351
|
Wang R, Zhu B, Young P, Luo Y, Taylor J, Cameron AJ, Squire CJ, Travas-Sejdic J. A Portable and Disposable Electrochemical Sensor Utilizing Laser-Scribed Graphene for Rapid SARS-CoV-2 Detection. BIOSENSORS 2023; 14:10. [PMID: 38248387 PMCID: PMC10813335 DOI: 10.3390/bios14010010] [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: 11/25/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
The COVID-19 pandemic caused by the virus SARS-CoV-2 was the greatest global threat to human health in the last three years. The most widely used methodologies for the diagnosis of COVID-19 are quantitative reverse transcription polymerase chain reaction (RT-qPCR) and rapid antigen tests (RATs). PCR is time-consuming and requires specialized instrumentation operated by skilled personnel. In contrast, RATs can be used in-home or at point-of-care but are less sensitive, leading to a higher rate of false negative results. In this work, we describe the development of a disposable, electrochemical, and laser-scribed graphene-based biosensor strips for COVID-19 detection that exploits a split-ester bond ligase system (termed 'EsterLigase') for immobilization of a virus-specific nanobody to maintain the out-of-plane orientation of the probe to ensure the efficacy of the probe-target recognition process. An anti-spike VHH E nanobody, genetically fused with the EsterLigase domain, was used as the specific probe for the spike receptor-binding domain (SP-RBD) protein as the target. The recognition between the two was measured by the change in the charge transfer resistance determined by fitting the electrochemical impedance spectroscopy (EIS) spectra. The developed LSG-based biosensor achieved a linear detection range for the SP-RBD from 150 pM to 15 nM with a sensitivity of 0.0866 [log(M)]-1 and a limit of detection (LOD) of 7.68 pM.
Collapse
Affiliation(s)
- Runzhong Wang
- Centre for Innovative Materials and Health, School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (R.W.); (B.Z.)
| | - Bicheng Zhu
- Centre for Innovative Materials and Health, School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (R.W.); (B.Z.)
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Paul Young
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (P.Y.); (J.T.); (C.J.S.)
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Yu Luo
- Micro- and Nano-Technology Research Center, State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China;
| | - John Taylor
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (P.Y.); (J.T.); (C.J.S.)
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Alan J. Cameron
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington 6012, New Zealand
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (P.Y.); (J.T.); (C.J.S.)
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Christopher J. Squire
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (P.Y.); (J.T.); (C.J.S.)
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1142, New Zealand
| | - Jadranka Travas-Sejdic
- Centre for Innovative Materials and Health, School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (R.W.); (B.Z.)
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
352
|
da Silva JL, de Souza BSW, de Albuquerque LZ, Aleixo SB, Resende GADS, de Oliveira DGB, dos Santos EN, Nogueira-Rodrigues A, Clara RO, Gaui MDFD, Mota ACDA, de Lima VCC, Rosa DD, Munhoz RR, Morbeck IAP, Gelatti ACZ, Mathias CMDC, de Melo AC. Factors influencing COVID-19 mortality among cancer patients: A Brazilian multi-institutional study. PLoS One 2023; 18:e0295597. [PMID: 38127882 PMCID: PMC10734930 DOI: 10.1371/journal.pone.0295597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE This study aimed to describe the demographic and clinical characteristics of cancer patients with COVID-19, exploring factors associated with adverse outcomes. PATIENTS AND METHODS This retrospective cohort study methodically extracted and curated data from electronic medical records (EMRs) of numerous healthcare institutions on cancer patients diagnosed with a confirmed SARS-CoV-2 infection between May 2020 and August 2021, to identify risk factors linked to extended hospitalization and mortality. The retrieved information encompassed the patients' demographic and clinical characteristics, including the incidence of prolonged hospitalization, acute complications, and COVID-19-related mortality. RESULTS A total of 1446 cancer patients with COVID-19 were identified (mean [Standard deviation] age, 59.2 [14.3] years). Most patients were female (913 [63.1%]), non-white (646 [44.7%]), with non-metastatic (818 [56.6%]) solid tumors (1318 [91.1%]), and undergoing chemotherapy (647 [44.7%]). The rate of extended hospitalization due to COVID-19 was 46% (n = 665), which was significantly impacted by age (p = 0.012), sex (p = 0.003), race and ethnicity (p = 0.049), the presence of two or more comorbidities (p = 0.006), hematologic malignancies (p = 0.013), metastatic disease (p = 0.002), and a performance status ≥ 2 (p = 0.001). The COVID-19-related mortality rate was 18.9% (n = 273), and metastatic disease (<0.001), performance status ≥2 (<0.001), extended hospitalization (p = 0.028), renal failure (p = 0.029), respiratory failure (p < 0.001), sepsis (p = 0.004), and shock (p = 0.040) significantly and negatively influenced survival. CONCLUSION The rate of extended hospitalization and COVID-19-specific death in cancer patients was notably high and could be influenced by comorbidities, cancer treatment status, and clinical fragility. These observations may aid in developing risk counseling strategies regarding COVID-19 in individuals diagnosed with cancer.
Collapse
Affiliation(s)
- Jessé Lopes da Silva
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Clinical Oncology, Galeao Air Force Hospital, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Lucas Zanetti de Albuquerque
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sabina Bandeira Aleixo
- Department of Clinical Oncology, Evangelical Hospital of Cachoeiro de Itapemirim, Cachoeiro de Itapemirim, Espírito Santo, Brazil
| | | | | | | | - Angélica Nogueira-Rodrigues
- Department of General Medicine UFMG, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Brazilian Society of Clinical Oncology, São Paulo, São Paulo, Brazil
| | | | | | | | | | - Daniela Dornelles Rosa
- Brazilian Society of Clinical Oncology, São Paulo, São Paulo, Brazil
- Department of Clinical Oncology, Hospital Moinhos de Vento, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | - Ana Caroline Zimmer Gelatti
- Oncoclinicas Group of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Brazilian Group of Thoracic Tumors, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Andréia Cristina de Melo
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
353
|
Rokni M, Rohani Bastami T, Meshkat Z, Reza Rahimi H, Zibaee S, Meshkat M, Fotouhi F, Serki E, Khoshakhlagh M, Dabirifar Z. Rapid and sensitive detection of SARS-CoV-2 virus in human saliva samples using glycan based nanozyme: a clinical study. Mikrochim Acta 2023; 191:36. [PMID: 38108890 DOI: 10.1007/s00604-023-06120-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/25/2023] [Indexed: 12/19/2023]
Abstract
A highly sensitive colorimetric method (glycan-based nano(e)zyme) was developed for sensitive and rapid detection of the SARS-CoV-2 virus based on N-acetyl neuraminic acid (sialic acid)-functionalized gold nanoparticles (SA-Au NZs). A number of techniques were used to characterize the prepared nanomaterials including XRD, FT-IR, UV-vis, DLS, and TEM. DLS analysis indicates an average hydrodynamic size of 34 nm, whereas TEM analysis indicates an average particle size of 15.78 nm. This observation confirms that water interacts with nanoparticle surfaces, resulting in a large hydrodynamic diameter. The peroxidase-like activity of SA-Au NZs was examined with SARS-CoV-2 and influenza viruses (influenza A (H1N1), influenza A (H3N2), and influenza B). UV-visible spectroscopy was used to monitor and record the results, as well as naked eye detection (photographs). SA-Au NZs exhibit a change in color from light red to purple when SARS-CoV-2 is present, and they exhibit a redshift in their spectrum. N-acetyl neuraminic acid interacts with SARS-CoV-2 spike glycoprotein, confirming its ability to bind glycans. As a result, SA-Au NZs can detect COVID-19 with sensitivity and specificity of over 95% and 98%, respectively. This method was approved by testing saliva samples from 533 suspected individuals at Ghaem Hospital of Mashhad, Mashhad, Iran. Sensitivity and specificity were calculated by comparing the results with the definitive results. The positive results were accompanied by a color change from bright red to purple within five minutes. Statistical analysis was performed based on variables such as age, gender, smoking, diabetes, hypertension, and lung involvement. In clinical trials, it was demonstrated that this method can be used to diagnose SARS-CoV-2 in a variety of places, such as medical centers, hospitals, airports, universities, and schools.
Collapse
Affiliation(s)
- Mehrdad Rokni
- Department of Chemical Engineering and Energy, Quchan University of Technology, Quchan, 94771-67335, Iran
| | - Tahereh Rohani Bastami
- Department of Chemical Engineering and Energy, Quchan University of Technology, Quchan, 94771-67335, Iran.
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Zibaee
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Mashhad, Iran
| | - Mojtaba Meshkat
- Department of Community Medicine, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Fatemeh Fotouhi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Serki
- Department of Clinical Biochemistry, Mashhad University of Medical Science, Mashhad, Iran Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdieh Khoshakhlagh
- Department of Clinical Biochemistry, Mashhad University of Medical Science, Mashhad, Iran Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeynab Dabirifar
- Department of Chemical Engineering and Energy, Quchan University of Technology, Quchan, 94771-67335, Iran
| |
Collapse
|
354
|
Mhamadi M, Mencattelli G, Gaye A, Ndiaye EH, Sow AA, Faye M, Ndione MHD, Diagne MM, Mhamadi M, Faye O, Weidmann M, Faye O, Diallo M, Diagne CT. Rapid On-Site Detection of Arboviruses by a Direct RT-qPCR Assay. BIOSENSORS 2023; 13:1035. [PMID: 38131795 PMCID: PMC10741549 DOI: 10.3390/bios13121035] [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: 09/17/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 12/23/2023]
Abstract
Arthropod-borne diseases currently constitute a source of major health concerns worldwide. They account for about 50% of global infectious diseases and cause nearly 700,000 deaths every year. Their rapid increase and spread constitute a huge challenge for public health, highlighting the need for early detection during epidemics, to curtail the virus spread, and to enhance outbreak management. Here, we compared a standard quantitative polymerase chain reaction (RT-qPCR) and a direct RT-qPCR assay for the detection of Zika (ZIKV), Chikungunya (CHIKV), and Rift Valley Fever (RVFV) viruses from experimentally infected-mosquitoes. The direct RT-qPCR could be completed within 1.5 h and required 1 µL of viral supernatant from homogenized mosquito body pools. Results showed that the direct RT-qPCR can detect 85.71%, 89%, and 100% of CHIKV, RVFV, and ZIKV samples by direct amplifications compared to the standard method. The use of 1:10 diluted supernatant is suggested for CHIKV and RVFV direct RT-qPCR. Despite a slight drop in sensitivity for direct PCR, our technique is more affordable, less time-consuming, and provides a better option for qualitative field diagnosis during outbreak management. It represents an alternative when extraction and purification steps are not possible because of insufficient sample volume or biosecurity issues.
Collapse
Affiliation(s)
- Moufid Mhamadi
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Giulia Mencattelli
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Alioune Gaye
- Department of Medical Zoology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (A.G.); (E.H.N.)
| | - El Hadji Ndiaye
- Department of Medical Zoology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (A.G.); (E.H.N.)
| | - Aïssatou Aïcha Sow
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Martin Faye
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Marie Henriette Dior Ndione
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Moussa Moïse Diagne
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Moundhir Mhamadi
- DIATROPIX, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (M.M.); (M.D.)
| | - Ousmane Faye
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Manfred Weidmann
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, 01968 Senftenberg, Germany;
| | - Oumar Faye
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
| | - Mawlouth Diallo
- DIATROPIX, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (M.M.); (M.D.)
| | - Cheikh Tidiane Diagne
- Department of Virology, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (G.M.); (A.A.S.); (M.F.); (M.H.D.N.); (M.M.D.); (O.F.); (O.F.)
- DIATROPIX, Fondation Institut Pasteur de Dakar 36, Avenue Pasteur, Dakar 220, Senegal; (M.M.); (M.D.)
| |
Collapse
|
355
|
Ishige T. Molecular biology of SARS-CoV-2 and techniques of diagnosis and surveillance. Adv Clin Chem 2023; 118:35-85. [PMID: 38280807 DOI: 10.1016/bs.acc.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
The World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19), a disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a global pandemic in March 2020. Reverse transcription-polymerase chain reaction (RT-PCR) is the reference technique for molecular diagnosis of SARS-CoV-2 infection. The SARS-CoV-2 virus is constantly mutating, and more transmissible variants have emerged, making genomic surveillance a crucial tool for investigating virus transmission dynamics, detecting novel genetic variants, and assessing mutation impact. The S gene, which encodes the spike protein, is frequently mutated, and it plays an important role in transmissibility. Spike protein mutations affect infectivity and vaccine effectiveness. SARS-CoV-2 variants are tracked using whole genome sequencing (WGS) and S-gene analysis. WGS, Sanger sequencing, and many S-gene-targeted RT-PCR methods have been developed. WGS and Sanger sequencing are standard methods for detecting mutations and can be used to identify known and unknown mutations. Melting curve analysis, endpoint genotyping assay, and S-gene target failure are used in the RT-PCR-based method for the rapid detection of specific mutations in SARS-CoV-2 variants. Therefore, these assays are suitable for high-throughput screening. The combinatorial use of RT-PCR-based assays, Sanger sequencing, and WGS enables rapid and accurate tracking of SARS-CoV-2 variants. In this review, we described RT-PCR-based detection and surveillance techniques for SARS-CoV-2.
Collapse
Affiliation(s)
- Takayuki Ishige
- Division of Laboratory Medicine, Chiba University Hospital, Chiba, Japan.
| |
Collapse
|
356
|
Shahbaz MA, Kuivanen S, Lampinen R, Mussalo L, Hron T, Závodná T, Ojha R, Krejčík Z, Saveleva L, Tahir NA, Kalapudas J, Koivisto AM, Penttilä E, Löppönen H, Singh P, Topinka J, Vapalahti O, Chew S, Balistreri G, Kanninen KM. Human-derived air-liquid interface cultures decipher Alzheimer's disease-SARS-CoV-2 crosstalk in the olfactory mucosa. J Neuroinflammation 2023; 20:299. [PMID: 38098019 PMCID: PMC10722731 DOI: 10.1186/s12974-023-02979-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The neurological effects of the coronavirus disease of 2019 (COVID-19) raise concerns about potential long-term consequences, such as an increased risk of Alzheimer's disease (AD). Neuroinflammation and other AD-associated pathologies are also suggested to increase the risk of serious SARS-CoV-2 infection. Anosmia is a common neurological symptom reported in COVID-19 and in early AD. The olfactory mucosa (OM) is important for the perception of smell and a proposed site of viral entry to the brain. However, little is known about SARS-CoV-2 infection at the OM of individuals with AD. METHODS To address this gap, we established a 3D in vitro model of the OM from primary cells derived from cognitively healthy and AD individuals. We cultured the cells at the air-liquid interface (ALI) to study SARS-CoV-2 infection under controlled experimental conditions. Primary OM cells in ALI expressed angiotensin-converting enzyme 2 (ACE-2), neuropilin-1 (NRP-1), and several other known SARS-CoV-2 receptor and were highly vulnerable to infection. Infection was determined by secreted viral RNA content and confirmed with SARS-CoV-2 nucleocapsid protein (NP) in the infected cells by immunocytochemistry. Differential responses of healthy and AD individuals-derived OM cells to SARS-CoV-2 were determined by RNA sequencing. RESULTS Results indicate that cells derived from cognitively healthy donors and individuals with AD do not differ in susceptibility to infection with the wild-type SARS-CoV-2 virus. However, transcriptomic signatures in cells from individuals with AD are highly distinct. Specifically, the cells from AD patients that were infected with the virus showed increased levels of oxidative stress, desensitized inflammation and immune responses, and alterations to genes associated with olfaction. These results imply that individuals with AD may be at a greater risk of experiencing severe outcomes from the infection, potentially driven by pre-existing neuroinflammation. CONCLUSIONS The study sheds light on the interplay between AD pathology and SARS-CoV-2 infection. Altered transcriptomic signatures in AD cells may contribute to unique symptoms and a more severe disease course, with a notable involvement of neuroinflammation. Furthermore, the research emphasizes the need for targeted interventions to enhance outcomes for AD patients with viral infection. The study is crucial to better comprehend the relationship between AD, COVID-19, and anosmia. It highlights the importance of ongoing research to develop more effective treatments for those at high risk of severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Muhammad Ali Shahbaz
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Suvi Kuivanen
- Department of Virology, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Virology, 10117, Berlin, Germany
| | - Riikka Lampinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Laura Mussalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Tomáš Hron
- Institute of Molecular Genetics, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Táňa Závodná
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Ravi Ojha
- Department of Virology, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Zdeněk Krejčík
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Numan Ahmad Tahir
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Juho Kalapudas
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210, Kuopio, Finland
| | - Anne M Koivisto
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210, Kuopio, Finland
- Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, 70210, Kuopio, Finland
- Department of Neurology and Geriatrics, Helsinki University Hospital and Neurosciences, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Elina Penttilä
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210, Kuopio, Finland
| | - Heikki Löppönen
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210, Kuopio, Finland
| | | | - Jan Topinka
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Sweelin Chew
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Giuseppe Balistreri
- Department of Virology, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
- The Queensland Brain Institute, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland.
