prescreening (initial contacts only). Specifically, we found a prevalence of 0.31% (95% CI 0.06%-0.58%) for individuals and 0.35% (95% CI 0.09%-0.61%) for households, and lower estimates with prescreening (0.07%, 95% CI 0.0%-0.15% for individuals and 0.02%, 95% CI 0.0%-0.06% for households). Asymptomatic infections occurred in 27% (3/11) of the positive cases with symptom data. The 2 arms without prescreening performed the best regarding effectiveness and accuracy. CONCLUSIONS This study showed that postal mailing of gargle sample kits and returning home-based self-collected liquid gargle samples followed by high-sensitivity RT-LAMP analysis is a feasible way to conduct active SARS-CoV-2 population surveillance without burdening routine diagnostic testing. Efforts to improve participation rates and integration into the public health system may increase the potential to monitor the course of the pandemic. TRIAL REGISTRATION Deutsches Register Klinischer Studien (DRKS) DRKS00023271; https://tinyurl.com/3xenz68a. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR2-10.1186/s13063-021-05619-5.
Collapse
Affiliation(s)
| | - Simon Anders
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | - Ivonne Morales
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Hoa Thi Nguyen
- Heidelberg Institute of Global Health, Heidelberg, Germany
| | | | | | - Matthias Meurer
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | - Robin Burk
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | - Dan Lou
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | - Lucia Brugnara
- evaplan GmbH at the University Hospital, Heidelberg, Germany
| | - Matthias Sand
- GESIS Leibniz-Institute for the Social Sciences, Mannheim, Germany
| | - Lisa Koeppel
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Lena Maier-Hein
- Division of Computer Assisted Medical Interventions, German Cancer Research Centre, Heidelberg, Germany
| | - Tobias Ross
- Division of Computer Assisted Medical Interventions, German Cancer Research Centre, Heidelberg, Germany
| | - Tim J Adler
- Division of Computer Assisted Medical Interventions, German Cancer Research Centre, Heidelberg, Germany
| | | | | | - Konrad Herbst
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | | | - Michael Marx
- evaplan GmbH at the University Hospital, Heidelberg, Germany
| | - Paul Schnitzler
- Center of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Knop
- Center for Molecular Biology Heidelberg, Heidelberg, Germany
| | | | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
470
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. RESEARCH SQUARE 2023:rs.3.rs-3154698. [PMID: 37645853 PMCID: PMC10462189 DOI: 10.21203/rs.3.rs-3154698/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | | | | | | | | | | | | | | | | |
Collapse
|
471
|
Hadynski JC, Diggins J, Goad Z, Joy M, Dunckel S, Kraus P, Lufkin T, Wriedt M. Metal-Organic Framework as a Fluorescent and Colorimetric Dual-Signal Readout Biosensor Platform for the Detection of a Genetic Sequence from the SARS-CoV-2 Genome. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38163-38170. [PMID: 37535905 DOI: 10.1021/acsami.3c03518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
The quest for the development of high-accuracy, point-of-care, and cost-effective testing platforms for SARS-CoV-2 infections is ongoing as current diagnostics rely on either assays based on costly yet accurate nucleic acid amplification tests (NAAT) or less selective and less sensitive but rapid and cost-effective antigen tests. As a potential solution, this work presents a fluorescence-based detection platform using a metal-organic framework (MOF) in an effective assay, demonstrating the potential of MOFs to recognize specific targets of the SARS-CoV-2 genome with high accuracy and rapid process turnaround time. As a highlight of this work, positive detection of SARS-CoV-2 is indicated by a visible color change of the MOF probe with ultrahigh detection selectivities down to single-base mismatch nucleotide sequences, thereby providing an alternative avenue for the development of innovative detection methods for diverse viral genomes.
Collapse
Affiliation(s)
- John C Hadynski
- Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Jaren Diggins
- Department of Chemistry, Texas Southern University, Houston, Texas 77004, United States
| | - Zachary Goad
- Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Monu Joy
- Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Steven Dunckel
- Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, New York 13699, United States
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, New York 13699, United States
| | - Mario Wriedt
- Department of Chemistry & Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| |
Collapse
|
472
|
López-Peñalver RS, Cañas-Cañas R, Casaña-Mohedo J, Benavent-Cervera JV, Fernández-Garrido J, Juárez-Vela R, Pellín-Carcelén A, Gea-Caballero V, Andreu-Fernández V. Predictive potential of SARS-CoV-2 RNA concentration in wastewater to assess the dynamics of COVID-19 clinical outcomes and infections. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 886:163935. [PMID: 37164095 PMCID: PMC10164651 DOI: 10.1016/j.scitotenv.2023.163935] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023]
Abstract
Coronavirus disease 2019 - caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) -, has triggered a worldwide pandemic resulting in 665 million infections and over 6.5 million deaths as of December 15, 2022. The development of different epidemiological tools have helped predict new outbreaks and assess the behavior of clinical variables in different health contexts. In this study, we aimed to monitor concentrations of SARS-CoV-2 in wastewater as a tool to predict the progression of clinical variables during Waves 3, 5, and 6 of the pandemic in the Spanish city of Xátiva from September 2020 to March 2022. We estimated SARS-CoV-2 RNA concentrations in 195 wastewater samples using the RT-PCR Diagnostic Panel validated by the Center for Disease Control and Prevention. We also compared the trends of several clinical variables (14-day cumulative incidence, positive cases, hospital cases and stays, critical cases and stays, primary care visits, and deaths) for each study wave against wastewater SARS-CoV-2 RNA concentrations using Pearson's product-moment correlations, a two-sided Mann-Whitney U test, and a cross-correlation analysis. We found strong correlations between SARS-CoV-2 concentrations with 14-day cumulative incidence and positive cases over time. Wastewater RNA concentrations showed strong correlations with these variables one and two weeks in advance. There were significant correlations with hospitalizations and critical care during Wave 3 and Wave 6; cross-correlations were stronger for hospitalization stays one week before during Wave 6. No association between vaccination percentages and wastewater viral concentrations was observed. Our findings support wastewater SARS-CoV-2 concentrations as a potential surveillance tool to anticipate infection and epidemiological data such as 14-day cumulative incidence, hospitalizations, and critical care stays. Public health authorities could use this epidemiological tool on a similar population as an aid for health care decision-making during an epidemic outbreak.
Collapse
Affiliation(s)
- Raimundo Seguí López-Peñalver
- Faculty of Health Sciences, Valencian International University (VIU), 46002, Valencia, Spain; Global Omnium, Valencia, Spain
| | | | - Jorge Casaña-Mohedo
- Faculty of Health Sciences, Valencian International University (VIU), 46002, Valencia, Spain; Faculty of Health Sciences, Universidad Católica de Valencia San Vicente Mártir, 46001, Valencia, Spain
| | | | - Julio Fernández-Garrido
- Consellería de Sanidad Universal y Salud Pública, Generalitat Valenciana, Department of Nursing, University of Valencia, 46001 Jaume Roig St, Valencia, Spain
| | - Raúl Juárez-Vela
- Faculty of Health Sciences, La Rioja University, 26006 Logroño, Spain
| | - Ana Pellín-Carcelén
- Faculty of Health Sciences, Valencian International University (VIU), 46002, Valencia, Spain
| | - Vicente Gea-Caballero
- Faculty of Health Sciences, Valencian International University (VIU), 46002, Valencia, Spain
| | - Vicente Andreu-Fernández
- Faculty of Health Sciences, Valencian International University (VIU), 46002, Valencia, Spain; Biosanitary Research Institute, Valencian International University (VIU), 46002, Valencia, Spain.
