151
|
Viermyr HK, Halvorsen B, Sagen EL, Michelsen AE, Barrat-Due A, Kåsine T, Nezvalova-Henriksen K, Dyrhol-Riise AM, Lerum TV, Müller F, Tonby K, Tveita A, Aukrust P, Trøseid M, Ueland T, Dahl TB. High viral loads combined with inflammatory markers predict disease severity in hospitalized COVID-19 patients: Results from the NOR-Solidarity trial. J Intern Med 2024; 296:249-259. [PMID: 39011800 DOI: 10.1111/joim.13820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
OBJECTIVES To investigate temporal changes in the association between SARS-CoV2 viral load (VL) and markers of inflammation during hospitalization, as well as the ability of these markers alone or in combination to predict severe outcomes. METHODS Serial oropharyngeal and blood samples were obtained from hospitalized COVID-19 patients (n = 160). Levels of inflammatory markers and oropharyngeal VL were measured during hospitalization (admission, days 3-5, and days 7-10) and related to severe outcomes (respiratory failure/intensive care unit admission). RESULTS Elevated admission levels of IL (interleukin)-6, IL-33, IL-8, monocyte chemoattractant protein-1 (MCP-1), interferon-γ-induced protein 10 (IP-10), IL-1β, and IL-1Ra were associated with severe outcomes during hospitalization. Although no inflammatory markers correlated with VL at baseline, there was a significant correlation between VL and levels of IP-10 and MCP-1 at days 3-5, accompanied by IL-8 and IL-6 at days 7-10. Finally, there was a seemingly additive effect of IP-10, MCP-1, and IL-6 in predicting severe outcomes when combined with high VL at baseline. CONCLUSIONS An increasing number of inflammatory markers were associated with VL during the first 10 days of hospitalization, and several of these markers were associated with severe outcomes, in particular when combined with elevated VL. Future studies should assess the potential for combining antiviral and immunomodulatory treatment, preferably guided by viral and inflammatory biomarkers, for the selection of high-risk patients.
Collapse
Affiliation(s)
- Hans-Kittil Viermyr
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ellen Lund Sagen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Barrat-Due
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
- Division of Critical Care and Emergencies, Oslo University Hospital, Oslo, Norway
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Trine Kåsine
- Division of Critical Care and Emergencies, Oslo University Hospital, Oslo, Norway
| | - Katerina Nezvalova-Henriksen
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- Hospital Pharmacies, South-Eastern Norway Enterprise, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Tøri Vigeland Lerum
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Fredrik Müller
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anders Tveita
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
152
|
Liu X, Xie R, Li K, Zhu Z, Huang X, He Q, Sun Z, He H, Ge Y, Zhang Q, Chen H, Wang Y. On-mask detection of SARS-CoV-2 related substances by surface enhanced Raman scattering. Talanta 2024; 277:126403. [PMID: 38878511 DOI: 10.1016/j.talanta.2024.126403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 06/09/2024] [Indexed: 07/19/2024]
Abstract
We have developed a convenient surface-enhanced Raman scattering (SERS) platform based on vertical standing gold nanowires (v-AuNWs) which enabled the on-mask detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) related substances such as the Spike-1 protein and the corresponding pseudo-virus. The Spike-1 protein was clearly distinguished from BSA protein with an accuracy above 99 %, and the detection limit could be achieved down to 0.01 μg/mL. Notably, a similar accuracy was achieved for the pseudo-SARS-CoV-2 (pSARS-2) virus as compared to the pseudo-influenza H7N9 (pH7N9) virus. The sensing strategy and setups could be easily adapted to the real SARS-CoV-2 virus and other highly contagious viruses. It provided a promising way to screen the virus carriers by a fast evaluation of their wearing v-AuNWs integrated face-mask which was mandatory during the pandemic.
Collapse
Affiliation(s)
- Xiaohu Liu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Jinlian Road 1, Wenzhou, 325001, China
| | - Ruifeng Xie
- School of Optoelectronic Engineering, Changchun University of Science and Technology, Weixing Road 7089, Changchun, 130013, China
| | - Kang Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Zhelei Zhu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Xi Huang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Qian He
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Zhe Sun
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Haiyang He
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China
| | - Yuancai Ge
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Jinlian Road 1, Wenzhou, 325001, China
| | - Qingwen Zhang
- Wenzhou Institute, University of Chinese Academy of Sciences, Jinlian Road 1, Wenzhou, 325001, China
| | - Hu Chen
- School of Materials Science and Engineering, Hunan Institute of Technology, Henghua Road 18, Hengyang, 421002, China.
| | - Yi Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Xueyuan Road 270, Wenzhou, 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Jinlian Road 1, Wenzhou, 325001, China; School of Optoelectronic Engineering, Changchun University of Science and Technology, Weixing Road 7089, Changchun, 130013, China.
| |
Collapse
|
153
|
Zhao Y, Qiu J, Jiang P, Wang M, Sun M, Fan G, Yang N, Huang N, Han Y, Han L, Zhang Y. RNA extraction-free reduced graphene oxide-based RT-LAMP fluorescence assay for highly sensitive SARS-CoV-2 detection. Talanta 2024; 277:126413. [PMID: 38876035 DOI: 10.1016/j.talanta.2024.126413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/14/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Infectious diseases have always been a seriously endanger for human life and health. A rapid, accurate and ultra-sensitive virus nucleic acid detection is still a challenge to deal with infectious diseases. Here, a RNA extraction-free reduced graphene oxide-based reverse transcription-loop-mediated isothermal amplification (EF-G-RT-LAMP) fluorescence assay was developed to achieve high-throughput, rapid and ultra-sensitive SARS-CoV-2 RNA detection. The whole detection process only took ∼36 min. The EF-G-RT-LAMP assay achieves a detection limit of 0.6 copies μL-1 with a wide dynamic range of aM-pM. A large number (up to 384) of samples can be detected simultaneously. Simulated detection of the COVID-19 pseudovirus and clinical samples in nasopharyngeal swabs demonstrated a high-throughput, rapid and ultra-sensitive practical detection capability of the EF-G-RT-LAMP assay. The results proved that the assay would be used as a rapid, easy-to-implement approach for epidemiologic diagnosis and could be extended to other nucleic acid detections.
Collapse
Affiliation(s)
- Yujuan Zhao
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Jiaoyan Qiu
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Peiqing Jiang
- Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, China
| | - Min Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Mingyuan Sun
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Guangpeng Fan
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Ningkai Yang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Ning Huang
- Shandong Lifei Biological Group, Qingdao, 266000, China
| | - Yunrui Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China; School of Integrated Circuits, Shandong University, Jinan, Shandong, 250100, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, 250100, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China; School of Integrated Circuits, Shandong University, Jinan, Shandong, 250100, China.
| |
Collapse
|
154
|
Leony LM, Vasconcelos LCM, Silva RSHD, Camelier AA, Bandeira AC, Costa DLS, Siqueira ICD, Santos FLN. Assessing the performance of commercial serological tests for SARS-CoV-2 diagnosis. IJID REGIONS 2024; 12:100383. [PMID: 38974172 PMCID: PMC11225813 DOI: 10.1016/j.ijregi.2024.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
Objectives The emergence of SARS-CoV-2 has triggered a global pandemic with profound implications for public health. Rapid changes in the pandemic landscape and limitations in in vitro diagnostics led to the introduction of numerous diagnostic devices with variable performance. In this study, we evaluated three commercial serological assays in Brazil for detecting anti-SARS-CoV-2 antibodies. Methods We collected 90 serum samples from SARS-CoV-2-negative blood donors and 352 from SARS-CoV-2-positive, unvaccinated patients, categorized by symptom onset. Subsequently, we assessed the diagnostic performance of three commercial enzyme immunoassays: GOLD ELISA (enzyme-linked immunosorbent assay) COVID-19 Ig (immunoglobulin) G + IgM, Anti-SARS-CoV-2 NCP IgM ELISA, and Anti-SARS-CoV-2 NCP IgG ELISA. Results Our findings revealed that the GOLD ELISA COVID-19 IgG + IgM exhibited the highest sensitivity (57.7%) and diagnostic odds ratio, surpassing the manufacturer's reported sensitivity in most analyzed time frames while maintaining exceptional specificity (98.9%). Conversely, the Anti-SARS-CoV-2 NCP IgG ELISA demonstrated lower sensitivity but aligned with independent evaluations, boasting a specificity of 100%. However, the Anti-SARS-CoV-2 NCP IgM ELISA exhibited lower sensitivity than claimed, particularly in samples collected shortly after positive reverse transcription polymerase chain reaction results. Performance improved 15-21 days after symptom onset and beyond 22 days, but in the first week, both Anti-SARS-CoV-2 NCP IgM ELISA and Anti-SARS-CoV-2 NCP IgG ELISA struggled to differentiate positive and negative samples. Conclusions Our study emphasizes the need for standardized validation protocols to address discrepancies between manufacturer-claimed and actual performance. These insights provide essential information for health care practitioners and policymakers regarding the diagnostic capabilities of these assays in various clinical scenarios.
Collapse
Affiliation(s)
- Leonardo Maia Leony
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
| | - Larissa Carvalho Medrado Vasconcelos
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
| | - Ricardo Sampaio Hein da Silva
- Laboratory of Investigation in Global Health and Neglected Diseases, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
| | | | | | | | - Isadora Cristina de Siqueira
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Laboratory of Investigation in Global Health and Neglected Diseases, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Integrated Translational Program in Chagas disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Rio de Janeiro, Brazil
| | - Fred Luciano Neves Santos
- Advanced Public Health Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Interdisciplinary Research Group in Biotechnology and Epidemiology of Infectious Diseases (GRUPIBE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Integrated Translational Program in Chagas disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Rio de Janeiro, Brazil
| |
Collapse
|
155
|
Zeng W, Chen W, Liu Y, Zhang T, Zhai C, Li W, Wang L, Zhang C, Zeng Q, Wang F, Ma L. Preamplification-free ultra-fast and ultra-sensitive point-of-care testing via LwaCas13a. Biosens Bioelectron 2024; 259:116400. [PMID: 38776799 DOI: 10.1016/j.bios.2024.116400] [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: 12/15/2023] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
CRISPR based nucleic acid detection technology provides a deployable approach to point of care testing. While, there remain challenges limiting its practical applications, such as the need for pre-amplification and the long turnaround time. Here, we present a self-cascade signal amplification method with LwaCas13a and an artificially designed "U" rich RNA of stem-loop structure (URH) for pre-amplification-free ultra-fast and ultra-sensitive point-of-care testing (PASSPORT). The PASSPORT system contains: URH, crRNA targeted the URH, crRNA targeted the interesting RNA, fluorescent RNA reporter and LwaCas13a. The assay realized the detection of 100 copies/mL, within 5 min. The PASSPORT platform was further adopted for the detection of marker gene from SASR-CoV-2 and Severe fever with thrombocytopenia syndrome virus (SFTSV), respectively, and 100% accuracy for the analysis of clinical specimens (100 SASR-CoV-2 specimens and 16 SFTSV specimens) was obtained. Integrated with a lateral flow assay device, this assay could provide an alternative platform for the development of point of care testing (POCT) biosensors. PASSPORT has the potential to enable sensitive, specific, user-friendly, rapid, affordable, equipment-free and point-of-care testing for the purpose of large-scale screening and in case of epidemic outbreak.
Collapse
Affiliation(s)
- Wanting Zeng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Wanping Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Yang Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Ting Zhang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China
| | - Chao Zhai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Wenqiang Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Longyu Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Cheng Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China
| | - Qili Zeng
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China
| | - Fei Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China.
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, China.
| |
Collapse
|
156
|
Silva T, Oliveira E, Oliveira A, Menezes A, Jeremias WDJ, Grenfell RF, Monte-Neto RLD, Pascoal-Xavier MA, Campos MA, Fernandes G, Alves P. Enhancing the epidemiological surveillance of SARS-CoV-2 using Sanger sequencing to identify circulating variants and recombinants. Braz J Microbiol 2024; 55:2085-2099. [PMID: 38802687 PMCID: PMC11405360 DOI: 10.1007/s42770-024-01387-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: 12/12/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Since the emergence of SARS-CoV-2 in December 2019, more than 12,000 mutations in the virus have been identified. These could cause changes in viral characteristics and directly impact global public health. The emergence of variants is a great concern due to the chance of increased transmissibility and infectivity. Sequencing for surveillance and monitoring circulating strains is extremely necessary as the early identification of new variants allows public health agencies to make faster and more effective decisions to contain the spread of the virus. In the present study, we identified circulating variants in samples collected in Belo Horizonte, Brazil, and detected a recombinant lineage using the Sanger method. The identification of lineages was done through gene amplification of SARS-CoV-2 by Reverse Transcription-Polymerase Chain Reaction (RT-PCR). By using these specific fragments, we were able to differentiate one variant of interest and five circulating variants of concern. We were also able to detect recombinants. Randomly selected samples were sequenced by either Sanger or Next Generation Sequencing (NGS). Our findings validate the effectiveness of Sanger sequencing as a powerful tool for monitoring variants. It is easy to perform and allows the analysis of a larger number of samples in countries that cannot afford NGS.
Collapse
Affiliation(s)
- Thaís Silva
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Eneida Oliveira
- Secretaria Municipal de Saúde, 2336, Afonso Pena Avenue, Belo Horizonte, Minas Gerais, 30130-007, Brazil
| | - Alana Oliveira
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - André Menezes
- Secretaria Municipal de Saúde, 2336, Afonso Pena Avenue, Belo Horizonte, Minas Gerais, 30130-007, Brazil
| | - Wander de Jesus Jeremias
- Department of Pharmacy, Federal University of Ouro Preto (UFOP), 27, Nine Street, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Rafaella Fq Grenfell
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, 501 D.W. Brooks Drive, Athens, GA, 30602, USA
| | - Rubens Lima do Monte-Neto
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Marcelo A Pascoal-Xavier
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
- Department of Anatomic Pathology, College of Medicine, Federal University of Minas Gerais, 6627, Presidente Antônio Carlos Avenue, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marco A Campos
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Gabriel Fernandes
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Pedro Alves
- Instituto René Rachou, Fundação Oswaldo Cruz, 1715, Augusto de Lima Avenue, Belo Horizonte, Minas Gerais, 30190-002, Brazil.
| |
Collapse
|
157
|
Shipley R, Seekings AH, Byrne AMP, Shukla S, James J, Goharriz H, Lean FZX, Núñez A, Fooks AR, McElhinney LM, Brookes SM. SARS-CoV-2 infection and transmission via the skin to oro-nasal route with the production of bioaerosols in the ferret model. J Gen Virol 2024; 105. [PMID: 39292223 PMCID: PMC11410047 DOI: 10.1099/jgv.0.002022] [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: 09/19/2024] Open
Abstract
Direct and indirect transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been attributed to virus survival in droplets, bioaerosols and on fomites including skin and surfaces. Survival of SARS-CoV-2 variants of concern (Alpha, Beta, Gamma, and Delta) on the skin and virus transference following rounds of skin-to-skin contact were assessed on porcine skin as a surrogate for human skin. SARS-CoV-2 variants were detectable on skin by RT-qPCR after 72 h at biologically relevant temperatures (35.2 °C) with viral RNA (vRNA) detected after ten successive skin-to-skin contacts. Skin-to-skin virus transmission to establish infection in ferrets as a model for mild/asymptomatic SARS-CoV-2 infection in mustelids and humans was also investigated and compared to intranasal ferret inoculation. Naïve ferrets exposed to Delta variant SARS-CoV-2 in a 'wet' or 'dry' form on porcine skin resulted in robust infection with shedding detectable for up to 14 days post-exposure, at comparable viral loads to ferrets inoculated intranasally. Transmission of SARS-CoV-2 to naïve ferrets in direct contact with infected ferrets was achieved, with environmental contamination detected from ferret fur swabs and air samples. Genetic substitutions were identified in bioaerosol samples acquired following single contact passage in ferrets, including Spike, ORF1ab, and ORF3a protein sequences, suggesting a utility for monitoring host adaptation and virus evolution via air sampling. The longevity of SARS-CoV-2 variants survival directly on the skin and skin-to-skin transference, enabling subsequent infection via the skin to oro-nasal contact route, could represent a pathway for SARS-CoV-2 infection with implications to public and veterinary health.