| |
Collapse
|
357
|
Hilligan KL, Namasivayam S, Clancy CS, Baker PJ, Old SI, Peluf V, Amaral EP, Oland SD, O'Mard D, Laux J, Cohen M, Garza NL, Lafont BAP, Johnson RF, Feng CG, Jankovic D, Lamiable O, Mayer-Barber KD, Sher A. Bacterial-induced or passively administered interferon gamma conditions the lung for early control of SARS-CoV-2. Nat Commun 2023; 14:8229. [PMID: 38086794 PMCID: PMC10716133 DOI: 10.1038/s41467-023-43447-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Type-1 and type-3 interferons (IFNs) are important for control of viral replication; however, less is known about the role of Type-2 IFN (IFNγ) in anti-viral immunity. We previously observed that lung infection with Mycobacterium bovis BCG achieved though intravenous (iv) administration provides strong protection against SARS-CoV-2 in mice yet drives low levels of type-1 IFNs but robust IFNγ. Here we examine the role of ongoing IFNγ responses to pre-established bacterial infection on SARS-CoV-2 disease outcomes in two murine models. We report that IFNγ is required for iv BCG induced reduction in pulmonary viral loads, an outcome dependent on IFNγ receptor expression by non-hematopoietic cells. Importantly, we show that BCG infection prompts pulmonary epithelial cells to upregulate IFN-stimulated genes with reported anti-viral activity in an IFNγ-dependent manner, suggesting a possible mechanism for the observed protection. Finally, we confirm the anti-viral properties of IFNγ by demonstrating that the recombinant cytokine itself provides strong protection against SARS-CoV-2 challenge when administered intranasally. Together, our data show that a pre-established IFNγ response within the lung is protective against SARS-CoV-2 infection, suggesting that concurrent or recent infections that drive IFNγ may limit the pathogenesis of SARS-CoV-2 and supporting possible prophylactic uses of IFNγ in COVID-19 management.
Collapse
Affiliation(s)
- Kerry L Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand.
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chad S Clancy
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Paul J Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samuel I Old
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand
| | - Victoria Peluf
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sandra D Oland
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Danielle O'Mard
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julie Laux
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Melanie Cohen
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicole L Garza
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bernard A P Lafont
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Reed F Johnson
- SARS-CoV2- Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carl G Feng
- Immunology and Host Defense Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia
- Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Immunoparasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Olivier Lamiable
- Malaghan Institute of Medical Research, Wellington, 6012, New Zealand
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
358
|
Nath S, Sarkar M, Maddheshiya A, De D, Paul S, Dey S, Pal K, Roy SK, Ghosh A, Sengupta S, Paine SK, Biswas NK, Basu A, Mukherjee S. Upper respiratory tract microbiome profiles in SARS-CoV-2 Delta and Omicron infected patients exhibit variant specific patterns and robust prediction of disease groups. Microbiol Spectr 2023; 11:e0236823. [PMID: 37905804 PMCID: PMC10715160 DOI: 10.1128/spectrum.02368-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE The role of the upper respiratory tract (URT) microbiome in predicting lung health has been documented in several studies. The dysbiosis in COVID patients has been associated with disease outcomes by modulating the host immune system. However, although it has been known that different SARS-CoV-2 variants manifest distinct transmissibility and mortality rates in human populations, their effect on the composition and diversity of the URT microbiome has not been studied to date. Unlike the older variant (Delta), the newer variant (Omicron) have become more transmissible with lesser mortality and the symptoms have also changed significantly. Hence, in the present study, we have investigated the change in the URT microbiome associated with Delta and Omicron variants and identified variant-specific signatures that will be useful in the assessment of lung health and can be utilized for nasal probiotic therapy in the future.
Collapse
Affiliation(s)
- Shankha Nath
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Mousumi Sarkar
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | - Debjit De
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Shouvik Paul
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Souradeep Dey
- Department of Community Medicine, College of Medicine and JNM Hospital, Kalyani, West Bengal, India
| | - Kuhu Pal
- Department of Microbiology, College of Medicine and JNM Hospital, Kalyani, West Bengal, India
| | - Suman Kr. Roy
- Department of Community Medicine, College of Medicine and JNM Hospital, Kalyani, West Bengal, India
| | - Ayan Ghosh
- Department of Community Medicine, College of Medicine and JNM Hospital, Kalyani, West Bengal, India
| | - Sharmila Sengupta
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | - Nidhan K. Biswas
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Analabha Basu
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Souvik Mukherjee
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| |
Collapse
|
359
|
Grootemaat AE, Wiersma N, van der Niet S, Schimmel IM, Florquin S, Reits EA, Miller SE, van der Wel NN. Nucleocapsid protein accumulates in renal tubular epithelium of a post-COVID-19 patient. Microbiol Spectr 2023; 11:e0302923. [PMID: 37975661 PMCID: PMC10715010 DOI: 10.1128/spectrum.03029-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE Even though the coronavirus disease 2019 (COVID-19) pandemic is slowly developing into a conventional infectious disease, the long-term effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infection are still not well understood. One of the problems is that many COVID-19 cases develop acute kidney injuries. Still, it is heavily debated whether SARS-CoV-2 virus enters and actively replicates in kidney tissue and if SARS-CoV-2 virus particles can be detected in kidney during or post-infection. Here, we demonstrated that nucleocapsid N protein was detected in kidney tubular epithelium of patients that already recovered form COVID-19. The presence of the abundantly produced N protein without signs of viral replication could have implications for the recurrence of kidney disease and have a continuing effect on the immune system.
Collapse
Affiliation(s)
- Anita E. Grootemaat
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Niek Wiersma
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sanne van der Niet
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Irene M. Schimmel
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sandrine Florquin
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Pathology, Amsterdam University Medical Centers (location University of Amsterdam), Amsterdam, the Netherlands
| | - Eric A. Reits
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sara E. Miller
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicole N. van der Wel
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
360
|
Matsumura Y, Nakazaki T, Kitamori K, Kure E, Shinohara K, Tsuchido Y, Yukawa S, Noguchi T, Yamamoto M, Nagao M. Development and evaluation of the automated multipurpose molecular testing system PCRpack for high-throughput SARS-CoV-2 testing. Microbiol Spectr 2023; 11:e0271623. [PMID: 37943047 PMCID: PMC10715159 DOI: 10.1128/spectrum.02716-23] [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: 07/01/2023] [Accepted: 10/03/2023] [Indexed: 11/10/2023] Open
Abstract
IMPORTANCE Accurate and fast molecular testing is important for the diagnosis and control of COVID-19. During patient surges in the COVID-19 pandemic, laboratories were challenged by a higher demand for molecular testing under skilled staff shortages. We developed an automated multipurpose molecular testing system, named PCRpack, for the rapid, high-throughput testing of infectious pathogens, including SARS-CoV-2. The system is provided in an all-in-one package, including a liquid handling instrument, a laboratory information management system, and other materials needed for testing operation; is highly customizable; and is easily implemented. PCRpack showed robust liquid handling performance, high clinical diagnostic performance, a shorter turn-around time with minimal hands-on time, and a high testing capacity. These features contribute to the rapid implementation of the high-performance and high-throughput molecular testing environment at any phase of the pandemic caused by SARS-CoV-2 or future emerging pathogens.
Collapse
Affiliation(s)
- Yasufumi Matsumura
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Kanako Kitamori
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Nippon Control System, Yokohama, Japan
| | - Eiki Kure
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Faculty of Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Koh Shinohara
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuhiro Tsuchido
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satomi Yukawa
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Taro Noguchi
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Yamamoto
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
361
|
Pérez P, Albericio G, Astorgano D, Flores S, Sánchez-Corzo C, Sánchez-Cordón PJ, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. Preclinical immune efficacy against SARS-CoV-2 beta B.1.351 variant by MVA-based vaccine candidates. Front Immunol 2023; 14:1264323. [PMID: 38155964 PMCID: PMC10754519 DOI: 10.3389/fimmu.2023.1264323] [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: 07/20/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
The constant appearance of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VoCs) has jeopardized the protective capacity of approved vaccines against coronavirus disease-19 (COVID-19). For this reason, the generation of new vaccine candidates adapted to the emerging VoCs is of special importance. Here, we developed an optimized COVID-19 vaccine candidate using the modified vaccinia virus Ankara (MVA) vector to express a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein, containing 3 proline (3P) substitutions in the S protein derived from the beta (B.1.351) variant, termed MVA-S(3Pbeta). Preclinical evaluation of MVA-S(3Pbeta) in head-to-head comparison to the previously generated MVA-S(3P) vaccine candidate, expressing a full-length prefusion-stabilized Wuhan S protein (with also 3P substitutions), demonstrated that two intramuscular doses of both vaccine candidates fully protected transgenic K18-hACE2 mice from a lethal challenge with SARS-CoV-2 beta variant, reducing mRNA and infectious viral loads in the lungs and in bronchoalveolar lavages, decreasing lung histopathological lesions and levels of proinflammatory cytokines in the lungs. Vaccination also elicited high titers of anti-S Th1-biased IgGs and neutralizing antibodies against ancestral SARS-CoV-2 Wuhan strain and VoCs alpha, beta, gamma, delta, and omicron. In addition, similar systemic and local SARS-CoV-2 S-specific CD4+ and CD8+ T-cell immune responses were elicited by both vaccine candidates after a single intranasal immunization in C57BL/6 mice. These preclinical data support clinical evaluation of MVA-S(3Pbeta) and MVA-S(3P), to explore whether they can diversify and potentially increase recognition and protection of SARS-CoV-2 VoCs.
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
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- 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
| | - 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
| | - Joanna Luczkowiak
- 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
|
362
|
Rowe CJ, Nwaolu U, Salinas D, Hong J, Nunez J, Lansford JL, McCarthy CF, Potter BK, Levi BH, Davis TA. Inhibition of focal adhesion kinase 2 results in a macrophage polarization shift to M2 which attenuates local and systemic inflammation and reduces heterotopic ossification after polysystem extremity trauma. Front Immunol 2023; 14:1280884. [PMID: 38116014 PMCID: PMC10728492 DOI: 10.3389/fimmu.2023.1280884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/16/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Heterotopic ossification (HO) is a complex pathology often observed in combat injured casualties who have sustained severe, high energy polytraumatic extremity injuries. Once HO has developed, prophylactic therapies are limited outside of surgical excision. Tourniquet-induced ischemia injury (IR) exacerbates trauma-mediated musculoskeletal tissue injury, inflammation, osteogenic progenitor cell development and HO formation. Others have shown that focal adhesion kinase-2 (FAK2) plays a key role in regulating early inflammatory signaling events. Therefore, we hypothesized that targeting FAK2 prophylactically would mitigate extremity trauma induced IR inflammation and HO formation. Methods We tested whether the continuous infusion of a FAK2 inhibitor (Defactinib, PF-573228; 6.94 µg/kg/min for 14 days) can mitigate ectopic bone formation (HO) using an established blast-related extremity injury model involving femoral fracture, quadriceps crush injury, three hours of tourniquet-induced limb ischemia, and hindlimb amputation through the fracture site. Tissue inflammation, infiltrating cells, osteogenic progenitor cell content were assessed at POD-7. Micro-computed tomography imaging was used to quantify mature HO at POD-56. Results In comparison to vehicle control-treated rats, FAK2 administration resulted in no marked wound healing complications or weight loss. FAK2 treatment decreased HO by 43%. At POD-7, marked reductions in tissue proinflammatory gene expression and assayable osteogenic progenitor cells were measured, albeit no significant changes in expression patterns of angiogenic, chondrogenic and osteogenic genes. At the same timepoint, injured tissue from FAK-treated rats had fewer infiltrating cells. Additionally, gene expression analyses of tissue infiltrating cells resulted in a more measurable shift from an M1 inflammatory to an M2 anti-inflammatory macrophage phenotype in the FAK2 inhibitor-treated group. Discussion Our findings suggest that FAK2 inhibition may be a novel strategy to dampen trauma-induced inflammation and attenuate HO in patients at high risk as a consequence of severe musculoskeletal polytrauma.
Collapse
Affiliation(s)
- Cassie J. Rowe
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Uloma Nwaolu
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Daniela Salinas
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Jonathan Hong
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Johanna Nunez
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Jefferson L. Lansford
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Conor F. McCarthy
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Benjamin K. Potter
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Benjamin H. Levi
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Thomas A. Davis
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
363
|
Beitari S, Agbayani G, Hewitt M, Duque D, Bavananthasivam J, Sandhu JK, Akache B, Hadžisejdić I, Tran A. Effectiveness of VSV vectored SARS-CoV-2 spike when administered through intranasal, intramuscular or a combination of both. Sci Rep 2023; 13:21390. [PMID: 38049498 PMCID: PMC10695950 DOI: 10.1038/s41598-023-48397-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/26/2023] [Indexed: 12/06/2023] Open
Abstract
A critical feature of the VSV vector platform is the ability to pseudotype the virus with different glycoproteins from other viruses, thus altering cellular tropism of the recombinant virus. The route of administration is critical in triggering local and systemic immune response and protection. Most of the vaccine platforms used at the forefront are administered by intramuscular injection. However, it is not known at what level ACE2 is expressed on the surface of skeletal muscle cells, which will have a significant impact on the efficiency of a VSV-SARS-CoV-2 spike vaccine to mount a protective immune response when administered intramuscularly. In this study, we investigate the immunogenicity and efficacy of a prime-boost immunization regimen administered intranasally (IN), intramuscularly (IM), or combinations of the two. We determined that the prime-boost combinations of IM followed by IN immunization (IM + IN) or IN followed by IN immunization (IN + IN) exhibited strong spike-specific IgG, IgA and T cell response in vaccinated K18 knock-in mice. Hamsters vaccinated with two doses of VSV expressing SARS-CoV-2 spike, both delivered by IN or IM + IN, showed strong protection against SARS-CoV-2 variants of concern Alpha and Delta. This protection was also observed in aged hamsters. Our study underscores the highly crucial role immunization routes have with the VSV vector platform to elicit a strong and protective immune response.
