51
|
Petrie J, Hay JA, Srimokla O, Panovska-Griffiths J, Whittaker C, Masel J. Enhanced testing can substantially improve defense against several types of respiratory virus pandemic. Epidemics 2025; 50:100812. [PMID: 39922066 DOI: 10.1016/j.epidem.2024.100812] [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: 04/29/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 02/10/2025] Open
Abstract
Mass testing to identify and isolate infected individuals is a promising approach for reducing harm from the next acute respiratory virus pandemic. It offers the prospect of averting hospitalizations and deaths whilst avoiding the need for indiscriminate social distancing measures. To understand scenarios where mass testing might or might not be a viable intervention, here we modeled how effectiveness depends both on characteristics of the pathogen (R0, time to peak viral load) and on the testing strategy (limit of detection, testing frequency, test turnaround time, adherence). We base time-dependent test sensitivity and time-dependent infectiousness on an underlying viral load trajectory model. We show that given moderately high public adherence, frequent testing can prevent as many transmissions as more costly interventions such as school or business closures. With very high adherence and fast, frequent, and sensitive testing, we show that most respiratory virus pandemics could be controlled with mass testing alone.
Collapse
Affiliation(s)
- James Petrie
- Pandemic Sciences Institute, University of Oxford, United Kingdom; Big Data Institute, University of Oxford, United Kingdom.
| | - James A Hay
- Pandemic Sciences Institute, University of Oxford, United Kingdom; Big Data Institute, University of Oxford, United Kingdom
| | - Oraya Srimokla
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Jasmina Panovska-Griffiths
- Pandemic Sciences Institute, University of Oxford, United Kingdom; Big Data Institute, University of Oxford, United Kingdom; UK Health Security Agency, United Kingdom; The Queen's College, University of Oxford, United Kingdom
| | - Charles Whittaker
- Department of Infectious Disease Epidemiology, Imperial College London, United Kingdom
| | - Joanna Masel
- Ecology & Evolutionary Biology, University of Arizona, United States of America
| |
Collapse
|
52
|
Wørzner K, Schmidt ST, Zimmermann J, Tami A, Polacek C, Fernandez-Antunez C, Hartmann KT, Jensen RF, Hansen JS, Illigen K, Isling LK, Erbs G, Jungersen G, Rosenkrands I, Offersgaard A, Gottwein J, Holmbeck K, Jensen HE, Ramirez S, Follmann F, Bukh J, Pedersen GK. Intranasal recombinant protein subunit vaccine targeting TLR3 induces respiratory tract IgA and CD8 T cell responses and protects against respiratory virus infection. EBioMedicine 2025; 113:105615. [PMID: 39983329 PMCID: PMC11893338 DOI: 10.1016/j.ebiom.2025.105615] [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: 06/10/2024] [Revised: 12/16/2024] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Intranasal vaccines against respiratory viruses are desired due to ease of administration and potential to protect against virus infection of the upper respiratory tract. METHODS We tested a cationic liposomal adjuvant delivering the TLR3 agonist Poly (I:C) (CAF®09b) for intranasal administration, by formulating this with SARS-CoV-2 spike trimeric protein and assessing airway mucosal immune responses in mice. The vaccine was further evaluated in SARS-CoV-2 virus challenge models, using mice expressing the human ACE2 receptor and Syrian hamsters. FINDINGS The intranasal vaccine elicited both serum neutralising antibody responses and IgA responses in the upper respiratory tract. Uniquely, it also elicited high-magnitude CD4 and CD8 T cell responses in the lung parenchyma and nasal-associated lymphoid tissue. In contrast, parenteral administration of the same vaccine, or the mRNA-1273 (Spikevax®) vaccine, led to systemic antibody responses and vaccine-induced CD4 T cells were mainly found in circulation. The intranasal vaccine protected against homologous SARS-CoV-2 (Wuhan-Hu-1) challenge in K18-hACE2 mice, preventing weight loss and virus infection in the upper and lower airways. In Syrian hamsters, the vaccine prevented weight loss and significantly reduced virus load after challenge with the homologous strain and Omicron BA.5. INTERPRETATION This study demonstrates that intranasal subunit vaccines containing TLR3-stimulating cationic liposomes effectively induce airway IgA and T cell responses, which could be utilised in future viral pandemics. FUNDING This work was primarily supported by the European Union Horizon 2020 research and innovation program under grant agreement no. 101003653.