| |
Collapse
|
473
|
Rahmanzadeh F, Malekpour N, Faramarzi A, Yusefzadeh H. Cost-effectiveness analysis of diagnostic strategies for COVID-19 in Iran. BMC Health Serv Res 2023; 23:861. [PMID: 37580701 PMCID: PMC10426176 DOI: 10.1186/s12913-023-09868-9] [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: 12/06/2022] [Accepted: 07/31/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Since 2020, COVID-19 has become a global public health issue and has caused problems worldwide. This infection can lead to a fever and respiratory problems. Asymptomatic carriers of the virus are a significant part of the spread of the disease, so early screening and diagnosis of suspected cases of COVID-19 are essential. Generally, standard diagnostic methods include lung imaging (CT), polymerase chain reaction (PCR), and corona antibody (IgM&IgG) testing. However, the costs of the above tests for the healthcare system cannot be ignored, and evaluating the incremental costs against the additional benefit is necessary. Therefore, this study aimed to determine the cost-effectiveness of diagnostic methods for COVID-19 patients. MATERIALS AND METHODS In this research, an economic evaluation analysis was conducted to reveal the cost-effectiveness of the diagnostic strategies for COVID-19 from the service provider's perspective. Basic information about the costs of CT, serology (IgG&IgM), and molecular (PCR) tests were collected from the Ministry of Health of Iran. The effectiveness data were calculated according to the sensitivity and specificity of the diagnostic tests for COVID-19. In this study, the incremental cost-effectiveness ratio (ICER) of the diagnostic strategies for COVID-19 was estimated, and the most cost-effective diagnostic strategy was determined. In calculating ICER and analyzing the sensitivity of the results, Treeage software was used. RESULTS According to the calculated incremental effectiveness cost ratio for scenarios with 5, 10, and 50% prevalence of COVID-19 and according to the threshold defined by the World Health Organization, in the study, PCR, PCR, and IgG&IgM strategies are the most cost-effective diagnostic methods of the corona. Also, the results were not sensitive to the desired parameters based on the results of one-way sensitivity analysis. CONCLUSION Nowadays there are various tests with different levels of accuracy in the diagnosis of COVID-19. In general, PCR tests are more cost-effective for low prevalence of Covid-19, while IgM&IgG tests are more cost-effective for high estimated prevalence. The results of this research can help policymakers and health system managers to validate the most accurate diagnostic method for COVID-19, considering the prevalence of the disease.
Collapse
Affiliation(s)
- F Rahmanzadeh
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - N Malekpour
- Health Education Department, Urmia University of Medical Sciences, Health deputy, Urmia, Iran
| | - A Faramarzi
- Department of Health Economics and Management, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran
| | - H Yusefzadeh
- Department of Health Economics and Management, School of Public Health, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
474
|
Štrbenc M, Kuhar U, Lainšček D, Orehek S, Slavec B, Krapež U, Malovrh T, Majdič G. Rehoming and Other Refinements and Replacement in Procedures Using Golden Hamsters in SARS-CoV-2 Vaccine Research. Animals (Basel) 2023; 13:2616. [PMID: 37627407 PMCID: PMC10451472 DOI: 10.3390/ani13162616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Effective vaccines are needed to fight the COVID-19 pandemic. Forty golden hamsters were inoculated with two promising vaccine candidates and eighteen animals were used in pilot trials with viral challenge. ELISA assays were performed to determine endpoint serum titres for specific antibodies and virus neutralisation tests were used to evaluate the efficacy of antibodies. All tests with serum from vaccinated hamsters were negative even after booster vaccinations and changes in vaccination protocol. We concluded that antibodies did not have sufficient neutralising properties. Refinements were observed at all steps, and the in vitro method (virus neutralisation test) presented a replacement measure and ultimately lead to a reduction in the total number of animals used in the project. The institutional animal welfare officer and institutional designated veterinarian approved the reuse or rehoming of the surplus animals. Simple socialization procedures were performed and ultimately 19 animals were rehomed, and feedback was collected. Recently, FELASA published recommendations for rehoming of animals used for scientific and educational purposes, with species-specific guidelines, including mice, rats, and rabbits. Based on our positive experience and feedback from adopters, we concluded that the rehoming of rodents, including hamsters, is not only possible, but highly recommended.
Collapse
Affiliation(s)
- Malan Štrbenc
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Urška Kuhar
- Institute for Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (T.M.)
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; (D.L.); (S.O.)
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; (D.L.); (S.O.)
| | - Brigita Slavec
- Institute of Poultry, Birds, Small Mammals and Reptiles, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.S.); (U.K.)
| | - Uroš Krapež
- Institute of Poultry, Birds, Small Mammals and Reptiles, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.S.); (U.K.)
| | - Tadej Malovrh
- Institute for Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (T.M.)
| | - Gregor Majdič
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| |
Collapse
|
475
|
Tan RMR, Ong GYK, Chong SL, Soo JSL, Ganapathy S, Tyebally A, Lee KP. Pretriage criteria for identifying COVID-19-positive patients in the paediatric emergency department. Singapore Med J 2023:384050. [PMID: 37675677 DOI: 10.4103/singaporemedj.smj-2021-381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Affiliation(s)
- Ronald Ming Ren Tan
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| | - Gene Yong-Kwang Ong
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| | - Shu-Ling Chong
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| | | | - Sashikumar Ganapathy
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| | - Arif Tyebally
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| | - Khai Pin Lee
- Department of Emergency Medicine, KK Women's and Children's Hospital; Duke-NUS Medical School, Singapore
| |
Collapse
|
476
|
Baek YH, Park MY, Lim HJ, Youm DJ, You Y, Ahn S, Park JE, Kim MJ, Lee SH, Sohn YH, Yang YJ. Evaluation of Rapid Multiplex Reverse Transcription-Quantitative Polymerase Chain Reaction Assays for SARS-CoV-2 Detection in Individual and Pooled Samples. Life (Basel) 2023; 13:1717. [PMID: 37629574 PMCID: PMC10455980 DOI: 10.3390/life13081717] [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: 07/13/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Although coronavirus disease 2019 (COVID-19) is no longer a Public Health Emergency of International Concern (PHEIC), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has had a vast impact to date. Hence, continuous management is required, given the uncertainty caused by the potential evolution of SARS-CoV-2. Reverse transcription-quantitative PCR (RT-qPCR) diagnosis has been fundamental in overcoming this issue. In this study, the performances of two rapid RT-qPCR assays (Real-Q Direct SARS-CoV-2 Detection Kit and Allplex™ SARS-CoV-2 fast PCR Assay) with short PCR times were comparatively evaluated using a STANDARD M nCoV Real-Time Detection Kit (STANDARD M, conventional RT-qPCR assay). All kits showed a limit of detection values (102-103 copies/reaction). The evaluation showed that the two rapid assay tests had ≥97.89% sensitivity and ≥99.51% specificity (κ = 0.98) for individual samples and ≥97.32% sensitivity and ≥97.67% specificity for pooled samples compared to STANDARD M. These results indicate that the two rapid RT-qPCR kits, which showed significant time reduction in performance, are as effective as a conventional RT-qPCR assay. They are likely to increase not only the number of tests that can be performed but also the efficiency of sustainable management of COVID-19 in the long term.
Collapse
Affiliation(s)
- Young-Hyun Baek
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Min-Young Park
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Ho-Jae Lim
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea;
| | - Dong-Jae Youm
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Youngshin You
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Seojin Ahn
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Jung-Eun Park
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea;
| | - Min-Jin Kim
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Sun-Hwa Lee
- Department of Laboratory Medicine, Seegene Medical Foundation, Seoul 04805, Republic of Korea;
| | - Yong-Hak Sohn
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| | - Yong-Jin Yang
- Department of Molecular Diagnostics, Seegene Medical Foundation, Seoul 04805, Republic of Korea; (Y.-H.B.); (M.-Y.P.); (H.-J.L.); (D.-J.Y.); (Y.Y.); (S.A.); (M.-J.K.); (Y.-H.S.)