Collapse
Affiliation(s)
- Saina Beitari
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Gerard Agbayani
- Immunomodulation, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Bassel Akache
- Immunomodulation, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Ita Hadžisejdić
- Clinical Department of Pathology and Cytology Clinical Hospital Center Rijeka, University of Rijeka, Rijeka, Croatia
| | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
364
|
Apaa T, Withers AJ, Mackenzie L, Staley C, Dessi N, Blanchard A, Bennett M, Bremner-Harrison S, Chadwick EA, Hailer F, Harrison SWR, Lambin X, Loose M, Mathews F, Tarlinton R. Lack of detection of SARS-CoV-2 in British wildlife 2020-21 and first description of a stoat ( Mustela erminea) Minacovirus. J Gen Virol 2023; 104:001917. [PMID: 38059490 PMCID: PMC10770931 DOI: 10.1099/jgv.0.001917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/23/2023] [Indexed: 12/08/2023] Open
Abstract
Repeat spillover of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into new hosts has highlighted the critical role of cross-species transmission of coronaviruses and establishment of new reservoirs of virus in pandemic and epizootic spread of coronaviruses. Species particularly susceptible to SARS-CoV-2 spillover include Mustelidae (mink, ferrets and related animals), cricetid rodents (hamsters and related animals), felids (domestic cats and related animals) and white-tailed deer. These predispositions led us to screen British wildlife with sarbecovirus-specific quantitative PCR and pan coronavirus PCR assays for SARS-CoV-2 using samples collected during the human pandemic to establish if widespread spillover was occurring. Fourteen wildlife species (n=402) were tested, including: two red foxes (Vulpes vulpes), 101 badgers (Meles meles), two wild American mink (Neogale vison), 41 pine marten (Martes martes), two weasels (Mustela nivalis), seven stoats (Mustela erminea), 108 water voles (Arvicola amphibius), 39 bank voles (Myodes glareolous), 10 field voles (Microtus agrestis), 15 wood mice (Apodemus sylvaticus), one common shrew (Sorex aranaeus), two pygmy shrews (Sorex minutus), two hedgehogs (Erinaceus europaeus) and 75 Eurasian otters (Lutra lutra). No cases of SARS-CoV-2 were detected in any animals, but a novel minacovirus related to mink and ferret alphacoronaviruses was detected in stoats recently introduced to the Orkney Islands. This group of viruses is of interest due to pathogenicity in ferrets. The impact of this virus on the health of stoat populations remains to be established.
Collapse
Affiliation(s)
- Ternenge Apaa
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
- Animal and Plant Health Agency, Addlestone, Surrey, UK
| | - Amy J. Withers
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
- Animal and Plant Health Agency, Addlestone, Surrey, UK
| | - Laura Mackenzie
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Ceri Staley
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | - Nicola Dessi
- National Wildlife Management Centre, Animal and Plant Health Agency, Sand Hutton, York, UK
| | - Adam Blanchard
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | - Malcolm Bennett
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | - Samantha Bremner-Harrison
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, UK
- Vincent Wildlife Trust, Eastnor, Ledbury, UK
| | | | - Frank Hailer
- Organisms and Environment, School of Biosciences, Cardiff University, Cardiff, UK
| | - Stephen W. R. Harrison
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, UK
| | - Xavier Lambin
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Matthew Loose
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Fiona Mathews
- School of Life Sciences, University of Sussex, Sussex, UK
| | - Rachael Tarlinton
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| |
Collapse
|
365
|
Moreira-Soto A, Bruno A, de Mora D, Paez M, Garces J, Wulf B, Sander AL, Olmedo M, Basantes Mantilla MJ, Gonzalez Gonzalez M, Orlando SA, Salgado Cisneros S, Zevallos JC, Drexler JF. Virological evidence of the impact of non-pharmaceutical interventions against COVID-19 in Ecuador, a resource-limited setting. Emerg Microbes Infect 2023; 12:2259001. [PMID: 37698611 PMCID: PMC10563623 DOI: 10.1080/22221751.2023.2259001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/10/2023] [Indexed: 09/13/2023]
Abstract
Ecuador had substantial COVID-19-mortality during 2020 despite early implementation of non-pharmaceutical interventions (NPIs). Resource-limited settings like Ecuador have high proportions of informal labour which entail high human mobility, questioning efficacy of NPIs. We performed a retrospective observational study in Ecuador's national reference laboratory for viral respiratory infections during March 2020-February 2021 using stored respiratory specimens from 1950 patients, corresponding to 2.3% of all samples analysed within the Ecuadorian national surveillance system per week. During 2020, detection of SARS-CoV-2 (Pearson correlation; r = -0.74; p = 0.01) and other respiratory viruses (Pearson correlation; r = -0.68; p = 0.02) by real-time RT-PCR correlated negatively with NPIs stringency. Among respiratory viruses, adenoviruses (Fisher's exact-test; p = 0.026), parainfluenzaviruses (p = 0.04), enteroviruses (p < 0.0001) and metapneumoviruses (p < 0.0001) occurred significantly more frequently during months of absent or non-stringent NPIs (characterized by <55% stringency according to the Oxford stringency index data for Ecuador). Phylogenomic analyses of 632 newly characterized SARS-CoV-2 genomes revealed 100 near-parallel SARS-CoV-2 introductions during early 2020 in the absence of NPIs. NPI stringency correlated negatively with the number of circulating SARS-CoV-2 lineages during 2020 (r = -0.69; p = 0.02). Phylogeographic reconstructions showed differential SARS-CoV-2 dispersion patterns during 2020, with more short-distance transitions potentially associated with recreational activity during non-stringent NPIs. There were also fewer geographic transitions during strict NPIs (n = 450) than during non-stringent or absent NPIs (n = 580). Virological evidence supports that NPIs had an effect on virus spread and distribution in Ecuador, providing a template for future epidemics in resource-limited settings and contributing to a balanced assessment of societal costs entailed by strict NPIs.
Collapse
Affiliation(s)
- Andres Moreira-Soto
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Institute of Virology, Berlin, Germany
- Tropical Disease Research Program, School of Veterinary Medicine, Universidad Nacional, Costa Rica, Costa Rica
| | - Alfredo Bruno
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
- Universidad Agraria del Ecuador, Guayaquil, Ecuador
| | - Doménica de Mora
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
| | - Michelle Paez
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
| | - Jimmy Garces
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
| | - Ben Wulf
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Anna-Lena Sander
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Maritza Olmedo
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
| | | | - Manuel Gonzalez Gonzalez
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
- Universidad ECOTEC, Km 13.5 Samborondon, Samborondon, EC092302, Ecuador
| | - Solon Alberto Orlando
- National Institute of Public Health Research (INSPI), Guayaquil, Ecuador
- Health Science Faculty, Universidad Espíritu Santo, Guayaquil, Ecuador
| | | | - Juan Carlos Zevallos
- Health Science Faculty, Universidad Espíritu Santo, Guayaquil, Ecuador
- Alianza para la Investigación de Enfermedades Emergentes (AIE), Quito, Ecuador
| | - Jan Felix Drexler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Institute of Virology, Berlin, Germany
- German Centre for Infection Research (DZIF), Associated Partner Site Charité, Berlin, Germany
| |
Collapse
|
366
|
Ghram A, Latiri I, Methnani J, Souissi A, Benzarti W, Toulgui E, Ben Saad H. Effects of cardiorespiratory rehabilitation program on submaximal exercise in patients with long-COVID-19 conditions: a systematic review of randomized controlled trials and recommendations for future studies. Expert Rev Respir Med 2023; 17:1095-1124. [PMID: 38063359 DOI: 10.1080/17476348.2023.2293226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/06/2023] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Long-COVID-19 patients (LC19Ps) often experience cardiovascular and respiratory complications. Cardiorespiratory rehabilitation programs (CRRPs) have emerged as promising interventions to enhance exercise capacity in this population. This systematic review aimed to assess the impact of CRRPs on submaximal exercise performance, specifically the 6-minute walk test (6MWT) outcomes, in LC19Ps through an analysis of available randomized controlled trials (RCTs). METHODS A systematic search was conducted in PubMed/Medline and Scopus to identify relevant RCTs. Six RCTs meeting inclusion criteria were included in this review, investigating the effects of CRRPs on 6MWT outcomes in LC19Ps. RESULTS The findings from the included RCTs provide compelling evidence supporting the effectiveness of CRRPs in improving submaximal exercise performance in LC19Ps. These results underscore the potential of CRRPs to enhance submaximal exercise capacity and overall functional well-being in this population. However, future research is imperative to determine optimal CRRPs, including duration, intensity, and specific intervention components. Additionally, the long-term sustainability and durability of CRRP-induced improvements warrant further exploration. Future studies should prioritize patient-centric outcomes and address potential implementation barriers. CONCLUSION CRRPs show promise in ameliorating submaximal exercise performance among LC19Ps. Further research is needed to refine these programs and ensure their lasting impact on this patient group. SYSTEMATIC REVIEW REGISTRATION https://doi.org/10.17605/OSF.IO/HMN38. [Figure: see text].
Collapse
Affiliation(s)
- Amine Ghram
- Department of Cardiac Rehabilitation, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Imed Latiri
- Heart Failure (LR12SP09) Research Laboratory, Farhat HACHED Hospital, Sousse, Tunisia
- Laboratory of Physiology, Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia
| | - Jabeur Methnani
- LR19ES09, Laboratoire de Physiologie de l'Exercice et Physiopathologie: de l'Intégré au Moléculaire 10 « Biologie, Médecine et Santé », Faculty of Medicine of Sousse, Sousse, Tunisia
| | - Amine Souissi
- Heart Failure (LR12SP09) Research Laboratory, Farhat HACHED Hospital, Sousse, Tunisia
| | - Wafa Benzarti
- Department of Pneumology, Farhat HACHED Hospital, Sousse, Tunisia
| | - Emna Toulgui
- Department of Physical Medicine and Rehabilitation, Sahloul Hospital, Sousse, Tunisia
| | - Helmi Ben Saad
- Heart Failure (LR12SP09) Research Laboratory, Farhat HACHED Hospital, Sousse, Tunisia
- Laboratory of Physiology, Faculty of Medicine of Sousse, University of Sousse, Sousse, Tunisia
- Department of Physiology and Functional Exploration, Farhat HACHED Hospital, Sousse, Tunisia
| |
Collapse
|
367
|
Machowski EE, Reyneke AE, Sher DE, Kana BD. Production and Performance Assessment of a Severe Acute Respiratory Syndrome Coronavirus 2 Biomimetic in a Verification Program for Pandemic Readiness. J Mol Diagn 2023; 25:907-912. [PMID: 37863192 DOI: 10.1016/j.jmoldx.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/28/2023] [Accepted: 08/24/2023] [Indexed: 10/22/2023] Open
Abstract
During the early stages of the 2019 coronavirus disease (COVID-19) pandemic in South Africa, one of many challenges included availability of control material for laboratory proficiency testing programs. Proficiency testing control material using live severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or RNA extracted from cell culture was either biohazardous or costly, particularly in resource-limited settings. This study reports the development and application of a noninfectious SARS-CoV-2 biomimetic Mycobacterium smegmatis strain that mimics a positive result in the GeneXpert SARS-CoV-2 Xpert Xpress cartridge. Nucleotide sequences located in genes encoding the RNA-dependent RNA polymerase, the nucleocapsid, and the envelope proteins were used. The resulting biomimetic strain was prepared as a positive proficiency testing control and distributed in South Africa for verification of laboratories before their testing of clinical specimens. Between April and December 2020, a total of 151 GeneXpert instruments with 2532 modules were verified to bring COVID-19 mass testing in South Africa online. An average concordance of 98.6% was noted in the entire laboratory network, allowing identification of false-positive/false-negative results and instrument errors. This noninfectious, easily scalable proficiency testing control material became available within 2 months after the start of the pandemic in South Africa and represents a useful approach to consider for other diseases and future pandemics.
Collapse
Affiliation(s)
- Edith E Machowski
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Anna E Reyneke
- SmartSpot Quality (Pty) Limited, Johannesburg, South Africa
| | - Dean E Sher
- SmartSpot Quality (Pty) Limited, Johannesburg, South Africa
| | - Bavesh D Kana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa.
| |
Collapse
|
368
|
Xia T, Fu X, Fulham M, Wang Y, Feng D, Kim J. CT-based Radiogenomics Framework for COVID-19 Using ACE2 Imaging Representations. J Digit Imaging 2023; 36:2356-2366. [PMID: 37553526 PMCID: PMC10584804 DOI: 10.1007/s10278-023-00895-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 which enters the body via the angiotensin-converting enzyme 2 (ACE2) and altering its gene expression. Altered ACE2 plays a crucial role in the pathogenesis of COVID-19. Gene expression profiling, however, is invasive and costly, and is not routinely performed. In contrast, medical imaging such as computed tomography (CT) captures imaging features that depict abnormalities, and it is widely available. Computerized quantification of image features has enabled 'radiogenomics', a research discipline that identifies image features that are associated with molecular characteristics. Radiogenomics between ACE2 and COVID-19 has yet to be done primarily due to the lack of ACE2 expression data among COVID-19 patients. Similar to COVID-19, patients with lung adenocarcinoma (LUAD) exhibit altered ACE2 expression and, LUAD data are abundant. We present a radiogenomics framework to derive image features (ACE2-RGF) associated with ACE2 expression data from LUAD. The ACE2-RGF was then used as a surrogate biomarker for ACE2 expression. We adopted conventional feature selection techniques including ElasticNet and LASSO. Our results show that: i) the ACE2-RGF encoded a distinct collection of image features when compared to conventional techniques, ii) the ACE2-RGF can classify COVID-19 from normal subjects with a comparable performance to conventional feature selection techniques with an AUC of 0.92, iii) ACE2-RGF can effectively identify patients with critical illness with an AUC of 0.85. These findings provide unique insights for automated COVID-19 analysis and future research.
Collapse
Affiliation(s)
- Tian Xia
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Xiaohang Fu
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael Fulham
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Dagan Feng
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jinman Kim
- School of Computer Science, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
369
|
Kallem P, Hegab HM, Alsafar H, Hasan SW, Banat F. SARS-CoV-2 detection and inactivation in water and wastewater: review on analytical methods, limitations and future research recommendations. Emerg Microbes Infect 2023; 12:2222850. [PMID: 37279167 PMCID: PMC10286680 DOI: 10.1080/22221751.2023.2222850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/03/2023] [Indexed: 06/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been detected in wastewater. Wastewater-based epidemiology (WBE) is a practical and cost-effective tool for the assessment and controlling of pandemics and probably for examining SARS-CoV-2 presence. Implementation of WBE during the outbreaks is not without limitations. Temperature, suspended solids, pH, and disinfectants affect the stability of viruses in wastewater. Due to these limitations, instruments and techniques have been utilized to detect SARS-CoV-2. SARS-CoV-2 has been detected in sewage using various concentration methods and computer-aided analyzes. RT-qPCR, ddRT-PCR, multiplex PCR, RT-LAMP, and electrochemical immunosensors have been employed to detect low levels of viral contamination. Inactivation of SARS-CoV-2 is a crucial preventive measure against coronavirus disease 2019 (COVID-19). To better assess the role of wastewater as a transmission route, detection, and quantification methods need to be refined. In this paper, the latest improvements in quantification, detection, and inactivation of SARS-CoV-2 in wastewater are explained. Finally, limitations and future research recommendations are thoroughly described.
Collapse
Affiliation(s)
- Parashuram Kallem
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Chemical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Environmental Health and Safety Program, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Hanaa M Hegab
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Chemical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology (BTC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Emirates Bio-research center, Ministry of Interior, Abu Dhabi, United Arab Emirates
| | - Shadi W. Hasan
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Chemical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fawzi Banat
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Chemical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
370
|
Storm L, Toftgaard Roensbo M, Jensen SK, Leth S, Jensen-Fangel S, Thomsen MK. Sampling from upper and/or lower respiratory tract when diagnosing coronavirus disease 2019 (COVID-19) - results from a Danish cohort study. Infect Dis (Lond) 2023; 55:831-838. [PMID: 37565740 DOI: 10.1080/23744235.2023.2245046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can replicate in the upper and lower respiratory tract. We aimed to evaluate whether symptom characteristics and symptom duration prior to sampling are associated with test positivity in upper or lower respiratory tract samples. METHODS We conducted a retrospective cohort study in the Central Denmark Region from 14 April 2020 to 2 November 2020 including hospitalised patients with SARS-CoV-2 reverse transcriptase-polymerase chain reaction samples from both the upper and lower respiratory tract within 48 h and at least one positive test result. RESULTS Of 122 patients, 101 were positive in both samples (83%), 7 (5%) were positive only in the upper respiratory tract sample, and 14 (11%) were only positive in the lower respiratory tract sample. The median number of symptoms was 4 (IQR 3, 5.75) and 5 (IQR 3, 7), respectively, in patients with only a positive upper respiratory tract sample and in concordant positive patients; while 1 (IQR 1, 3) in patients with only a positive upper respiratory tract sample. 98% (120/122) of patients would have been diagnosed with coronavirus disease 2019 if supplemental sampling from the lower respiratory tract was guided by lower respiratory tract symptoms. No substantial difference in the duration of symptoms was observed across the three patient groups. CONCLUSIONS The presence of lower respiratory tract symptoms could have been used to determine whether supplemental sampling from the lower respiratory tract was necessary. Symptom duration was not associated with test positivity in the upper or lower respiratory tract.