Collapse
Affiliation(s)
- Katharina Wørzner
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Signe Tandrup Schmidt
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Julie Zimmermann
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ahmad Tami
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Charlotta Polacek
- Virus Research & Development Laboratory, Department of Virology and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Carlota Fernandez-Antunez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Katrine Top Hartmann
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rune Fledelius Jensen
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Julia Sid Hansen
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Kristin Illigen
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Louise Krag Isling
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Gitte Erbs
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anna Offersgaard
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Judith Gottwein
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Kenn Holmbeck
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark; Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
53
|
Ana-Sosa-Batiz F, Verma SK, Shafee N, Miller R, Conner C, Hastie KM, Timis J, Maule E, Nguyen MN, Tran L, Varghese K, Madany H, Street AE, Zandonatti M, Moi ML, Jarnagin K, Webb DR, Saphire EO, Kim K, Shresta S. A humanised ACE2, TMPRSS2, and FCGRT mouse model reveals the protective efficacy of anti-receptor binding domain antibodies elicited by SARS-CoV-2 hybrid immunity. EBioMedicine 2025; 113:105619. [PMID: 40020261 PMCID: PMC11910679 DOI: 10.1016/j.ebiom.2025.105619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite the importance of vaccination- and infection-elicited antibodies (Abs) to SARS-CoV-2 immunity, current mouse models do not fully capture the dynamics of Ab-mediated immunity in vivo, including potential contributions of the neonatal Fc receptor, encoded by FCGRT. METHODS We generated triple knock-in (TKI) mice expressing human ACE2, TMPRSS2, and FCGRT; and evaluated the protective efficacy of anti-SARS-CoV-2 monoclonal Abs (mAbs) and plasma from individuals with immunity elicited by vaccination alone plus SARS-CoV-2 infection-induced (hybrid) immunity. FINDINGS A human anti-SARS-CoV-2 mAb harbouring a half-life-extending mutation, but not the wild-type mAb, exhibited prolonged half-life in TKI mice and protected against lung infection with Omicron BA.2, validating the utility of these mice for evaluating therapeutic Abs. Pooled plasma from individuals with hybrid immunity to Delta, but not from vaccinated-only individuals, cleared infectious Delta from the lungs of TKI mice (P < 0.01), even though the two plasma pools had similar Delta-binding and -neutralising Ab titres in vitro. Similarly, plasma from individuals with hybrid Omicron BA.1/2 immunity, but not hybrid Delta immunity, decreased lung infection (P < 0.05) with BA.5 in TKI mice, despite the plasma pools having comparable BA.5-binding and -neutralising titres in vitro. Depletion of receptor-binding domain-targeting Abs from hybrid immune plasma abrogated their protection against infection. INTERPRETATION These results demonstrate the utility of TKI mice as a tool for the development of anti-SARS-CoV-2 mAb therapeutics, show that in vitro neutralisation assays do not accurately predict in vivo protection, and highlight the importance of hybrid immunity for eliciting protective anti-receptor-binding domain Abs. FUNDING This work was funded by grants from the e-Asia Joint Research Program (N10A650706 and N10A660577 to MLM, in collaboration with SS); the NIH (U19 AI142790-02S1 to EOS and SS and R44 AI157900 to KJ); the GHR Foundation (to SS and EOS); the Overton family (to SS and EOS); the Arvin Gottlieb Foundation (to SS and EOS), the Prebys Foundation (to SS); and the American Association of Immunologists Fellowship Program for Career Reentry (to FASB).
Collapse
Affiliation(s)
| | - Shailendra Kumar Verma
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Norazizah Shafee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA; Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Robyn Miller
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Chris Conner
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Michael N Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Linda Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Krithik Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Henry Madany
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Michelle Zandonatti
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kurt Jarnagin
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - David R Webb
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- Histopathology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA; Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
54
|
Chen YY, Yang MH, Lai JZ, Chen JW, Wang YL, Hung CS, Kow CD, Lin CL, Hou SM, Wu HS, Wei ST. Transfusion safety concerns during the COVID-19 pandemic in Taiwan: Altered by evolving control strategies. J Formos Med Assoc 2025; 124:218-226. [PMID: 39306515 DOI: 10.1016/j.jfma.2024.09.003] [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] [Received: 05/28/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND In 2022, the SARS-CoV-2 Omicron surge affected 8.8 million people in Taiwan. This study delves into how the transition from containment to mitigation strategies in COVID-19 control has altered concerns regarding transfusion safety. METHODS Blood donations during 2020-2022 in Taiwan were included. Donation details and post-donation information (PDI) were retrieved to assess donation fluctuations and incidences of various PDI. The main effects of PDI reporting were assessed using chi-square test and logistic regression. Additionally, from April to August 2022, we collected disease information from COVID-19 donors, and tested their repository specimens for SARS-CoV-2 RNA and antibodies. RESULTS Before 2022, when containment measures were in place, only 8 blood donors with COVID-19 reported PDI. However, by mid-2021, there was a significant decrease in blood donations. In 2022, with mitigation strategies implemented, a total of 3483 donations reported COVID-19 PDI. The incidence of all cause PDI increased from 10.5 per 10,000 donations in 2020-2021 to 29.9 per 10,000 in 2022, with nearly 70% of PDI being related to COVID-19. Female donors reported more PDI events. Additionally, the incidence significantly decreased with age. A total of 1148 repository specimens from COVID-19 donor were tested, revealing no detection of SARS-CoV-2 RNA. The seroprevalence rates of anti-nucleocapsid(N) and anti-spike(S) antibodies were 0.61% and 98.4%, respectively. CONCLUSION Transfusion safety concerns in Taiwan progressed alongside the evolution of control strategies, with a one-year delay following the pandemic started. The absence of RNAemia among COVID-19 donors indicates that precautionary measures were commensurate with the risk.