Collapse
Affiliation(s)
- Rebecca Shipley
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Amanda H Seekings
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Alexander M P Byrne
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
- Present address: Worldwide Influenza Centre, The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
| | - Shweta Shukla
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Joe James
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Hooman Goharriz
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Fabian Z X Lean
- Pathology and Animal Sciences Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - Alejandro Núñez
- Pathology and Animal Sciences Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Anthony R Fooks
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Lorraine M McElhinney
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| | - Sharon M Brookes
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone, Surrey, KT15 3NB, UK
| |
Collapse
|
158
|
Fernández-Bastit L, Montalvo T, Franco S, Barahona L, López-Bejar M, Carbajal A, Casas-Díaz E, Closa-Sebastià F, Segalés J, Vergara-Alert J. Monitoring SARS-CoV-2 infection in urban and peri-urban wildlife species from Catalonia (Spain). ONE HEALTH OUTLOOK 2024; 6:15. [PMID: 39217373 PMCID: PMC11366145 DOI: 10.1186/s42522-024-00109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Human activities including deforestation, urbanization, and wildlife exploitation increase the risk of transmission of zoonotic diseases. Urban and peri-urban wildlife species often flourish in human-altered environments, with their survival and behavior heavily influenced by human-generated food and waste. In Catalonia, Spain, and other Mediterranean regions, species of rodents, including the house mouse (Mus musculus), black rat (Rattus rattus), Norway rat (Rattus norvegicus), as well as wild boar (Sus scrofa) are common in urban and peri-urban areas. These species host numerous infectious agents, including coronaviruses (CoVs), posing potential human health risks. During the coronavirus disease 2019 (COVID-19) pandemic, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolved to infect previously non-susceptible species, with variants capable of infecting rodents, emphasizing their importance in surveillance studies. METHODS The present study assessed SARS-CoV-2 presence and/or exposure in 232 rodents, 313 wild boar, and 37 Vietnamese Pot-bellied pigs in Catalonia during the pandemic period (2020-2023). RESULTS All the animals tested for acute SARS-CoV-2 infection (232 rodents and 29 wild boar) were negative. For SARS-CoV-2 exposure, 3 out of 313 (0.96%) wild boar tested positive by ELISA, while the remaining 32 rodents, 310 wild boar, and 37 Vietnamese Pot-bellied pigs were all negative. Cross-reactivity with other CoVs was predicted for ELISA-positive samples, as the 3 wild boar tested negative by the virus neutralization assay, considered as the gold standard technique. CONCLUSIONS The absence of SARS-CoV-2 exposure or acute infection in wild boar and rodent species supports their negligible role in viral spread or transmission during the COVID-19 pandemic in Catalonia. However, their proximity to humans and the ongoing genetic evolution of SARS-CoV-2 underline the need for continued monitoring. Surveillance of SARS-CoV-2 infection in animal species can contribute to design measures to control the emergence of new animal reservoirs or intermediate hosts that could facilitate viral spillover events.
Collapse
Affiliation(s)
- Leira Fernández-Bastit
- Unitat Mixta d'investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Campus, Bellaterra, 08193, Catalonia, Spain
- Programa de Sanitat Animal, IRTA, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Campus, Bellaterra, 08193, Catalonia, Spain
| | - Tomás Montalvo
- Agència de Salut Pública de Barcelona, Barcelona, Spain
- CIBER de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, 08041, Spain
| | - Sandra Franco
- Agència de Salut Pública de Barcelona, Barcelona, Spain
| | - Laura Barahona
- Agència de Salut Pública de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, Barcelona, 08041, Spain
| | - Manel López-Bejar
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Annais Carbajal
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | | | | | - Joaquim Segalés
- Unitat Mixta d'investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Campus, Bellaterra, 08193, Catalonia, Spain.
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.
| | - Júlia Vergara-Alert
- Unitat Mixta d'investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Campus, Bellaterra, 08193, Catalonia, Spain.
- Programa de Sanitat Animal, IRTA, Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona (UAB), Campus, Bellaterra, 08193, Catalonia, Spain.
| |
Collapse
|
159
|
D'Souza N, Porter AM, Rose JB, Dreelin E, Peters SE, Nowlin PJ, Carbonell S, Cissell K, Wang Y, Flood MT, Rachmadi AT, Xi C, Song P, Briggs S. Public health use and lessons learned from a statewide SARS-CoV-2 wastewater monitoring program (MiNET). Heliyon 2024; 10:e35790. [PMID: 39220928 PMCID: PMC11363850 DOI: 10.1016/j.heliyon.2024.e35790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/27/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
The global SARS-CoV-2 monitoring effort has been extensive, resulting in many states and countries establishing wastewater-based epidemiology programs to address the spread of the virus during the pandemic. Challenges for programs include concurrently optimizing methods, training new laboratories, and implementing successful surveillance programs that can rapidly translate results for public health, and policy making. Surveillance in Michigan early in the pandemic in 2020 highlights the importance of quality-controlled data and explores correlations with wastewater and clinical case data aggregated at the state level. The lessons learned and potential measures to improve public utilization of results are discussed. The Michigan Network for Environmental Health and Technology (MiNET) established a network of laboratories that partnered with local health departments, universities, wastewater treatment plants (WWTPs) and other stakeholders to monitor SARS-CoV-2 in wastewater at 214 sites in Michigan. MiNET consisted of nineteen laboratories, twenty-nine local health departments, 6 Native American tribes, and 60 WWTPs monitoring sites representing 45 % of Michigan's population from April 6 and December 29, 2020. Three result datasets were created based on quality control criteria. Wastewater results that met all quality assurance criteria (Dataset Mp) produced strongest correlations with reported clinical cases at 16 days lag (rho = 0.866, p < 0.05). The project demonstrated the ability to successfully track SARS-CoV-2 on a large, state-wide scale, particularly data that met the outlined quality criteria and provided an early warning of increasing COVID-19 cases. MiNET is currently poised to leverage its competency to complement public health surveillance networks through environmental monitoring for new and emerging pathogens of concern and provides a valuable resource to state and federal agencies to support future responses.
Collapse
Affiliation(s)
- Nishita D'Souza
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
| | - Alexis M. Porter
- Annis Water Resources Insititute, Grand Valley State University, Muskegon, MI, USA
| | - Joan B. Rose
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
| | - Erin Dreelin
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
| | - Susan E. Peters
- Michigan Department of Health and Human Services, Lansing, MI, USA
| | | | - Samantha Carbonell
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | - Yili Wang
- University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew T. Flood
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
| | | | - Chuanwu Xi
- University of Michigan, Ann Arbor, Michigan, USA
| | - Peter Song
- University of Michigan, Ann Arbor, Michigan, USA
| | - Shannon Briggs
- Michigan Department of Environment, Great Lakes, and Energy, Lansing, MI, USA
| | - the Michigan Network for Environmental Health and Technology (MiNET) consortium
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
- Annis Water Resources Insititute, Grand Valley State University, Muskegon, MI, USA
- Michigan Department of Health and Human Services, Lansing, MI, USA
- Northern Michigan Regional Laboratory, Gaylord, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
- Saginaw Valley State University, Michigan, USA
- University of Michigan, Ann Arbor, Michigan, USA
- Institute of Environmental Science and Research (ESR), New Zealand
- Michigan Department of Environment, Great Lakes, and Energy, Lansing, MI, USA
| |
Collapse
|
160
|
Babaei M, Heidari B, Sadeghi Haddad Zavareh M, Ahmadnia Z, Ghorbani H, Rouhi S. Serum tumor necrosis factor-alpha status in hospitalized patients with coronavirus disease-2019 (COVID-19). CASPIAN JOURNAL OF INTERNAL MEDICINE 2024; 15:601-605. [PMID: 39359436 PMCID: PMC11444111 DOI: 10.22088/cjim.15.4.601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/08/2022] [Indexed: 10/04/2024]
Abstract
Background Tumor necrosis factor alpha (TNF-α) produces an inflammatory process and plays a critical role against infection and in the control of viral infection. The present study was conducted to determine the status of serum TNF-α in hospitalized patients with coronavirus disease-2019 (COVID-19). Methods In this cross-sectional study the serum TNF-α level, sex, and age, were determined in patients with COVID-19. The association between variables was determined using the student t-test, analysis of variance (ANOVA) test, multiple logistic regression analysis, and the statistical package for the Social Sciences (SPSS)-18 (p < 0.05). Results A total of 91 (women 41.75%, and men 58.24%) patients with a mean serum TNF-α level of 9.9 picograms per milliliter (pg/mL) were considered. In all (100%) patients, the TNF-α serum level was more than the normal limit (P=0.95). 95.60% of patients suffered severe COVID-19, with a TNF-a serum level of 10.20 pg/mL (P=0.87). Mean TNF-α serum levels in women and men were 11.37 pg/mL and 8.8 pg/mL, respectively (P= 0.17). In the age group of > 70 years (11.30 pg/mL), serum TNF-α concentration was higher than the other age groups (p>0.05). Conclusion A significant proportion of women and men patients with COVID-19 in the middle and old age had a high concentration of serum TNF-α which may indicate the severity of the disease. Serum TNF-α level is different in women and men of different ages, so it can contribute to treatment strategies.
Collapse
Affiliation(s)
- Mansour Babaei
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Behzad Heidari
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | | | - Zahra Ahmadnia
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Hossein Ghorbani
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Samaneh Rouhi
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
161
|
Pérez P, Astorgano D, Albericio G, Flores S, Sánchez-Corzo C, Noriega MA, Sánchez-Cordón PJ, Labiod N, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. MVA-based vaccine candidates expressing SARS-CoV-2 prefusion-stabilized spike proteins of the Wuhan, Beta or Omicron BA.1 variants protect transgenic K18-hACE2 mice against Omicron infection and elicit robust and broad specific humoral and cellular immune responses. Front Immunol 2024; 15:1420304. [PMID: 39267752 PMCID: PMC11390564 DOI: 10.3389/fimmu.2024.1420304] [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: 04/19/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Despite the decrease in mortality and morbidity due to SARS-CoV-2 infection, the incidence of infections due to Omicron subvariants of SARS-CoV-2 remains high. The mutations acquired by these subvariants, mainly concentrated in the receptor-binding domain (RBD), have caused a shift in infectivity and transmissibility, leading to a loss of effectiveness of the first authorized COVID-19 vaccines, among other reasons, by neutralizing antibody evasion. Hence, the generation of new vaccine candidates adapted to Omicron subvariants is of special interest in an effort to overcome this immune evasion. Here, an optimized COVID-19 vaccine candidate, termed MVA-S(3P_BA.1), was developed using a modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein from the Omicron BA.1 variant. The immunogenicity and efficacy induced by MVA-S(3P_BA.1) were evaluated in mice in a head-to-head comparison with the previously generated vaccine candidates MVA-S(3P) and MVA-S(3Pbeta), which express prefusion-stabilized S proteins from Wuhan strain and Beta variant, respectively, and with a bivalent vaccine candidate composed of a combination of MVA-S(3P) and MVA-S(3P_BA.1). The results showed that all four vaccine candidates elicited, after a single intramuscular dose, protection of transgenic K18-hACE2 mice challenged with SARS-CoV-2 Omicron BA.1, reducing viral loads, histopathological lesions, and levels of proinflammatory cytokines in the lungs. They also elicited anti-S IgG and neutralizing antibodies against various Omicron subvariants, with MVA-S(3P_BA.1) and the bivalent vaccine candidate inducing higher titers. Additionally, an intranasal immunization in C57BL/6 mice with all four vaccine candidates induced systemic and mucosal S-specific CD4+ and CD8+ T-cell and humoral immune responses, and the bivalent vaccine candidate induced broader immune responses, eliciting antibodies against the ancestral Wuhan strain and different Omicron subvariants. These results highlight the use of MVA as a potent and adaptable vaccine vector against new emerging SARS-CoV-2 variants, as well as the promising feature of combining multivalent MVA vaccine candidates.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- Mice, Transgenic
- Immunity, Humoral
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Humans
- Immunity, Cellular
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Female
- Vaccines, DNA/immunology
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sara Flores
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Cristina Sánchez-Corzo
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María A. Noriega
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Nuria Labiod
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - José M. Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
162
|
Sivertsen A, Mortensen N, Solem U, Valen E, Bullita MF, Wensaas KA, Litleskare S, Rørtveit G, Grewal HMS, Ulvestad E. Comprehensive contact tracing during an outbreak of alpha-variant SARS-CoV-2 in a rural community reveals less viral genomic diversity and higher household secondary attack rates than expected. mSphere 2024; 9:e0011424. [PMID: 39109863 DOI: 10.1128/msphere.00114-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/03/2024] [Indexed: 08/29/2024] Open
Abstract
Sequencing of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genomes throughout the COVID-19 pandemic has generated a wealth of data on viral evolution across populations, but only a few studies have so far explored SARS-CoV-2 evolution across large connected transmission networks. Here, we couple data from SARS-CoV-2 sequencing with contact tracing data from an outbreak with a single origin in a rural Norwegian community where samples from all exposed persons were collected prospectively. A total of 134 nasopharyngeal samples were positive by PCR. Among the 121 retrievable genomes, 81 were identical to the genome of the introductor, thus demonstrating that genomics beyond clustering genotypically similar viral genomes to confirm relatedness offers limited additional value to manual contact tracing. In the cases where mutations were discovered, five small genetic clusters were identified. We observed a household secondary attack rate of 77%, with 92% of household members infected among households with secondary transmission, suggesting that SARS-CoV-2 introduction into large families is likely to affect all household members. IMPORTANCE In outbreak investigations, obtaining a full overview of infected individuals within a population is seldom achieved. We here present an example where a single introduction of B1.1.7 SARS-CoV-2 within a rural community allowed for tracing of the virus from an introductor via dissemination through larger gatherings into households. The outbreak occurred before widespread vaccination, allowing for a "natural" outbreak development with community lockdown. We show through sequencing that the virus can infect up to five consecutive persons without gaining mutations, thereby showing that contact tracing seems more important than sequencing for local outbreak investigations in settings with few alternative introductory transmission pathways. We also show how larger households where a child introduced transmission appeared more likely to promote further spread of the virus compared to households with an adult as the primary introductor.
Collapse
Affiliation(s)
- Audun Sivertsen
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Nicolay Mortensen
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | | | - Eivind Valen
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | | | - Knut-Arne Wensaas
- NORCE Norwegian Research Centre, Research Unit for General Practice, Bergen, Norway
| | - Sverre Litleskare
- NORCE Norwegian Research Centre, Research Unit for General Practice, Bergen, Norway
| | - Guri Rørtveit
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Harleen M S Grewal
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Elling Ulvestad
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Bergen Integrated Diagnostic Stewardship Cluster, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
163
|
Thaler M, Ofman TP, Kok K, Heming JJA, Moran E, Pickles I, Leijs AA, van den Nieuwendijk AMC, van den Berg RJBHN, Ruijgrok G, Armstrong Z, Salgado-Benvindo C, Ninaber DK, Snijder EJ, van Boeckel CAA, Artola M, Davies GJ, Overkleeft HS, van Hemert MJ. Epi-Cyclophellitol Cyclosulfate, a Mechanism-Based Endoplasmic Reticulum α-Glucosidase II Inhibitor, Blocks Replication of SARS-CoV-2 and Other Coronaviruses. ACS CENTRAL SCIENCE 2024; 10:1594-1608. [PMID: 39220688 PMCID: PMC11363342 DOI: 10.1021/acscentsci.4c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024]
Abstract
The combined inhibition of endoplasmic reticulum (ER) α-glucosidases I and II has been shown to inhibit replication of a broad range of viruses that rely on ER protein quality control. We found, by screening a panel of deoxynojirimycin and cyclitol glycomimetics, that the mechanism-based ER α-glucosidase II inhibitor, 1,6-epi-cyclophellitol cyclosulfate, potently blocks SARS-CoV-2 replication in lung epithelial cells, halting intracellular generation of mature spike protein, reducing production of infectious progeny, and leading to reduced syncytium formation. Through activity-based protein profiling, we confirmed ER α-glucosidase II inhibition in primary airway epithelial cells, grown at the air-liquid interface. 1,6-epi-Cyclophellitol cyclosulfate inhibits early pandemic and more recent SARS-CoV-2 variants, as well as SARS-CoV and MERS-CoV. The reported antiviral activity is comparable to the best-in-class described glucosidase inhibitors, all competitive inhibitors also targeting ER α-glucosidase I and other glycoprocessing enzymes not involved in ER protein quality control. We propose selective blocking ER-resident α-glucosidase II in a covalent and irreversible manner as a new strategy in the search for effective antiviral agents targeting SARS-CoV-2 and other viruses that rely on ER protein quality control.