Collapse
Affiliation(s)
- Line Storm
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus N, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, Denmark
| | | | - Simon Kok Jensen
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Denmark
| | - Steffen Leth
- Department of Infectious Diseases and Internal Medicine, Gødstrup Hospital, Herning, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Søren Jensen-Fangel
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, Denmark
| | - Marianne Kragh Thomsen
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
371
|
Yousefi M, Lee WS, Chan WOY, He W, Mah MG, Yong CL, Deerain JM, Wang L, Arcinas C, Yan B, Tan D, Sia WR, Gamage AM, Yang J, Hsu ACY, Li S, Linster M, Yang X, Ghosh S, Anderson DE, Smith GJD, Tan CW, Wang LF, Ooi YS. Betacoronaviruses SARS-CoV-2 and HCoV-OC43 infections in IGROV-1 cell line require aryl hydrocarbon receptor. Emerg Microbes Infect 2023; 12:2256416. [PMID: 37672505 PMCID: PMC10512916 DOI: 10.1080/22221751.2023.2256416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
The emergence of novel betacoronaviruses has posed significant financial and human health burdens, necessitating the development of appropriate tools to combat future outbreaks. In this study, we have characterized a human cell line, IGROV-1, as a robust tool to detect, propagate, and titrate betacoronaviruses SARS-CoV-2 and HCoV-OC43. IGROV-1 cells can be used for serological assays, antiviral drug testing, and isolating SARS-CoV-2 variants from patient samples. Using time-course transcriptomics, we confirmed that IGROV-1 cells exhibit a robust innate immune response upon SARS-CoV-2 infection, recapitulating the response previously observed in primary human nasal epithelial cells. We performed genome-wide CRISPR knockout genetic screens in IGROV-1 cells and identified Aryl hydrocarbon receptor (AHR) as a critical host dependency factor for both SARS-CoV-2 and HCoV-OC43. Using DiMNF, a small molecule inhibitor of AHR, we observed that the drug selectively inhibits HCoV-OC43 infection but not SARS-CoV-2. Transcriptomic analysis in primary normal human bronchial epithelial cells revealed that DiMNF blocks HCoV-OC43 infection via basal activation of innate immune responses. Our findings highlight the potential of IGROV-1 cells as a valuable diagnostic and research tool to combat betacoronavirus diseases.
Collapse
Affiliation(s)
- Meisam Yousefi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Wai Suet Lee
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Wharton O. Y. Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Wei He
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Marcus G. Mah
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Cythia Lingli Yong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Joshua M. Deerain
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lijin Wang
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Camille Arcinas
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Biaoguo Yan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Dewei Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Wan Rong Sia
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Akshamal M. Gamage
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jinxuan Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, People’s Republic of China
| | - Alan Chen-Yu Hsu
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, Australia
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Shang Li
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Martin Linster
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Xinglou Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, People’s Republic of China
| | - Sujoy Ghosh
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Danielle E. Anderson
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Gavin J. D. Smith
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Chee Wah Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Infectious Diseases Translation Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lin-Fa Wang
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yaw Shin Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
372
|
Yi L, Peng S, Liu P, Liang X, Gao Q. A novel auxiliary diagnostic model for COVID-19 screening using enzymes based detection of serum biomarkers and clinical characteristics. Clin Exp Med 2023; 23:4967-4974. [PMID: 37925683 DOI: 10.1007/s10238-023-01233-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
The screening for the coronavirus disease 2019 (COVID-19) based on virus nuclear acid detection and radiology has encountered unprecedented difficulties due to the shortage of kits and facilities, and the lack of sensitivity and specificity, especially for developing countries. The study aimed to develop an auxiliary diagnostic score based on age, biomarkers, clinical characteristics (ABC) to rapidly and accurately screen COVID-19. Serum biomarkers were detected by enzymes catalyzed reaction method which is rapid and accurate. A retrospective case-control study among Chinese patients with laboratory-confirmed COVID-19 and those without COVID-19 was conducted. Data of age, sex, signs and symptoms, history of disease, complete blood counts, and serum biochemical items such as ALT and AST were used to establish the diagnostic model. ALT/AST was detected by enzymes based biochemical reaction method. Stepwise logistic regression and random forests with variable selection process were used to establish the model. Ten-fold cross-validation and out-of-bag error were used to assess the accuracy of the models. Decision curve analysis was used to compare different models. A total of 279 cases and 253 controls were recruited, with mean age of 60.7 ± 13.7 and 42.6 ± 20.2, respectively. The regression model selected nine variables with Kappa of 0.77, sensitivity of 0.90, and specificity of 0.87. The random forests retained eight variables with Kappa of 0.76, sensitivity of 0.87, and specificity of 0.89. Based on decision curve analysis, the ABC-based diagnostic score provided the largest net benefit compared to other models with less data input. Subgroup analysis by excluding severely ill cases showed a decreased but acceptable sensitivity (0.75) and unchanged overall accuracy. The ABC-based diagnostic score provides a rapid and valid method for COVID-19 screening, especially for regions with a shortage of facilities. The model performs less well in the diagnosis of mild-to-moderate COVID-19.
Collapse
Affiliation(s)
- Lang Yi
- Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Peilin Liu
- Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xianghui Liang
- Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Gao
- Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
373
|
Shiohara A, Wojnilowicz M, Lyu Q, Pei Y, Easton CD, Chen Y, White JF, McAuley A, Prieto‐Simon B, Thissen H, Voelcker NH. SARS-CoV-2 Virus Detection Via a Polymeric Nanochannel-Based Electrochemical Biosensor. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205281. [PMID: 36585382 PMCID: PMC9880620 DOI: 10.1002/smll.202205281] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/14/2022] [Indexed: 06/17/2023]
Abstract
The development of simple, cost-effective, rapid, and quantitative diagnostic tools remains critical to monitor infectious COVID-19 disease. Although numerous diagnostic platforms, including rapid antigen tests, are developed and used, they suffer from limited accuracy, especially when tested with asymptomatic patients. Here, a unique approach to fabricate a nanochannel-based electrochemical biosensor that can detect the entire virion instead of virus fragments, is demonstrated. The sensing platform has uniform nanoscale channels created by the convective assembly of polystyrene (PS) beads on gold electrodes. The PS beads are then functionalized with bioreceptors while the gold surface is endowed with anti-fouling properties. When added to the biosensor, SARS-CoV-2 virus particles block the nanochannels by specific binding to the bioreceptors. The nanochannel blockage hinders the diffusion of a redox probe; and thus, allows quantification of the viral load by measuring the changes in the oxidation current before and after virus incubation. The biosensor shows a low limit of detection of ≈1.0 viral particle mL-1 with a wide detection range up to 108 particles mL-1 in cell culture media. Moreover, the biosensor is able to differentiate saliva samples with SARS-CoV-2 from those without, demonstrating the potential of this technology for translation into a point-of-care biosensor product.
Collapse
Affiliation(s)
- Amane Shiohara
- Drug Delivery, Deposition,and DynamicsMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoria3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
- Melbourne Centre of NanofabricationVictorian Node of the Australian National Fabrication FacilityClaytonVictoria3168Australia
| | - Marcin Wojnilowicz
- Drug Delivery, Deposition,and DynamicsMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoria3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Quanxia Lyu
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Yi Pei
- Drug Delivery, Deposition,and DynamicsMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoria3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Christopher D. Easton
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Yu Chen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Jacinta F White
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Alexander McAuley
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Beatriz Prieto‐Simon
- Department of Electronic EngineeringUniversitat Rovira i VirgiliTarragona43007Spain
- ICREAPg. Lluís Companys 23Barcelona08010Spain
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
| | - Nicolas H Voelcker
- Drug Delivery, Deposition,and DynamicsMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoria3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVictoria3168Australia
- Melbourne Centre of NanofabricationVictorian Node of the Australian National Fabrication FacilityClaytonVictoria3168Australia
| |
Collapse
|
374
|
Nguyen HT, Denkinger CM, Brenner S, Koeppel L, Brugnara L, Burk R, Knop M, Bärnighausen T, Deckert A, De Allegri M. Cost and cost-effectiveness of four different SARS-CoV-2 active surveillance strategies: evidence from a randomised control trial in Germany. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2023; 24:1545-1559. [PMID: 36656403 PMCID: PMC9850332 DOI: 10.1007/s10198-022-01561-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
INTRODUCTION The COVID-19 pandemic has entered its third year and continues to affect most countries worldwide. Active surveillance, i.e. testing individuals irrespective of symptoms, presents a promising strategy to accurately measure the prevalence of SARS-CoV-2. We aimed to identify the most cost-effective active surveillance strategy for COVID-19 among the four strategies tested in a randomised control trial between 18th November 2020 and 23rd December 2020 in Germany. The four strategies included: (A1) direct testing of individuals; (A2) direct testing of households; (B1) testing conditioned on upstream COVID-19 symptom pre-screening of individuals; and (B2) testing conditioned on upstream COVID-19 symptom pre-screening of households. METHODS We adopted a health system perspective and followed an activity-based approach to costing. Resource consumption data were collected prospectively from a digital individual database, daily time records, key informant interviews and direct observations. Our cost-effectiveness analysis compared each strategy with the status quo and calculated the average cost-effective ratios (ACERs) for one primary outcome (sample tested) and three secondary outcomes (responder recruited, case detected and asymptomatic case detected). RESULTS Our results showed that A2, with cost per sample tested at 52,89 EURO, had the lowest ACER for the primary outcome, closely followed by A1 (63,33 EURO). This estimate was much higher for both B1 (243,84 EURO) and B2 (181,06 EURO). CONCLUSION A2 (direct testing at household level) proved to be the most cost-effective of the four evaluated strategies and should be considered as an option to strengthen the routine surveillance system in Germany and similar settings.
Collapse
Affiliation(s)
- Hoa Thi Nguyen
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany.
| | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
- German Center for Infection Research (DZIF), Im Neuenheimer Feld 344, Heidelberg, Germany
| | - Stephan Brenner
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Lisa Koeppel
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Lucia Brugnara
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- evaplan GmbH at the University Hospital Heidelberg, Ringstr.19B, 69115, Heidelberg, Germany
| | - Robin Burk
- Center for Molecular Biology (ZMBH), Heidelberg University, Im Neuenheimer Feld 282, 69120, Heidelberg, Germany
| | - Michael Knop
- Center for Molecular Biology (ZMBH), Heidelberg University, Im Neuenheimer Feld 282, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), ZMBH Alliance, 69120, Heidelberg, Germany
| | - Till Bärnighausen
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Andreas Deckert
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Manuela De Allegri
- Heidelberg Institute of Global Health, University Hospital and Medical Faculty, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| |
Collapse
|
375
|
Flagg M, Goldin K, Pérez-Pérez L, Singh M, Williamson BN, Pruett N, Hoang CD, de Wit E. Low level of tonic interferon signalling is associated with enhanced susceptibility to SARS-CoV-2 variants of concern in human lung organoids. Emerg Microbes Infect 2023; 12:2276338. [PMID: 37883246 PMCID: PMC10732190 DOI: 10.1080/22221751.2023.2276338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
There is tremendous heterogeneity in the severity of COVID-19 disease in the human population, and the mechanisms governing the development of severe disease remain incompletely understood. The emergence of SARS-CoV-2 variants of concern (VOC) Delta (B.1.617.2) and Omicron (B.1.1.529) further compounded this heterogeneity. Virus replication and host cell damage in the distal lung is often associated with severe clinical disease, making this an important site to consider when evaluating pathogenicity of SARS-CoV-2 VOCs. Using distal human lung organoids (hLOs) derived from multiple human donors, we compared the fitness and pathogenicity of SARS-CoV-2 VOC Delta and Omicron, along with an ancestral clade B variant D614G, and evaluated donor-dependent differences in susceptibility to infection. We observed substantial attenuation of Omicron in hLOs and demonstrated enhanced susceptibility to Omicron and D614G replication in hLOs from one donor. Transcriptomic analysis revealed that increased susceptibility to SARS-CoV-2 infection in these hLOs was associated with reduced tonic interferon signaling activity at baseline. We show that hLOs can be used to model heterogeneity of SARS-CoV-2 pathogenesis in humans, and propose that variability in tonic interferon signaling set point may impact susceptibility to SARS-CoV-2 VOCs and subsequent COVID-19 disease progression.
Collapse
Affiliation(s)
- Meaghan Flagg
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kerry Goldin
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Lizzette Pérez-Pérez
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Manmeet Singh
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Brandi N. Williamson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Nathanael Pruett
- Thoracic Surgery Branch, Division of Intramural Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chuong D. Hoang
- Thoracic Surgery Branch, Division of Intramural Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
376
|
Vargas-De-León C, Cureño-Díaz MA, Salazar MI, Cruz-Cruz C, Loyola-Cruz MÁ, Durán-Manuel EM, Zamora-Pacheco ER, Bravata-Alcántara JC, Lugo-Zamudio GE, Fernández-Sánchez V, Bello-López JM, Ibáñez-Cervantes G. Neutralizing Antibodies against SARS-CoV-2: Importance of Comorbidities in Health Personnel against Reinfections. Viruses 2023; 15:2354. [PMID: 38140595 PMCID: PMC10747730 DOI: 10.3390/v15122354] [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: 09/30/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
One of the priority lines of action to contain the SARS-CoV-2 pandemic was vaccination programs for healthcare workers. However, with the emergence of highly contagious strains, such as the Omicron variant, it was necessary to know the serological status of health personnel to make decisions for the application of reinforcements. The aim of this work was to determine the seroprevalence against SARS-CoV-2 in healthcare workers in a Mexican hospital after six months of the administration of the Pfizer-BioNTech vaccine (two doses, 4 weeks apart) and to investigate the association between comorbidities, response to the vaccine, and reinfections. Neutralizing antibodies against SARS-CoV-2 were determined using ELISA assays for 262 employees of Hospital Juárez de México with and without a history of COVID-19. A beta regression analysis was performed to study the associated comorbidities and their relationship with the levels of antibodies against SARS-CoV-2. Finally, an epidemiological follow-up was carried out to detect reinfections in this population. A significant difference in SARS-CoV-2 seroprevalence was observed in workers with a history of COVID-19 prior to vaccination compared to those without a history of the disease (MD: 0.961 and SD: 0.049; <0.001). Beta regression showed that workers with a history of COVID-19 have greater protection compared to those without a history of the infection. Neutralizing antibodies were found to be decreased in alcoholic and diabetic subjects (80.1%). Notably, eight cases of Omicron reinfections were identified, and gender and obesity were associated with the presence of reinfections (6.41 OR; 95% BCa CI: 1.15, 105.0). The response to the vaccine was influenced by the history of SARS-CoV-2 infection and associated comorbidities. The above highlights the importance of prioritizing this segment of the population for reinforcements in periods of less than one year to guarantee their effectiveness against new variants.
Collapse
Affiliation(s)
- Cruz Vargas-De-León
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
- Laboratorio de Modelación Bioestadística para la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | | | - Ma. Isabel Salazar
- Laboratorio Nacional de Vacunología y Virus Tropicales, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional, Ciudad de México 11340, México;
| | - Clemente Cruz-Cruz
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
| | - Miguel Ángel Loyola-Cruz
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
| | - Emilio Mariano Durán-Manuel
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
| | - Edwin Rodrigo Zamora-Pacheco
- Laboratorio de Modelación Bioestadística para la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | | | | | - Verónica Fernández-Sánchez
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
| | - Juan Manuel Bello-López
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
| | - Gabriela Ibáñez-Cervantes
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (C.V.-D.-L.); (C.C.-C.); (M.Á.L.-C.); (E.M.D.-M.)