Collapse
Affiliation(s)
- Yun-Yuan Chen
- Head Office, Taiwan Blood Services Foundation, Taipei, Taiwan
| | - Min-Hui Yang
- Hsinchu Blood Center, Taiwan Blood Services Foundation, Hsinchu, Taiwan
| | - Jou-Zhen Lai
- Hsinchu Blood Center, Taiwan Blood Services Foundation, Hsinchu, Taiwan
| | - Jen-Wei Chen
- Head Office, Taiwan Blood Services Foundation, Taipei, Taiwan
| | - Yun-Long Wang
- Hsinchu Blood Center, Taiwan Blood Services Foundation, Hsinchu, Taiwan
| | - Cheng-Shen Hung
- Head Office, Taiwan Blood Services Foundation, Taipei, Taiwan; Taipei Blood Center, Taiwan Blood Services Foundation, Taipei, Taiwan
| | - Chang-Der Kow
- Hsinchu Blood Center, Taiwan Blood Services Foundation, Hsinchu, Taiwan
| | - Chi-Ling Lin
- Taichung Blood Center, Taiwan Blood Services Foundation, Taichung, Taiwan
| | - Sheng-Mou Hou
- Head Office, Taiwan Blood Services Foundation, Taipei, Taiwan; Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ho-Sheng Wu
- Hsinchu Blood Center, Taiwan Blood Services Foundation, Hsinchu, Taiwan; School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan.
| | - Sheng-Tang Wei
- Head Office, Taiwan Blood Services Foundation, Taipei, Taiwan.
| |
Collapse
|
55
|
Chung HY, Jian MJ, Chang CK, Perng CL, Hung KS, Chiu CH, Shang HS. Enhancing public health outcomes with AI-powered clinical surveillance: Precise detection of COVID-19 variants using qPCR and nanopore sequencing. J Infect Public Health 2025; 18:102663. [PMID: 39862804 DOI: 10.1016/j.jiph.2025.102663] [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: 06/18/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND We aimed to evaluate the efficacy of integrating the Varia5 multiplex assay (qPCR) and whole genome sequencing (WGS) for monitoring SARS-CoV-2, focusing on their overall performance in identifying various virus variants. METHODS This study included 140 naso-pharyngeal swab samples from individuals with suspected COVID-19. We utilized our self-developed Varia5 multiplex assay, which targets five viral genes linked to COVID-19 mutations, in conjunction with comprehensive genomic analysis performed through whole genome sequencing (WGS) using the Oxford Nanopore system. Machine learning was integrated to optimize the qPCR conditions and enhance the detection efficiency. RESULTS The Varia5 assay identified the prevalent BA.2.75 variant in 92 samples compared to that in 81 samples detected via WGS. The BA.5.2 variant, indicative of higher viral loads, was identified in 15 samples via Varia5 and in 14 samples via WGS.Furthermore, rare variants, such as BA.2.10, were identified. The mean Ct value was 18.36, with significant viral load differences noted between specific variants. CONCLUSION Our findings demonstrate that while WGS offers enhanced sensitivity and specificity for variant detection, qPCR remains crucial for large-scale testing because of its cost and time efficiency. The integrated approach, which combines both techniques, represents a more comprehensive monitoring algorithm that can improve public health strategies against pandemics such as COVID-19.
Collapse
Affiliation(s)
- Hsing-Yi Chung
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan.
| | - Ming-Jr Jian
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Chih-Kai Chang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Cherng-Lih Perng
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Kuo-Sheng Hung
- Center for Precision Medicine and Genomics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Chun-Hsiang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
56
|
da Silva Junior LC, Wailante DF, Bueno MG, Moura PEB, Pauvolid-Corrêa A, Novaes RLM, da Costa-Neto SF, Veríssimo I, Bertocchi NA, Moratelli R, Gentile R, Motta FC, de Araújo MF, Brown D, Resende PC, de Siqueira MAMT, Ogrzewalska M. Cross-Species Surveillance of Respiratory Viruses in Domestic and Wild Mammals of an Urban Atlantic Forest from Brazil. ECOHEALTH 2025; 22:11-28. [PMID: 39904935 PMCID: PMC11890330 DOI: 10.1007/s10393-024-01691-w] [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: 01/22/2024] [Revised: 08/16/2024] [Accepted: 11/07/2024] [Indexed: 02/06/2025]
Abstract
Our aim was to investigate respiratory viruses circulating in animals from the urban Atlantic Forest, which is located in the most densely populated area near Rio de Janeiro in Brazil. This study focused on the detection of Influenza A viruses and diverse coronaviruses, including SARS-CoV-2, in domestic and wild animals, including bats, nonhuman primates, rodents, and marsupials. From August 2020 to September 2022, biological samples were collected from a total of 72 pets, 66 primates, 20 rodents, 36 marsupials, and 390 bats. Samples were tested using RT-PCR for Influenza A and coronaviruses, and positive samples were sequenced. When blood samples were available, they were tested for SARS-CoV-2 antibodies. All terrestrial animals were negative for evidence of infection with SARS-CoV-2 and Influenza A viruses. However, samples from 17 phyllostomid bats, including Great fruit-eating bat (Artibeus lituratus), Silky short-tailed bat (Carollia brevicauda), Seba's short-tailed bat (Carollia perspicillata), Common big-eared bat (Micronycteris microtis), Greater spear-nosed bat (Phyllostomus hastatus), White-lined broad-nosed bat (Platyrrhinus lineatus), Little yellow-shouldered bat (Sturnira lilium), Greater round-eared bat (Tonatia bidens), and Common vampire bat (Desmodus rotundus), were positive for bat coronaviruses classified as Alphacoronavirus. Our study adds new information on the occurrence of coronaviruses in bats and contributes to a long-term program of Influenza surveillance. Developing active surveillance for viruses in wildlife species, as implemented in this study, is crucial for understanding zoonotic risks and preventing future global pandemics.