Collapse
Affiliation(s)
- Melissa Thaler
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Tim P. Ofman
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Ken Kok
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | | | - Elisha Moran
- Department
of Chemistry, University of York, York YO10 5DD, United Kingdom
| | - Isabelle Pickles
- Department
of Chemistry, University of York, York YO10 5DD, United Kingdom
| | - Anouk A. Leijs
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | | - Gijs Ruijgrok
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Zachary Armstrong
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Clarisse Salgado-Benvindo
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Dennis K. Ninaber
- Department
of Pulmonology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - Eric J. Snijder
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Marta Artola
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Gideon J. Davies
- Department
of Chemistry, University of York, York YO10 5DD, United Kingdom
| | - Herman S. Overkleeft
- Leiden
Institute of Chemistry, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Martijn J. van Hemert
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
164
|
Wee EGT, Kempster S, Ferguson D, Hall J, Ham C, Morris S, Crook A, Gilbert SC, Korber B, Almond N, Hanke T. Design, Immunogenicity and Preclinical Efficacy of the ChAdOx1.COVconsv12 Pan-Sarbecovirus T-Cell Vaccine. Vaccines (Basel) 2024; 12:965. [PMID: 39339997 PMCID: PMC11436245 DOI: 10.3390/vaccines12090965] [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: 07/29/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
During the COVID-19 pandemic, antibody-based vaccines targeting the SARS-CoV-2 spike glycoprotein were the focus for development because neutralizing antibodies were associated with protection against the SARS-CoV-2 infection pre-clinically and in humans. While deploying these spike-based vaccines saved millions of lives worldwide, it has become clear that the immunological mechanisms of protection against severe disease are multifaceted and involve non-neutralizing antibody components. Here, we describe a novel pan-sarbecovirus T-cell vaccine, ChAdOx1.COVconsv12, designed to complement and broaden the protection of spike vaccines. The vaccine immunogen COVconsv12 employs the two regions in the viral proteome most conserved among sarbecoviruses, which are delivered by replication-deficient vector ChAdOx1. It directs T cells towards epitopes shared among sarbecoviruses including evolving SARS-CoV-2 variants. Here, we show that ChAdOx1.COVconsv12 induced broad T-cell responses in the BALB/c and C57BL/6 mice. In the Syrian hamster challenge model, ChAdOx1.COVconsv12 alone did not protect against the SARS-CoV-2 infection, but when co-administered with 1/50th of the ChAdOx1 nCoV-19 spike vaccine protective dose, faster recovery and lower oral swab viral load were observed. Induction of CD8+ T cells may decrease COVID-19 severity and extend the T-cell response coverage of variants to match the known (and as yet unknown) members of the β-coronavirus family.
Collapse
Affiliation(s)
- Edmund G.-T. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah Kempster
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Deborah Ferguson
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Joanna Hall
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Claire Ham
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah C. Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Bette Korber
- New Mexico Consortium, Los Alamos, NM 87544, USA;
| | - Neil Almond
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
165
|
Pang Z, Shi L, Liu W, Liu W, Tian X, Wang M, Tao J. Development of a Novel Electrostatic-Based Bioaerosol Sampler. MICROMACHINES 2024; 15:1068. [PMID: 39337728 PMCID: PMC11434052 DOI: 10.3390/mi15091068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
On-site bioaerosol monitoring is essential for estimating microbial biomass and mitigating the risk of infection induced by aerosol transmission. This study introduces a novel electrostatic bioaerosol sampler, which is fabricated by the use of 3D printing, for rapid bioaerosol collection. Aerosol particles were charged and enriched in the sampler. Relationships between particle sizes and collection efficiencies under varying charging voltages were established using a charging model. The design of the sampler was optimized using commercial software, incorporating electrostatic field analysis, computational fluid dynamics (CFD), and particle trajectory simulations. To validate the sampler's collection efficiency, polystyrene (PS) spheres in an aerosol dispenser were atomized into an aerosol. The sampler collection efficiency exceeded 90% for particles larger than 1.2 μm under an applied voltage of 4.7 kV and an airflow rate of 2 L/min. The enrichment capacity was greater than 153,000 for particles larger than 1.2 μm under an applied voltage of 4.7 kV and an airflow rate of 8 L/min. With the merits of low cost, miniaturization, and high collection efficiency, the sampler can be used to collect samples on-site and in remote areas to verify the pathogens and reduce the risk of infection through aerosol transmission.
Collapse
Affiliation(s)
- Zirui Pang
- Key Laboratory of Laser and Infrared System Ministry of Education, Shandong University, Qingdao 266237, China
| | - Lulu Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Wei Liu
- Qingdao Institute of Measurement Technology, Qingdao 266000, China
| | - Wenru Liu
- Qingdao Institute of Measurement Technology, Qingdao 266000, China
| | - Xin Tian
- School of Physics and Electronic Information, Weifang University, Weifang 261061, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jifang Tao
- Key Laboratory of Laser and Infrared System Ministry of Education, Shandong University, Qingdao 266237, China
- School of Information Science and Engineering, Shandong University, Qingdao 266237, China
| |
Collapse
|
166
|
Segura-Ulate I, Apú N, Cortés B, Querol-Audi J, Zaldívar Y, Ortega CA, Flores-Mora F, Gatica-Arias A, Madrigal-Redondo G. Cross comparison of alternative diagnostic protocols including substitution to the clinical sample, RNA extraction method and nucleic acid amplification technology for COVID-19 diagnosis. Front Mol Biosci 2024; 11:1445142. [PMID: 39247206 PMCID: PMC11377848 DOI: 10.3389/fmolb.2024.1445142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Background the gold-standard diagnostic protocol (GSDP) for COVID-19 consists of a nasopharyngeal swab (NPS) sample processed through traditional RNA extraction (TRE) and amplified with retrotranscription quantitative polymerase chain reaction (RT-qPCR). Multiple alternatives were developed to decrease time/cost of GSDP, including alternative clinical samples, RNA extraction methods and nucleic acid amplification. Thus, we carried out a cross comparison of various alternatives methods against GSDP and each other. Methods we tested alternative diagnostic methods using saliva, heat-induced RNA release (HIRR) and a colorimetric retrotranscription loop-mediated isothermal amplification (RT-LAMP) as substitutions to the GSDP. Results RT-LAMP using NPS processed by TRE showed high sensitivity (96%) and specificity (97%), closely matching GSDP. When saliva was processed by TRE and amplified with both RT-LAMP and RT-qPCR, RT-LAMP yielded high diagnostic parameters (88%-96% sensitivity and 95%-100% specificity) compared to RT-qPCR. Nonetheless, when saliva processed by TRE and detected by RT-LAMP was compared against the GSDP, the resulting diagnostic values for sensitivity (78%) and specificity (87%) were somewhat high but still short of those of the GSDP. Finally, saliva processed with HIRR and detected via RT-LAMP was the simplest and fastest method, but its sensitivity against GSDP was too low (56%) for any clinical application. Also, in this last method, the acidity of a large percentage of saliva samples (9%-22%) affected the pH-sensitive colorimetric indicator used in the test, requiring the exclusion of these acidic samples or an extra step for pH correction. Discussion our comparison shows that RT-LAMP technology has diagnostic performance on par with RT-qPCR; likewise, saliva offers the same diagnostic functionality as NPS when subjected to a TRE method. Nonetheless, use of direct saliva after a HIRR and detected with RT-LAMP does not produce an acceptable diagnostic performance.
Collapse
Affiliation(s)
- Ismael Segura-Ulate
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Navilla Apú
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Bernal Cortés
- Agencia Costarricense de Investigaciones Biomédicas (ACIB) - Fundación INCIENSA (FUNIN), San José, Costa Rica
| | - Jordi Querol-Audi
- Laboratorio de Microbiología Experimental y Aplicada (LAMEXA), Universidad de Panamá, Ciudad de Panamá, Panama
- Sistema Nacional de Investigación (SNI), SENACYT, Ciudad de Panamá, Panama
| | - Yamitzel Zaldívar
- Instituto Conmemorativo Gorgas de Estudio de la Salud, Ciudad de Panamá, Panama
| | - Carlos Alexander Ortega
- Sección de Virología, Facultad de Medicina, Universidad de El Salvador, San Salvador, El Salvador
| | - Fernando Flores-Mora
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Andrés Gatica-Arias
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
- Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
| | - Germán Madrigal-Redondo
- Instituto de Investigaciones Farmacéuticas (INIFAR), Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
167
|
Claeys M, Al Obaidi S, Bruyland K, Vandecandelaere I, Vandesompele J. Assessment of DNA/RNA Defend Pro: An Inactivating Sample Collection Buffer for Enhanced Stability, Extraction-Free PCR, and Rapid Antigen Testing of Nasopharyngeal Swab Samples. Int J Mol Sci 2024; 25:9097. [PMID: 39201783 PMCID: PMC11354787 DOI: 10.3390/ijms25169097] [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: 08/02/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
This study comprehensively evaluated the DNA/RNA Defend Pro (DRDP) sample collection buffer, designed to inactivate and stabilize patient samples. The primary objectives were to assess DRDP's efficacy in ensuring sample stability, facilitating extraction-free polymerase chain reaction (PCR), and ensuring compatibility with rapid antigen testing (RAT). Ninety-five diagnostic nasopharyngeal swab samples tested for influenza virus (influenza A), respiratory syncytial virus (RSV A), and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were 10-fold diluted with DRDP and anonymized. Initial characterization and retesting of these samples using cobas Liat confirmed 88 samples as positive, validating the presence of viral targets. Results from rapid antigen testing showed lower sensitivity compared to nucleic acid amplification testing (NAAT) but maintained perfect specificity, with 40 out of 88 positive samples by cobas Liat also testing positive for RAT. Direct RT-qPCR of DRDP-diluted samples demonstrated robust compatibility, with 72 out of 88 samples positive for cobas Liat also testing positive by direct RT-qPCR. Non-concordant results could be explained by the 200-fold lower input of extraction-free NAAT. Stability testing involved incubating 31 positive samples at 4 °C, 20 °C, and 37 °C for 7 days, with extraction-free NAAT. DRDP guaranteed viral RNA stability at all temperatures for influenza A, SARS-CoV-2, and RSV A, showing stability up to 7 days at 4 °C. In conclusion, DRDP is an effective stabilizing medium compatible with direct RT-qPCR and rapid antigen testing and shows great potential for optimizing diagnostic processes, particularly in resource-limited or time-sensitive scenarios.
Collapse
Affiliation(s)
- Mikhail Claeys
- InActiv Blue, Industriepark Oost 2A, 8730 Beernem, Belgium; (M.C.); (S.A.O.)
| | - Saif Al Obaidi
- InActiv Blue, Industriepark Oost 2A, 8730 Beernem, Belgium; (M.C.); (S.A.O.)
- Campus Brugge Station, Howest University of Applied Sciences, Rijselstraat 5, 8200 Brugge, Belgium
| | - Karen Bruyland
- Medisch Labo Bruyland, Beneluxpark 2, 8500 Kortrijk, Belgium (I.V.)
| | | | - Jo Vandesompele
- InActiv Blue, Industriepark Oost 2A, 8730 Beernem, Belgium; (M.C.); (S.A.O.)
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000 Gent, Belgium
| |
Collapse
|
168
|
Thamsborg KKM, Hansen MS, Scheutz C, Klintø K, Kjeldsen P, Kvisgaard LK, Jensen HE, Hjerpe FB, Lohse L, Rasmussen TB, Rasmussen LD, Bedsted AE, Belsham GJ, Leisner JJ, Dalsgaard A. Microbiological and decomposition analysis of mass mink burial sites during the COVID-19 pandemic. Sci Rep 2024; 14:19440. [PMID: 39169071 PMCID: PMC11339334 DOI: 10.1038/s41598-024-69902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
In 2020, Denmark buried approximately four million culled, farmed mink in mass graves treated with slaked lime due to widespread SARS-CoV-2 infections. After six months, environmental concerns prompted the exhumation of these cadavers. Our analysis encompassed visual inspections, soil pH measurements, and gas emission assessments of the grave environment. Additionally, we evaluated carcasses for decay status, cadaverine content, and the presence of various pathogens, including SARS-CoV-2 and mink coronavirus. Our findings revealed minimal microbial activity and limited carcass decomposition. Although viral RNA from SARS-CoV-2 and mink coronavirus, along with DNA from Aleutian mink disease virus, were detected, the absence of infectious SARS-CoV-2 in cell culture assays suggests slow natural degradation processes. This study provides critical insights for future considerations in managing mass burial scenarios during outbreaks of livestock-associated zoonotic pathogens.
Collapse
Affiliation(s)
- Kristian Key Milan Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Mette Sif Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Charlotte Scheutz
- Department of Environmental and Resource Engineering, Technical University of Denmark, Bygningstorvet, Building 115, 2800 Kgs, Lyngby, Denmark
| | - Kasper Klintø
- Danish Veterinary and Food Administration, Stationsparken 31-33, 2600, Glostrup, Denmark
| | - Peter Kjeldsen
- Department of Environmental and Resource Engineering, Technical University of Denmark, Bygningstorvet, Building 115, 2800 Kgs, Lyngby, Denmark
| | - Lise Kirstine Kvisgaard
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Freja Broe Hjerpe
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Louise Lohse
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Thomas Bruun Rasmussen
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Lasse Dam Rasmussen
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Amalie Ehlers Bedsted
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Graham J Belsham
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Jørgen J Leisner
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark
| | - Anders Dalsgaard
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870, Frederiksberg C., Denmark.
| |
Collapse
|
169
|
van Doremalen N, Bushmaker T, Fischer RJ, Okumura A, Figueroa Acosta DM, McMinn RJ, Letko M, Scott D, Saturday G, Munster VJ. Transmission dynamics of MERS-CoV in a transgenic human DPP4 mouse model. NPJ VIRUSES 2024; 2:36. [PMID: 40295821 PMCID: PMC11721671 DOI: 10.1038/s44298-024-00048-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/20/2024] [Indexed: 04/30/2025]
Abstract
Since 2002, three novel coronavirus outbreaks have occurred: severe acute respiratory syndrome coronavirus (SARS-CoV-1), Middle East respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2. A better understanding of the transmission potential of coronaviruses will result in adequate infection control precautions and an early halt of transmission within the human population. Experiments on the stability of coronaviruses in the environment, as well as transmission models, are thus pertinent.Here, we show that transgenic mice expressing human DPP4 can be infected with MERS-CoV via the aerosol route. Exposure to 5 × 106 TCID50 and 5 × 104 TCID50 MERS-CoV per cage via fomites resulted in transmission in 15 out of 20 and 11 out of 18 animals, respectively. Exposure of sentinel mice to donor mice one day post inoculation with 105 TCID50 MERS-CoV resulted in transmission in 1 out of 38 mice via direct contact and 4 out of 54 mice via airborne contact. Exposure to donor mice inoculated with 104 TCID50 MERS-CoV resulted in transmission in 0 out of 20 pairs via direct contact and 0 out of 5 pairs via the airborne route. Our model shows limited transmission of MERS-CoV via the fomite, direct contact, and airborne routes. The hDPP4 mouse model will allow assessment of the ongoing evolution of MERS-CoV in the context of acquiring enhanced human-to-human transmission kinetics and will inform the development of other transmission models.