- Laboratorio de Modelación Bioestadística para la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| |
Collapse
|
377
|
Zumbrun EE, Zak SE, Lee ED, Bowling PA, Ruiz SI, Zeng X, Koehler JW, Delp KL, Bakken RR, Hentschel SS, Bloomfield HA, Ricks KM, Clements TL, Babka AM, Dye JM, Herbert AS. SARS-CoV-2 Aerosol and Intranasal Exposure Models in Ferrets. Viruses 2023; 15:2341. [PMID: 38140582 PMCID: PMC10747480 DOI: 10.3390/v15122341] [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: 11/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the worldwide COVID-19 pandemic. Animal models are extremely helpful for testing vaccines and therapeutics and for dissecting the viral and host factors that contribute to disease severity and transmissibility. Here, we report the assessment and comparison of intranasal and small particle (~3 µm) aerosol SARS-CoV-2 exposure in ferrets. The primary endpoints for analysis were clinical signs of disease, recovery of the virus in the upper respiratory tract, and the severity of damage within the respiratory tract. This work demonstrated that ferrets were productively infected with SARS-CoV-2 following either intranasal or small particle aerosol exposure. SARS-CoV-2 infection of ferrets resulted in an asymptomatic disease course following either intranasal or small particle aerosol exposure, with no clinical signs, significant weight loss, or fever. In both aerosol and intranasal ferret models, SARS-CoV-2 replication, viral genomes, and viral antigens were detected within the upper respiratory tract, with little to no viral material detected in the lungs. The ferrets exhibited a specific IgG immune response to the SARS-CoV-2 full spike protein. Mild pathological findings included inflammation, necrosis, and edema within nasal turbinates, which correlated to positive immunohistochemical staining for the SARS-CoV-2 virus. Environmental sampling was performed following intranasal exposure of ferrets, and SARS-CoV-2 genomic material was detected on the feeders and nesting areas from days 2-10 post-exposure. We conclude that both intranasal and small particle aerosol ferret models displayed measurable parameters that could be utilized for future studies, including transmission studies and testing SARS-CoV-2 vaccines and therapeutics.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Samantha E. Zak
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Eric D. Lee
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Philip A. Bowling
- Division of Veterinary Medicine, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Sara I. Ruiz
- Division of Bacteriology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA;
| | - Xiankun Zeng
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Jeffrey W. Koehler
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Korey L. Delp
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Russel R. Bakken
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Shannon S. Hentschel
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Holly A. Bloomfield
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - Keersten M. Ricks
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - Tamara L. Clements
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (J.W.K.); (K.L.D.); (K.M.R.); (T.L.C.)
| | - April M. Babka
- Division of Pathology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (E.D.L.); (X.Z.); (H.A.B.); (A.M.B.)
| | - John M. Dye
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| | - Andrew S. Herbert
- Division of Virology, United States Army Medical Research Institute of Infectious Disease, Frederick, MD 21702, USA; (S.E.Z.); (R.R.B.); (S.S.H.); (J.M.D.); (A.S.H.)
| |
Collapse
|
378
|
Qi L, Zhang Z, Wang M, Ke Z, Mao H, Deng G, Wang J. One-plasmid double-expression system for preparation of MS2 virus-like particles packaging SARS-CoV-2 RNA. Front Cell Infect Microbiol 2023; 13:1238543. [PMID: 38094745 PMCID: PMC10716189 DOI: 10.3389/fcimb.2023.1238543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
COVID-19 is a disease caused by a virus named SARS-CoV-2. SARS-CoV-2 is a single-stranded positive-sense RNA virus. Reverse transcription quantitative PCR (RT-qPCR) assays are the gold standard molecular test for detection of RNA viruses. The aim of this study was to construct an RNA-positive control based on MS2 phage-like particles (MS2 VLPs) to detect SARS-CoV-2 RNA. pCDFDuet-1 was used as a one-plasmid double-expression system to construct MS2 VLPs containing ssRNA of SARS-CoV-2. The sequence encoding one copy of maturase, His-tag and coat protein dimer was cloned and inserted into MCS1 of the plasmid; the fragment encoding protein N and ORF1ab from SARS-CoV-2 was cloned and inserted into MCS2. The prepared plasmid was transformed into Escherichia coli strain BL2 (DE3), and expression of the construct was induced by 1 mM isopropyl-L-thio-D-galactopyranoside (IPTG) at 30°C for 12 hours. MS2 VLPs were purified and collected with Ni-NTA affinity chromatography columns. The size and shape of the MS2 VLPs were verified by transmission electron microscopy, and the stability of MS2 VLP packaged RNA was evaluated by treatment with RNase A. Effects of storage temperature and buffer on MS2 VLP stability were also investigated. The results showed that SARS-CoV-2 MS2 VLPs could be successfully produced by this one-plasmid double-expression system. MS2 VLPs showed high stability and may be used as a positive control in molecular diagnosis of COVID-19.
Collapse
Affiliation(s)
- Lili Qi
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Zheng Zhang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Mengting Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Zhijian Ke
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Haiguang Mao
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| | - Gang Deng
- Blood Transfusion Research Institute, Ningbo Central Blood Station, Ningbo, Zhejiang, China
| | - Jinbo Wang
- School of Biological and Chemical Engineering, NingboTech University, Ningbo, Zhejiang, China
| |
Collapse
|
379
|
Ashmore P, Sherwood E. An overview of COVID-19 global epidemiology and discussion of potential drivers of variable global pandemic impacts. J Antimicrob Chemother 2023; 78:ii2-ii11. [PMID: 37995358 PMCID: PMC10666997 DOI: 10.1093/jac/dkad311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023] Open
Abstract
With a WHO-estimated excess mortality burden of 14.9 million over the course of 2020 and 2021, the COVID-19 pandemic has had a major human impact so far. It has also affected a range of disciplines, systems and practices from mathematical modelling to behavioural sciences, pharmaceutical development to health system management. This article explores these developments and, to set the scene, this paper summarizes the global epidemiology of COVID-19 from January 2020 to June 2021 and considers some potential drivers of variation.
Collapse
Affiliation(s)
- Polly Ashmore
- Health Education England, Stewart House, 32 Russell Square, London WC1B 5DN, UK
| | - Emma Sherwood
- Health Education England, Stewart House, 32 Russell Square, London WC1B 5DN, UK
| |
Collapse
|
380
|
Kang M, Jeong E, Kim JY, Yun SA, Jang MA, Jang JH, Kim TY, Huh HJ, Lee NY. Optimization of extraction-free protocols for SARS-CoV-2 detection using a commercial rRT-PCR assay. Sci Rep 2023; 13:20364. [PMID: 37990045 PMCID: PMC10663557 DOI: 10.1038/s41598-023-47645-0] [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: 08/16/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023] Open
Abstract
In the ongoing global fight against coronavirus disease 2019 (COVID-19), the sample preparation process for real-time reverse transcription polymerase chain reaction (rRT-PCR) faces challenges due to time-consuming steps, labor-intensive procedures, contamination risks, resource demands, and environmental implications. However, optimized strategies for sample preparation have been poorly investigated, and the combination of RNase inhibitors and Proteinase K has been rarely considered. Hence, we investigated combinations of several extraction-free protocols incorporating heat treatment, sample dilution, and Proteinase K and RNase inhibitors, and validated the effectiveness using 120 SARS-CoV-2 positive and 62 negative clinical samples. Combining sample dilution and heat treatment with Proteinase K and RNase inhibitors addition exhibited the highest sensitivity (84.26%) with a mean increase in cycle threshold (Ct) value of + 3.8. Meanwhile, combined sample dilution and heat treatment exhibited a sensitivity of 79.63%, accounting for a 38% increase compared to heat treatment alone. Our findings highlight that the incorporation of Proteinase K and RNase inhibitors with sample dilution and heat treatment contributed only marginally to the improvement without yielding statistically significant differences. Sample dilution significantly impacts SARS-CoV-2 detection, and sample conditions play a crucial role in the efficiency of extraction-free methods. Our findings may provide insights for streamlining diagnostic testing, enhancing its accessibility, cost-effectiveness, and sustainability.
Collapse
Affiliation(s)
- Minhee Kang
- Smart Healthcare Research Institute, Biomedical Engineering Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Eunjung Jeong
- Smart Healthcare Research Institute, Biomedical Engineering Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Ji-Yeon Kim
- Samsung Biomedical Research Institute, Center for Clinical Medicine, Samsung Medical Center, Seoul, South Korea
| | - Sun Ae Yun
- Samsung Biomedical Research Institute, Center for Clinical Medicine, Samsung Medical Center, Seoul, South Korea
| | - Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tae Yeul Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Hee Jae Huh
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Nam Yong Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
381
|
Russo M, Mendes-Corrêa MC, Lins BB, Kersten V, Pernambuco Filho PCA, Martins TR, Tozetto-Mendoza TR, Vilas Boas LS, Gomes BM, Dati LMM, Duarte-Neto AN, Reigado GR, Frederico ABT, de Brito e Cunha DRDA, de Paula AV, da Silva JIG, Vasconcelos CFM, Chambergo FS, Nunes VA, Ano Bom APD, Castilho LR, Martins RAP, Hirata MH, Mirotti L. Intranasal Liposomal Formulation of Spike Protein Adjuvanted with CpG Protects and Boosts Heterologous Immunity of hACE2 Transgenic Mice to SARS-CoV-2 Infection. Vaccines (Basel) 2023; 11:1732. [PMID: 38006064 PMCID: PMC10675295 DOI: 10.3390/vaccines11111732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Mucosal vaccination appears to be suitable to protect against SARS-CoV-2 infection. In this study, we tested an intranasal mucosal vaccine candidate for COVID-19 that consisted of a cationic liposome containing a trimeric SARS-CoV-2 spike protein and CpG-ODNs, a Toll-like receptor 9 agonist, as an adjuvant. In vitro and in vivo experiments indicated the absence of toxicity following the intranasal administration of this vaccine formulation. First, we found that subcutaneous or intranasal vaccination protected hACE-2 transgenic mice from infection with the wild-type (Wuhan) SARS-CoV-2 strain, as shown by weight loss and mortality indicators. However, when compared with subcutaneous administration, the intranasal route was more effective in the pulmonary clearance of the virus and induced higher neutralizing antibodies and anti-S IgA titers. In addition, the intranasal vaccination afforded protection against gamma, delta, and omicron virus variants of concern. Furthermore, the intranasal vaccine formulation was superior to intramuscular vaccination with a recombinant, replication-deficient chimpanzee adenovirus vector encoding the SARS-CoV-2 spike glycoprotein (Oxford/AstraZeneca) in terms of virus lung clearance and production of neutralizing antibodies in serum and bronchial alveolar lavage (BAL). Finally, the intranasal liposomal formulation boosted heterologous immunity induced by previous intramuscular vaccination with the Oxford/AstraZeneca vaccine, which was more robust than homologous immunity.
Collapse
Affiliation(s)
- Momtchilo Russo
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Maria Cássia Mendes-Corrêa
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Bruna B. Lins
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Victor Kersten
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Paulo C. A. Pernambuco Filho
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Toni Ricardo Martins
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas (UFAM), Manaus 69080-900, Brazil
| | - Tânia Regina Tozetto-Mendoza
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Lucy Santos Vilas Boas
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - Brisa Moreira Gomes
- Department of Immunology, Institute of Biomedical Science, University of São Paulo (ICB-USP), São Paulo 05508-000, Brazil
| | - Livia Mendonça Munhoz Dati
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciências Farmacêuticas da Universidade de Sao Paulo (FCF-USP), São Paulo 05508-000, Brazil (M.H.H.)
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil
| | - Gustavo Roncoli Reigado
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Ana Beatriz T. Frederico
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Danielle R. de A. de Brito e Cunha
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Anderson Vicente de Paula
- Laboratório de Virologia (LIM52), Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina da Universidade de São Paulo (FM-USP), São Paulo 05403-000, Brazil; (M.C.M.-C.); (T.R.M.)
| | - José Igor G. da Silva
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Carlos F. Moreira Vasconcelos
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Felipe S. Chambergo
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Viviane Abreu Nunes
- Laboratório de Biotecnologia, Escola de Artes, Ciências e Humanidades, Universidade de São Paulo (EACH-USP), São Paulo 03828-000, Brazil (F.S.C.); (V.A.N.)
| | - Ana Paula Dinis Ano Bom
- Immunological Technology Laboratory, Institute of Immunobiological Technology (Bio-Manguinhos), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil (A.P.D.A.B.)
| | - Leda R. Castilho
- Cell Culture Engineering Laboratory, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-598, Brazil;
| | - Rodrigo A. P. Martins
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil (R.A.P.M.)
| | - Mario Hiroyuki Hirata
- Departamento de Analises Clinicas e Toxicologicas, Faculdade de Ciências Farmacêuticas da Universidade de Sao Paulo (FCF-USP), São Paulo 05508-000, Brazil (M.H.H.)
| | - Luciana Mirotti
- Institute of Science and Technology in Biomodels (ICTB), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
382
|
Kotwa JD, Lobb B, Massé A, Gagnier M, Aftanas P, Banerjee A, Banete A, Blais-Savoie J, Bowman J, Buchanan T, Chee HY, Kruczkiewicz P, Nirmalarajah K, Soos C, Vernygora O, Yip L, Lindsay LR, McGeer AJ, Maguire F, Lung O, Doxey AC, Pickering B, Mubareka S. Genomic and transcriptomic characterization of delta SARS-CoV-2 infection in free-ranging white-tailed deer ( Odocoileus virginianus). iScience 2023; 26:108319. [PMID: 38026171 PMCID: PMC10665813 DOI: 10.1016/j.isci.2023.108319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/12/2023] [Accepted: 10/20/2023] [Indexed: 11/29/2023] Open
Abstract
White-tailed deer (WTD) are susceptible to SARS-CoV-2 and represent an important species for surveillance. Samples from WTD (n = 258) collected in November 2021 from Québec, Canada were analyzed for SARS-CoV-2 RNA. We employed viral genomics and host transcriptomics to further characterize infection and investigate host response. We detected Delta SARS-CoV-2 (B.1.617.2) in WTD from the Estrie region; sequences clustered with human sequences from October 2021 from Vermont, USA, which borders this region. Mutations in the S-gene and a deletion in ORF8 were detected. Host expression patterns in SARS-CoV-2 infected WTD were associated with the innate immune response, including signaling pathways related to anti-viral, pro- and anti-inflammatory signaling, and host damage. We found limited correlation between genes associated with innate immune response from human and WTD nasal samples, suggesting differences in responses to SARS-CoV-2 infection. Our findings provide preliminary insights into host response to SARS-CoV-2 infection in naturally infected WTD.
Collapse
Affiliation(s)
| | - Briallen Lobb
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ariane Massé
- Ministère de l’Environnement, de la Lutte contre les changements climatiques, de la Faune et des Parcs, Québec City, QC G1S 4X4, Canada
| | - Marianne Gagnier
- Ministère de l’Environnement, de la Lutte contre les changements climatiques, de la Faune et des Parcs, Québec City, QC G1S 4X4, Canada
| | | | - Arinjay Banerjee
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Andra Banete
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | | | - Jeff Bowman
- Wildlife Research and Monitoring Section, Ontario Ministry of Natural Resources and Forestry, Peterborough, ON K9J 8M5, Canada
| | - Tore Buchanan
- Wildlife Research and Monitoring Section, Ontario Ministry of Natural Resources and Forestry, Peterborough, ON K9J 8M5, Canada
| | - Hsien-Yao Chee
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Global Health Research Center and Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, Jiangsu 215316, China
| | - Peter Kruczkiewicz
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
| | | | - Catherine Soos
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, Saskatoon, SK S7N 3H5, Canada
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Oksana Vernygora
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
| | - Lily Yip
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - L. Robbin Lindsay
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Allison J. McGeer
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Finlay Maguire
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Community Health & Epidemiology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Shared Hospital Laboratory, Toronto, ON M4N 3M5, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Andrew C. Doxey
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Bradley Pickering
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
- Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
383
|
Agarwal S, Hamidizadeh M, Bier FF. Detection of Reverse Transcriptase LAMP-Amplified Nucleic Acid from Oropharyngeal Viral Swab Samples Using Biotinylated DNA Probes through a Lateral Flow Assay. BIOSENSORS 2023; 13:988. [PMID: 37998163 PMCID: PMC10669123 DOI: 10.3390/bios13110988] [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: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
This study focuses on three key aspects: (a) crude throat swab samples in a viral transport medium (VTM) as templates for RT-LAMP reactions; (b) a biotinylated DNA probe with enhanced specificity for LFA readouts; and (c) a digital semi-quantification of LFA readouts. Throat swab samples from SARS-CoV-2 positive and negative patients were used in their crude (no cleaning or pre-treatment) forms for the RT-LAMP reaction. The samples were heat-inactivated but not treated for any kind of nucleic acid extraction or purification. The RT-LAMP (20 min processing time) product was read out by an LFA approach using two labels: FITC and biotin. FITC was enzymatically incorporated into the RT-LAMP amplicon with the LF-LAMP primer, and biotin was introduced using biotinylated DNA probes, specifically for the amplicon region after RT-LAMP amplification. This assay setup with biotinylated DNA probe-based LFA readouts of the RT-LAMP amplicon was 98.11% sensitive and 96.15% specific. The LFA result was further analysed by a smartphone-based IVD device, wherein the T-line intensity was recorded. The LFA T-line intensity was then correlated with the qRT-PCR Ct value of the positive swab samples. A digital semi-quantification of RT-LAMP-LFA was reported with a correlation coefficient of R2 = 0.702. The overall RT-LAMP-LFA assay time was recorded to be 35 min with a LoD of three RNA copies/µL (Ct-33). With these three advancements, the nucleic acid testing-point of care technique (NAT-POCT) is exemplified as a versatile biosensor platform with great potential and applicability for the detection of pathogens without the need for sample storage, transportation, or pre-processing.