Collapse
Affiliation(s)
- Leonardo Corrêa da Silva Junior
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Deborah Fernandes Wailante
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Marina Galvao Bueno
- Laboratório de Virologia Comparada e Ambiental, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Patricia Emilia Bento Moura
- Laboratório de Virologia Comparada e Ambiental, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Alex Pauvolid-Corrêa
- Laboratório de Virologia Veterinária de Viçosa, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa, MG, 36570-900, Brazil
| | | | | | - Iuri Veríssimo
- Fundação Oswaldo Cruz, Fiocruz Mata Atlântica, Rio de Janeiro, RJ, 22713-570, Brazil
| | | | - Ricardo Moratelli
- Fundação Oswaldo Cruz, Fiocruz Mata Atlântica, Rio de Janeiro, RJ, 22713-570, Brazil
| | - Rosana Gentile
- Laboratório de Biologia e Parasitologia de Mamíferos Silvestres Reservatórios, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Fernando Couto Motta
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Mia Ferreira de Araújo
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - David Brown
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Paola Cristina Resende
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil
| | | | - Maria Ogrzewalska
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Fundação Oswaldo Cruz, IOC, Rio de Janeiro, RJ, 21040-900, Brazil.
| |
Collapse
|
57
|
Maru B, Edinboro A, Katolik A, El-Khoury R, Basran K, Wahba A, Damha M, Luedtke N, McKeague M. Fluorogenic oligonucleotide cleavage probes with a branched linker improve RNA detection. Nucleic Acids Res 2025; 53:gkaf141. [PMID: 40052819 PMCID: PMC11886808 DOI: 10.1093/nar/gkaf141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/25/2025] [Accepted: 02/16/2025] [Indexed: 03/10/2025] Open
Abstract
Fluorescent probe-based quantitative polymerase chain reaction (qPCR) is essential for DNA/RNA quantification widely used in research and clinical diagnostics. The performance of fluorogenic probes depends heavily on their design, particularly the identities of the fluorophore and quencher moieties, and the linkers used to attach them to oligonucleotides. Here we report a highly modular, three-way branched glycerol 'X' linker in fluorogenic TaqMan® type oligonucleotide probes for multiplexed, reverse transcription qPCR (RT-qPCR). The flexible 'X' linker served as an internal attachment point for various quenchers (BHQ1, BHQ2) in probes containing a variable fluorophore at the 5' end (Flu, Hex, Cy5, Cy5.5). A four-color RT-qPCR 'tetraplex' assay was thereby developed for distinguishing between RNA genomes from SARS-CoV-2, influenza A, and influenza B viruses in a single reaction. The 'X' linker exhibited superior performance with single-molecule detection limits approaching four copies, compared to an internal arabinoside-based (ara) linker strategy, demonstrating the presence of competing processes during primer extension, one where Taq exonuclease activity cleaves the fluorogenic X probe leading to productive fluorescence, and the second where the ara probe is displaced from the PCR template without cleavage. Together these results demonstrate the importance of linker structure selection in oligonucleotides for developing highly effective fluorogenic probes for qPCR.
Collapse
Affiliation(s)
- Bruktawit Maru
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Ayodele Edinboro
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
| | - Adam Katolik
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
| | - Roberto El-Khoury
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
| | - Kaleena Basran
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
| | - Alexander S Wahba
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
- McGill Chemistry Characterization (MC), McGill University, Montreal H3A 0B8, Canada
| | - Masad J Damha
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
- Centre de recherche en biologie structurale, McGill University, Montreal H3G 0B1, Canada
| | - Nathan W Luedtke
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
- Centre de recherche en biologie structurale, McGill University, Montreal H3G 0B1, Canada
| | - Maureen McKeague
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
- Department of Chemistry, McGill University, Montreal H3A 0B8, Canada
- Centre de recherche en biologie structurale, McGill University, Montreal H3G 0B1, Canada
| |
Collapse
|
58
|
Khalili Fakhrabadi A, Shahbazzadeh MJ, Jalali N, Eslami M. A hybrid inception-dilated-ResNet architecture for deep learning-based prediction of COVID-19 severity. Sci Rep 2025; 15:6490. [PMID: 39987169 PMCID: PMC11846838 DOI: 10.1038/s41598-025-91322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 02/19/2025] [Indexed: 02/24/2025] Open
Abstract
Chest computed tomography (CT) scans are essential for accurately assessing the severity of the novel Coronavirus (COVID-19), facilitating appropriate therapeutic interventions and monitoring disease progression. However, determining COVID-19 severity requires a radiologist with significant expertise. This study introduces a pioneering utilization of deep learning (DL) for evaluate COVID-19 severity using lung CT images, presenting a novel and effective method for assessing the severity of pulmonary manifestations in COVID-19 patients. Inception-Residual networks (Inception-ResNet), advanced hybrid models known for their compactness and effectiveness, were used to extract relevant features from CT scans. Inception-ResNet incorporates the dilated mechanism into its ResNet component, enhancing its ability to accurately classify lung involvement stages. This study demonstrates that dilated residual networks (dResNet) outperform their non-dilated counterparts in image classification tasks, as their architectural designs allow the systems to acquire comprehensive global data by expanding their receptive fields. Our study utilized an initial dataset of 1548 human thoracic CT scans, meticulously annotated by two experienced specialists. Lung involvement was determined by calculating a percentage based on observations made at each scan. The hybrid methodology successfully distinguished the ten distinct severity levels associated with COVID-19, achieving a maximum accuracy of 96.40%. This system demonstrates its effectiveness as a diagnostic framework for assessing lung involvement in COVID-19-affected individuals, facilitating disease progression tracking.