Collapse
Affiliation(s)
- Neeltje van Doremalen
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Trenton Bushmaker
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Robert J Fischer
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Atsushi Okumura
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Dania M Figueroa Acosta
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebekah J McMinn
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Michael Letko
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Dana Scott
- Division of Intramural Research, Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Greg Saturday
- Division of Intramural Research, Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Division of Intramural Research, Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
170
|
Länsivaara A, Lehto KM, Hyder R, Janhonen ES, Lipponen A, Heikinheimo A, Pitkänen T, Oikarinen S. Comparison of Different Reverse Transcriptase-Polymerase Chain Reaction-Based Methods for Wastewater Surveillance of SARS-CoV-2: Exploratory Study. JMIR Public Health Surveill 2024; 10:e53175. [PMID: 39158943 PMCID: PMC11369532 DOI: 10.2196/53175] [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: 09/28/2023] [Revised: 04/09/2024] [Accepted: 05/30/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Many countries have applied the wastewater surveillance of the COVID-19 pandemic to their national public health monitoring measures. The most used methods for detecting SARS-CoV-2 in wastewater are quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) and reverse transcriptase-droplet digital polymerase chain reaction (RT-ddPCR). Previous comparison studies have produced conflicting results, thus more research on the subject is required. OBJECTIVE This study aims to compare RT-qPCR and RT-ddPCR for detecting SARS-CoV-2 in wastewater. It also aimed to investigate the effect of changes in the analytical pipeline, including the RNA extraction kit, RT-PCR kit, and target gene assay, on the results. Another aim was to find a detection method for low-resource settings. METHODS We compared 2 RT-qPCR kits, TaqMan RT-qPCR and QuantiTect RT-qPCR, and RT-ddPCR based on sensitivity, positivity rates, variability, and correlation of SARS-CoV-2 gene copy numbers in wastewater to the incidence of COVID-19. Furthermore, we compared 2 RNA extraction methods, column- and magnetic-bead-based. In addition, we assessed 2 target gene assays for RT-qPCR, N1 and N2, and 2 target gene assays for ddPCR N1 and E. Reverse transcription strand invasion-based amplification (RT-SIBA) was used to detect SARS-CoV-2 from wastewater qualitatively. RESULTS Our results indicated that the most sensitive method to detect SARS-CoV-2 in wastewater was RT-ddPCR. It had the highest positivity rate (26/30), and its limit of detection was the lowest (0.06 gene copies/µL). However, we obtained the best correlation between COVID-19 incidence and SARS-CoV-2 gene copy number in wastewater using TaqMan RT-qPCR (correlation coefficient [CC]=0.697, P<.001). We found a significant difference in sensitivity between the TaqMan RT-qPCR kit and the QuantiTect RT-qPCR kit, the first having a significantly lower limit of detection and a higher positivity rate than the latter. Furthermore, the N1 target gene assay was the most sensitive for both RT-qPCR kits, while no significant difference was found between the gene targets using RT-ddPCR. In addition, the use of different RNA extraction kits affected the result when the TaqMan RT-qPCR kit was used. RT-SIBA was able to detect SARS-CoV-2 RNA in wastewater. CONCLUSIONS As our study, as well as most of the previous studies, has shown RT-ddPCR to be more sensitive than RT-qPCR, its use in the wastewater surveillance of SARS-CoV-2 should be considered, especially if the amount of SARS-CoV-2 circulating in the population was low. All the analysis steps must be optimized for wastewater surveillance as our study showed that all the analysis steps including the compatibility of the RNA extraction, the RT-PCR kit, and the target gene assay influence the results. In addition, our study showed that RT-SIBA could be used to detect SARS-CoV-2 in wastewater if a qualitative result is sufficient.
Collapse
Affiliation(s)
- Annika Länsivaara
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi-Maarit Lehto
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Rafiqul Hyder
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Anssi Lipponen
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, Kuopio, Finland
| | - Annamari Heikinheimo
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Finnish Food Authority - Ruokavirasto, Seinäjoki, Finland
| | - Tarja Pitkänen
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, Kuopio, Finland
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
171
|
Ramos BD, Hudson NR, Gonzales DE, Brown AN, White MG, Browde RJ, McNeary-Garvin AM, Knight CE, Pham KC, Sweatt RJ, Phan LM, Ly E, Garcia AR. Evaluation of Self-collected Saliva Samples Without Viral Transport Media for SARS-CoV-2 Testing via RT-PCR and Comparison of Amplicon Sequences Against Commonly Used Primers in Diagnostic Assays. Mil Med 2024; 189:184-189. [PMID: 39160794 DOI: 10.1093/milmed/usae070] [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: 11/24/2023] [Revised: 01/26/2024] [Accepted: 02/16/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Mass screening for SARS-CoV-2 using nasopharyngeal swabs (NPS) is costly, uncomfortable for patients, and increases the chance of virus exposure to health care workers. Therefore, this study focused on determining if self-collected unpreserved saliva can be an effective alternative to NPS collection in COVID-19 surveillance. MATERIALS AND METHODS In this study, patients being tested for SARS-CoV-2 using NPS were asked to provide a saliva sample to compare their results. NPS samples were evaluated for SARS-CoV-2 using BioFire® FilmArray® Torch® or Cepheid® GeneXpert® systems while saliva samples were evaluated using an in-house developed reverse transcriptase polymerase chain reaction (RT-PCR) which targeted the Envelope (E) and Nucleocapsid (N) genes. RESULTS Detection of SARS-CoV-2 using self-collected saliva was found to be only slightly less accurate (<5%) than testing using NPS. In addition, initial saliva RT-PCR identified 27 positive subjects, 18 of which provided amplicons sufficient for confirmatory sequencing. The sequencing data showed a genetic shift in the virus within our population sometime between 22 June and July 8, 2021 from Alpha to Delta variant. CONCLUSIONS The saliva sample collection method identifies the E gene in SARS COVID-2 samples which provides an alternative specimen source to the NPS. This identifies the S gene and ORF1ab. Saliva collection is more convenient to the patient, yields comparable results to NPS collection, and potentially increases Covid-19 surveillance.
Collapse
Affiliation(s)
- Benjamin D Ramos
- Department of Pathology, 60th Medical Group (MDG), Travis Air Force Base (AFB), Fairfield, CA 94535, USA
| | - N Ryan Hudson
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Diane E Gonzales
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Ashleigh N Brown
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Matthew G White
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Ryan J Browde
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | | | - Celynn E Knight
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Kevin C Pham
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Robert J Sweatt
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Liem Minh Phan
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Eileen Ly
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| | - Andrew R Garcia
- Clinical Investigation Facility, 60MDG, Fairfield, CA 94535, USA
| |
Collapse
|
172
|
Vu Manh TP, Gouin C, De Wolf J, Jouneau L, Pascale F, Bevilacqua C, Ar Gouilh M, Da Costa B, Chevalier C, Glorion M, Hannouche L, Urien C, Estephan J, Magnan A, Le Guen M, Marquant Q, Descamps D, Dalod M, Schwartz-Cornil I, Sage E. SARS-CoV2 infection in whole lung primarily targets macrophages that display subset-specific responses. Cell Mol Life Sci 2024; 81:351. [PMID: 39147987 PMCID: PMC11335275 DOI: 10.1007/s00018-024-05322-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 08/17/2024]
Abstract
Deciphering the initial steps of SARS-CoV-2 infection, that influence COVID-19 outcomes, is challenging because animal models do not always reproduce human biological processes and in vitro systems do not recapitulate the histoarchitecture and cellular composition of respiratory tissues. To address this, we developed an innovative ex vivo model of whole human lung infection with SARS-CoV-2, leveraging a lung transplantation technique. Through single-cell RNA-seq, we identified that alveolar and monocyte-derived macrophages (AMs and MoMacs) were initial targets of the virus. Exposure of isolated lung AMs, MoMacs, classical monocytes and non-classical monocytes (ncMos) to SARS-CoV-2 variants revealed that while all subsets responded, MoMacs produced higher levels of inflammatory cytokines than AMs, and ncMos contributed the least. A Wuhan lineage appeared to be more potent than a D614G virus, in a dose-dependent manner. Amidst the ambiguity in the literature regarding the initial SARS-CoV-2 cell target, our study reveals that AMs and MoMacs are dominant primary entry points for the virus, and suggests that their responses may conduct subsequent injury, depending on their abundance, the viral strain and dose. Interfering on virus interaction with lung macrophages should be considered in prophylactic strategies.
Collapse
Affiliation(s)
- Thien-Phong Vu Manh
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France.
| | - Carla Gouin
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Julien De Wolf
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Luc Jouneau
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Florentina Pascale
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Claudia Bevilacqua
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Meriadeg Ar Gouilh
- Department of Virology, Univ Caen Normandie, Dynamicure INSERM UMR 1311, CHU Caen, 14000, Caen, France
| | - Bruno Da Costa
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | | | - Matthieu Glorion
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Laurent Hannouche
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Céline Urien
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Jérôme Estephan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Antoine Magnan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
| | - Morgan Le Guen
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Anesthesiology, Foch Hospital, 92150, Suresnes, France
| | - Quentin Marquant
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
- Delegation to Clinical Research and Innovation, Foch Hospital, 92150, Suresnes, France
| | - Delphyne Descamps
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Marc Dalod
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
| | | | - Edouard Sage
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| |
Collapse
|
173
|
D’Amato M, Grignano MA, Iadarola P, Rampino T, Gregorini M, Viglio S. The Impact of Serum/Plasma Proteomics on SARS-CoV-2 Diagnosis and Prognosis. Int J Mol Sci 2024; 25:8633. [PMID: 39201322 PMCID: PMC11354567 DOI: 10.3390/ijms25168633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
While COVID-19's urgency has diminished since its emergence in late 2019, it remains a significant public health challenge. Recent research reveals that the molecular intricacies of this virus are far more complex than initially understood, with numerous post-translational modifications leading to diverse proteoforms and viral particle heterogeneity. Mass spectrometry-based proteomics of patient serum/plasma emerges as a promising complementary approach to traditional diagnostic methods, offering insights into SARS-CoV-2 protein dynamics and enhancing understanding of the disease and its long-term consequences. This article highlights key findings from three years of pandemic-era proteomics research. It delves into biomarker discovery, diagnostic advancements, and drug development efforts aimed at monitoring COVID-19 onset and progression and exploring treatment options. Additionally, it examines global protein abundance and post-translational modification profiling to elucidate signaling pathway alterations and protein-protein interactions during infection. Finally, it explores the potential of emerging multi-omics analytic strategies in combatting SARS-CoV-2.
Collapse
Affiliation(s)
- Maura D’Amato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.D.); (S.V.)
| | - Maria Antonietta Grignano
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (M.A.G.); (T.R.); (M.G.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (M.A.G.); (T.R.); (M.G.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Marilena Gregorini
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (M.A.G.); (T.R.); (M.G.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Simona Viglio
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.D.); (S.V.)
- Lung Transplantation Unit, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| |
Collapse
|
174
|
Wang Z, He Y, He Z, Guo Y, Zhao Y, Zhang Y. Development of highly adaptable RT-PCR methods for identifying Delta and BA.1 variants in inactivated COVID-19 vaccines. Mol Biol Rep 2024; 51:892. [PMID: 39110319 DOI: 10.1007/s11033-024-09799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/16/2024] [Indexed: 02/06/2025]
Abstract
Background The emergence and rapid spread of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), poses a significant threat to human health and public safety. While next-generation sequencing (NGS) is capable of detecting and tracking new COVID-19 variants for disease diagnosis and prevention, its high cost and time-consuming nature limit its widespread use. In this study, our aim was to develop a highly adaptable and accurate RT-PCR method for identifying the Delta or BA.1 variants in inactivated COVID-19 vaccine. We devised three two-plex RT-PCR methods targeting specific mutation sites: S: Δ156-157, S: N211-, L212I, and S: Δ142-144, Y145D. The RT-PCR method targeting the S: Δ156-157 mutation site was able to distinguish the Delta variant from other COVID-19 virus strains, while the RT-PCR methods targeting the S: N211-, L212I or S: Δ142-144, Y145D mutation sites were able to distinguish the BA.1 variant from other COVID-19 virus strains. We separately validated these three two-plex RT-PCR methods, and the results demonstrated good linearity, repeatability, reproducibility, and specificity for each method. Moreover, all three methods can be applied in the production of SARS-CoV-2 variant inactivated vaccines, enabling the identification of Delta or BA.1 variants in virus cultures as well as in inactivated vaccine stocks. This study presents a systematic approach to identify COVID-19 variants using multiple RT-PCR methods. We successfully developed three two-plex RT-PCR methods that can identify Delta and BA.1 variants based on specific mutation sites, and we completed the validation of these three methods.
Collapse
Affiliation(s)
- Zhanhui Wang
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China
| | - Yao He
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China
| | - Zhenyu He
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China
| | - Yancen Guo
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China
| | - Yuxiu Zhao
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China.
| | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing, 100176, China.
| |
Collapse
|
175
|
Liu S, Chai X, Liu C, Bai J, Meng J, Tian H, Han X, Han G, Li Q, Xu X. Sensitivity analysis of RT-qPCR and RT-ddPCR for SARS-CoV-2 detection with mutations on N1 and E primer-probe region. Microbiol Spectr 2024; 12:e0429223. [PMID: 38916349 PMCID: PMC11302302 DOI: 10.1128/spectrum.04292-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/16/2024] [Indexed: 06/26/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an RNA virus that undergoes rapid mutation. Based on viral whole genome sequencing analysis in Hebei Province, China, we identified several essential single nucleotide variants (SNVs) on primer-probe regions accumulating within some Omicron variants' genomes. In this study, we focused on three SNVs, C28290T, T28297C, and C28311T emerging on 2019-nCoV-N1 (CDC-N1) primer-probe regions, recommended by CDC in 2020, and two SNVs, C26270T, A26275G emerging on E (Charité-E) primer-probe regions recommended by Charité, Germany. Our findings revealed that the presence of one or two SNVs in the primer or probe region affected the sensitivity of reverse transcription-quantitative polymerase chain reaction and droplet digital PCR to varying extents. This discovery underscores the importance of continuously monitoring the whole genome sequences of SARS-CoV-2 variants, especially the primer-probe targeting regions, and correspondingly updating commercial test kits or recommended primer-probe sequence sets. IMPORTANCE The emergence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has resulted in a growing number of mutations in its genome, presenting new challenges for the diagnosis of SARS-CoV-2 using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and droplet digital PCR (RT-ddPCR) methods. There is an urgent need to develop refined methods for modifying primers and probes to improve the detection of these emerging variants. In this study, our focus was on the SNVs that have emerged in the CDC-N1 and Charité-E primer-probe regions. Our research has confirmed that the presence of these SNVs in the primer or probe region can significantly affect the results of coronavirus disease 2019 tests. we have developed and validated a modified detection method that can provide higher sensitivity and specificity. This study emphasizes the importance of refining the primer-probe sets to ensure the diagnostic accuracy of RT-qPCR and RT-ddPCR detection.