Collapse
Affiliation(s)
- Saloni Agarwal
- Institute for Biochemistry and Biology, Chair of Molecular Bioanalysis and Bioelectronics, University of Potsdam, Karl-Liebknecht-Strasse 24/25, 14476 Potsdam, Germany; (S.A.); (M.H.)
| | - Mojdeh Hamidizadeh
- Institute for Biochemistry and Biology, Chair of Molecular Bioanalysis and Bioelectronics, University of Potsdam, Karl-Liebknecht-Strasse 24/25, 14476 Potsdam, Germany; (S.A.); (M.H.)
| | - Frank F. Bier
- Institute for Biochemistry and Biology, Chair of Molecular Bioanalysis and Bioelectronics, University of Potsdam, Karl-Liebknecht-Strasse 24/25, 14476 Potsdam, Germany; (S.A.); (M.H.)
- Institute for Molecular Diagnostics and Bioanalysis-IMDB gGmbH, Am Mühlenberg 10, 14476 Potsdam, Germany
| |
Collapse
|
384
|
Biava M, Notari S, Grassi G, Bordi L, Tartaglia E, Agrati C, Cimini E, Sberna G, Nicastri E, Antinori A, Girardi E, Vaia F, Maggi F, Lalle E. In Vitro and In Vivo Crosstalk between Type I IFN and IL-8 Responses in SARS-CoV-2 Infection. Microorganisms 2023; 11:2787. [PMID: 38004798 PMCID: PMC10672883 DOI: 10.3390/microorganisms11112787] [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: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
COVID-19 patients show characteristic over-expression of different cytokines that may interfere with the interferon (IFN) response, delaying its production. Within the overexpressed cytokines, IL-8 plays a key role, and it may impede IFN-I activation. PBMC from eight healthy donors were exposed to 2019-nCoV/Italy-INMI1 isolate and supernatants/cells were collected at different time points; the production of either IFN-alpha or IL-8 was assessed. The same analysis was performed on plasma samples obtained from 87 COVID-19 patients. Antagonism between IFN-alpha and IL-8 was observed, since in those PBMC with medium or high IL-8 levels, IFN-α levels were low. The same scenario was observed in SARS-CoV-2-infected patients that were divided into three groups based on IL-8 low, medium and high levels; the correlation between low levels of IFN-α and high levels of IL-8 was statistically significant in both the IL-8 medium and IL-8 high group. Overall, our results showed a crosstalk/antagonism between IL-8 and IFN-alpha in PBMC from healthy donors challenged with SARS-CoV-2 and inversely proportional IFN-alpha levels to IL-8 concentrations detected in plasma samples from COVID-19 patients, suggesting that the impairment of the innate immune response in COVID-19 patients may be linked to a dysregulated cytokine response, namely through IL-8 production.
Collapse
Affiliation(s)
- Mirella Biava
- Laboratory of Virology and Biosafety Laboratories, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy (G.S.)
| | - Stefania Notari
- Cellular Immunology Laboratory, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Germana Grassi
- Cellular Immunology Laboratory, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Licia Bordi
- Laboratory of Virology and Biosafety Laboratories, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy (G.S.)
| | - Eleonora Tartaglia
- Cellular Immunology Laboratory, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Chiara Agrati
- Unit of Pathogen Specific Immunity, Ospedale Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Giuseppe Sberna
- Laboratory of Virology and Biosafety Laboratories, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy (G.S.)
| | - Emanuele Nicastri
- Clinical and Research Department, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Andrea Antinori
- Clinical and Research Department, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Enrico Girardi
- Scientific Direction, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Francesco Vaia
- General and Health Management Direction, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology and Biosafety Laboratories, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy (G.S.)
| | - Eleonora Lalle
- Laboratory of Virology and Biosafety Laboratories, National Institute of Infectious Diseases “L. Spallanzani”, IRCCS, 00149 Rome, Italy (G.S.)
| |
Collapse
|
385
|
Marques GVDL, Marques DPDA, Clarindo FA, Avendaño-Villarreal JA, Guerra FS, Fernandes PD, Dos Santos EN, Gusevskaya EV, Kohlhoff M, Moreira FDA, Andrade LAF, Fonseca FGD, Dos-Reis JGAC, Oliveira RBD. Synthesis of cannabidiol-based compounds as ACE2 inhibitors with potential application in the treatment of COVID-19. Eur J Med Chem 2023; 260:115760. [PMID: 37657273 DOI: 10.1016/j.ejmech.2023.115760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023]
Abstract
Cannabis is a general name for plants of the genus Cannabis. Used as fiber, medicine, drug, for religious, therapeutic, and hedonistic purposes along the millenia, it is mostly known for its psychoactive properties. One of its major constituents, cannabidiol (CBD), a non-psychoactive substance, among many other biological activities, has shown potential as an anti-SARS-CoV-2 drug. In this work, three derivatives and an analogue of CBD were synthesized, and cell viability and antiviral activities were evaluated. None of the compounds showed cytotoxicity up to a maximum concentration of 100 μM and, in contrast, displayed a significant antiviral activity, superior to remdesivir and nafamostat mesylate, with IC50 values ranging from 9.4 to 1.9 μM. In order to search for a possible molecular target, the inhibitory activity of the compounds against ACE2 was investigated, with expressive results (IC50 ranging from 3.96 μM to 0.01 μM).
Collapse
Affiliation(s)
- Gabriel Vitor de Lima Marques
- Universidade Federal de Minas Gerais, Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Belo Horizonte, Brazil
| | | | - Felipe Alves Clarindo
- Universidade Federal de Minas Gerais, Departamento de Microbiologia, Instituto de Ciências Biológicas, Belo Horizonte, Brazil
| | | | - Fabiana Sélos Guerra
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pesquisa em Descoberta de Fármacos, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brazil
| | - Patrícia Dias Fernandes
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pesquisa em Descoberta de Fármacos, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brazil
| | - Eduardo Nicolau Dos Santos
- Universidade Federal de Minas Gerais, Departamento de Química, Instituto de Ciências Exatas, Belo Horizonte, Brazil
| | - Elena Vitalievna Gusevskaya
- Universidade Federal de Minas Gerais, Departamento de Química, Instituto de Ciências Exatas, Belo Horizonte, Brazil
| | - Markus Kohlhoff
- Instituto René Rachou (IRR) - FIOCRUZ Minas, Química de Produtos Naturais Bioativos (QPNB), Belo Horizonte, Brazil
| | - Fabrício de Araújo Moreira
- Universidade Federal de Minas Gerais, Departamento de Farmacologia, Instituto de Ciências Biológicas, Belo Horizonte, Brazil
| | - Luis Adan Flores Andrade
- Universidade Federal de Minas Gerais, Departamento de Microbiologia, Instituto de Ciências Biológicas, Belo Horizonte, Brazil; Universidade Federal de Minas Gerais, Centro de Tecnologia de Vacinas - CT Vacinas, Belo Horizonte, Brazil
| | - Flávio Guimarães da Fonseca
- Universidade Federal de Minas Gerais, Departamento de Microbiologia, Instituto de Ciências Biológicas, Belo Horizonte, Brazil; Universidade Federal de Minas Gerais, Centro de Tecnologia de Vacinas - CT Vacinas, Belo Horizonte, Brazil
| | | | - Renata Barbosa de Oliveira
- Universidade Federal de Minas Gerais, Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Belo Horizonte, Brazil.
| |
Collapse
|
386
|
Zhu D, Huang J, Hu B, Cao D, Chen D, Song X, Chen J, Zhou H, Cen A, Hou T. Trial of the Pluslife SARS-CoV-2 Nucleic Acid Rapid Test Kit: Prospective Cohort Study. JMIR Public Health Surveill 2023; 9:e48107. [PMID: 37962934 PMCID: PMC10650960 DOI: 10.2196/48107] [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: 04/11/2023] [Revised: 06/28/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND In response to the SARS-CoV-2 epidemic, a convenient, rapid, and sensitive diagnostic method for detecting COVID-19 is crucial for patient control and timely treatment. OBJECTIVE This study aimed to validate the detection of SARS-CoV-2 with the Pluslife SARS-CoV-2 rapid test kit developed based on a novel thermostatic amplification technique called RNase hybridization-assisted amplification. METHODS From November 25 to December 8, 2022, patients with suspected or confirmed COVID-19, close contacts, and health care workers at high risk of exposure were recruited from 3 hospitals and 1 university. Respiratory specimens were collected for testing with the Pluslife SARS-CoV-2 rapid test kit and compared with reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and a commercial antigen assay kit. Samples from 1447 cases were obtained from 3 "ready-to-test" scenarios in which samples were collected on site and tested immediately, and samples from 503 cases were obtained from a "freeze-thaw test" scenario in which samples were collected, frozen, and thawed for testing. RESULTS Pluslife SARS-CoV-2 rapid testing of samples from the "ready-to-test" scenario was found to be accurate (overall sensitivity and specificity of 98.3% and 99.3%, respectively) and diagnostically useful (positive and negative likelihood ratios of 145.45 and 0.02, respectively). Pluslife SARS-CoV-2 rapid testing of samples from the "freeze-thaw test" scenario was also found to be accurate (overall sensitivity and specificity of 71.2% and 98.6%, respectively) and diagnostically useful (positive and negative likelihood ratios of 51.01 and 0.67, respectively). Our findings demonstrated that the time efficiency and accuracy of the results in a "ready-to-test" scenario were better. The time required from sample preparation to the seeing the result of the Pluslife SARS-CoV-2 rapid test was 10 to 38 minutes, which was substantially shorter than that of RT-qPCR (at least 90 minutes). In addition, the diagnostic efficacy of the Pluslife SARS-CoV-2 rapid test was better than that of a commercial antigen assay kit. CONCLUSIONS The developed RNase hybridization-assisted amplification assay provided rapid, sensitive, and convenient detection of SARS-CoV-2 infection and may be useful for enhanced detection of COVID-19 in homes, high-risk industries, and hospitals.
Collapse
Affiliation(s)
- Dandan Zhu
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Huang
- Fever Clinic, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bei Hu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Dingqiang Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinqiang Song
- Party Committee Office and President Office, Guangdong University of Finance and Economics, Guangzhou, China
| | - Jialing Chen
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hao Zhou
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Aiqun Cen
- Fever Clinic, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Tieying Hou
- Hospital Office, Huazhong University of Science and Technology Union Shenzhen Hospital/Shenzhen Nanshan People's Hospital, Shenzhen, China
| |
Collapse
|
387
|
Alewaidat H, Bataineh Z, Bani-Ahmad M, Alali M, Almakhadmeh A. Investigation of the diagnostic importance and accuracy of CT in the chest compared to the RT-PCR test for suspected COVID-19 patients in Jordan. F1000Res 2023; 12:741. [PMID: 37822316 PMCID: PMC10562777 DOI: 10.12688/f1000research.130388.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 10/13/2023] Open
Abstract
This article aims to synthesize the existing literature on the implementation of public policies to incentivize the development of treatments for rare diseases, (diseases with very low prevalence and therefore with low commercial interest) otherwise known as orphan drugs. The implementation of these incentives in the United States (US), Japan, and in the European Union (EU) seems to be related to a substantial increase in treatments for these diseases, and has influenced the way the pharmaceutical research & development (R&D) system operates beyond this policy area. Despite the success of the Orphan Drug model, the academic literature also highlights the negative implications that these public policies have on affordability and access to orphan drugs, as well as on the prioritization of certain disease rare areas over others. The synthesis focuses mostly on the United States' Orphan Drug Act (ODA) as a model for subsequent policies in other regions and countries. It starts with a historical overview of the creation of the term "rare diseases", continues with a summary of the evidence available on the US ODA's positive and negative impacts, and provides a summary of the different proposals to reform these incentives in light of the negative outcomes described. Finally, it describes some key aspects of the Japanese and European policies, as well as some of the challenges captured in the literature related to their impact in Low- and Middle-Income Countries (LMICs).
Collapse
Affiliation(s)
- Haytham Alewaidat
- Applied Medical Sciences, Jordan University of Science and Technology, irbid, 22110, Jordan
| | - Ziad Bataineh
- Anatomy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad Bani-Ahmad
- Medical Laboratory Science, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Manar Alali
- Medical Laboratory Science, Zarqa University, Zarqa, Jordan
| | - Ali Almakhadmeh
- Radiologic Technology, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
388
|
Boby ML, Fearon D, Ferla M, Filep M, Koekemoer L, Robinson MC, Chodera JD, Lee AA, London N, von Delft A, von Delft F. Open science discovery of potent noncovalent SARS-CoV-2 main protease inhibitors. Science 2023; 382:eabo7201. [PMID: 37943932 PMCID: PMC7615835 DOI: 10.1126/science.abo7201] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 10/09/2023] [Indexed: 11/12/2023]
Abstract
We report the results of the COVID Moonshot, a fully open-science, crowdsourced, and structure-enabled drug discovery campaign targeting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease. We discovered a noncovalent, nonpeptidic inhibitor scaffold with lead-like properties that is differentiated from current main protease inhibitors. Our approach leveraged crowdsourcing, machine learning, exascale molecular simulations, and high-throughput structural biology and chemistry. We generated a detailed map of the structural plasticity of the SARS-CoV-2 main protease, extensive structure-activity relationships for multiple chemotypes, and a wealth of biochemical activity data. All compound designs (>18,000 designs), crystallographic data (>490 ligand-bound x-ray structures), assay data (>10,000 measurements), and synthesized molecules (>2400 compounds) for this campaign were shared rapidly and openly, creating a rich, open, and intellectual property-free knowledge base for future anticoronavirus drug discovery.