| |
Collapse
|
477
|
Millward GG, Popelka SM, Gutierrez AG, Kowallis WJ, von Tersch RL, Yerramilli SV. A novel strategy to avoid sensitivity loss in pooled testing for SARS-CoV-2 surveillance: validation using nasopharyngeal swab and saliva samples. Front Public Health 2023; 11:1190308. [PMID: 37637813 PMCID: PMC10450028 DOI: 10.3389/fpubh.2023.1190308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
At the peak of the COVID-19 pandemic, pooled surveillance strategies were employed to alleviate the overwhelming demand for clinical testing facilities. A major drawback of most pooled-testing methods is the dilution of positive samples, which leads to a loss of detection sensitivity and the potential for false negatives. We developed a novel pooling strategy that compensates for the initial dilution with an appropriate concentration during nucleic acid extraction and real-time PCR. We demonstrated the proof of principle using laboratory-created 10-sample pools with one positive and corresponding individual positive samples by spiking a known amount of heat-inactivated SARS-CoV-2 into viral transport medium (VTM) or pooled negative saliva. No Ct difference was observed between a 10-sample pool with one positive vs. the corresponding individually analyzed positive sample by this method, suggesting that there is no detectable loss of sensitivity. We further validated this approach by using nasopharyngeal swab (NPS) specimens and showed that there is no loss of sensitivity. Serial dilutions of the virus were spiked into VTM and pooled with negative saliva in simulated 10-sample pools containing one positive to determine the LOD and process efficiency of this pooling methodology. The LOD of this approach was 10 copies/PCR, and the process efficiencies are ~95%-103% for N1 and ~87%-98% for N2 with samples in different matrices and with two different master mixes tested. Relative to TaqPath 1-step master mix, the TaqMan Fast Virus 1-Step master mix showed better sensitivity for the N2 assay, while the N1 assay showed no Ct difference. Our pooled testing strategy can facilitate large-scale, cost-effective SARS-CoV-2 surveillance screening and maintain the same level of sensitivity when analyzed individually or in a pool. This approach is highly relevant for public health surveillance efforts aimed at mitigating SARS-CoV-2 spread.
Collapse
Affiliation(s)
| | | | | | | | | | - Subrahmanyam V. Yerramilli
- Emerging Biological Threats Branch, Molecular Biology Division, Laboratory Sciences, Defense Centers for Public Health - Aberdeen “Formerly the Army Public Health Center”, Aberdeen Proving Ground, Edgewood, MD, United States
| |
Collapse
|
478
|
Shukla N, Srivastava N, Gupta R, Srivastava P, Narayan J. COVID Variants, Villain and Victory: A Bioinformatics Perspective. Microorganisms 2023; 11:2039. [PMID: 37630599 PMCID: PMC10459809 DOI: 10.3390/microorganisms11082039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 08/27/2023] Open
Abstract
The SARS-CoV-2 virus, a novel member of the Coronaviridae family, is responsible for the viral infection known as Coronavirus Disease 2019 (COVID-19). In response to the urgent and critical need for rapid detection, diagnosis, analysis, interpretation, and treatment of COVID-19, a wide variety of bioinformatics tools have been developed. Given the virulence of SARS-CoV-2, it is crucial to explore the pathophysiology of the virus. We intend to examine how bioinformatics, in conjunction with next-generation sequencing techniques, can be leveraged to improve current diagnostic tools and streamline vaccine development for emerging SARS-CoV-2 variants. We also emphasize how bioinformatics, in general, can contribute to critical areas of biomedicine, including clinical diagnostics, SARS-CoV-2 genomic surveillance and its evolution, identification of potential drug targets, and development of therapeutic strategies. Currently, state-of-the-art bioinformatics tools have helped overcome technical obstacles with respect to genomic surveillance and have assisted in rapid detection, diagnosis, and delivering precise treatment to individuals on time.
Collapse
Affiliation(s)
- Nityendra Shukla
- CSIR Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (N.S.); (R.G.)
| | - Neha Srivastava
- Amity Institute of Biotechnology, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow 226010, India; (N.S.); (P.S.)
| | - Rohit Gupta
- CSIR Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (N.S.); (R.G.)
| | - Prachi Srivastava
- Amity Institute of Biotechnology, Amity University, Uttar Pradesh, Lucknow Campus, Lucknow 226010, India; (N.S.); (P.S.)
| | - Jitendra Narayan
- CSIR Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (N.S.); (R.G.)
| |
Collapse
|
479
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.06.23286853. [PMID: 37609199 PMCID: PMC10441495 DOI: 10.1101/2023.03.06.23286853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
|
480
|
Hazem M, Ali SI, AlAlwan QM, Al Jabr IK, Alshehri SAF, AlAlwan MQ, Alsaeed MI, Aldawood M, Turkistani JA, Amin YA. Diagnostic Performance of the Radiological Society of North America Consensus Statement for Reporting COVID-19 Chest CT Findings: A Revisit. J Clin Med 2023; 12:5180. [PMID: 37629222 PMCID: PMC10455816 DOI: 10.3390/jcm12165180] [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: 05/14/2023] [Revised: 07/24/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious respiratory disease that leads to variable degrees of illness, and which may be fatal. We evaluated the diagnostic performance of each chest computed tomography (CT) reporting category recommended by the Expert Consensus of the Radiological Society of North America (RSNA) in comparison with that of reverse transcription polymerase chain reaction (RT-PCR). We aimed to add an analysis of this form of reporting in the Middle East, as few studies have been performed there. Between July 2021 and February 2022, 184 patients with a mean age of 55.56 ± 16.71 years and probable COVID-19 infections were included in this retrospective study. Approximately 64.67% (119 patients) were male, while 35.33% (65 patients) were female. Within 7 days, all patients underwent CT and RT-PCR examinations. According to a statement by the RSNA, the sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and accuracy of each CT reporting category were calculated, and the RT-PCR results were used as a standard reference. The RT-PCR results confirmed a final diagnosis of COVID-19 infection in 60.33% of the patients. For COVID-19 diagnoses, the typical category (n = 88) had a sensitivity, specificity, PPV, and accuracy of 74.8%, 93.2%, 94.3%, and 92.5%, respectively. For non-COVID-19 diagnoses, the PPVs for the atypical (n = 22) and negative (n = 46) categories were 81.8% and 89.1%, respectively. The PPV for the indeterminate (n = 28) category was 67.9%, with a low sensitivity of 17.1%. However, the RSNA's four chest CT reporting categories provide a strong diagnostic foundation and are highly correlated with the RT-PCR results for the typical, atypical, and negative categories, but they are weaker for the indeterminate category.
Collapse
Affiliation(s)
- Mohammed Hazem
- Department of Surgery, Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia; (I.K.A.J.); (S.A.F.A.)
- Diagnostic and Interventional Radiology Department, Faculty of Medicine, Sohag University, Sohag 82524, Egypt;
| | - Sayed Ibrahim Ali
- Department of Family and Community Medicine, Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia; (S.I.A.); (J.A.T.)
- Educational Psychology Department, College of Education, Helwan University, Cairo 11795, Egypt
| | - Qasem M. AlAlwan
- Department of Radiology, King Fahd Hospital Hofuf, Al-Ahsa 36441, Saudi Arabia; (Q.M.A.); (M.Q.A.)
| | - Ibrahim Khalid Al Jabr
- Department of Surgery, Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia; (I.K.A.J.); (S.A.F.A.)
| | - Sarah Abdulrahman F. Alshehri
- Department of Surgery, Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia; (I.K.A.J.); (S.A.F.A.)
| | - Mohammed Q. AlAlwan
- Department of Radiology, King Fahd Hospital Hofuf, Al-Ahsa 36441, Saudi Arabia; (Q.M.A.); (M.Q.A.)
| | | | - Mohammed Aldawood
- Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
| | - Jamela A. Turkistani
- Department of Family and Community Medicine, Collage of Medicine, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia; (S.I.A.); (J.A.T.)