Collapse
Affiliation(s)
- Shiyou Liu
- Hebei Key Laboratory of Pathogens and Epidemiology of Infectious Diseases, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xiaoru Chai
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Chao Liu
- Shijiazhuang Qiaodong Sewage Treatment Plant, Shijiazhuang, China
| | - Jiaxuan Bai
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Juntao Meng
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Hong Tian
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xu Han
- Hebei Key Laboratory of Pathogens and Epidemiology of Infectious Diseases, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China
| | - Guangyue Han
- Hebei Key Laboratory of Pathogens and Epidemiology of Infectious Diseases, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China
| | - Qi Li
- Hebei Key Laboratory of Pathogens and Epidemiology of Infectious Diseases, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xiangdong Xu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| |
Collapse
|
176
|
Ndiaye D, Diatta G, Bassene H, Cortaredona S, Sambou M, Ndiaye AJS, Bedotto-Buffet M, Edouard S, Mediannikov O, Sokhna C, Fenollar F. Prevalence of Respiratory Pathogens in Nasopharyngeal Swabs of Febrile Patients with or without Respiratory Symptoms in the Niakhar Area of Rural Senegal. Pathogens 2024; 13:655. [PMID: 39204255 PMCID: PMC11357141 DOI: 10.3390/pathogens13080655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Acute respiratory tract infections are one of the leading causes of morbidity and mortality worldwide. More data are needed on circulating respiratory microorganisms in different geographical areas and ecosystems. We analyzed nasopharyngeal swabs from 500 febrile patients living in the Niakhar area (Senegal), using FTDTM multiplex qPCR and simplex qPCR to target a panel of 25 microorganisms. We detected at least one microorganism for 366/500 patients (73.2%), at least one virus for 193/500 (38.6%), and at least one bacterium for 324/500 (64.8%). The most frequently detected microorganisms were Streptococcus pneumoniae (36.8%), Haemophilus influenzae (35.8%), adenovirus (11.8%), influenza viruses (6.4%), rhinovirus (5.0%), SARS-CoV-2 (4.0%), and RSV (4.0%). The main microorganisms significantly associated with respiratory symptoms, with a p-value ≤ 0.05, were influenza virus (11.9% in patients with respiratory symptoms versus 2.9% in patients without), RSV (6.5% versus 2.6%), metapneumovirus (5.4% versus 1.3%), HPIVs (7.6% versus 1.0%), S. pneumoniae (51.9% versus 28.0%), and H. influenzae (54.6% versus 24.5%). Co-infections were significantly associated with respiratory symptoms (65.4% versus 32.9%). All the epidemiological data show a high level of circulation of respiratory pathogens among febrile patients, including those preventable by vaccination such as S. pneumoniae, raising the question of the serotypes currently circulating. Furthermore, the availability of affordable real-time etiological diagnostic tools would enable management to be adapted as effectively as possible.
Collapse
Affiliation(s)
- Dame Ndiaye
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
- EMR MINES, Campus Commun UCAD-IRD of Hann, IRD, Dakar 1386, Senegal; (G.D.); (H.B.); (M.S.)
| | - Georges Diatta
- EMR MINES, Campus Commun UCAD-IRD of Hann, IRD, Dakar 1386, Senegal; (G.D.); (H.B.); (M.S.)
| | - Hubert Bassene
- EMR MINES, Campus Commun UCAD-IRD of Hann, IRD, Dakar 1386, Senegal; (G.D.); (H.B.); (M.S.)
| | - Sébastien Cortaredona
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
- Campus Santé Timone, Aix Marseille University, IRD, MINES, 13005 Marseille, France
| | - Masse Sambou
- EMR MINES, Campus Commun UCAD-IRD of Hann, IRD, Dakar 1386, Senegal; (G.D.); (H.B.); (M.S.)
| | - Anna Julienne Selbe Ndiaye
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
| | | | - Sophie Edouard
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
| | - Oleg Mediannikov
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
- Campus Santé Timone, Aix Marseille University, AP-HM, MEPHI, 13005 Marseille, France
- IRD, 13002 Marseille, France
| | - Cheikh Sokhna
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
- EMR MINES, Campus Commun UCAD-IRD of Hann, IRD, Dakar 1386, Senegal; (G.D.); (H.B.); (M.S.)
| | - Florence Fenollar
- Campus Santé Timone, Aix Marseille University, AP-HM, SSA, RITMES, 13005 Marseille, France; (D.N.); (S.C.); (A.J.S.N.); (S.E.); (C.S.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France;
| |
Collapse
|
177
|
Sandoval-Ramírez CM, Ballesteros N, Pinilla JC, Hernández C, Muñoz M, Ramírez JD. SARS-CoV-2 Mu variant in dogs visiting veterinary clinics during the third pandemic peak in Eastern Colombia. Vet Res Commun 2024; 48:2657-2662. [PMID: 38598117 DOI: 10.1007/s11259-024-10374-4] [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: 02/13/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024]
Abstract
The global presence of SARS-CoV-2 in household pets is acknowledged, yet documentation remains scarce, leaving many regions unexplored. Thus, our study sought to fill this gap by investigating SARS-CoV-2 presence in dogs visiting veterinary clinics during the third pandemic peak in eastern Colombia. We collected and analyzed 43 oropharyngeal and rectal swabs using real-time PCR assays targeting the Envelope Gene of SARS-CoV-2. Out of these, two dogs tested positive, indicating an infection rate of 4.7%. Further examination through complete sequencing and phylogenetic analysis revealed the lineage B.1.621 for the SARS-CoV-2 genome. Consequently, our study unveils the first documented cases of Canis lupus familiaris infected with the Mu variant of SARS-CoV-2, the variant with the most death burden during the whole pandemic in Colombia. Remarkably, these cases presented mild and reversible respiratory and gastrointestinal symptoms, or no clinical manifestations at all. This sheds light on the virus's interaction with our four-legged companions, offering valuable insights into its transmission dynamics and potential effects on animal health.
Collapse
Affiliation(s)
- Claudia Magaly Sandoval-Ramírez
- Facultad de Ciencias Exactas, Naturales y Agropecuarias, Grupo de Investigación en Ciencias Básicas y Aplicadas para la Sostenibilidad (CIBAS), Universidad de Santander, Calle 70 N° 55-210, Bucaramanga, Santander, Colombia.
| | - Nathalia Ballesteros
- Centro de Investigaciones en Microbiología y Biotecnología (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan Carlos Pinilla
- Facultad de Ciencias Exactas, Naturales y Agropecuarias, Grupo de Investigación en Ciencias Agropecuarias (GICA), Universidad de Santander, Bucaramanga, Santander, Colombia
| | - Carolina Hernández
- Centro de Investigaciones en Microbiología y Biotecnología (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Centro de Tecnología en Salud (CETESA), Innovaseq SAS, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
178
|
Kantonen JN, Kuivanen S, Smura T, Puttonen H, Kekäläinen E, Sajantila A, Myllykangas L, Kantele A, Vapalahti O, Mäyränpää MI, Carpén O. Infective SARS-CoV-2 in Skull Sawdust at Autopsy, Finland. Emerg Infect Dis 2024; 30:1735-1737. [PMID: 39043418 PMCID: PMC11286058 DOI: 10.3201/eid3008.240145] [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: 07/25/2024] Open
Abstract
We assessed the distribution of SARS-CoV-2 at autopsy in 22 deceased persons with confirmed COVID-19. SARS-CoV-2 was found by PCR (2/22, 9.1%) and by culture (1/22, 4.5%) in skull sawdust, suggesting that live virus is present in tissues postmortem, including bone. Occupational exposure risk is low with appropriate personal protective equipment.
Collapse
|
179
|
Shirani K, Hajihashemi M, Mortazavi A, Assadi A, Baradaran A, Ataei B, Badei H. Diagnostic value of antibody testing in comparison with lung scan and PCR in patients suspected of having COVID-19. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:509-514. [PMID: 39267938 PMCID: PMC11389766 DOI: 10.18502/ijm.v16i4.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Background and Objectives SARS-CoV-2 is a newly discovered viral infection. It's still unclear how antibodies react in infected individuals, and there is not enough evidence to support the clinical use of antibody examination. This study evaluates the diagnostic value of serologic tests for diagnosing COVID-19. Materials and Methods 32 patients for whom serologic testing was performed within 7 to 21 days from symptom onset and whether they were diagnosed with COVID-19 by both PCR and lung HRCT as gold standard tests at the same time, were included in the study. Results Serologic tests (IgM / IgG) compared to PCR and lung HRCT scan to diagnose COVID-19, were 89.3% specific and 59.6% sensitive. Positive predictive value (PPV) was 95% and negative predictive value (NPV) was 37%. The diagnostic accuracy index of the serologic test was 0.745 (CI 0.651-0.838) (p-value <0.001). Conclusion Serologic testing can be a complementary alternative for SARA-CoV-2 nucleic acid RT-PCR, although it cannot replace it completely. IgG/IgM combo test kits and RT-PCR together can give more insight into the diagnosis of SARS-CoV-2.
Collapse
Affiliation(s)
- Kiana Shirani
- Department of Infectious Diseases, Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Milad Hajihashemi
- Department of Infectious Diseases, Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ashkan Mortazavi
- Department of Infectious Diseases, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Assadi
- Department of Infectious Diseases, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azar Baradaran
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrooz Ataei
- Department of Infectious Diseases, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Badei
- Department of Infectious Diseases, Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
180
|
Shahbazi E, Moradi A, Mollasalehi H, Mohebbi SR. Unravelling the diagnostic methodologies for SARS-CoV-2; the Indispensable need for developing point-of-care testing. Talanta 2024; 275:126139. [PMID: 38696900 DOI: 10.1016/j.talanta.2024.126139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/13/2024] [Accepted: 04/20/2024] [Indexed: 05/04/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused COVID-19 pandemic that continues to be a global menace and since its emergence in the late 2019, SARS-CoV-2 has been vigorously spreading throughout the globe putting the whole world into a multidimensional calamity. The suitable diagnosis strategies are on the front line of the battle against preventing the spread of infections. Since the clinical manifestation of COVID-19 is shared between various diseases, detection of the unique impacts of the pathogen on the host along with the diagnosis of the virus itself should be addressed. Employing the most suitable approaches to specifically, sensitively and effectively recognize the infected cases may be a real game changer in controlling the outbreak and the crisis management. In that matter, point-of-care assays (POC) appears to be the potential option, due to sensitivity, specificity, affordable, and availability. Here we brief the most recent findings about the virus, its variants, and the conventional methods that have been used for its detection, along with the POC strategies that have been applied to the virus diagnosis and the developing technologies which can accelerate the diagnosis procedure yet maintain its efficiency.
Collapse
Affiliation(s)
- Erfan Shahbazi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Asma Moradi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Hamidreza Mollasalehi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Seyed Reza Mohebbi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
181
|
Essex M, Millet Pascual-Leone B, Löber U, Kuhring M, Zhang B, Brüning U, Fritsche-Guenther R, Krzanowski M, Fiocca Vernengo F, Brumhard S, Röwekamp I, Anna Bielecka A, Lesker TR, Wyler E, Landthaler M, Mantei A, Meisel C, Caesar S, Thibeault C, Corman VM, Marko L, Suttorp N, Strowig T, Kurth F, Sander LE, Li Y, Kirwan JA, Forslund SK, Opitz B. Gut microbiota dysbiosis is associated with altered tryptophan metabolism and dysregulated inflammatory response in COVID-19. NPJ Biofilms Microbiomes 2024; 10:66. [PMID: 39085233 PMCID: PMC11291933 DOI: 10.1038/s41522-024-00538-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
The clinical course of COVID-19 is variable and often unpredictable. To test the hypothesis that disease progression and inflammatory responses associate with alterations in the microbiome and metabolome, we analyzed metagenome, metabolome, cytokine, and transcriptome profiles of repeated samples from hospitalized COVID-19 patients and uninfected controls, and leveraged clinical information and post-hoc confounder analysis. Severe COVID-19 was associated with a depletion of beneficial intestinal microbes, whereas oropharyngeal microbiota disturbance was mainly linked to antibiotic use. COVID-19 severity was also associated with enhanced plasma concentrations of kynurenine and reduced levels of several other tryptophan metabolites, lysophosphatidylcholines, and secondary bile acids. Moreover, reduced concentrations of various tryptophan metabolites were associated with depletion of Faecalibacterium, and tryptophan decrease and kynurenine increase were linked to enhanced production of inflammatory cytokines. Collectively, our study identifies correlated microbiome and metabolome alterations as a potential contributor to inflammatory dysregulation in severe COVID-19.
Collapse
Affiliation(s)
- Morgan Essex
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Belén Millet Pascual-Leone
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mathias Kuhring
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Core Unit Bioinformatics, Berlin, Germany
| | - Bowen Zhang
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ulrike Brüning
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
| | | | - Marta Krzanowski
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Facundo Fiocca Vernengo
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Röwekamp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Christian Meisel
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sandra Caesar
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor M Corman
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Lajos Marko
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Till Strowig
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Yang Li
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Jennifer A Kirwan
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- University of Nottingham School of Veterinary Medicine and Science, Loughborough, UK
| | - Sofia K Forslund
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany.
- German Center for Lung Research (DZL), Berlin, Germany.
| |
Collapse
|
182
|
Lopes TRR, Silva Júnior JVJ, Trindade PDA, Gregianini TS, Weiblen R, Flores EF. An end-point multiplex RT-PCR for SARS-CoV-2, Influenza A and B detection, including simultaneous RNAse P amplification: a timely tool for more accessible differential diagnosis. J Med Microbiol 2024; 73. [PMID: 39140993 DOI: 10.1099/jmm.0.001868] [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: 08/15/2024] Open
Abstract
The multiplex molecular diagnostic assays described for severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2), influenza A (IAV) and B (IBV) viruses have been mainly based on real-time reaction, which limits their access to many laboratories or diagnostic institutions. To contribute to available strategies and expand access to differential diagnosis, we describe an end-point multiplex RT-PCR targeting SARS-CoV-2, IAV and IBV with simultaneous endogenous control amplification. Initially, we looked for well-established primers sets for SARS-CoV-2, IAV, IBV and RNAse P whose amplicons could be distinguished on agarose gel. The multiplex assay was then standardized by optimizing the reaction mix and cycle conditions. The limit of detection (LoD) was determined using titrated viruses (for SARS-CoV-2 and IAV) and by dilution from a pool of IBV-positive samples. The diagnostic performance of the multiplex was evaluated by testing samples with different RNAse P and viral loads, previously identified as positive or negative for the target viruses. The amplicons of IAV (146 bp), SARS-CoV-2 (113 bp), IBV (103 bp) and RNAse P (65 bp) were adequately distinguished in our multiplex. The LoD for SARS-CoV-2, IAV and IBV was 0.02 TCID50/ml, 0.07 TCID50/ml and 10-3 from a pool of positive samples, respectively. All samples positive for SARS-CoV-2 (n=70, Ct 17.2-36.9), IAV (n=53, Ct 14-34.9) and IBV (n=12, Ct 23.9-31.9) remained positive in our multiplex assay. RNAse P from negative samples (n=40, Ct 25.2-30.2) was also amplified in the multiplex. Overall, our assay is a timely and alternative tool for detecting SARS-CoV-2 and influenza viruses in laboratories with limited access to supplies/equipment.
Collapse
Affiliation(s)
- Thaísa Regina Rocha Lopes
- Programa de Pós-graduação em Medicina Veterinária, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - José Valter Joaquim Silva Júnior
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
- Setor de Virologia, Instituto Keizo Asami, Universidade Federal de Pernambuco, Pernambuco, Brazil
- Laboratório NB3 de Neuroimunologia, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
- Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - Priscila de Arruda Trindade
- Laboratório de Biologia Molecular e Bioinformática Aplicada à Microbiologia Clínica, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - Tatiana Schäffer Gregianini
- Laboratório Central de Saúde, Centro Estadual de Vigilância em Saúde da Secretaria de Saúde do estado do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Rudi Weiblen
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - Eduardo Furtado Flores
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
183
|
Ding X, Wang Y, Gui Y, Yang C. Two-Stage Mixed-Dye-Based Isothermal Amplification with Ribonuclease-Cleavable Enhanced Probes for Dual-Visualization Detection of SARS-CoV-2 Variants of Interest. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401988. [PMID: 38829265 PMCID: PMC11304323 DOI: 10.1002/advs.202401988] [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] [Received: 02/25/2024] [Revised: 05/17/2024] [Indexed: 06/05/2024]
Abstract
Rapid and visual detection of SARS-CoV-2 variants is vital for timely assessment of variant transmission in resource-limited settings. Here, a closed-tube, two-stage, mixed-dye-based isothermal amplification method with ribonuclease-cleavable enhanced probes (REP), termed REP-TMAP, for dual-visualization detection of SARS-CoV-2 variants including JN.1, BA.2, BA.4/5, and Delta is introduced. The first stage of REP-TMAP is reverse transcription recombinase polymerase amplification and the second stage is dual-visualization detection synergistically mediated by the REP and the mixed dyes of cresol red and hydroxy naphthol blue. In REP-TMAP reaction, the color change under ambient light indicates SARS-CoV-2 infection, while the fluorescence change under blue light excitation specifies variant type. On detecting transcribed RNA of SARS-CoV-2 spike gene, this assay is rapid (within 40 min), highly sensitive (10-200 copies per reaction), and highly specific (identification of single-base mutations). Furthermore, the assay has been clinically validated to accurately detect JN.1, BA.2, and BA.4/5 variants from 102 human oropharyngeal swabs. The proposed assay therefore holds great potentials to provide a rapid, dual-visualization, sensitive, specific, point-of-care detection of SARS-CoV-2 variants and beyond.