Collapse
Affiliation(s)
- Ali Khalili Fakhrabadi
- Department of Electrical Engineering, Kerman Branch, Islamic Azad University, Kerman, Iran
| | | | - Nazanin Jalali
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Neurology Department, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mahdiyeh Eslami
- Department of Electrical Engineering, Kerman Branch, Islamic Azad University, Kerman, Iran
| |
Collapse
|
59
|
Juárez E, Vázquez-Pérez JA, Carreto-Binaghi LE, Martínez-Sanabria CA, Salgado-Cantú MG, Sarabia C, Herrera MT, Guzmán-Beltrán S, Gutiérrez-González LH, González Y. COVID-19 extracellular vesicles display heterogeneity based on viral and host RNA expression: implications for host immune response. J Leukoc Biol 2025; 117:qiae212. [PMID: 39327799 DOI: 10.1093/jleuko/qiae212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024] Open
Abstract
Viral RNA and miRNAs released by immune cells contribute to inflammation in COVID-19 patients. Here, we investigated the role of SARS-CoV2 RNA and host miRNAs carried within extracellular vesicles (EVs) in modulating inflammation. EVs were classified as positive or negative depending on their viral RNA cargo. To assess the function of viral RNA, EVs, and lipopolysaccharide (LPS) were used to stimulate whole blood samples from healthy subjects, and the secretion of 27 serum analytes was measured. EVs alone did not induce cytokines, chemokines, or growth factors. However, under LPS stimulation, (SARS-CoV2+) EVs increased IL-12 and decreased IL-13 secretion, while (SARS-CoV2-) EVs increased MIP-1α and IL-1β secretion. Host miR-19a-3p, -192-5p, -let-7c-5p, and -92b-3a were differentially expressed in association with viral RNA. EVs from COVID-19 patients exhibited differences in viral RNA and miRNA expression profiles that modulate LPS responses. This knowledge sheds light on the immunopathology of COVID-19.
Collapse
Affiliation(s)
- Esmeralda Juárez
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Joel A Vázquez-Pérez
- Laboratory of Molecular Biology of Emergent Diseases and COPD, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Laura E Carreto-Binaghi
- Laboratory of Immunobiology of Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Claudia A Martínez-Sanabria
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
- School of Medicine, Benemérita Universidad Autónoma de Puebla, 4 Sur 104, 72000 Puebla, Mexico
| | - Manuel G Salgado-Cantú
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Carmen Sarabia
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - María Teresa Herrera
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Silvia Guzmán-Beltrán
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Luis H Gutiérrez-González
- Laboratory of Transcriptomics and Molecular Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| | - Yolanda González
- Department of Microbiology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, 14080 Mexico City, Mexico
| |
Collapse
|
60
|
de
Lima IL, Cataldi TR, Brites C, Labate MT, Vaz SN, Deminco F, da Cunha GS, Labate CA, Eberlin MN. 4D-DIA Proteomics Uncovers New Insights into Host Salivary Response Following SARS-CoV-2 Omicron Infection. J Proteome Res 2025; 24:499-514. [PMID: 39803891 PMCID: PMC11812090 DOI: 10.1021/acs.jproteome.4c00630] [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: 07/23/2024] [Revised: 12/04/2024] [Accepted: 12/30/2024] [Indexed: 02/08/2025]
Abstract
Since late 2021, Omicron variants have dominated the epidemiological scenario as the most successful severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sublineages, driving new and breakthrough infections globally over the past two years. In this study, we investigated for the first time the host salivary response of COVID-19 patients infected with Omicron variants (BA.1, BA.2, and BA.4/5) by using an untargeted four-dimensional data-independent acquisition (4D-DIA)-based proteomics approach. We identified 137 proteins whose abundance levels differed between the COVID-19 positive and negative groups. Salivary signatures were mainly enriched in ribosomal proteins, linked to mRNAviral translation, protein synthesis and processing, immune innate, and antiapoptotic signaling. The higher abundance of 14-3-3 proteins (YWHAG, YWHAQ, YWHAE, and SFN) in saliva, first reported here, may be associated with increased infectivity and improved viral replicative fitness. We also identified seven proteins (ACTN1, H2AC2, GSN, NDKA, CD109, GGH, and PCYOX) that yielded comprehension into Omicron infection and performed outstandingly in screening patients with COVID-19 in a hospital setting. This panel also presented an enhanced anti-COVID-19 and anti-inflammatory signature, providing insights into disease severity, supported by comparisons with other proteome data sets. The salivary signature provided valuable insights into the host's response to SARS-CoV-2 Omicron infection, shedding light on the pathophysiology of COVID-19, particularly in cases associated with mild disease. It also underscores the potential clinical applications of saliva for disease screening in hospital settings. Data are available via ProteomeXchange with the identifier PXD054133.