| | - Yasser Abdelkarim Amin
- Diagnostic and Interventional Radiology Department, Faculty of Medicine, Sohag University, Sohag 82524, Egypt;
| |
Collapse
|
481
|
Ehrlich M, Madden C, McBride DS, Nolting JM, Huey D, Kenney S, Wang Q, Saif LJ, Vlasova A, Dennis P, Lombardi D, Gibson S, McLaine A, Lauterbach S, Yaxley P, Winston JA, Diaz-Campos D, Pesapane R, Flint M, Flint J, Junge R, Faith SA, Bowman AS, Hale VL. Lack of SARS-CoV-2 Viral RNA Detection among a Convenience Sampling of Ohio Wildlife, Companion, and Agricultural Animals, 2020-2021. Animals (Basel) 2023; 13:2554. [PMID: 37627345 PMCID: PMC10451347 DOI: 10.3390/ani13162554] [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: 06/24/2023] [Revised: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in humans in late 2019 and spread rapidly, becoming a global pandemic. A zoonotic spillover event from animal to human was identified as the presumed origin. Subsequently, reports began emerging regarding spillback events resulting in SARS-CoV-2 infections in multiple animal species. These events highlighted critical links between animal and human health while also raising concerns about the development of new reservoir hosts and potential viral mutations that could alter the virulence and transmission or evade immune responses. Characterizing susceptibility, prevalence, and transmission between animal species became a priority to help protect animal and human health. In this study, we coalesced a large team of investigators and community partners to surveil for SARS-CoV-2 in domestic and free-ranging animals around Ohio between May 2020 and August 2021. We focused on species with known or predicted susceptibility to SARS-CoV-2 infection, highly congregated or medically compromised animals (e.g., shelters, barns, veterinary hospitals), and animals that had frequent contact with humans (e.g., pets, agricultural animals, zoo animals, or animals in wildlife hospitals). This included free-ranging deer (n = 76 individuals), free-ranging mink (n = 57), multiple species of bats (n = 59), and other wildlife in addition to domestic cats (n = 275) and pigs (n = 184). In total, we tested 792 individual animals (34 species) via rRT-PCR for SARS-CoV-2 RNA. SARS-CoV-2 viral RNA was not detected in any of the tested animals despite a major peak in human SARS-CoV-2 cases that occurred in Ohio subsequent to the peak of animal samplings. Importantly, we did not test for SARS-CoV-2 antibodies in this study, which limited our ability to assess exposure. While the results of this study were negative, the surveillance effort was critical and remains key to understanding, predicting, and preventing the re-emergence of SARS-CoV-2 in humans or animals.
Collapse
Affiliation(s)
- Margot Ehrlich
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Christopher Madden
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Dillon S. McBride
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Jacqueline M. Nolting
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Devra Huey
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Scott Kenney
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Qiuhong Wang
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J. Saif
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Anastasia Vlasova
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Patricia Dennis
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Cleveland Metroparks Zoo, Cleveland, OH 44109, USA
- Cleveland Metroparks, Cleveland, OH 44144, USA
| | | | | | - Alexis McLaine
- Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah Lauterbach
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Page Yaxley
- Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jenessa A. Winston
- Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Center of Microbiome Science, The Ohio State University, Columbus, OH 43210, USA
| | - Dubraska Diaz-Campos
- Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Risa Pesapane
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- School of Environment and Natural Resources, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Mark Flint
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Jaylene Flint
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Randy Junge
- Columbus Zoo & Aquarium, Powell, OH 43065, USA
| | - Seth A. Faith
- Center of Microbiome Science, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew S. Bowman
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
| | - Vanessa L. Hale
- Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA (A.V.)
- Center of Microbiome Science, The Ohio State University, Columbus, OH 43210, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
482
|
Li Y, Chen J, Wei J, Liu X, Yu L, Yu L, Ding D, Yang Y. Metallic nanoplatforms for COVID-19 diagnostics: versatile applications in the pandemic and post-pandemic era. J Nanobiotechnology 2023; 21:255. [PMID: 37542245 PMCID: PMC10403867 DOI: 10.1186/s12951-023-01981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/03/2023] [Indexed: 08/06/2023] Open
Abstract
The COVID-19 pandemic, which originated in Hubei, China, in December 2019, has had a profound impact on global public health. With the elucidation of the SARS-CoV-2 virus structure, genome type, and routes of infection, a variety of diagnostic methods have been developed for COVID-19 detection and surveillance. Although the pandemic has been declared over, we are still significantly affected by it in our daily lives in the post-pandemic era. Among the various diagnostic methods, nanomaterials, especially metallic nanomaterials, have shown great potential in the field of bioanalysis due to their unique physical and chemical properties. This review highlights the important role of metallic nanosensors in achieving accurate and efficient detection of COVID-19 during the pandemic outbreak and spread. The sensing mechanisms of each diagnostic device capable of analyzing a range of targets, including viral nucleic acids and various proteins, are described. Since SARS-CoV-2 is constantly mutating, strategies for dealing with new variants are also suggested. In addition, we discuss the analytical tools needed to detect SARS-CoV-2 variants in the current post-pandemic era, with a focus on achieving rapid and accurate detection. Finally, we address the challenges and future directions of metallic nanomaterial-based COVID-19 detection, which may inspire researchers to develop advanced biosensors for COVID-19 monitoring and rapid response to other virus-induced pandemics based on our current achievements.
Collapse
Affiliation(s)
- Yuqing Li
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Mate-Rials & Devices, Soochow University, Suzhou, 215123, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinchao Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xueliang Liu
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Yu
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Linqi Yu
- Department of Immunization Program, Jing'an District Center for Disease Control and Prevention, Shanghai, 200072, China.
| | - Ding Ding
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yu Yang
- Institute of Molecular Medicine (IMM), School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
483
|
Mousavi SM, Kalashgrani MY, Gholami A, Omidifar N, Binazadeh M, Chiang WH. Recent Advances in Quantum Dot-Based Lateral Flow Immunoassays for the Rapid, Point-of-Care Diagnosis of COVID-19. BIOSENSORS 2023; 13:786. [PMID: 37622872 PMCID: PMC10452855 DOI: 10.3390/bios13080786] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023]
Abstract
The COVID-19 pandemic has spurred demand for efficient and rapid diagnostic tools that can be deployed at point of care to quickly identify infected individuals. Existing detection methods are time consuming and they lack sensitivity. Point-of-care testing (POCT) has emerged as a promising alternative due to its user-friendliness, rapidity, and high specificity and sensitivity. Such tests can be conveniently conducted at the patient's bedside. Immunodiagnostic methods that offer the rapid identification of positive cases are urgently required. Quantum dots (QDs), known for their multimodal properties, have shown potential in terms of combating or inhibiting the COVID-19 virus. When coupled with specific antibodies, QDs enable the highly sensitive detection of viral antigens in patient samples. Conventional lateral flow immunoassays (LFAs) have been widely used for diagnostic testing due to their simplicity, low cost, and portability. However, they often lack the sensitivity required to accurately detect low viral loads. Quantum dot (QD)-based lateral flow immunoassays have emerged as a promising alternative, offering significant advancements in sensitivity and specificity. Moreover, the lateral flow immunoassay (LFIA) method, which fulfils POCT standards, has gained popularity in diagnosing COVID-19. This review focuses on recent advancements in QD-based LFIA for rapid POCT COVID-19 diagnosis. Strategies to enhance sensitivity using QDs are explored, and the underlying principles of LFIA are elucidated. The benefits of using the QD-based LFIA as a POCT method are highlighted, and its published performance in COVID-19 diagnostics is examined. Overall, the integration of quantum dots with LFIA holds immense promise in terms of revolutionizing COVID-19 detection, treatment, and prevention, offering a convenient and effective approach to combat the pandemic.
Collapse
Affiliation(s)
- Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan;
| | - Masoomeh Yari Kalashgrani
- Biotechnology Research Center, Shiraz University of Medical Science, Shiraz 71468-64685, Iran; (M.Y.K.); (A.G.)