Collapse
Affiliation(s)
- Xiong Ding
- Key Laboratory of Environmental Medicine and EngineeringMinistry of EducationDepartment of Nutrition and Food HygieneSchool of Public Health, Southeast UniversityNanjing210009P. R. China
| | - Yaru Wang
- Key Laboratory of Environmental Medicine and EngineeringMinistry of EducationDepartment of Nutrition and Food HygieneSchool of Public Health, Southeast UniversityNanjing210009P. R. China
| | - Yuxin Gui
- Key Laboratory of Environmental Medicine and EngineeringMinistry of EducationDepartment of Nutrition and Food HygieneSchool of Public Health, Southeast UniversityNanjing210009P. R. China
| | - Chuankun Yang
- Center of Clinical Laboratory MedicineZhongda Hospital, Southeast UniversityNanjing210009P. R. China
| |
Collapse
|
184
|
Sibounheuang B, Boutthasavong L, Chommanam D, Phommasone K, Panapruksachat S, Praphasiri V, Bouttavong S, Sisavath H, Christy NCV, Letizia AG, Mayxay M, Vongsouvath M, Ashley EA, Dubot-Pérès A. Dry Swabs and Dried Saliva as Alternative Samples for SARS-CoV-2 Detection in Remote Areas in Lao PDR. Open Forum Infect Dis 2024; 11:ofae433. [PMID: 39145142 PMCID: PMC11322834 DOI: 10.1093/ofid/ofae433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Background Surveillance of SARS-CoV-2 circulation is mainly based on real-time reverse transcription-polymerase chain reaction, which requires laboratory facilities and cold chain for sample transportation. This is difficult to achieve in remote rural areas of resource-limited settings. The use of dried blood spots shipped at room temperature has shown good efficiency for the detection of arboviral RNA. Using a similar approach, we conducted a study at 3 provincial hospitals in Laos to compare the detection of SARS-CoV-2 from neat and dried spot samples. Methods Between January 2022 and March 2023, patients with respiratory symptoms were recruited. Nasopharyngeal/oropharyngeal swabs in virus transport medium (VTM), dry swabs, saliva, and dried saliva spotted on filter paper were collected. All samples were tested by SARS-CoV-2 real-time reverse transcription-polymerase chain reaction. Results In total, 479 participants were included. The VTM samples tested positive for 288 (60.1%). High positive percent agreements were observed for dry swab (84.8%; 95% CI, 80.2%-88.8%) and saliva (89.2%; 95% CI, 85.1%-92.6%) as compared with VTM. There was a loss of sensitivity when saliva was dried on filter paper (73.6%; 95% CI, 68.1%-78.6%) as compared with saliva. SARS-CoV-2 variant (Delta or Omicron) had no significant impact on the performance of the different sample types. Conclusions Our findings suggest that dry swabs could be a good alternative for sample collection and permit easy shipping at ambient temperature for subsequent viral SARS-CoV-2 RNA purification and molecular investigation. This is a useful tool to consider for a rapid implementation of large-scale surveillance of SARS-CoV-2 in remote areas, which could be extrapolated to other respiratory targets during routine surveillance or in the case of a novel emerging pandemic.
Collapse
Affiliation(s)
- Bountoy Sibounheuang
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | - Latsaniphone Boutthasavong
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | - Danoy Chommanam
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | - Koukeo Phommasone
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | - Siribun Panapruksachat
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | | | | | | | - Nathaniel C V Christy
- Naval Medical Research Unit INDO PACIFIC, Emerging Infectious Disease Department, Singapore, Singapore
| | - Andrew G Letizia
- Naval Medical Research Unit INDO PACIFIC, Emerging Infectious Disease Department, Singapore, Singapore
| | - Mayfong Mayxay
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
- Institute of Research and Education Development, University of Health Sciences, Vientiane, Lao PDR
| | - Manivanh Vongsouvath
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
| | - Elizabeth A Ashley
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Audrey Dubot-Pérès
- Microbiology Laboratory, Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Lao PDR
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), Marseille, France
| |
Collapse
|
185
|
Cabrera LE, Jokiranta ST, Mäki S, Miettinen S, Kant R, Kareinen L, Sironen T, Pietilä JP, Kantele A, Kekäläinen E, Lindgren H, Mattila P, Kipar A, Vapalahti O, Strandin T. The assembly of neutrophil inflammasomes during COVID-19 is mediated by type I interferons. PLoS Pathog 2024; 20:e1012368. [PMID: 39172744 PMCID: PMC11340896 DOI: 10.1371/journal.ppat.1012368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/24/2024] [Indexed: 08/24/2024] Open
Abstract
The severity of COVID-19 is linked to excessive inflammation. Neutrophils represent a critical arm of the innate immune response and are major mediators of inflammation, but their role in COVID-19 pathophysiology remains poorly understood. We conducted transcriptomic profiling of neutrophils obtained from patients with mild and severe COVID-19, as well as from SARS-CoV-2 infected mice, in comparison to non-infected healthy controls. In addition, we investigated the inflammasome formation potential in neutrophils from patients and mice upon SARS-CoV-2 infection. Transcriptomic analysis of polymorphonuclear cells (PMNs), consisting mainly of mature neutrophils, revealed a striking type I interferon (IFN-I) gene signature in severe COVID-19 patients, contrasting with mild COVID-19 and healthy controls. Notably, low-density granulocytes (LDGs) from severe COVID-19 patients exhibited an immature neutrophil phenotype and lacked this IFN-I signature. Moreover, PMNs from severe COVID-19 patients showed heightened nigericin-induced caspase1 activation, but reduced responsiveness to exogenous inflammasome priming. Furthermore, IFN-I emerged as a priming stimulus for neutrophil inflammasomes. These findings suggest a potential role for neutrophil inflammasomes in driving inflammation during severe COVID-19. Altogether, these findings open promising avenues for targeted therapeutic interventions to mitigate the pathological processes associated with the disease.
Collapse
Affiliation(s)
- Luz E. Cabrera
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
| | - Suvi T. Jokiranta
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sanna Mäki
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
| | - Simo Miettinen
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Ravi Kant
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Tropical Parasitology, Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Lauri Kareinen
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tarja Sironen
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Meilahti Vaccine Research Center MeVac, Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Anu Kantele
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Meilahti Vaccine Research Center MeVac, Department of Infectious Diseases, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eliisa Kekäläinen
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Division of Virology and Immunology, HUSLAB Clinical Microbiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Lindgren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pirkko Mattila
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anja Kipar
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Olli Vapalahti
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Division of Virology and Immunology, HUSLAB Clinical Microbiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tomas Strandin
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
186
|
Boom TT, de Hoog MLA, Westerhof I, Jaddoe V, Heuvelman VD, Fourie E, Sluiter-Post JGC, Badoux P, Euser S, Herpers B, Sanders EAM, Eggink D, Reusken C, Bont LJ, Wildenbeest JG, van Houten MA, Duijts L, Bruijning-Verhagen PCJL. Age-specific SARS-CoV-2 transmission differed from human rhinovirus in households during the early COVID-19 pandemic. J Infect 2024; 89:106218. [PMID: 38950866 DOI: 10.1016/j.jinf.2024.106218] [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: 03/21/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVES Children are generally considered main drivers of transmission for respiratory viruses, but the emergence of SARS-CoV-2 challenged this paradigm. Human rhinovirus (RV) continued to co-circulate throughout the pandemic, allowing for direct comparison of age-specific infectivity and susceptibility within households between these viruses during a time of low SARS-CoV-2 population immunity. METHODS Households with children were prospectively monitored for ≥23 weeks between August 2020 and July 2021. Upon onset of respiratory symptoms in a household, an outbreak study was initiated, including questionnaires and repeated nasal self-sampling in all household members. Swabs were tested by PCR. Age-stratified within-household secondary attack rates (SARs) were compared between SARS-CoV-2 and RV. RESULTS A total of 307 households participated, including 582 children and 627 adults. Overall, SAR was lower for SARS-CoV-2 than for RV (aOR 0.55) and age distributions differed between both viruses (p < 0.001). Following household exposure, children were significantly less likely to become infected with SARS-CoV-2 compared to RV (aOR 0.16), whereas this was opposite in adults (aOR 1.71). CONCLUSION In households, age-specific susceptibility to SARS-CoV-2 and RV differs and drives differences in household transmission between these pathogens. This highlights the importance of characterizing age-specific transmission risks, particularly for emerging infections, to guide appropriate infection control interventions.
Collapse
Affiliation(s)
- Trisja T Boom
- Julius Centre for Health Sciences and Primary Care, Department of Epidemiology, University Medical Centre Utrecht, Utrecht, the Netherlands.
| | - Marieke L A de Hoog
- Julius Centre for Health Sciences and Primary Care, Department of Epidemiology, University Medical Centre Utrecht, Utrecht, the Netherlands.
| | - Ilse Westerhof
- Julius Centre for Health Sciences and Primary Care, Department of Epidemiology, University Medical Centre Utrecht, Utrecht, the Netherlands.
| | - Vincent Jaddoe
- Erasmus MC - Sophia Children's Hospital, Erasmus University Medical Center, Department of Pediatrics, Rotterdam, the Netherlands.
| | - Valerie D Heuvelman
- Erasmus MC - Sophia Children's Hospital, Erasmus University Medical Center, Department of Pediatrics, Rotterdam, the Netherlands.
| | - Elandri Fourie
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, the Netherlands.
| | | | - Paul Badoux
- Regional Public Health Laboratory Kennemerland, Haarlem, the Netherlands.
| | - Sjoerd Euser
- Regional Public Health Laboratory Kennemerland, Haarlem, the Netherlands.
| | - Bjorn Herpers
- Regional Public Health Laboratory Kennemerland, Haarlem, the Netherlands.
| | - Elisabeth A M Sanders
- Centre for Infectious Disease Control, WHO COVID-19 Reference Laboratory, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Dirk Eggink
- Centre for Infectious Disease Control, WHO COVID-19 Reference Laboratory, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Chantal Reusken
- Centre for Infectious Disease Control, WHO COVID-19 Reference Laboratory, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Louis J Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Joanne G Wildenbeest
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Marlies A van Houten
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, the Netherlands; Spaarne Gasthuis, Department of Pediatrics, Hoofddorp, Haarlem, the Netherlands.
| | - Liesbeth Duijts
- Erasmus MC - Sophia Children's Hospital, Erasmus University Medical Center, Department of Pediatrics, Rotterdam, the Netherlands.
| | - Patricia C J L Bruijning-Verhagen
- Julius Centre for Health Sciences and Primary Care, Department of Epidemiology, University Medical Centre Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
187
|
Lehrer LW, Lewis AM, Tolliver S, Degen M, Singh R, Houser S, Rao J. An evaluation of a new rapid qPCR test for the detection of 2019-novel coronavirus nucleocapsid (N1) gene in wastewater in Roanoke and Salem VA sewersheds. JOURNAL OF WATER AND HEALTH 2024; 22:1419-1428. [PMID: 39212279 DOI: 10.2166/wh.2024.085] [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] [Received: 02/29/2024] [Accepted: 07/20/2024] [Indexed: 09/04/2024]
Abstract
The COVID-19 pandemic initiated public interest in wastewater-based epidemiology (WBE). Public and private entities responded to the need to produce timely and accurate data. LuminUltra and Hach partnered to provide a rapid, field-based quantitative polymerase chain reaction (qPCR) test for detecting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in wastewater. This study evaluates the Hach GeneCount SARS-CoV-2 Wastewater RT-qPCR Assay Kit and LuminUltra GeneCount® Q-16 RT-PCR instrument. The Hach LuminUltra methods were compared to the Promega Wizard® Enviro Total Nucleic Acid kit and Bio-Rad CFX Opus 96 Real-time PCR Detection System. Over a 12-week period, wastewater samples were collected weekly from seven locations in the Roanoke/Salem, VA sewersheds. Concentration and extraction of the viral RNA were followed by qPCR analysis. The target gene for detection was the nucleocapsid gene (N1) of the SARS-CoV-2 virus. Costs, ease of use, time to produce results, sample preparation, and data comparisons were considered. The comparison determined that the Hach LuminUltra method and instrument were more affordable, consumed less time, and required less technical expertise. While the new method was specific, it had low sensitivity. This evaluation suggests the Hach LuminUltra method should be reserved for limited situations requiring onsite field analysis where data accuracy is not essential.
Collapse
Affiliation(s)
- Lia Willow Lehrer
- Radford University Carilion (RUC), Roanoke, VA, USA; Equal first authors
| | - Anna Marie Lewis
- Radford University Carilion (RUC), Roanoke, VA, USA; Equal first authors
| | - Susan Tolliver
- Carilion Roanoke Community Hospital, Carilion Clinic Basic Science Research Lab (BSRL), Roanoke, VA, USA
| | - Marcia Degen
- Virginia Department of Health (VDH), Richmond, VA, USA
| | - Rekha Singh
- Virginia Department of Health (VDH), Richmond, VA, USA
| | - Sara Houser
- Radford University Carilion (RUC), Roanoke, VA, USA E-mail:
| | - Jayasimha Rao
- Carilion Roanoke Community Hospital, Carilion Clinic Basic Science Research Lab (BSRL), Roanoke, VA, USA; Internal Medicine, Division of Infectious Disease, Carilion Medical Center, Roanoke, VA, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA; Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, VA, USA
| |
Collapse
|
188
|
Yani H, Yuan TD, Lubis AD, Iswara LK, Lubis IN. Comparison of RT-PCR cycle threshold values between individual and pooled SARS-CoV-2 infected nasopharyngeal swab specimens. NARRA J 2024; 4:e765. [PMID: 39280312 PMCID: PMC11391988 DOI: 10.52225/narra.v4i2.765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/17/2024] [Indexed: 09/18/2024]
Abstract
The molecular reverse transcription-polymerase chain reaction (RT-PCR) testing of respiratory tract swabs has become mandatory to confirm the diagnosis of coronavirus disease 2019 (COVID-19). However, RT-PCR tests are expensive, require standardized equipment, and relatively long testing times, and the sample pooling method has been introduced to solve this issue. The aim of this study was to compare the cycle threshold (Ct) values of the individual sample and pooled sample methods to assess how accurate the pooling method was. Repeat RT-PCR examinations were initially performed to confirm the Ct values for each sample before running the pooled test procedure. Sample extraction and amplification were performed in both assays to detect ORF1ab, N, and E genes with a cut-off point value of Ct <38. Overall, there was no difference in Ct values between individual sample and pooled sample groups at all concentrations (p=0.259) and for all pooled sizes. Only pooled size of five could detect the Ct value in the pooled samples for all concentration samples, including low-concentration sample (Ct values 36 to 38). This study highlighted that pooled RT-PCR testing strategy did not reduce the quality of individually measured RT-PCR Ct values. A pool size of five could provide a practical technique to expand the screening capacity of RT-PCR.