Collapse
Affiliation(s)
- Iasmim Lopes de
Lima
- PPGEMN,
School of Engineering, Mackenzie Presbyterian University & MackGraphe
- Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo, São
Paulo 01302-907, Brazil
| | - Thais Regiani Cataldi
- Department
of Genetics, “Luiz de Queiroz”
College of Agriculture, University of São Paulo/ESALQ, Piracicaba, São Paulo 13418-900, Brazil
| | - Carlos Brites
- LAPI
- Laboratory of Research in Infectology, University Hospital Professor
Edgard Santos (HUPES), Federal University
of Bahia (UFBA), Salvador, Bahia 40110-060, Brazil
| | - Mônica Teresa
Veneziano Labate
- Department
of Genetics, “Luiz de Queiroz”
College of Agriculture, University of São Paulo/ESALQ, Piracicaba, São Paulo 13418-900, Brazil
| | - Sara Nunes Vaz
- LAPI
- Laboratory of Research in Infectology, University Hospital Professor
Edgard Santos (HUPES), Federal University
of Bahia (UFBA), Salvador, Bahia 40110-060, Brazil
| | - Felice Deminco
- LAPI
- Laboratory of Research in Infectology, University Hospital Professor
Edgard Santos (HUPES), Federal University
of Bahia (UFBA), Salvador, Bahia 40110-060, Brazil
| | - Gustavo Santana da Cunha
- PPGEMN,
School of Engineering, Mackenzie Presbyterian University & MackGraphe
- Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo, São
Paulo 01302-907, Brazil
| | - Carlos Alberto Labate
- Department
of Genetics, “Luiz de Queiroz”
College of Agriculture, University of São Paulo/ESALQ, Piracicaba, São Paulo 13418-900, Brazil
| | - Marcos Nogueira Eberlin
- PPGEMN,
School of Engineering, Mackenzie Presbyterian University & MackGraphe
- Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo, São
Paulo 01302-907, Brazil
| |
Collapse
|
61
|
Ouyang T, Tang Y, Klimes F, Vogel-Claussen J, Voskrebenzev A, Yang Q. Phase-resolved Functional Lung (PREFUL) MRI May Reveal Distinct Pulmonary Perfusion Defects in Postacute COVID-19 Syndrome: Sex, Hospitalization, and Dyspnea Heterogeneity. J Magn Reson Imaging 2025; 61:851-862. [PMID: 38887850 DOI: 10.1002/jmri.29458] [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: 03/28/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Pulmonary perfusion defects have been observed in patients with coronavirus disease 2019 (COVID-19). Currently, there is a need for further data on non-contrast-enhanced MRI in COVID patients. The early identification of heterogeneity in pulmonary perfusion defects among COVID-19 patients is beneficial for their timely clinical intervention and management. PURPOSE To investigate the utility of phase-resolved functional lung (PREFUL) MRI in detecting pulmonary perfusion disturbances in individuals with postacute COVID-19 syndrome (PACS). STUDY TYPE Prospective. SUBJECTS Forty-four participants (19 females, mean age 64.1 years) with PACS and 44 healthy subjects (19 females, mean age 59.5 years). Moreover, among the 44 patients, there were 19 inpatients and 25 outpatients; 19 were female and 25 were male; 18 with non-dyspnea and 26 with dyspnea. FIELD STRENGTH/SEQUENCE 3-T, two-dimensional (2D) spoiled gradient-echo sequence. ASSESSMENT Ventilation and perfusion-weighted maps were extracted from five coronal slices using PREFUL analysis. Subsequently, perfusion defect percentage (QDP), ventilation defect percentage (VDP), and ventilation-perfusion match healthy (VQM) were calculated based on segmented lung parenchyma ventilation and perfusion-weighted maps. Additionally, clinical features, including demographic data (such as sex and age) and serum biomarkers (such as D-dimer levels), were evaluated. STATISTICAL TESTS Spearman correlation coefficients to explore relationships between clinical features and QDP, VDP, and VQM. Propensity score matching analysis to reduce the confounding bias between patients with PACS and healthy controls. The Mann-Whitney U tests and Chi-squared tests to detect differences between groups. Multivariable linear regression analyses to identify factors related to QDP, VDP, and VQM. A P-value <0.05 was considered statistically significant. RESULTS QDP significantly exceeded that of healthy controls in individuals with PACS (39.8% ± 15.0% vs. 11.0% ± 4.9%) and was significantly higher in inpatients than in outpatients (46.8% ± 17.0% vs. 34.5% ± 10.8%). Moreover, males exhibited pulmonary perfusion defects significantly more frequently than females (43.9% ± 16.8% vs. 34.4% ± 10.2%), and dyspneic participants displayed significantly higher perfusion defects than non-dyspneic patients (44.8% ± 15.8% vs. 32.6% ± 10.3%). QDP showed a significant positive relationship with age (β = 0.50) and D-dimer level (β = 0.72). DATA CONCLUSION PREFUL MRI may show pulmonary perfusion defects in patients with PACS. Furthermore, perfusion impairments may be more pronounced in males, inpatients, and dyspneic patients. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Key Lab. of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yichen Tang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Key Lab. of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Filip Klimes