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Science, Shiraz 71468-64685, Iran; (M.Y.K.); (A.G.)
| | - Navid Omidifar
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran;
| | - Mojtaba Binazadeh
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz 71557-13876, Iran;
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City 106335, Taiwan;
| |
Collapse
|
484
|
Marin LM, Katselis GS, Chumala P, Sanche S, Julseth L, Penz E, Skomro R, Siqueira WL. Identification of SARS-CoV-2 biomarkers in saliva by transcriptomic and proteomics analysis. Clin Proteomics 2023; 20:30. [PMID: 37537537 PMCID: PMC10398966 DOI: 10.1186/s12014-023-09417-w] [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: 03/07/2022] [Accepted: 06/20/2023] [Indexed: 08/05/2023] Open
Abstract
The detection of SARS-CoV-2 biomarkers by real time PCR (rRT-PCR) has shown that the sensitivity of the test is negatively affected by low viral loads and the severity of the disease. This limitation can be overcome by the use of more sensitive approaches such as mass spectrometry (MS), which has not been explored for the detection of SARS-CoV-2 proteins in saliva. Thus, this study aimed at assessing the translational applicability of mass spectrometry-based proteomics approaches to identify viral proteins in saliva from people diagnosed with COVID-19 within fourteen days after the initial diagnosis, and to compare its performance with rRT-PCR. After ethics approval, saliva samples were self-collected by 42 COVID-19 positive and 16 healthy individuals. Samples from people positive for COVID-19 were collected on average on the sixth day (± 4 days) after initial diagnosis. Viable viral particles in saliva were heat-inactivated followed by the extraction of total proteins and viral RNA. Proteins were digested and then subjected to tandem MS analysis (LC-QTOF-MS/MS) using a data-dependent MS/MS acquisition qualitative shotgun proteomics approach. The acquired spectra were queried against a combined SARS-CoV-2 and human database. The qualitative detection of SARS-CoV-2 specific RNA was done by rRT-PCR. SARS-CoV-2 proteins were identified in all COVID-19 samples (100%), while viral RNA was detected in only 24 out of 42 COVID-19 samples (57.1%). Seven out of 18 SARS-CoV-2 proteins were identified in saliva from COVID-19 positive individuals, from which the most frequent were replicase polyproteins 1ab (100%) and 1a (91.3%), and nucleocapsid (45.2%). Neither viral proteins nor RNA were detected in healthy individuals. Our mass spectrometry approach appears to be more sensitive than rRT-PCR for the detection of SARS-CoV-2 biomarkers in saliva collected from COVID-19 positive individuals up to 14 days after the initial diagnostic test. Based on the novel data presented here, our MS technology can be used as an effective diagnostic test of COVID-19 for initial diagnosis or follow-up of symptomatic cases, especially in patients with reduced viral load.
Collapse
Affiliation(s)
- Lina M Marin
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - George S Katselis
- Canadian Centre for Health and Safety in Agriculture, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 2Z4, Canada
| | - Paulos Chumala
- Canadian Centre for Health and Safety in Agriculture, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 2Z4, Canada
| | - Stephen Sanche
- Division of Infectious Diseases, Department of Medicine, and Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Lucas Julseth
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
- Canadian Centre for Health and Safety in Agriculture, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 2Z4, Canada
| | - Erika Penz
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Robert Skomro
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0X8, Canada
| | - Walter L Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
485
|
Adler JM, Martin Vidal R, Voß A, Kunder S, Nascimento M, Abdelgawad A, Langner C, Vladimirova D, Osterrieder N, Gruber AD, Kunec D, Trimpert J. A non-transmissible live attenuated SARS-CoV-2 vaccine. Mol Ther 2023; 31:2391-2407. [PMID: 37263272 PMCID: PMC10214529 DOI: 10.1016/j.ymthe.2023.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/23/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Live attenuated vaccines (LAVs) administered via the mucosal route may offer better control of the COVID-19 pandemic than non-replicating vaccines injected intramuscularly. Conceptionally, LAVs have several advantages, including presentation of the entire antigenic repertoire of the virus, and the induction of strong mucosal immunity. Thus, immunity induced by LAV could offer superior protection against future surges of COVID-19 cases caused by emerging SARS-CoV-2 variants. However, LAVs carry the risk of unintentional transmission. To address this issue, we investigated whether transmission of a SARS-CoV-2 LAV candidate can be blocked by removing the furin cleavage site (FCS) from the spike protein. The level of protection and immunity induced by the attenuated virus with the intact FCS was virtually identical to the one induced by the attenuated virus lacking the FCS. Most importantly, removal of the FCS completely abolished horizontal transmission of vaccine virus between cohoused hamsters. Furthermore, the vaccine was safe in immunosuppressed animals and showed no tendency to recombine in vitro or in vivo with a SARS-CoV-2 field strain. These results indicate that removal of the FCS from SARS-CoV-2 LAV is a promising strategy to increase vaccine safety and prevent vaccine transmission without compromising vaccine efficacy.
Collapse
Affiliation(s)
- Julia M Adler
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany
| | | | - Anne Voß
- Institut für Tierpathologie, Freie Universität Berlin, 14163 Berlin, Germany
| | - Sandra Kunder
- Institut für Tierpathologie, Freie Universität Berlin, 14163 Berlin, Germany
| | | | - Azza Abdelgawad
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany
| | - Christine Langner
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany
| | - Daria Vladimirova
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany
| | | | - Achim D Gruber
- Institut für Tierpathologie, Freie Universität Berlin, 14163 Berlin, Germany
| | - Dusan Kunec
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany
| | - Jakob Trimpert
- Institut für Virologie, Freie Universität Berlin, 14163 Berlin, Germany.
| |
Collapse
|
486
|
Wang Z, Yi J, Yu Q, Liu Y, Zhang R, Zhang D, Yang W, Xu Y, Chen Y. Performance evaluation of QuantStudio 1 plus real-time PCR instrument for clinical laboratory analysis: A proof-of-concept study. Pract Lab Med 2023; 36:e00330. [PMID: 37649547 PMCID: PMC10462677 DOI: 10.1016/j.plabm.2023.e00330] [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: 07/23/2023] [Revised: 08/13/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023] Open
Abstract
Objective The real-time PCR system is one of the most powerful research tools available in the life sciences field. The aim of this study was to preliminarily evaluate the analytical performance of QuantStudio 1 Plus real-time PCR system (QS 1 plus) for clinical procedures. Methods The consistency of QS 1 plus with the reference system in terms of various clinical procedures was evaluated. For qualitative data, the Kappa test was used to analyze the agreement of the results. For the quantitative data, Passing-Bablok regression analysis and Bland-Altman plot analysis were used to assess the concordance between QS 1 plus and the reference instrument. Results Passing-Bablok regression showed an excellent agreement between the QS 1 plus and LC 480 systems for HBV DNA quantification (y = 0.928 + 0.970x), whereas Bland-Altman plot analysis showed very small mean deviations between the two systems. The QS 1 plus yielded perfectly consistent results with the reference instrument for methylenetetrahydrofolate reductase (MTHFR) C677T melting curve genotyping analysis, MTHFR C677T genotyping analysis, Norovirus RNA negative/positive analysis, influenza B virus (Flu B) RNA negative/positive analysis, Mycobacterium tuberculosis (MTB) DNA negative/positive analysis, Human Papillomavirus (HPV) genotyping analysis, epidermal growth factor receptor (EGFR) gene mutation analysis. Both the relative quantitative analysis and the relative quantitative analysis (standard curve) confirmed the satisfactory concordance between the QS 1 plus instrument and the ABI 7500 instrument by Passing-Bablok regression analysis (y = 0.180 + 0.817x and y = 0.012 + 1.000x, respectively) and Bland-Altman plot analysis. Conclusions Our research has proven that QS 1 plus is adaptable to most test procedures in the clinical laboratory. This may provide the basis for its further application.
Collapse
Affiliation(s)
- Ziran Wang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Yi
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Yu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiwei Liu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dong Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenhang Yang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Chen
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
487
|
Ma L, Zhao W, Huang T, Jin E, Wu G, Zhao W, Bao Y. On the collection and integration of SARS-CoV-2 genome data. BIOSAFETY AND HEALTH 2023; 5:204-210. [PMID: 40078223 PMCID: PMC11894986 DOI: 10.1016/j.bsheal.2023.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 03/14/2025] Open
Abstract
Genome data of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is essential for virus diagnosis, vaccine development, and variant surveillance. To archive and integrate worldwide SARS-CoV-2 genome data, a series of resources have been constructed, serving as a fundamental infrastructure for SARS-CoV-2 research, pandemic prevention and control, and coronavirus disease 2019 (COVID-19) therapy. Here we present an overview of extant SARS-CoV-2 resources that are devoted to genome data deposition and integration. We review deposition resources in data accessibility, metadata standardization, data curation and annotation; review integrative resources in data source, de-redundancy processing, data curation and quality assessment, and variant annotation. Moreover, we address issues that impede SARS-CoV-2 genome data integration, including low-complexity, inconsistency and absence of isolate name, sequence inconsistency, asynchronous update of genome data, and mismatched metadata. We finally provide insights into data standardization consensus and data submission guidelines, to promote SARS-CoV-2 genome data sharing and integration.