Collapse
Affiliation(s)
- Handa Yani
- Department of Pediatric, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Toh D Yuan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| | - Aridamuriany D Lubis
- Department of Pediatric, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Lia K Iswara
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Inke Nd Lubis
- Department of Pediatric, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
189
|
Els F, Kleynhans J, Wolter N, du Plessis M, Moosa F, Tempia S, Makhasi M, Nel J, Dawood H, Meiring S, von Gottberg A, Cohen C, Walaza S. Comparing adults with severe SARS-CoV-2 or influenza infection: South Africa, 2016-2021. S Afr J Infect Dis 2024; 39:574. [PMID: 39114258 PMCID: PMC11304391 DOI: 10.4102/sajid.v39i1.574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/26/2023] [Indexed: 08/10/2024] Open
Abstract
Background Comparisons of the characteristics of individuals hospitalised with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or seasonal influenza in low-to middle-income countries with high human immunodeficiency virus (HIV) prevalence are limited. Objectives Determine the epidemiological differences with those hospitalised with influenza or SARS-CoV-2 infection. Method We investigated hospitalised individuals ≥18 years of age testing positive for seasonal influenza (2016-2019) or SARS-CoV-2 (2020-2021). We used random effects multivariable logistic regression, controlling for clustering by site, to evaluate differences among adults hospitalised with influenza or SARS-CoV-2 infection. Results Compared to individuals with influenza, individuals with SARS-CoV-2 infection were more likely to be diabetic (adjusted odds ratio [aOR]: 1.70, 95% confidence interval [CI]: 1.11-2.61) or die in hospital (aOR: 2.57, 95% CI: 1.61-4.12). Additionally, those with SARS-CoV-2 infection were less likely to be living with HIV (not immunosuppressed) (aOR: 0.50, 95% CI: 0.34-0.73) or living with HIV (immunosuppressed) (aOR: 0.27, 95% CI: 0.18-0.39) compared to not living with HIV and less likely to be asthmatic (aOR: 0.21, 95% CI: 0.13-0.33) rather than those living with influenza. Conclusion Individuals hospitalised with SARS-CoV-2 had different characteristics to individuals hospitalised with influenza before the coronavirus disease 2019 (COVID-19) pandemic. Risk factors should be considered in health management especially as we move into an era of co-circulation of SARS-CoV-2 and influenza pathogens. Contribution Identifying groups at high risk of severe disease could help to better monitor, prevent and control SARS-CoV-2 or influenza severe disease.
Collapse
Affiliation(s)
- Fiona Els
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- South African Field Epidemiology Training Programme (SAFETP), Division of Public Health, Surveillance and Response (DPHSR), National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Jackie Kleynhans
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nicole Wolter
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mignon du Plessis
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fahima Moosa
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stefano Tempia
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mvuyo Makhasi
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Jeremy Nel
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Halima Dawood
- Department of Medicine, Greys Hospital, Pietermaritzburg and Centre for the Aids programme of research in South Africa, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Susan Meiring
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sibongile Walaza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
190
|
Sharma S, Caputi M, Asghar W. Development of a Diagnostic Microfluidic Chip for SARS-CoV-2 Detection in Saliva and Nasopharyngeal Samples. Viruses 2024; 16:1190. [PMID: 39205164 PMCID: PMC11360425 DOI: 10.3390/v16081190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
The novel coronavirus SARS-CoV-2 was first isolated in late 2019; it has spread to all continents, infected over 700 million people, and caused over 7 million deaths worldwide to date. The high transmissibility of the virus and the emergence of novel strains with altered pathogenicity and potential resistance to therapeutics and vaccines are major challenges in the study and treatment of the virus. Ongoing screening efforts aim to identify new cases to monitor the spread of the virus and help determine the danger connected to the emergence of new variants. Given its sensitivity and specificity, nucleic acid amplification tests (NAATs) such as RT-qPCR are the gold standard for SARS-CoV-2 detection. However, due to high costs, complexity, and unavailability in low-resource and point-of-care (POC) settings, the available RT-qPCR assays cannot match global testing demands. An alternative NAAT, RT-LAMP-based SARS-CoV-2 detection offers scalable, low-cost, and rapid testing capabilities. We have developed an automated RT-LAMP-based microfluidic chip that combines the RNA isolation, purification, and amplification steps on the same device and enables the visual detection of SARS-CoV-2 within 40 min from saliva and nasopharyngeal samples. The entire assay is executed inside a uniquely designed, inexpensive disposable microfluidic chip, where assay components and reagents have been optimized to provide precise and qualitative results and can be effectively deployed in POC settings. Furthermore, this technology could be easily adapted for other novel emerging viruses.
Collapse
Affiliation(s)
- Sandhya Sharma
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
| | - Massimo Caputi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Waseem Asghar
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431, USA
- Department of Biological Sciences (Courtesy Appointment), Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
191
|
Stahl-Hennig C, Peter AS, Cordsmeier A, Stolte-Leeb N, Vestweber R, Socher E, Merida SA, Sauermann U, Bleyer M, Fraedrich K, Grunwald T, Winkler TH, Ensser A, Jäck HM, Überla K. Genetic barrier to resistance: a critical parameter for efficacy of neutralizing monoclonal antibodies against SARS-CoV-2 in a nonhuman primate model. J Virol 2024; 98:e0062824. [PMID: 38899895 PMCID: PMC11265388 DOI: 10.1128/jvi.00628-24] [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: 04/12/2024] [Accepted: 05/12/2024] [Indexed: 06/21/2024] Open
Abstract
The potency of antibody neutralization in cell culture has been used as the key criterion for selection of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) for clinical development. As other aspects may also influence the degree of protection in vivo, we compared the efficacy of two neutralizing monoclonal antibodies (TRES6 and 4C12) targeting different epitopes of the receptor binding domain (RBD) of SARS-CoV-2 in a prophylactic setting in rhesus monkeys. All four animals treated with TRES6 had reduced viral loads in the upper respiratory tract 2 days after naso-oropharyngeal challenge with the Alpha SARS-CoV-2 variant. Starting 2 days after challenge, mutations conferring resistance to TRES6 were dominant in two of the rhesus monkeys, with both animals failing to maintain reduced viral loads. Consistent with its lower serum neutralization titer at the day of challenge, prophylaxis with 4C12 tended to suppress viral load at day 2 less efficiently than TRES6. However, a week after challenge, mean viral loads in the lower respiratory tract in 4C12-treated animals were lower than in the TRES6 group and no mutations conferring resistance to 4C12 could be detected in viral isolates from nasal or throat swabs. Thus, genetic barrier to resistance seems to be a critical parameter for the efficacy of prophylaxis with monoclonal antibodies against SARS-CoV-2. Furthermore, comparison of antibody concentrations in respiratory secretions to those in serum shows reduced distribution of the 4C12 antibody into respiratory secretions and a delay in the appearance of antibodies in bronchoalveolar lavage fluid compared to their appearance in secretions of the upper respiratory tract.IMPORTANCEMonoclonal antibodies are a powerful tool for the prophylaxis and treatment of acute viral infections. Hence, they were one of the first therapeutic agents licensed for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Oftentimes, the main criterion for the selection of antibodies for clinical development is their potency of neutralization in cell culture. By comparing two antibodies targeting the Spike protein of SARS-CoV-2, we now observed that the antibody that neutralized SARS-CoV-2 more efficiently in cell culture suppressed viral load in challenged rhesus monkeys to a lesser extent. Extraordinary rapid emergence of mutants of the challenge virus, which had lost their sensitivity to the antibody, was identified as the major reason for the reduced efficacy of the antibody in rhesus monkeys. Therefore, the viral genetic barrier to resistance to antibodies also affects their efficacy.
Collapse
Affiliation(s)
| | - Antonia Sophia Peter
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arne Cordsmeier
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ramona Vestweber
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Eileen Socher
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ulrike Sauermann
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Martina Bleyer
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Kirsten Fraedrich
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Grunwald
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Internal Medicine III, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
192
|
Fernández-Bastit L, Cano-Terriza D, Caballero-Gómez J, Beato-Benítez A, Fernández A, García-Párraga D, Domingo M, Sierra C, Canales R, Borragan S, de la Riva-Fraga M, Molina-López R, Cabezón Ó, Puig-Ribas M, Espunyes J, Vázquez-Calero DB, Vergara-Alert J, García-Bocanegra I, Segalés J. Survey of severe acute respiratory syndrome coronavirus 2 in captive and free-ranging wildlife from Spain. Vet Res 2024; 55:90. [PMID: 39030652 PMCID: PMC11264983 DOI: 10.1186/s13567-024-01348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/24/2024] [Indexed: 07/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), considered a zoonotic agent of wildlife origin, can infect various animal species, including wildlife in free-range and captive environments. Detecting susceptible species and potential reservoirs is crucial for preventing the transmission, spread, genetic evolution, and further emergence of viral variants that are major threats to global health. This study aimed to detect exposure or acute infection by SARS-CoV-2 in 420 animals from 40 different wildlife species, including terrestrial and aquatic mammals, from different regions of Spain during the 2020-2023 coronavirus disease 19 (COVID-19) pandemic. In total, 8/137 animals were positive for SARS-CoV-2 antibodies against the receptor binding domain and/or viral nucleoprotein according to independent ELISAs. However, only one ELISA-positive sample of a captive bottlenose dolphin (Tursiops truncatus) tested positive for SARS-CoV-2 neutralizing antibodies with a low titre (SNT50 38.15) according to a virus neutralization test. Cetaceans are expected to have a high risk of infection with SARS-CoV-2 according to early predictive studies due to the similarity of their angiotensin converting enzyme 2 cell receptor to that of humans. Moreover, of 283 animals analysed for SARS-CoV-2 RNA using RT-qPCR, none tested positive. Our results reinforce the importance of considering cetaceans at risk for SARS-CoV-2 infection and support taking preventive biosecurity measures when interacting with them, especially in the presence of individuals with suspected or confirmed COVID-19. Although most animals in this study tested negative for acute infection or viral exposure, ongoing surveillance of wildlife species and potentially susceptible animals is important to prevent future spillover events and detect potential novel reservoirs.
Collapse
Affiliation(s)
- Leira Fernández-Bastit
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Bellaterra, Barcelona, Spain
| | - David Cano-Terriza
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, 14014, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Javier Caballero-Gómez
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, 14014, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Maimonides Institute for Biomedical Research of Cordoba, Reina Sofía University Hospital, University of Córdoba, 14004, Córdoba, Spain
| | - Adrián Beato-Benítez
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, 14014, Córdoba, Spain
| | - Antonio Fernández
- Atlantic Cetacean Research Center, Institute of Animal Health, University of Las Palmas de Gran Canaria, 35001, Las Palmas, Trasmontaña, Spain
| | - Daniel García-Párraga
- Research Department, Fundación Oceanografic de la Comunitat Valenciana, Ciudad de las Artes y las Ciencias, 46013, Valencia, Spain
| | - Mariano Domingo
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain
- Veterinary Pathology Diagnostic Service, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain
- Departament de Sanitat I Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Cecilia Sierra
- Selwo Aventura, 29680, Estepona, Málaga, Spain
- Selwo Marina, 29630, Benalmádena, Málaga, Spain
| | | | - Santiago Borragan
- Parque de la Naturaleza de Cabárceno, 39690, Obregón, Cantabria, Spain
| | | | - Rafael Molina-López
- Centre de Fauna de Torreferrussa, Àrea de Gestió Ambiental Servei de Fauna I Flora, Forestal Catalana, 08130, Santa Perpètua de Mogoda, Barcelona, Spain
| | - Óscar Cabezón
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain
- Wildlife Conservation Medicine Research Group (WildCoM), Departament de Medicina I Cirugia Animals, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Maria Puig-Ribas
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain
- Wildlife Conservation Medicine Research Group (WildCoM), Departament de Medicina I Cirugia Animals, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Johan Espunyes
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain
- Wildlife Conservation Medicine Research Group (WildCoM), Departament de Medicina I Cirugia Animals, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | | | - Júlia Vergara-Alert
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain.
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08193, Bellaterra, Barcelona, Spain.
| | - Ignacio García-Bocanegra
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, 14014, Córdoba, Spain.
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Joaquim Segalés
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal. Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08139, Bellaterra, Barcelona, Spain.
- Veterinary Pathology Diagnostic Service, Autonomous University of Barcelona, 08193, Bellaterra, Barcelona, Spain.
- Departament de Sanitat I Anatomia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| |
Collapse
|
193
|
Myeni SK, Leijs AA, Bredenbeek PJ, Morales ST, Linger ME, Fougeroux C, van Zanen-Gerhardt S, Zander SAL, Sander AF, Kikkert M. Protection of K18-hACE2 Mice against SARS-CoV-2 Challenge by a Capsid Virus-like Particle-Based Vaccine. Vaccines (Basel) 2024; 12:766. [PMID: 39066404 PMCID: PMC11281552 DOI: 10.3390/vaccines12070766] [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: 05/19/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The SARS-CoV-2 pandemic and the emergence of novel virus variants have had a dramatic impact on public health and the world economy, underscoring the need for detailed studies that explore the high efficacy of additional vaccines in animal models. In this study, we confirm the pathogenicity of the SARS-CoV-2/Leiden_008 isolate (GenBank accession number MT705206.1) in K18-hACE2 transgenic mice. Using this isolate, we show that a vaccine consisting of capsid virus-like particles (cVLPs) displaying the receptor-binding domain (RBD) of SARS-CoV-2 (Wuhan strain) induces strong neutralizing antibody responses and sterilizing immunity in K18-hACE2 mice. Furthermore, we demonstrate that vaccination with the RBD-cVLP vaccine protects mice from both a lethal infection and symptomatic disease. Our data also indicate that immunization significantly reduces inflammation and lung pathology associated with severe disease in mice. Additionally, we show that the survival of naïve animals significantly increases when sera from animals vaccinated with RBD-cVLP are passively transferred, prior to a lethal virus dose. Finally, the RBD-cVLP vaccine has a similar antigen composition to the clinical ABNCOV2 vaccine, which has shown non-inferiority to the Comirnaty mRNA vaccine in phase I-III trials. Therefore, our study provides evidence that this vaccine design is highly immunogenic and confers full protection against severe disease in mice.
Collapse
Affiliation(s)
- Sebenzile K. Myeni
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Anouk A. Leijs
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Peter J. Bredenbeek
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Shessy Torres Morales
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marissa E. Linger
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Sophie van Zanen-Gerhardt
- Experimental Pathology Services Laboratory, Central Animal and Transgenic Facility, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Serge A. L. Zander
- Experimental Pathology Services Laboratory, Central Animal and Transgenic Facility, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Experimental Animal Pathology Facility, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Adam F. Sander
- AdaptVac Aps, Ole Maaløes Vej 3, 2200 Copenhagen, Denmark; (C.F.)
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Marjolein Kikkert
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
194
|
Silva-Santos Y, Pagni RL, Gamon THM, de Azevedo MSP, Bielavsky M, Darido MLG, de Oliveira DBL, de Souza EE, Wrenger C, Durigon EL, Luvizotto MCR, Ackerman HC, Marinho CRF, Epiphanio S, Carvalho LJM. Lisinopril increases lung ACE2 levels and SARS-CoV-2 viral load and decreases inflammation but not disease severity in experimental COVID-19. Front Pharmacol 2024; 15:1414406. [PMID: 39070798 PMCID: PMC11282493 DOI: 10.3389/fphar.2024.1414406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 07/30/2024] Open
Abstract
COVID-19 causes more severe and frequently fatal disease in patients with pre-existing comorbidities such as hypertension and heart disease. SARS-CoV-2 virus enters host cells through the angiotensin-converting enzyme 2 (ACE2), which is fundamental in maintaining arterial pressure through the renin-angiotensin system (RAS). Hypertensive patients commonly use medications such as angiotensin-converting enzyme inhibitors (ACEi), which can modulate the expression of ACE2 and, therefore, potentially impact the susceptibility and severity of SARS-CoV-2 infection. Here we assessed whether treatment of ACE2-humanized (K18-hACE2) mice with the ACEi Lisinopril affects lung ACE2 levels and the outcome of experimental COVID-19. K18-hACE2 mice were treated for 21 days with Lisinopril 10 mg/kg and were then infected with 105 PFU of SARS-CoV-2 (Wuhan strain). Body weight, clinical score, respiratory function, survival, lung ACE2 levels, viral load, lung histology, and cytokine (IL-6, IL-33, and TNF-α) levels were assessed. Mice treated with Lisinopril for 21 days showed increased levels of ACE2 in the lungs. Infection with SARS-CoV-2 led to massive decrease in lung ACE2 levels at 3 days post-infection (dpi) in treated and untreated animals, but Lisinopril-treated mice showed a fast recovery (5dpi) of ACE2 levels. Higher ACE2 levels in Lisinopril-treated mice led to remarkably higher lung viral loads at 3 and 6/7dpi. Lisinopril-treated mice showed decreased levels of the pro-inflammatory cytokines IL-6 and TNF-α in the serum and lungs at 6/7dpi. Marginal improvements in body weight, clinical score and survival were observed in Lisinopril-treated mice. No differences between treated and untreated infected mice were observed in respiratory function and lung histology. Lisinopril treatment showed both deleterious (higher viral loads) and beneficial (anti-inflammatory and probably anti-constrictory and anti-coagulant) effects in experimental COVID-19. These effects seem to compensate each other, resulting in marginal beneficial effects in terms of outcome for Lisinopril-treated animals.