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research, Hanover, Germany
| | - Jens Vogel-Claussen
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research, Hanover, Germany
| | - Andreas Voskrebenzev
- Institute of Diagnostic and Interventional Radiology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research, Hanover, Germany
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Key Lab. of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
62
|
Fraenkel C, Thuresson S, Medstrand P, Alsved M, Löndahl J. Comparison of Airborne SARS-CoV-2 Omicron and Pre-Delta Variants Around Infected Patients. J Med Virol 2025; 97:e70258. [PMID: 39977450 PMCID: PMC11841929 DOI: 10.1002/jmv.70258] [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: 08/01/2024] [Revised: 01/06/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Transmissibility has increased during the evolution of SARS-CoV-2, possibly by improved airborne transmission. An increased transmission was noted also in many hospitals. We analyzed SARS-CoV-2 in room air of hospitalized Omicron infected patients and compared results with previous findings with pre-Delta variants to study if SARS-CoV-2 was more prevalent in patient rooms after the introduction of Omicron. Only 4 of 75 (5%) air samples, from 3 of 43 included patients, were positive during the early Omicron wave, compared to 14/120 (12%), from 10 of 60 included patients during the initial wave. No certain statistical difference between virus variants could be established, but the tendency was a lower occurrence at Omicron infected patients, also when adjusting for relevant confounders. These finding do not support the initial hypothesis that increased SARS-CoV-2 aerosol emission from diagnosed patients with Omicron could explain any increased risk of hospital transmission.
Collapse
Affiliation(s)
- Carl‐Johan Fraenkel
- Division of Infection Medicine, Department of Clinical SciencesLund UniversityLundSweden
- Department of Clinical Microbiology and Infection Prevention and ControlSkåne University HospitalLundSweden
| | - Sara Thuresson
- Division of Ergonomics and Aerosol Technology, Department of Design SciencesLund UniversityLundSweden
| | - Patrik Medstrand
- Department of Translational Medicine, Clinical VirologyLund UniversityLundSweden
| | - Malin Alsved
- Division of Ergonomics and Aerosol Technology, Department of Design SciencesLund UniversityLundSweden
| | - Jakob Löndahl
- Division of Ergonomics and Aerosol Technology, Department of Design SciencesLund UniversityLundSweden
| |
Collapse
|
63
|
Zeigler Z, Acevedo A, Mews I, Lesser D, Koornneef A. Effect of wearing N95 masks for 10 hours on ambulatory blood pressure in healthy adults. Blood Press Monit 2025; 30:18-23. [PMID: 39436209 DOI: 10.1097/mbp.0000000000000729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The impact of wearing a face mask for an extended duration is unknown. This study aimed to determine if wearing a face mask for 10 h impacts blood pressure (BP) and arterial stiffness. Subjects received an ambulatory blood pressure cuff and were asked to wear it for 10 h while readings were taken every 15 min. During the face mask trial, subjects wore an N95 mask for 10 h. During the control, subjects did not wear a mask. Subjects were randomized to start their trial. An accelerometer was given to ensure no physical activity differences. Linear mixed models were used to determine group differences, and McNemar test was used to assess frequency differences when determining BP load. Twelve college-aged (20.5 ± 1.5 years) male ( n = 5) and female ( n = 7) individuals with normal BP participated in this study. There were no differences in time spent in any physical activity domain (all P > 0.05). There was no difference in brachial SBP ( P = 0.688), brachial DBP ( P = 0.063), central SBP ( P = 0.875), central DBP ( P = 0.246), heart rate ( P = 0.125), and augmentation pressure ( P = 0.158) between conditions. During mask condition, augmentation pressure was reduced by 5.2 ± 3.1% compared to control ( P < 0.001). There were no frequency differences in the number of BP readings above 140 mmHg for SBP ( P = 0.479) and >90 mmHg for DBP ( P = 0.212). The current study found that wearing an N95 mask for 10 h did not affect brachial or central BP but significantly decreased augmentation pressure.