Collapse
Affiliation(s)
- Lina Ma
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Zhao
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianhao Huang
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Enhui Jin
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gangao Wu
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenming Zhao
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiming Bao
- China National Center for Bioinformation, Beijing 100101, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
488
|
Gwarinda HB, Tessema SK, Raman J, Greenhouse B, Birkholtz LM. Population structure and genetic connectivity of Plasmodium falciparum in pre-elimination settings of Southern Africa. FRONTIERS IN EPIDEMIOLOGY 2023; 3:1227071. [PMID: 38455947 PMCID: PMC10910941 DOI: 10.3389/fepid.2023.1227071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/17/2023] [Indexed: 03/09/2024]
Abstract
To accelerate malaria elimination in the Southern African region by 2030, it is essential to prevent cross-border malaria transmission. However, countries within the region are highly interconnected due to human migration that aids in the movement of the parasite across geographical borders. It is therefore important to better understand Plasmodium falciparum transmission dynamics in the region, and identify major parasite source and sink populations, as well as cross-border blocks of high parasite connectivity. We performed a meta-analysis using collated parasite allelic data generated by microsatellite genotyping of malaria parasites from Namibia, Eswatini, South Africa, and Mozambique (N = 5,314). The overall number of unique alleles was significantly higher (P ≤ 0.01) in Namibia (mean A = 17.3 ± 1.46) compared to South Africa (mean A = 12.2 ± 1.22) and Eswatini (mean A = 13.3 ± 1.27, P ≤ 0.05), whilst the level of heterozygosity was not significantly different between countries. The proportion of polyclonal infections was highest for Namibia (77%), and lowest for Mozambique (64%). A was significant population structure was detected between parasites from the four countries, and patterns of gene flow showed that Mozambique was the major source area and Eswatini the major sink area of parasites between the countries. This study showed strong signals of parasite population structure and genetic connectivity between malaria parasite populations across national borders. This calls for strengthening the harmonization of malaria control and elimination efforts between countries in the southern African region. This data also proves its potential utility as an additional surveillance tool for malaria surveillance on both a national and regional level for the identification of imported cases and/or outbreaks, as well as monitoring for the potential spread of anti-malarial drug resistance as countries work towards malaria elimination.
Collapse
Affiliation(s)
- Hazel B. Gwarinda
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Sofonias K. Tessema
- EppiCenter, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Jaishree Raman
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research (ARMMOR), Centre for Emerging Zoonotic and Parasitic Diseases, A Division of the National Health Laboratory Service, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, Wits Research Institute for Malaria, University of Witwatersrand, Johannesburg, South Africa
| | - Bryan Greenhouse
- EppiCenter, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Lyn-Marié Birkholtz
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
489
|
Zheng P, Zhou C, Ding Y, Liu B, Lu L, Zhu F, Duan S. Nanopore sequencing technology and its applications. MedComm (Beijing) 2023; 4:e316. [PMID: 37441463 PMCID: PMC10333861 DOI: 10.1002/mco2.316] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
Since the development of Sanger sequencing in 1977, sequencing technology has played a pivotal role in molecular biology research by enabling the interpretation of biological genetic codes. Today, nanopore sequencing is one of the leading third-generation sequencing technologies. With its long reads, portability, and low cost, nanopore sequencing is widely used in various scientific fields including epidemic prevention and control, disease diagnosis, and animal and plant breeding. Despite initial concerns about high error rates, continuous innovation in sequencing platforms and algorithm analysis technology has effectively addressed its accuracy. During the coronavirus disease (COVID-19) pandemic, nanopore sequencing played a critical role in detecting the severe acute respiratory syndrome coronavirus-2 virus genome and containing the pandemic. However, a lack of understanding of this technology may limit its popularization and application. Nanopore sequencing is poised to become the mainstream choice for preventing and controlling COVID-19 and future epidemics while creating value in other fields such as oncology and botany. This work introduces the contributions of nanopore sequencing during the COVID-19 pandemic to promote public understanding and its use in emerging outbreaks worldwide. We discuss its application in microbial detection, cancer genomes, and plant genomes and summarize strategies to improve its accuracy.
Collapse
Affiliation(s)
- Peijie Zheng
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
| | - Chuntao Zhou
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
| | - Yuemin Ding
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
- Institute of Translational Medicine, School of MedicineZhejiang University City CollegeHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineZhejiang University City CollegeHangzhouChina
| | - Bin Liu
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
| | - Liuyi Lu
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
| | - Feng Zhu
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
| | - Shiwei Duan
- Department of Clinical MedicineSchool of MedicineZhejiang University City CollegeHangzhouChina
- Institute of Translational Medicine, School of MedicineZhejiang University City CollegeHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineZhejiang University City CollegeHangzhouChina
| |
Collapse
|
490
|
Rockenfeller R, Günther M, Mörl F. Reports of deaths are an exaggeration: all-cause and NAA-test-conditional mortality in Germany during the SARS-CoV-2 era. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221551. [PMID: 37538740 PMCID: PMC10394418 DOI: 10.1098/rsos.221551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/10/2023] [Indexed: 08/05/2023]
Abstract
Counts of SARS-CoV-2-related deaths have been key numbers for justifying severe political, social and economical measures imposed by authorities world-wide. A particular focus thereby was the concomitant excess mortality (EM), i.e. fatalities above the expected all-cause mortality (AM). Recent studies, inter alia by the WHO, estimated the SARS-CoV-2-related EM in Germany between 2020 and 2021 as high as 200 000. In this study, we attempt to scrutinize these numbers by putting them into the context of German AM since the year 2000. We propose two straightforward, age-cohort-dependent models to estimate German AM for the 'Corona pandemic' years, as well as the corresponding flu seasons, out of historic data. For Germany, we find overall negative EM of about -18 500 persons for the year 2020, and a minor positive EM of about 7000 for 2021, unveiling that officially reported EM counts are an exaggeration. In 2022, the EM count is about 41 200. Further, based on NAA-test-positive related death counts, we are able to estimate how many Germans have died due to rather than with CoViD-19; an analysis not provided by the appropriate authority, the RKI. Through 2020 and 2021 combined, our due estimate is at no more than 59 500. Varying NAA test strategies heavily obscured SARS-CoV-2-related EM, particularly within the second year of the proclaimed pandemic. We compensated changes in test strategies by assuming that age-cohort-specific NAA-conditional mortality rates during the first pandemic year reflected SARS-CoV-2-characteristic constants.
Collapse
Affiliation(s)
- R. Rockenfeller
- Mathematical Institute, University of Koblenz, Koblenz, Germany
| | - M. Günther
- Computational Biophysics and Biorobotics, Institute for Modelling and Simulation of Biomechanical Systems, University of Stuttgart, Stuttgart, Germany
- Friedrich–Schiller–Universität, Jena, Germany
| | - F. Mörl
- Forschungsgesellschaft für Angewandte Systemsicherheit und Arbeitsmedizin mbH, AG Biomechanik and Ergonomie, Erfurt, Germany
| |
Collapse
|
491
|
Liang Y, Xiao M, Xie J, Li J, Zhang Y, Liu H, Zhang Y, He J, Zhang G, Wei N, Peng LM, Ke Y, Zhang ZY. Amplification-Free Detection of SARS-CoV-2 Down to Single Virus Level by Portable Carbon Nanotube Biosensors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208198. [PMID: 37046180 DOI: 10.1002/smll.202208198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/18/2023] [Indexed: 06/19/2023]
Abstract
The rapid and sensitive detection of trace-level viruses in a simple and reliable way is of great importance for epidemic prevention and control. Here, a multi-functionalized floating gate carbon nanotube field effect transistor (FG-CNT FET) based biosensor is reported for the single virus level detection of SARS-CoV-2 virus antigen and RNA rapidly with a portable sensing platform. The aptamers functionalized sensors can detect SARS-CoV-2 antigens from unprocessed nasopharyngeal swab samples within 1 min. Meanwhile, enhanced by a multi-probe strategy, the FG-CNT FET-based biosensor can detect the long chain RNA directly without amplification down to single virus level within 1 min. The device, constructed with packaged sensor chips and a portable sensing terminal, can distinguish 10 COVID-19 patients from 10 healthy individuals in clinical tests both by the RNAs and antigens by a combination detection strategy with an combined overall percent agreement (OPA) close to 100%. The results provide a general and simple method to enhance the sensitivity of FET-based biochemical sensors for the detection of nucleic acid molecules and demonstrate that the CNT FG FET biosensor is a versatile and reliable integrated platform for ultrasensitive multibiomarker detection without amplification and has great potential for point-of-care (POC) clinical tests.