Collapse
Affiliation(s)
- Yasmin Silva-Santos
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
- Laboratory of Malaria Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Roberta Liberato Pagni
- Immunology Laboratory, Heart Institute, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Thais Helena Martins Gamon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Marcela Santiago Pacheco de Azevedo
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Mônica Bielavsky
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | - Maria Laura Goussain Darido
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | - Danielle Bruna Leal de Oliveira
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Luiz Durigon
- Laboratory of Clinical and Molecular Virology, Institute of Biomedical Sciences, Department of Microbiology, University of São Paulo, São Paulo, Brazil
| | | | - Hans Christian Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Claudio Romero Farias Marinho
- Laboratory of Experimental Immunoparasitology, Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Laboratory of Malaria Cellular and Molecular Immunopathology, Faculty of Pharmaceutical Sciences, Department of Clinical and Toxicological Analysis, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
195
|
Perez-Restrepo LS, Ciuoderis K, Usuga J, Moreno I, Vargas V, Arévalo-Arbelaez AJ, Berg MG, Cloherty GA, Hernández-Ortiz JP, Osorio JE. Mayaro Virus as the cause of Acute Febrile Illness in the Colombian Amazon Basin. Front Microbiol 2024; 15:1419637. [PMID: 39044955 PMCID: PMC11263195 DOI: 10.3389/fmicb.2024.1419637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Mayaro Fever (MF) is a tropical disease caused by the Mayaro virus (MAYV), with outbreaks documented in Latin America. Methods A hospital-based fever surveillance in Leticia, Colombian Amazon, collected sera from 1,460 patients aged 5-89 between December 2020 and April 2023. Results Dengue and malaria were the main diagnoses (19.4 and 5.8%, respectively), leaving 71.4% of cases unidentified after testing. Metagenomic sequencing and real-time RT-qPCR testing identified MAYV in two patients (25-year-old male and an 80-year-old female) exhibiting typical symptoms, of MF including rash, joint pain, and fever. Phylogenetics analysis of these two viruses revealed a close relationship to Peruvian strains within the MAYV D genotype. Discussion The study of AFI in Leticia, Colombia, identified dengue as prevalent, with malaria, COVID-19, Influenza, and Zika viruses also detected. Despite extensive testing, most cases remained unexplained until metagenomic sequencing revealed MAYV, previously unseen in Colombia but known in neighboring countries. Conclusion This study presents the first near full-length genomes of MAYV in Colombia, highlighting the need for further seroprevalence studies and enhanced surveillance to understand and control the spread of the virus in the region.
Collapse
Affiliation(s)
- Laura S. Perez-Restrepo
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Karl Ciuoderis
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Jaime Usuga
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Isabel Moreno
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Vanessa Vargas
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Angela J. Arévalo-Arbelaez
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
| | - Michael G. Berg
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
- Infectious Diseases Research, Abbott Diagnostics, Abbott Park, IL, United States
| | - Gavin A. Cloherty
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
- Infectious Diseases Research, Abbott Diagnostics, Abbott Park, IL, United States
| | - Juan Pablo Hernández-Ortiz
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
- Faculty of Life Sciences, Universidad Nacional de Colombia, Medellín, Colombia
| | - Jorge E. Osorio
- GHI One Health Colombia, Universidad Nacional de Colombia, Medellín, Colombia
- Abbott Pandemic Defense Coalition, Chicago, IL, United States
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
- Global Health Institute, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
196
|
Daigle L, Khalid H, Gagnon CA, Arsenault J, Bienzle D, Bisson SK, Blais MC, Denis-Robichaud J, Forest C, Grenier St-Sauveur V, Koszegi M, MacNicol J, Nantel-Fortier N, Nury C, Prystajecky N, Fraser E, Carabin H, Aenishaenslin C. High prevalence of SARS-CoV-2 antibodies and low prevalence of SARS-CoV-2 RNA in cats recently exposed to human cases. BMC Vet Res 2024; 20:304. [PMID: 38982461 PMCID: PMC11232172 DOI: 10.1186/s12917-024-04150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND The primary objective of this cross-sectional study, conducted in Québec and Bristish Columbia (Canada) between February 2021 and January 2022, was to measure the prevalence of viral RNA in oronasal and rectal swabs and serum antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) amongst cats living in households with at least one confirmed human case. Secondary objectives included a description of potential risk factors for the presence of SARS-CoV-2 antibodies and an estimation of the association between the presence of viral RNA in swabs as well as SARS-CoV-2 antibodies and clinical signs. Oronasal and rectal swabs and sera were collected from 55 cats from 40 households at most 15 days after a human case confirmation, and at up to two follow-up visits. A RT-qPCR assay and an ELISA were used to detect SARS-CoV-2 RNA in swabs and serum SARS-CoV-2 IgG antibodies, respectively. Prevalence and 95% Bayesian credibility intervals (BCI) were calculated, and associations were evaluated using prevalence ratio and 95% BCI obtained from Bayesian mixed log-binomial models. RESULTS Nine (0.16; 95% BCI = 0.08-0.28) and 38 (0.69; 95% BCI = 0.56-0.80) cats had at least one positive RT-qPCR and at least one positive serological test result, respectively. No risk factor was associated with the prevalence of SARS-CoV-2 serum antibodies. The prevalence of clinical signs suggestive of COVID-19 in cats, mainly sneezing, was 2.12 (95% BCI = 1.03-3.98) times higher amongst cats with detectable viral RNA compared to those without. CONCLUSIONS We showed that cats develop antibodies to SARS-CoV-2 when exposed to recent human cases, but detection of viral RNA on swabs is rare, even when sampling occurs soon after confirmation of a human case. Moreover, cats with detectable levels of virus showed clinical signs more often than cats without signs, which can be useful for the management of such cases.
Collapse
Affiliation(s)
- Laurence Daigle
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada.
- Groupe de recherche en épidémiologie des zoonoses et santé publique (GREZOSP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada.
- Centre de recherche en santé publique de l'Université de Montréal et du CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montréal, Qc, Canada.
| | - Hattaw Khalid
- BC Centre for Disease Control, Vancouver, BC, Canada
- School of Population and Public Health, UBC Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| | - Carl A Gagnon
- Swine and Poultry Infectious Diseases Research Center - FRQ, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Molecular Diagnostic Laboratory (MDL), Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Julie Arsenault
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Groupe de recherche en épidémiologie des zoonoses et santé publique (GREZOSP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Dorothee Bienzle
- Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Sarah-Kim Bisson
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Marie-Claude Blais
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - José Denis-Robichaud
- Groupe de recherche en épidémiologie des zoonoses et santé publique (GREZOSP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Independent Researcher, Amqui, QC, Canada
| | - Caroline Forest
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Valérie Grenier St-Sauveur
- Molecular Diagnostic Laboratory (MDL), Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Marika Koszegi
- Molecular Diagnostic Laboratory (MDL), Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Jennifer MacNicol
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Nicolas Nantel-Fortier
- Molecular Diagnostic Laboratory (MDL), Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Charlotte Nury
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
| | - Natalie Prystajecky
- BC Centre for Disease Control, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Erin Fraser
- BC Centre for Disease Control, Vancouver, BC, Canada
- School of Population and Public Health, UBC Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| | - Hélène Carabin
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Groupe de recherche en épidémiologie des zoonoses et santé publique (GREZOSP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Centre de recherche en santé publique de l'Université de Montréal et du CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montréal, Qc, Canada
| | - Cécile Aenishaenslin
- Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Groupe de recherche en épidémiologie des zoonoses et santé publique (GREZOSP), Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Qc, Canada
- Centre de recherche en santé publique de l'Université de Montréal et du CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montréal, Qc, Canada
| |
Collapse
|
197
|
Awad H, El-Brolossy TA, Abdallah T, Osman A, Negm S, Mansour OI, Girgis SA, Hafez HM, Zaki AM, Talaat H. Accurate and reliable surface-enhanced Raman spectroscopy assay for early detection of SARS-CoV-2 RNA with exceptional sensitivity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124184. [PMID: 38608556 DOI: 10.1016/j.saa.2024.124184] [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: 10/25/2023] [Revised: 02/28/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024]
Abstract
This research proposes a highly sensitive and simple surface-enhanced Raman spectroscopy (SERS) assay for the detection of SARS-CoV-2 RNA using suitably designed probes specific for RdRp and N viral genes attached to a Raman marker. The sensitivity of the assay was optimized through precise adjustments to the conditions of immobilization and hybridization processes of the target RNA, including modifications to factors such as time and temperature. The assay achieved a remarkable sensitivity down to 58.39 copies/mL, comparable to or lower than the sensitivities reported for commercial fluorescent polymerase chain reaction (PCR) based methods. It has good selectivity in discriminating SARS-CoV-2 RNA against other respiratory viruses, respiratory syncytial virus (RSV), and influenza A virus. The reliability of the assay was validated by testing 24 clinical samples, including 12 positive samples with varying cycle threshold (Ct) values and 12 negative samples previously tested using real-time PCR. The assay consistently predicted true results that were in line with the PCR results for all samples. Furthermore, the assay demonstrated a notable limit of detection (LOD) of Ct (38 for RdRp gene and 37.5 for N-gene), indicating its capability to detect low concentrations of the target analyte and potentially facilitating early detection of the pathogen.
Collapse
Affiliation(s)
- Hend Awad
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Tamer Abdallah
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ahmed Osman
- Institute of Basic and Applied Science - Egpt-Japan University of Science and Technology (E-JUST), Egypt
| | - Sohair Negm
- Department of Physics and Mathematics, Banha University, Banha, Egypt
| | | | | | - Hala M Hafez
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali M Zaki
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hassan Talaat
- Physics Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
198
|
Kulma M, Šakanović A, Bedina-Zavec A, Caserman S, Omersa N, Šolinc G, Orehek S, Hafner-Bratkovič I, Kuhar U, Slavec B, Krapež U, Ocepek M, Kobayashi T, Kwiatkowska K, Jerala R, Podobnik M, Anderluh G. Sequestration of membrane cholesterol by cholesterol-binding proteins inhibits SARS-CoV-2 entry into Vero E6 cells. Biochem Biophys Res Commun 2024; 716:149954. [PMID: 38704887 DOI: 10.1016/j.bbrc.2024.149954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Membrane lipids and proteins form dynamic domains crucial for physiological and pathophysiological processes, including viral infection. Many plasma membrane proteins, residing within membrane domains enriched with cholesterol (CHOL) and sphingomyelin (SM), serve as receptors for attachment and entry of viruses into the host cell. Among these, human coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use proteins associated with membrane domains for initial binding and internalization. We hypothesized that the interaction of lipid-binding proteins with CHOL in plasma membrane could sequestrate lipids and thus affect the efficiency of virus entry into host cells, preventing the initial steps of viral infection. We have prepared CHOL-binding proteins with high affinities for lipids in the plasma membrane of mammalian cells. Binding of the perfringolysin O domain four (D4) and its variant D4E458L to membrane CHOL impaired the internalization of the receptor-binding domain of the SARS-CoV-2 spike protein and the pseudovirus complemented with the SARS-CoV-2 spike protein. SARS-CoV-2 replication in Vero E6 cells was also decreased. Overall, our results demonstrate that the integrity of CHOL-rich membrane domains and the accessibility of CHOL in the membrane play an essential role in SARS-CoV-2 cell entry.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Apolonija Bedina-Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Urška Kuhar
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Brigita Slavec
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Matjaž Ocepek
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan; UMR 7021 CNRS, Université de Strasbourg, F-67401, Illkirch, France
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia.
| |
Collapse
|
199
|
Bennet R, Rinder MR, George E, Hertting O, Luthander J, Åkefeldt SO, Hammas B, Allander T, Eriksson M. Pre-admission virus detection during the COVID-19 pandemic in children with and without symptoms of infection. Acta Paediatr 2024; 113:1679-1684. [PMID: 38445712 DOI: 10.1111/apa.17195] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
AIM Pre-admission viral screening is used only in exceptional situations such as pandemics. We therefore evaluated pre-admission screening for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respiratory syncytial virus (RSV) and influenza during the COVID-19 pandemic, comparing epidemiology and clinical features of admitted children. METHODS Children were screened at a paediatric emergency department from 1 March 2020 to 30 June 2022 by nasopharyngeal sampling and polymerase chain reaction kit. We retrospectively retrieved positive results from the laboratory and scrutinised charts of admitted children. RESULTS Out of 15 927 screened children, 522, 127 and 572 were positive and admitted with RSV, influenza A or SARS-CoV-2, respectively. Of these, 29 (5.6%), 26 (24.1%) and 245 (44.8%) were incidental findings, lacking symptoms of infection. RSV and influenza A were initially absent but re-emerged in the autumn of 2021. The rate of COVID-19 rose when the Omicron variant emerged in December 2021. The median age of children with RSV was 0.3 years, of those with influenza A 6.7 years and of those with COVID-19 1.6 years. Major complications were rare. CONCLUSION Frequent incidental detections of SARS-CoV-2 likely reflected widespread presence of a mild infection. Clinically, COVID-19 was like other viral respiratory infections in children.
Collapse
Affiliation(s)
- Rutger Bennet
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| | - Malin Ryd Rinder
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| | - Eric George
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| | - Olof Hertting
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| | - Joachim Luthander
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| | | | - Berit Hammas
- Department of Medical Microbiology, Karolinska University Hospital, Solna, Sweden
| | - Tobias Allander
- Department of Medical Microbiology, Karolinska University Hospital, Solna, Sweden
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Eriksson
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
200
|
Gilbert-Girard S, Piret J, Carbonneau J, Hénaut M, Goyette N, Boivin G. Viral interference between severe acute respiratory syndrome coronavirus 2 and influenza A viruses. PLoS Pathog 2024; 20:e1012017. [PMID: 39038029 PMCID: PMC11293641 DOI: 10.1371/journal.ppat.1012017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/01/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024] Open
Abstract
Some respiratory viruses can cause a viral interference through the activation of the interferon (IFN) pathway that reduces the replication of another virus. Epidemiological studies of coinfections between SARS-CoV-2 and other respiratory viruses have been hampered by non-pharmacological measures applied to mitigate the spread of SARS-CoV-2 during the COVID-19 pandemic. With the ease of these interventions, SARS-CoV-2 and influenza A viruses can now co-circulate. It is thus of prime importance to characterize their interactions. In this work, we investigated viral interference effects between an Omicron variant and a contemporary influenza A/H3N2 strain, in comparison with an ancestral SARS-CoV-2 strain and the 2009 pandemic influenza A/H1N1 virus. We infected nasal human airway epitheliums with SARS-CoV-2 and influenza, either simultaneously or 24 h apart. Viral load was measured by RT-qPCR and IFN-α/β/λ1/λ2 proteins were quantified by immunoassay. Expression of four interferon-stimulated genes (ISGs; OAS1/IFITM3/ISG15/MxA) was also measured by RT-droplet digital PCR. Additionally, susceptibility of each virus to IFN-α/β/λ2 recombinant proteins was determined. Our results showed that influenza A, and especially A/H3N2, interfered with both SARS-CoV-2 viruses, but that SARS-CoV-2 did not significantly interfere with A/H3N2 or A/H1N1. Consistently with these results, influenza, and particularly the A/H3N2 strain, caused a higher production of IFN proteins and expression of ISGs than SARS-CoV-2. SARS-CoV-2 induced a marginal IFN production and reduced the IFN response during coinfections with influenza. All viruses were susceptible to exogenous IFNs, with the ancestral SARS-CoV-2 and Omicron being less susceptible to type I and type III IFNs, respectively. Thus, influenza A causes a viral interference towards SARS-CoV-2 most likely through an IFN response. The opposite is not necessarily true, and a concurrent infection with both viruses leads to a lower IFN response. Taken together, these results help us to understand how SARS-CoV-2 interacts with another major respiratory pathogen.
Collapse
Affiliation(s)
| | - Jocelyne Piret
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Julie Carbonneau
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Mathilde Hénaut
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Nathalie Goyette
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Guy Boivin
- Research Center of the CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|