Collapse
Affiliation(s)
- Melissa L. Boby
- Pharmacology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Program in Chemical Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Program in Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daren Fearon
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, UK
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Matteo Ferla
- Oxford Biomedical Research Centre, National Institute for Health Research, University of Oxford, Oxford, UK
| | - Mihajlo Filep
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Lizbé Koekemoer
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - John D. Chodera
- Program in Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Nir London
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Annette von Delft
- Oxford Biomedical Research Centre, National Institute for Health Research, University of Oxford, Oxford, UK
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Frank von Delft
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, UK
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg 2006, South Africa
| |
Collapse
|
389
|
Schröder S, Richter A, Veith T, Emanuel J, Gudermann L, Friedmann K, Jeworowski LM, Mühlemann B, Jones TC, Müller MA, Corman VM, Drosten C. Characterization of intrinsic and effective fitness changes caused by temporarily fixed mutations in the SARS-CoV-2 spike E484 epitope and identification of an epistatic precondition for the evolution of E484A in variant Omicron. Virol J 2023; 20:257. [PMID: 37940989 PMCID: PMC10633978 DOI: 10.1186/s12985-023-02154-4] [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: 06/23/2023] [Accepted: 08/08/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Intrinsic fitness costs are likely to have guided the selection of lineage-determining mutations during emergence of variants of SARS-CoV-2. Whereas changes in receptor affinity and antibody neutralization have been thoroughly mapped for individual mutations in spike, their influence on intrinsic replicative fitness remains understudied. METHODS We analyzed mutations in immunodominant spike epitope E484 that became temporarily fixed over the pandemic. We engineered the resulting immune escape mutations E484K, -A, and -Q in recombinant SARS-CoV-2. We characterized viral replication, entry, and competitive fitness with and without immune serum from humans with defined exposure/vaccination history and hamsters monospecifically infected with the E484K variant. We additionally engineered a virus containing the Omicron signature mutations N501Y and Q498R that were predicted to epistatically enhance receptor binding. RESULTS Multistep growth kinetics in Vero-, Calu-3, and NCI-H1299 were identical between viruses. Synchronized entry experiments based on cold absorption and temperature shift identified only an insignificant trend toward faster entry of the E484K variant. Competitive passage experiments revealed clear replicative fitness differences. In absence of immune serum, E484A and E484Q, but not E484K, were replaced by wildtype (WT) in competition assays. In presence of immune serum, all three mutants outcompeted WT. Decreased E484A fitness levels were over-compensated for by N501Y and Q498R, identifying a putative Omicron founder background that exceeds the intrinsic and effective fitness of WT and matches that of E484K. Critically, the E484A/Q498R/N501Y mutant and E484K have equal fitness also in presence of pre-Omicron vaccinee serum, whereas the fitness gain by E484K is lost in the presence of serum raised against the E484K variant in hamsters. CONCLUSIONS The emergence of E484A and E484Q prior to widespread population immunity may have been limited by fitness costs. In populations already exposed to the early immune escape epitope E484K, the Omicron founder background may have provided a basis for alternative immune escape evolution via E484A. Studies of major antigenic epitope changes with and without their epistatic context help reconstruct the sequential adjustments of intrinsic fitness versus neutralization escape during the evolution of major SARS-CoV-2 variants in an increasingly immune human population.
Collapse
Affiliation(s)
- Simon Schröder
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Richter
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Talitha Veith
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jackson Emanuel
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Luca Gudermann
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kirstin Friedmann
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lara M Jeworowski
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Mühlemann
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Terry C Jones
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing St, CB2 3EJ, Cambridge, U.K
| | - Marcel A Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
390
|
Ahmad I, Merla A, Ali F, Shah B, AlZubi AA, AlZubi MA. A deep transfer learning approach for COVID-19 detection and exploring a sense of belonging with Diabetes. Front Public Health 2023; 11:1308404. [PMID: 38026271 PMCID: PMC10657998 DOI: 10.3389/fpubh.2023.1308404] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
COVID-19 is an epidemic disease that results in death and significantly affects the older adult and those afflicted with chronic medical conditions. Diabetes medication and high blood glucose levels are significant predictors of COVID-19-related death or disease severity. Diabetic individuals, particularly those with preexisting comorbidities or geriatric patients, are at a higher risk of COVID-19 infection, including hospitalization, ICU admission, and death, than those without Diabetes. Everyone's lives have been significantly changed due to the COVID-19 outbreak. Identifying patients infected with COVID-19 in a timely manner is critical to overcoming this challenge. The Real-Time Polymerase Chain Reaction (RT-PCR) diagnostic assay is currently the gold standard for COVID-19 detection. However, RT-PCR is a time-consuming and costly technique requiring a lab kit that is difficult to get in crises and epidemics. This work suggests the CIDICXR-Net50 model, a ResNet-50-based Transfer Learning (TL) method for COVID-19 detection via Chest X-ray (CXR) image classification. The presented model is developed by substituting the final ResNet-50 classifier layer with a new classification head. The model is trained on 3,923 chest X-ray images comprising a substantial dataset of 1,360 viral pneumonia, 1,363 normal, and 1,200 COVID-19 CXR images. The proposed model's performance is evaluated in contrast to the results of six other innovative pre-trained models. The proposed CIDICXR-Net50 model attained 99.11% accuracy on the provided dataset while maintaining 99.15% precision and recall. This study also explores potential relationships between COVID-19 and Diabetes.
Collapse
Affiliation(s)
- Ijaz Ahmad
- Digital Transition, Innovation and Health Service, Leonardo da Vinci Telematic University, Chieti, Italy
| | - Arcangelo Merla
- Department of Engineering and Geology (INGEO) University "G. d’Annunzio" Chieti-Pescara, Pescara, Italy
| | - Farman Ali
- Department of Computer Science and Engineering, School of Convergence, College of Computing and Informatics, Sungkyunkwan University, Seoul, Republic of Korea
| | - Babar Shah
- College of Technological Innovation, Zayed University, Dubai, United Arab Emirates
| | - Ahmad Ali AlZubi
- Department of Computer Science, Community College, King Saud University, Riyadh, Saudi Arabia
| | - Mallak Ahmad AlZubi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
391
|
Batta I, Kaur T, Agrawal DK. Distinguishing Swine Flu (H1N1) from COVID-19: Clinical, Virological, and Immunological Perspectives. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2023; 7:271-280. [PMID: 37994372 PMCID: PMC10664801 DOI: 10.26502/ami.936500125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
This article provides an in-depth examination on the differences between the influenza A strain, H1N1 (also called Swine Flu) and Covid-19 focusing on the immune response and clinical symptoms. Flu symptoms due to influenza A strain, H1N1, were initially discovered in 2009. This variant of influenza A is believed to have emerged through reassortment, a process where the resulting virus inherits gene segments from each of its parental viruses. This reassortment event has resulted in a variant with altered characteristics, potentially affecting the level of immunity in humans. The symptoms of this strain typically manifest 1-4 days after exposure and include fever, cough, sore throat, runny/stuffy nose, body aches, fatigue, and gastrointestinal symptoms such as diarrhea. The transmission dynamics of this new variant, including human-to-human transmission, are still under investigation by health authorities. Individuals with weakened immune systems are generally more susceptible to severe illness. Risk factors associated with swine flu can include older adults, young children, pregnant women, and individuals with obesity. Historical variants of swine flu, such as the 2015 variant in India, have been associated with significant case numbers and deaths, often due to respiratory failure. Since the epidemic of Covid-19 due to SARS-CoV2 in early 2020, several symptoms of COVID-19 and swine flu overlap. In this article, we critically reviewed the differences and similarities in the immune response and clinical symptoms due to H1N1 virus and SARS-CoV2 in human.
Collapse
Affiliation(s)
- Irene Batta
- Bothell High School, Bothell, Washington, USA
| | - Tejinder Kaur
- Department of Zoology, DAV University, Jallandhar, Punjab, India
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
392
|
Nogareda F, Regan AK, Couto P, Fowlkes AL, Gharpure R, Loayza S, Leite JA, Rodríguez A, Vicari A, Azziz-Baumgartner E, Salas D. Effectiveness of COVID-19 vaccines against hospitalisation in Latin America during three pandemic waves, 2021-2022: a test-negative case-control design. LANCET REGIONAL HEALTH. AMERICAS 2023; 27:100626. [PMID: 38035125 PMCID: PMC10682274 DOI: 10.1016/j.lana.2023.100626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/10/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023]
Abstract
Background Vaccine effectiveness (VE) is essential to monitor the performance of vaccines and generate strategic information to guide decision making. We pooled data from six Latin American countries to estimate the effectiveness of COVID-19 vaccines in preventing laboratory-confirmed SARS-CoV-2 hospitalisation during three different pandemic waves from February 2021 to September 2022. Methods We used a test-negative case-control design in hospitalised adults in Chile, Costa Rica, Ecuador, Guatemala, Paraguay, and Uruguay. We estimated adjusted VE by age group (18-64 and ≥65 years), vaccine type and product for primary series vaccination and booster vaccination and by time since last dose during the Omicron variant dominant period. We used mixed effects logistic regression models adjusting for sex, age, week of onset of symptom onset and pre-existing conditions with country fit as a random effect term. Findings We included 15,241 severe acute respiratory infection (SARI) patients in the analysis. Among adults 18-64 years, VE estimates for primary series vaccination during pre-Delta and Delta periods ranged by product from 66.5% to 95.1% and from 33.5% to 88.2% for older adults. During the Omicron period, VE estimates for primary series were lower and decreased by time since last vaccination, but VE increased to between 26.4% and 57.4% when a booster was administered. Interpretation mRNA and viral vector vaccines presented higher VE for both primary series and booster. While VE decreased over time, protection against severe COVID-19-associated hospitalisation increased when booster doses were administered. Vaccination with additional doses should be recommended, particularly for persons at increased risk of developing severe COVID-19. Funding This work was supported by a grant from the U.S. Centers for Disease Control and Prevention (CDC) through cooperative agreements with the Pan American Health Organization/World Health Organization.
Collapse
Affiliation(s)
| | - Annette K. Regan
- Pan American Health Organization, Washington, DC, USA
- School of Nursing and Health Professions, University of San Francisco, USA
- Fielding School of Public Health, University of California Los Angeles, USA
| | - Paula Couto
- Pan American Health Organization, Washington, DC, USA
| | - Ashley L. Fowlkes
- National Center for Immunization and Respiratory Diseases, U.S. CDC, USA
| | - Radhika Gharpure
- National Center for Immunization and Respiratory Diseases, U.S. CDC, USA
| | - Sergio Loayza
- Pan American Health Organization, Washington, DC, USA
| | | | | | - Andrea Vicari
- Pan American Health Organization, Washington, DC, USA
| | | | - Daniel Salas
- Pan American Health Organization, Washington, DC, USA
| |
Collapse
|
393
|
Gómez‐Rojas S, Segura GP, Ollé J, Carreño Gómez‐Tarragona G, Medina JG, Aguado JM, Guerrero EV, Santaella MP, Martínez‐López J. A machine learning tool for the diagnosis of SARS-CoV-2 infection from hemogram parameters. J Cell Mol Med 2023; 27:3423-3430. [PMID: 37882471 PMCID: PMC10660618 DOI: 10.1111/jcmm.17864] [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: 01/12/2023] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 10/27/2023] Open
Abstract
Monocytes and neutrophils play key roles in the cytokine storm triggered by SARS-CoV-2 infection, which changes their conformation and function. These changes are detectable at the cellular and molecular level and may be different to what is observed in other respiratory infections. Here, we applied machine learning (ML) to develop and validate an algorithm to diagnose COVID-19 using blood parameters. In this retrospective single-center study, 49 hemogram parameters from 12,321 patients with clinical suspicion of COVID-19 and tested by RT-PCR (4239 positive and 8082 negative) were analysed. The dataset was randomly divided into training and validation sets. Blood cell parameters and patient age were used to construct the predictive model with the support vector machine (SVM) tool. The model constructed from the training set (5936 patients) achieved an accuracy for diagnosis of SARS-CoV-2 infection of 0.952 (95% CI: 0.875-0.892). Test sensitivity and specificity was 0.868 and 0.899, respectively, with a positive (PPV) and negative (NPV) predictive value of 0.896 and 0.872, respectively (prevalence 0.50). The validation set model (4964 patients) achieved an accuracy of 0.894 (95% CI: 0.883-0.903). Test sensitivity and specificity was 0.8922 and 0.8951, respectively, with a positive (PPV) and negative (NPV) predictive value of 0.817 and 0.94, respectively (prevalence 0.34). The area under the receiver operating characteristic curve was 0.952 for the algorithm performance. This algorithm may allow to rule out COVID-19 diagnosis with 94% of probability. This represents a great advance for early diagnostic orientation and guiding clinical decisions.
Collapse
Affiliation(s)
- S. Gómez‐Rojas
- Department of HematologyHospital Universitario 12 octubreMadridSpain
| | - G. Pérez Segura
- Department of HematologyHospital Universitario 12 octubreMadridSpain
| | - J. Ollé
- Conceptos Claros CoBarcelonaSpain
| | | | - J. González Medina
- Department of HematologyHospital Universitario Fundación Jiménez DíazMadridSpain
| | - J. M. Aguado
- Unit of Infectious DiseasesHospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (i+12), CIBERINFEC, ISCIIIMadridSpain
- Department of Medicine, School of MedicineUniversidad ComplutenseMadridSpain
| | - E. Vera Guerrero
- Department of HematologyHospital Universitario 12 octubreMadridSpain
| | - M. Poza Santaella
- Department of HematologyHospital Universitario 12 octubreMadridSpain
| | - J. Martínez‐López
- Department of HematologyHospital Universitario 12 octubreMadridSpain
- Department of Medicine, School of MedicineUniversidad ComplutenseMadridSpain
| |
Collapse
|
394
|
Yang H, Hu J, Tan BK, Wong KH, Huang JJ, Cheung PC, Lin S. Lesson learned from COVID-19 pandemic for the future of food industry. Heliyon 2023; 9:e22479. [PMID: 38045130 PMCID: PMC10689951 DOI: 10.1016/j.heliyon.2023.e22479] [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: 06/06/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023] Open
Abstract
With WHO announcing COVID-19 no longer as a public health emergency of international concern (PHEIC) on May 5, 2023, coupled with the fact that the majority of the countries of the world have dropped strict city lockdown or border closure, this perhaps signals the end of the COVID-19 crisis caused by the SARS-CoV-2 virus. However, the COVID-19 pandemic has resulted in far-reaching effects affecting nearly every aspect of our lives and society. Notably, the food industry including agriculture, food manufacturers, food logistics, distributors and retailers have all felt the profound impact and had experienced significant stress during the pandemic. Therefore, it is essential to retrospect the lessons that can be learned from this pandemic for the food industry. This short review aims to address the food safety issues related to the COVID-19 pandemic by focusing on its foodborne transmission potential, innovations of virus detection strategies suitable for food industry; development of phathogenicaidal methods and devices to inactivate SARS-CoV-2 virus (particularly in industrial scale); and the set-up of related food regulations and guidelines as preventive and control measures for preventing the spread of SARS-CoV-2 virus through the food supply chain during the pandemic. This article may provide useful references for the food industry to minimize the food safety impact of COVID-19 (as well as other respiratory virus) and allows them to better prepare for similar future challenges.
Collapse
Affiliation(s)
- Haoqing Yang
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China
| | - Jiamiao Hu
- Diabetes Research Centre, Leicester General Hospital, Leicester LE5 4PW, United Kingdom
| | - Bee K. Tan
- Diabetes Research Centre, Leicester General Hospital, Leicester LE5 4PW, United Kingdom
| | - Ka-hing Wong
- Department of Applied Biology and Chemical Technology, The Hongkong Polytechnic University, Hongkong SAR, China
| | - Jim Junhui Huang
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Republic of Singapore
| | - Peter C.K. Cheung
- Food Research Centre, School of Life Sciences, The Chinese University of Hongkong, Hongkong SAR, China
| | - Shaoling Lin
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian 350002, China
| |
Collapse
|
395
|
Tedbury PR, Manfredi C, Degenhardt F, Conway J, Horwath MC, McCracken C, Sorscher AJ, Moreau S, Wright C, Edwards C, Brewer J, Guarner J, de Wit E, Williamson BN, Suthar MS, Ong YT, Roback JD, Alter DN, Holter JC, Karlsen TH, Sacchi N, Romero-Gómez M, Invernizzi P, Fernández J, Buti M, Albillos A, Julià A, Valenti L, Asselta R, Banales JM, Bujanda L, de Cid R, Sarafianos SG, Hong JS, Sorscher EJ, Ehrhardt A. Mechanisms by which the cystic fibrosis transmembrane conductance regulator may influence SARS-CoV-2 infection and COVID-19 disease severity. FASEB J 2023; 37:e23220. [PMID: 37801035 PMCID: PMC10760435 DOI: 10.1096/fj.202300077r] [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: 03/29/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
Patients with cystic fibrosis (CF) exhibit pronounced respiratory damage and were initially considered among those at highest risk for serious harm from SARS-CoV-2 infection. Numerous clinical studies have subsequently reported that individuals with CF in North America and Europe-while susceptible to severe COVID-19-are often spared from the highest levels of virus-associated mortality. To understand features that might influence COVID-19 among patients with cystic fibrosis, we studied relationships between SARS-CoV-2 and the gene responsible for CF (i.e., the cystic fibrosis transmembrane conductance regulator, CFTR). In contrast to previous reports, we found no association between CFTR carrier status (mutation heterozygosity) and more severe COVID-19 clinical outcomes. We did observe an unexpected trend toward higher mortality among control individuals compared with silent carriers of the common F508del CFTR variant-a finding that will require further study. We next performed experiments to test the influence of homozygous CFTR deficiency on viral propagation and showed that SARS-CoV-2 production in primary airway cells was not altered by the absence of functional CFTR using two independent protocols. On the contrary, experiments performed in vitro strongly indicated that virus proliferation depended on features of the mucosal fluid layer known to be disrupted by absent CFTR in patients with CF, including both low pH and increased viscosity. These results point to the acidic, viscous, and mucus-obstructed airways in patients with cystic fibrosis as unfavorable for the establishment of coronaviral infection. Our findings provide new and important information concerning relationships between the CF clinical phenotype and severity of COVID-19.