Collapse
Affiliation(s)
- Yuqi Liang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
| | - Mengmeng Xiao
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Hunan, 411105, China
| | - Jing Xie
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China
| | - Jiahao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjia Lake West Road, Wuhan, 430065, China
| | - Yuyan Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
| | - Haiyang Liu
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
| | - Yang Zhang
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Hunan, 411105, China
| | - Jianping He
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Hunan, 411105, China
| | - Guojun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjia Lake West Road, Wuhan, 430065, China
| | - Nan Wei
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
| | - Lian-Mao Peng
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Hunan, 411105, China
| | - Yuehua Ke
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China
| | - Zhi-Yong Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, China
- Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Hunan, 411105, China
| |
Collapse
|
492
|
Trivedi S, Javed NB, Desai RS, Issar P. Diagnostic efficacy of chest CT imaging in diagnosis of COVID-19 cases based on duration of symptoms. Niger J Clin Pract 2023; 26:1171-1175. [PMID: 37635613 DOI: 10.4103/njcp.njcp_103_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Background Chest computed tomography (CT) imaging provides results more rapidly and with higher sensitivity than reverse transcription polymerase chain reaction in diagnosis of COVID-19. Aim To evaluate diagnostic efficacy of chest CT imaging in diagnosis of COVID-19 cases based on age and duration of symptoms. Materials and Methods A retrospective study conducted during December 2020 to June 2021 in a tertiary care hospital, India. Total 495 patients with typical clinical symptoms of COVID-19, reverse transcription polymerase chain reaction positive for COVID-19 and had undergone chest CT imaging were included. Descriptive statistical analysis was performed for all the variables. Receiver operating characteristic curve analysis was used to determine threshold value of chest CT severity score (CT_SS) based on duration of symptoms and age to diagnose COVID-19. Results Mean age of patients was 61.86 ± 10.77 years and 367 (71.4%) patients were male. Ground glass opacities were observed in 456 (92.1%) patients and in 332 (67.1%) patients, multilobes were affected. Total CT_SS showed positive correlation with age (r = 0.257) and duration of symptoms (r = 0.625). Total CT_SS >6 after a duration of 2 days of symptoms identified COVID-19 cases with sensitivity 90.8% (95% confidence interval [CI]: 87.5%-93.5%) and specificity 84.6% (95% CI: 76.2%-90.9%). Total CT_SS >11 in patients aged more than 60 years identified COVID-19 cases with sensitivity 47.4% (95% CI: 41.2%-53.6%) and specificity 87.3% (95% CI: 82.3%-91.4%). Conclusion Threshold value of CT_SS determined will help to expedite diagnosis of COVID-19 patients by the clinicians in an early stage especially in India and other developing countries which have a high patient volume and limited health resources.
Collapse
Affiliation(s)
- S Trivedi
- Department of Respiratory Medicine, Jawaharlal Nehru Hospital and Research Center, Bhilai Nagar, Chhattisgarh, India
| | - N B Javed
- Department of Public Health, College of Health Science, Saudi Electronic University, Dammam, Saudi Arabia
| | - R S Desai
- Department of Respiratory Medicine, Jawaharlal Nehru Hospital and Research Center, Bhilai Nagar, Chhattisgarh, India
| | - P Issar
- Department of Radiology, Jawaharlal Nehru Hospital and Research Center, Bhilai Nagar, Chhattisgarh, India
| |
Collapse
|
493
|
George A, Murugan T, Sampath S, N S M. Epidemiology of COVID-19 and the Utility of Cycle Threshold (Ct) Values in Predicting the Severity of Disease. Cureus 2023; 15:e43679. [PMID: 37724229 PMCID: PMC10505265 DOI: 10.7759/cureus.43679] [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: 08/14/2023] [Indexed: 09/20/2023] Open
Abstract
OBJECTIVES Advanced molecular diagnostic methods like real-time polymerase chain reaction (PCR) play a vital role in the early recognition of viral infections, including the coronavirus disease 2019 (COVID-19). Therefore, in the context of the recent COVID-19 pandemic, this study aimed to determine the correlation of cycle threshold (Ct) values with symptoms in COVID-19-positive patients. MATERIALS AND METHODS A retrospective study was conducted in a virus research diagnostic laboratory (VRDL) at a COVID-19-dedicated tertiary care hospital in South India. A total of 5563 COVID-19-positive patients were analyzed for symptom spectrum and duration of illness with Ct values of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). RESULTS Around 80% (n= 4401) of the patients were symptomatic and the rest were asymptomatic. Among the symptomatic patients, fever (66%) was the most common symptom. About 44% of symptomatic patients had a low Ct value (Ct ≤ 24). There was a significant difference in symptoms among patients with low, medium, and high Ct values. In the subpopulation of symptomatic patients analyzed for the association of Ct value and duration of illness, the mean duration of illness was three days and almost 88% of the patients were tested within five days of onset of symptoms. It has been observed that a shorter duration of illness lowers the Ct values. A significant association was seen between the mean Ct value and days since symptom onset (p-value = 0.016). CONCLUSION Most of the symptomatic patients had lower Ct values in comparison to the asymptomatic patients. A significant association between low Ct values and the duration of symptoms observed in our study explains the viral dynamics, i.e., higher viral shedding at the onset of symptoms and declines thereafter.
Collapse
Affiliation(s)
- Anuja George
- Microbiology, Indira Gandhi Medical College & Research Institute, Puducherry, IND
| | - Thamizharasi Murugan
- Microbiology, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry, IND
| | - Srinivasan Sampath
- Microbiology, Indira Gandhi Medical College & Research Institute, Puducherry, IND
| | - Madhusudhan N S
- Microbiology, Indira Gandhi Medical College & Research Institute, Puducherry, IND
| |
Collapse
|
494
|
Yang YP, Jiesisibieke ZL, Tung TH. Association Between Rapid Antigen Detection Tests and Real-Time Reverse Transcription-Polymerase Chain Reaction Assay for SARS-CoV-2: A Systematic Review and Meta-Analyses. Int J Public Health 2023; 68:1605452. [PMID: 37588042 PMCID: PMC10425602 DOI: 10.3389/ijph.2023.1605452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 07/11/2023] [Indexed: 08/18/2023] Open
Abstract
Objectives: We aimed to assess the association between rapid antigen detection tests and real-time reverse transcription-polymerase chain reaction assay for severe acute respiratory syndrome coronavirus 2. Methods: We searched PubMed, Cochrane Library, EMBASE, and the Web of Science from their inception to 31 May 2023. A random-effects meta-analysis was used to estimate false positives in the RADTs group, relative to those in the RT-PCR group, and subgroup analyses were conducted based on the different Ct value cut-offs (<40 or ≥40). We performed this study in accordance with the guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Results: Fifty-one studies were included and considered to be of moderate quality. We found a satisfactory overall false positive rate (0.01, 95% CI: 0.00-0.01) for the RADTs compared to RT-PCR. In the stratified analysis, we also found that the false positive rates of the RADTs did not increase when Ct values of RT-PCR (Ct < 40, 0.01, 95% CI: 0.00-0.01; Ct ≥ 40, 0.01, 95% CI: 0.00-0.01). Conclusion: In conclusion, the best available evidence supports an association between RADTs and RT-PCR. When Ct-values were analyzed using cut-off <40 or ≥40, this resulted in an estimated false positive rate of only 1%.
Collapse
Affiliation(s)
- Yu-Pei Yang
- Department of Hematology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zhu Liduzi Jiesisibieke
- School of Public Health, The University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong, Hong Kong SAR, China
| | - Tao-Hsin Tung
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
495
|
Ferreira MD, López LZ, da Silva FP, Miléo FC, Bortoluzzi MC, Dos Santos FA. COVID-19 hospitalized patients and oral changes: a case-control study. Clin Oral Investig 2023; 27:4481-4491. [PMID: 37191716 PMCID: PMC10185958 DOI: 10.1007/s00784-023-05070-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVES To investigate the association between hospitalization for COVID-19 and oral changes, and to evaluate whether oral changes can indicate a higher risk of disease progression to death. MATERIALS AND METHODS This case-control study analyzed patients hospitalized (university hospital), including those in intensive care unit and clinical wards. The study group comprised 69 COVID-19 positive patients (PCR-test), while the control group included 43 COVID-19 negative patients. A dentist performed oral evaluations, and salivary samples were collected for calcium, phosphatase, and pH analysis. Sociodemographic data, hospitalization information, and hematological test results were collected from electronic-medical records. The presence of oral changes was assessed using chi-square tests, and the predicted risk of death was analyzed using binary logistic regression. RESULTS COVID-19 positive patients had a significantly higher prevalence of oral changes compared to COVID-19 negative patients. The presence of any oral changes in COVID-19 positive patients indicated a 13-fold higher risk of mortality. "Bleeding ulcers," "pressure ulcers," and "angular cheilitis" were significantly associated with hospitalization for COVID-19. CONCLUSION There may be an association between hospitalization for COVID-19 and the development of oral changes, including bleeding ulcers, pressure ulcers. and angular cheilitis. These oral changes may serve as potential indicator for disease progression an increased risk of death. CLINICAL RELEVANCE COVID-19 hospitalized patients have a higher prevalence of oral changes, which indicate an increased risk of mortality. Oral medicine staff should be included in multidisciplinary teams to detect and treat these oral changes promptly.
Collapse
Affiliation(s)
- Marceli Dias Ferreira
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil
| | - Lourdes Zeballos López
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil
| | - Fernanda Pereira da Silva
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil
| | - Fernanda Couto Miléo
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil
| | - Marcelo Carlos Bortoluzzi
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil
| | - Fábio André Dos Santos
- Department of Dentistry, Universidade Estadual de Ponta Grossa, CEP, Av. Carlos Cavalcanti, N. 4748, Bloco M; Sala 13, Ponta Grossa, PR, 84030-900, Brazil.
| |
Collapse
|
496
|
Liu Y, Potts JL, Bloch D, Nian K, McCormick CA, Fanari O, Rouhanifard SH. Paired Capture and FISH Detection of Individual Virions Enable Cell-Free Determination of Infectious Titers. ACS Sens 2023; 8:2563-2571. [PMID: 37368999 PMCID: PMC10621038 DOI: 10.1021/acssensors.3c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Early detection of viruses can prevent the uncontrolled spread of viral infections. Determination of viral infectivity is also critical for determining the dosage of gene therapies, including vector-based vaccines, CAR T-cell therapies, and CRISPR therapeutics. In both cases, for viral pathogens and viral vector delivery vehicles, fast and accurate measurement of infectious titers is desirable. The most common methods for virus detection are antigen-based (rapid but not sensitive) and polymerase chain reaction (PCR)-based (sensitive but not rapid). Current viral titration methods heavily rely on cultured cells, which introduces variability within labs and between labs. Thus, it is highly desirable to directly determine the infectious titer without using cells. Here, we report the development of a direct, fast, and sensitive assay for virus detection (dubbed rapid capture fluorescence in situ hybridization (FISH) or rapture FISH) and cell-free determination of infectious titers. Importantly, we demonstrate that the virions captured are "infectious," thus serving as a more consistent proxy of infectious titers. This assay is unique because it first captures viruses bearing an intact coat protein using an aptamer and then detects genomes directly in individual virions using fluorescence in situ hybridization (FISH); thus, it is selective for infectious particles (i.e., positive for coat proteins and positive for genomes).
Collapse
Affiliation(s)
- Yifang Liu
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jacob L. Potts
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Dylan Bloch
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Keqing Nian
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Caroline A. McCormick
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Oleksandra Fanari
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Sara H. Rouhanifard
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
497
|
de Melo GD, Perraud V, Alvarez F, Vieites-Prado A, Kim S, Kergoat L, Coleon A, Trüeb BS, Tichit M, Piazza A, Thierry A, Hardy D, Wolff N, Munier S, Koszul R, Simon-Lorière E, Thiel V, Lecuit M, Lledo PM, Renier N, Larrous F, Bourhy H. Neuroinvasion and anosmia are independent phenomena upon infection with SARS-CoV-2 and its variants. Nat Commun 2023; 14:4485. [PMID: 37495586 PMCID: PMC10372078 DOI: 10.1038/s41467-023-40228-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
Anosmia was identified as a hallmark of COVID-19 early in the pandemic, however, with the emergence of variants of concern, the clinical profile induced by SARS-CoV-2 infection has changed, with anosmia being less frequent. Here, we assessed the clinical, olfactory and neuroinflammatory conditions of golden hamsters infected with the original Wuhan SARS-CoV-2 strain, its isogenic ORF7-deletion mutant and three variants: Gamma, Delta, and Omicron/BA.1. We show that infected animals develop a variant-dependent clinical disease including anosmia, and that the ORF7 of SARS-CoV-2 contributes to the induction of olfactory dysfunction. Conversely, all SARS-CoV-2 variants are neuroinvasive, regardless of the clinical presentation they induce. Taken together, this confirms that neuroinvasion and anosmia are independent phenomena upon SARS-CoV-2 infection. Using newly generated nanoluciferase-expressing SARS-CoV-2, we validate the olfactory pathway as a major entry point into the brain in vivo and demonstrate in vitro that SARS-CoV-2 travels retrogradely and anterogradely along axons in microfluidic neuron-epithelial networks.
Collapse
Affiliation(s)
- Guilherme Dias de Melo
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Victoire Perraud
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Flavio Alvarez
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, F-75015, Paris, France
- Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Alba Vieites-Prado
- Institut du Cerveau et de la Moelle Épinière, Laboratoire de Plasticité Structurale, , Sorbonne Université, INSERM U1127, CNRS UMR7225, 75013, Paris, France
| | - Seonhee Kim
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Lauriane Kergoat
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Anthony Coleon
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Bettina Salome Trüeb
- Institute of Virology and Immunology (IVI), Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Magali Tichit
- Institut Pasteur, Université Paris Cité, Histopathology Platform, F-75015, Paris, France
| | - Aurèle Piazza
- Institut Pasteur, Université Paris Cité, Spatial Regulation of Genomes Laboratory, F-75015, Paris, France
| | - Agnès Thierry
- Institut Pasteur, Université Paris Cité, Spatial Regulation of Genomes Laboratory, F-75015, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, F-75015, Paris, France
| | - Nicolas Wolff
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, F-75015, Paris, France
| | - Sandie Munier
- Institut Pasteur, Université Paris Cité, Molecular Genetics of RNA viruses Unit, F-75015, Paris, France
| | - Romain Koszul
- Institut Pasteur, Université Paris Cité, Spatial Regulation of Genomes Laboratory, F-75015, Paris, France
| | - Etienne Simon-Lorière
- Institut Pasteur, Université Paris Cité, Evolutionary Genomics of RNA Viruses Group, F-75015, Paris, France
| | - Volker Thiel
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Marc Lecuit
- Institut Pasteur, Université Paris Cité, Inserm U1117, Biology of Infection Unit, 75015, Paris, France
- Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, APHP, Institut Imagine, 75006, Paris, France
| | - Pierre-Marie Lledo
- Institut Pasteur, Université Paris Cité, Perception and Memory Unit, F-75015 Paris, France; CNRS UMR3571, 75015, Paris, France
| | - Nicolas Renier
- Institut du Cerveau et de la Moelle Épinière, Laboratoire de Plasticité Structurale, , Sorbonne Université, INSERM U1127, CNRS UMR7225, 75013, Paris, France
| | - Florence Larrous
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France
| | - Hervé Bourhy
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015, Paris, France.
| |
Collapse
|
498
|
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
|
499
|
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
|
500
|
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
|