Collapse
Affiliation(s)
- Philip R. Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Candela Manfredi
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Joseph Conway
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | - Michael C. Horwath
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Courtney McCracken
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Adam J. Sorscher
- Dartmouth University School of Medicine, Hanover, New Hampshire, United States
| | - Sandy Moreau
- Elliot Hospital, Manchester, New Hampshire, United States
| | | | - Carolina Edwards
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | - Jo Brewer
- Northeast Georgia Medical Center, Gainesville, Georgia, United States
| | | | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, NIAID, National Institutes of Health, Hamilton, Montana, United States
| | - Brandi N. Williamson
- Laboratory of Virology, Division of Intramural Research, NIAID, National Institutes of Health, Hamilton, Montana, United States
| | - Mehul S. Suthar
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Yee T. Ong
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - John D. Roback
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - David N. Alter
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jan C. Holter
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tom H. Karlsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute for Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Gastroenterology, Department of Transplantation Medicine, Division for Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Manuel Romero-Gómez
- Hospital Universitario Virgen del Rocío de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
- University of Sevilla, Sevilla, Spain
- Digestive Diseases Unit, Virgen del Rocio University Hospital, Institute of Biomedicine of Seville, University of Seville, Seville, Spain
| | - Pietro Invernizzi
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Javier Fernández
- Hospital Clinic, University of Barcelona, and IDIBAPS, Barcelona, Spain
- European Foundation for the Study of Chronic Liver Failure (EF-CLIF), Barcelona, Spain
| | - Maria Buti
- Liver Unit. Hospital Universitario Valle Hebron and CIBEREHD del Instituto Carlos III. Barcelona, Spain
| | - Agustin Albillos
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Gastroenterology, Hospital Universitario Ramón y Cajal, University of Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Antonio Julià
- Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Barcelona, Spain
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Biological Resorce Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milan Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Jesus M. Banales
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute – Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Rafael de Cid
- Genomes for Life-GCAT lab. German Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | | | - Stefan G. Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Jeong S. Hong
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Eric J. Sorscher
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Annette Ehrhardt
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
396
|
Moreno-Contreras J, Espinoza MA, Cantú-Cuevas MA, Madrid-González DA, Barón-Olivares H, Ortiz-Orozco OD, Guzmán-Rodríguez C, Arias CF, Lopez S. Saliva sampling and its direct lysis is an excellent option for SARS-CoV-2 diagnosis in paediatric patients: comparison with the PanBio COVID-19 antigen rapid test in symptomatic and asymptomatic children. J Med Microbiol 2023; 72. [PMID: 38014762 DOI: 10.1099/jmm.0.001779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Introduction. Lateral flow test (LFTs) have been used as an alternative to reverse transcription quantitative PCR (RT-qPCR) in point-of-care testing. Despite their benefits, the sensitivity of LFTs may be low and is affected by several factors. We have previously reported the feasibility of using direct lysis of individual or pools of saliva samples from symptomatic and asymptomatic patients as a source of viral genomes for detection by RT-qPCR.Hypothesis. Direct lysed saliva is more sensitive than antigen tests to detect severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in samples from children.Aim. Our goals here were to valuate the specificity and sensitivity of the PanBio COVID-19 antigen rapid test device (Ag-RTD) compared with RT-qPCR of direct lysed saliva.Methodology. We evaluated the performance of the PanBio COVID-19 Ag-RTD in comparison to RT-qPCR direct lysed saliva from paired samples of 256 symptomatic and 242 asymptomatic paediatric patients.Results. Overall, although there were no differences in the specificity (96.6%), we found a lower sensitivity (64.3%) of the PanBio Ag-test RTD compared to saliva in both symptomatic and asymptomatic patients. In addition, the sensitivity of PanBio was not correlated with the viral load present in the samples.Conclusion. Our data highlight the benefits of using RT-qPCR and saliva samples for SARS-CoV-2 detection, particularly in paediatric patients.
Collapse
Affiliation(s)
- Joaquín Moreno-Contreras
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología UNAM, Av. Universidad 2001, Col. Chamilpa, Cuernavaca, Morelos, Mexico
| | - Marco A Espinoza
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología UNAM, Av. Universidad 2001, Col. Chamilpa, Cuernavaca, Morelos, Mexico
| | - Marco A Cantú-Cuevas
- Secretaría de Salud del Edo. de Morelos, Ajusco #2 Col. Buena Vista, Cuernavaca, Morelos, Mexico
| | - Daniel A Madrid-González
- Secretaría de Salud del Edo. de Morelos, Ajusco #2 Col. Buena Vista, Cuernavaca, Morelos, Mexico
| | - Héctor Barón-Olivares
- Servicios de Salud del Edo. de Morelos, Callejón Borda 3 Col. Centro, Cuernavaca, Morelos, Mexico
| | - Oscar D Ortiz-Orozco
- Servicios de Salud del Edo. de Morelos, Callejón Borda 3 Col. Centro, Cuernavaca, Morelos, Mexico
| | - Cecilia Guzmán-Rodríguez
- Servicios de Salud del Edo. de Morelos, Callejón Borda 3 Col. Centro, Cuernavaca, Morelos, Mexico
| | - Carlos F Arias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología UNAM, Av. Universidad 2001, Col. Chamilpa, Cuernavaca, Morelos, Mexico
| | - Susana Lopez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología UNAM, Av. Universidad 2001, Col. Chamilpa, Cuernavaca, Morelos, Mexico
| |
Collapse
|
397
|
Bello-Perez M, Hurtado-Tamayo J, Mykytyn AZ, Lamers MM, Requena-Platek R, Schipper D, Muñoz-Santos D, Ripoll-Gómez J, Esteban A, Sánchez-Cordón PJ, Enjuanes L, Haagmans BL, Sola I. SARS-CoV-2 ORF8 accessory protein is a virulence factor. mBio 2023; 14:e0045123. [PMID: 37623322 PMCID: PMC10653805 DOI: 10.1128/mbio.00451-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/04/2023] [Indexed: 08/26/2023] Open
Abstract
IMPORTANCE The relevance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ORF8 in the pathogenesis of COVID-19 is unclear. Virus natural isolates with deletions in ORF8 were associated with wild milder disease, suggesting that ORF8 might contribute to SARS-CoV-2 virulence. This manuscript shows that ORF8 is involved in inflammation and in the activation of macrophages in two experimental systems: humanized K18-hACE2 transgenic mice and organoid-derived human airway cells. These results identify ORF8 protein as a potential target for COVID-19 therapies.
Collapse
Affiliation(s)
- M. Bello-Perez
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J. Hurtado-Tamayo
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - A. Z. Mykytyn
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - M. M. Lamers
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - R. Requena-Platek
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - D. Schipper
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - D. Muñoz-Santos
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J. Ripoll-Gómez
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - A. Esteban
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - P. J. Sánchez-Cordón
- Veterinary Pathology Department, Animal Health Research Center (CISA), National Institute of Research, Agricultural and Food Technology, Valdeolmos, Spain
| | - L. Enjuanes
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - B. L. Haagmans
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - I. Sola
- Department of Molecular and Cell Biology, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
398
|
Schoen A, Hölzer M, Müller MA, Wallerang KB, Drosten C, Marz M, Lamp B, Weber F. Functional comparisons of the virus sensor RIG-I from humans, the microbat Myotis daubentonii, and the megabat Rousettus aegyptiacus, and their response to SARS-CoV-2 infection. J Virol 2023; 97:e0020523. [PMID: 37728614 PMCID: PMC10653997 DOI: 10.1128/jvi.00205-23] [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: 02/07/2023] [Accepted: 07/09/2023] [Indexed: 09/21/2023] Open
Abstract
IMPORTANCE A common hypothesis holds that bats (order Chiroptera) are outstanding reservoirs for zoonotic viruses because of a special antiviral interferon (IFN) system. However, functional studies about key components of the bat IFN system are rare. RIG-I is a cellular sensor for viral RNA signatures that activates the antiviral signaling chain to induce IFN. We cloned and functionally characterized RIG-I genes from two species of the suborders Yangochiroptera and Yinpterochiroptera. The bat RIG-Is were conserved in their sequence and domain organization, and similar to human RIG-I in (i) mediating virus- and IFN-activated gene expression, (ii) antiviral signaling, (iii) temperature dependence, and (iv) recognition of RNA ligands. Moreover, RIG-I of Rousettus aegyptiacus (suborder Yinpterochiroptera) and of humans were found to recognize SARS-CoV-2 infection. Thus, members of both bat suborders encode RIG-Is that are comparable to their human counterpart. The ability of bats to harbor zoonotic viruses therefore seems due to other features.
Collapse
Affiliation(s)
- Andreas Schoen
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Martin Hölzer
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- European Virus Bioinformatics Center, Jena, Germany
| | - Marcel A. Müller
- German Centre for Infection Research (DZIF), Partner Sites Giessen and Charité, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kai B. Wallerang
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Christian Drosten
- European Virus Bioinformatics Center, Jena, Germany
- German Centre for Infection Research (DZIF), Partner Sites Giessen and Charité, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- European Virus Bioinformatics Center, Jena, Germany
| | - Benjamin Lamp
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, Giessen, Germany
- European Virus Bioinformatics Center, Jena, Germany
- German Centre for Infection Research (DZIF), Partner Sites Giessen and Charité, Berlin, Germany
| |
Collapse
|
399
|
Lin C, Liu Z, Fang F, Zhao S, Li Y, Xu M, Peng Y, Chen H, Yuan F, Zhang W, Zhang X, Teng Z, Xiao R, Yang Y. Next-Generation Rapid and Ultrasensitive Lateral Flow Immunoassay for Detection of SARS-CoV-2 Variants. ACS Sens 2023; 8:3733-3743. [PMID: 37675933 DOI: 10.1021/acssensors.3c01019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic highlighted the need for rapid and accurate viral detection at the point-of-care testing (POCT). Compared with nucleic acid detection, lateral flow immunoassay (LFIA) is a rapid and flexible method for POCT detection. However, the sensitivity of LFIA limits its use for early identification of patients with COVID-19. Here, an innovative surface-enhanced Raman scattering (SERS)-LFIA platform based on two-dimensional black phosphorus decorated with Ag nanoparticles as important antigen-capturing and Raman-signal-amplification unit was developed for detection of SARS-CoV-2 variants within 5-20 min. The novel SERS-LFIA platform realized a limit of detection of 0.5 pg/mL and 100 copies/mL for N protein and SARS-CoV-2, demonstrating 1000 times more sensitivity than the commercial LFIA strips. It could reliably detect seven different SARS-CoV-2 variants with cycle threshold (Ct) < 38, with sensitivity and specificity of 97 and 100%, respectively, exhibiting the same sensitivity with q-PCR. Furthermore, the detection results for 48 SARS-CoV-2-positive nasopharyngeal swabs (Ct = 19.8-38.95) and 96 negative nasopharyngeal swabs proved the reliability of the strips in clinical application. The method also had good specificity in double-blind experiments involving several other coronaviruses, respiratory viruses, and respiratory medications. The results showed that the innovative SERS-LFIA platform is expected to be the next-generation antigen detection technology. The inexpensive amplification-free assay combines the advantages of rapid low-cost POCT and highly sensitive nucleic acid detection, and it is suitable for rapid detection of SARS-CoV-2 variants and other pathogens. Thus, it could replace existing antigens and nucleic acids to some extent.
Collapse
Affiliation(s)
- Chenglong Lin
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zhenzhen Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, No.20 Dongdajie, Fengtai District, Beijing 100101, People's Republic of China
| | - Fanghao Fang
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Shuai Zhao
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yanyan Li
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Meimei Xu
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yusi Peng
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Graduate School of the Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Hongyou Chen
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Fang Yuan
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Wanju Zhang
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Xi Zhang
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Zheng Teng
- Shanghai Municipal Centre for Disease Control and Prevention, No. 1380, Zhongshan West Road, Shanghai 200336, People's Republic of China
| | - Rui Xiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, No.20 Dongdajie, Fengtai District, Beijing 100101, People's Republic of China
| | - Yong Yang
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
400
|
Rashid SA, Nazakat R, Muhamad Robat R, Ismail R, Suppiah J, Rajendran K, Raj Louis Masalamany ASS, Muhamad Hendri NA, Mohamad N, Khairul Hasni NA, Suib FA, Nik Hassan NMN, Pahrol MA, Shaharudin R. Droplet digital PCR application for the detection of SARS-CoV-2 in air sample. Front Public Health 2023; 11:1208348. [PMID: 37965510 PMCID: PMC10641526 DOI: 10.3389/fpubh.2023.1208348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may transmit through airborne route particularly when the aerosol particles remain in enclosed spaces with inadequate ventilation. There has been no standard recommended method of determining the virus in air due to limitations in pre-analytical and technical aspects. Furthermore, the presence of low virus loads in air samples could result in false negatives. Our study aims to explore the feasibility of detecting SARS-CoV-2 ribonucleic acid (RNA) in air samples using droplet digital polymerase chain reaction (ddPCR). Active and passive air sampling was conducted between December 2021 and February 2022 with the presence of COVID-19 confirmed cases in two hospitals and a quarantine center in Klang Valley, Malaysia. SARS-CoV-2 RNA in air was detected and quantified using ddPCR and real-time reverse transcriptase-polymerase chain reaction (RT-PCR). The comparability of two different digital PCR platforms (QX200 and QIAcuity) to RT-PCR were also investigated. Additionally negative staining transmission electron microscopy was performed to visualize virus ultrastructure. Detection rates of SARS-CoV-2 in air samples using ddPCR were higher compared to RT-PCR, which were 15.2% (22/145) and 3.4% (5/145), respectively. The sensitivity and specificity of ddPCR was 100 and 87%, respectively. After excluding 17 negative samples (50%) by both QX200 and QIAcuity, 15% samples (5/34) were found to be positive both ddPCR and dPCR. There were 23.5% (8/34) samples that were detected positive by ddPCR but negative by dPCR. In contrast, there were 11.7% (4/34) samples that were detected positive by dPCR but negative by ddPCR. The SARS-CoV-2 detection method by ddPCR is precise and has a high sensitivity for viral RNA detection. It could provide advances in determining low viral titter in air samples to reduce false negative reports, which could complement detection by RT-PCR.
Collapse
Affiliation(s)
- Siti Aishah Rashid
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Raheel Nazakat
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Rosnawati Muhamad Robat
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Rohaida Ismail
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Jeyanthi Suppiah
- Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Kamesh Rajendran
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - A. S. Santhana Raj Louis Masalamany
- Special Resource Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Nur Afrina Muhamad Hendri
- Special Resource Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Nadia Mohamad
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Nurul Amalina Khairul Hasni
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Fatin Amirah Suib
- Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Nik Muhamad Nizam Nik Hassan
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Muhammad Alfatih Pahrol
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Rafiza Shaharudin
- Environmental Health Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| |
Collapse
|