Collapse
Affiliation(s)
- Zachary Zeigler
- Exercise and Nutritional Sciences, College of Natural Science, Grand Canyon University, Phoenix, Arizona, USA
| | | | | | | | | |
Collapse
|
64
|
Nagy A, Czitrovszky A, Lehoczki A, Farkas Á, Füri P, Osán J, Groma V, Kugler S, Micsinai A, Horváth A, Ungvári Z, Müller V. Creating respiratory pathogen-free environments in healthcare and nursing-care settings: a comprehensive review. GeroScience 2025; 47:543-571. [PMID: 39392557 PMCID: PMC11872867 DOI: 10.1007/s11357-024-01379-7] [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: 08/02/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
Hospital- and nursing-care-acquired infections are a growing problem worldwide, especially during epidemics, posing a significant threat to older adults in geriatric settings. Intense research during the COVID-19 pandemic highlighted the prominent role of aerosol transmission of pathogens. Aerosol particles can easily adsorb different airborne pathogens, carrying them for a long time. Understanding the dynamics of airborne pathogen transmission is essential for controlling the spread of many well-known pathogens, like the influenza virus, and emerging ones like SARS-CoV-2. Particles smaller than 50 to 100 µm remain airborne and significantly contribute to pathogen transmission. This review explores the journey of pathogen-carrying particles from formation in the airways, through airborne travel, to deposition in the lungs. The physicochemical properties of emitted particles depend on health status and emission modes, such as breathing, speaking, singing, coughing, sneezing, playing wind instruments, and medical interventions. After emission, sedimentation and evaporation primarily determine particle fate. Lung deposition of inhaled aerosol particles can be studied through in vivo, in vitro, or in silico methods. We discuss several numerical lung models, such as the Human Respiratory Tract Model, the LUng Dose Evaluation Program software (LUDEP), the Stochastic Lung Model, and the Computational Fluid Dynamics (CFD) techniques, and real-time or post-evaluation methods for detecting and characterizing these particles. Various air purification methods, particularly filtration, are reviewed for their effectiveness in healthcare settings. In the discussion, we analyze how this knowledge can help create environments with reduced PM2.5 and pathogen levels, enhancing safety in healthcare and nursing-care settings. This is particularly crucial for protecting older adults, who are more vulnerable to infections due to weaker immune systems and the higher prevalence of chronic conditions. By implementing effective airborne pathogen control measures, we can significantly improve health outcomes in geriatric settings.
Collapse
Affiliation(s)
- Attila Nagy
- Department of Applied and Nonlinear Optics, HUN-REN Wigner Research Centre for Physics, Konkoly-Thege Miklós St. 29-33, 1121, Budapest, Hungary.
| | - Aladár Czitrovszky
- Department of Applied and Nonlinear Optics, HUN-REN Wigner Research Centre for Physics, Konkoly-Thege Miklós St. 29-33, 1121, Budapest, Hungary
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Árpád Farkas
- Environmental Physics Department, HUN-REN Centre for Energy Research, Budapest, Hungary
| | - Péter Füri
- Environmental Physics Department, HUN-REN Centre for Energy Research, Budapest, Hungary
| | - János Osán
- Environmental Physics Department, HUN-REN Centre for Energy Research, Budapest, Hungary
| | - Veronika Groma
- Environmental Physics Department, HUN-REN Centre for Energy Research, Budapest, Hungary
| | - Szilvia Kugler
- Environmental Physics Department, HUN-REN Centre for Energy Research, Budapest, Hungary
| | | | - Alpár Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Zoltán Ungvári
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
65
|
Ji X, Bian Y, Zhang C, Zhong Z, Wang Y. Making Accessible and Attractive Porosities in Block Copolymer Nanofibers for Highly Permeable and Durable Air Filtration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410692. [PMID: 39723691 DOI: 10.1002/smll.202410692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Submicron particulate matter (PM) can penetrate deeply into human tissue, posing a serious threat to human health. However, the electrostatic charge of commercial respirators is easily dissipated, making it difficult to maintain long-term filtration. Herein, a hierarchically porous filter based on nanofibers with accessible porosity and particulate-attractive surfaces, achieving significant filtration performance is developed through polarity-driven interactions. This is achieved by selective swelling of electrospun nanofibers of the block copolymer of polysulfone and poly(ethylene glycol) (PSF-b-PEG), in which the originally solid nanofibers are 3D perforated with the PEG chains lined along the pore walls. Thus-produces nanofiber filters exhibit a long-term continuous filtration with an efficiency of over 95% for PM0.3 and a low pressure drop of only 40 Pa. In particular, it maintains superior filtration performance even under high particle concentrations and high humidity conditions. Additionally, the filter exhibits high air permeability (10814 m3 m-2 h-1 kPa-1) and water vapor transmission rate (3707 g m-2 d-1). This work provides new strategies and understandings on the development of porous structures simultaneously exhibiting high gas permeability and efficient particulate rejection.
Collapse
Affiliation(s)
- Xuzheng Ji
- School of Energy and Environment, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Ye Bian
- School of Energy and Environment, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Chencheng Zhang
- School of Energy and Environment, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Zhaoxiang Zhong
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing, Jiangsu, 211816, China
| | - Yong Wang
- School of Energy and Environment, Southeast University, Nanjing, Jiangsu, 210096, China
| |
Collapse
|
66
|
França TDS, da Silva JFA, da Silva GCN, dos Santos BO, Silva SA, Linhares JHR, da Silva MAN, Barreto-Vieira DF, de Paula VS, de Morais LM, Santos RT, Trindade GF. Subgenomic RNA Detection in SARS-CoV-2 Assessing Replication and Inactivation Through Serial Passages, RT-qPCR, and Electron Microscopy. Int J Mol Sci 2025; 26:1281. [PMID: 39941049 PMCID: PMC11818844 DOI: 10.3390/ijms26031281] [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: 12/02/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Subgenomic RNAs (sgRNAs) are potential markers of active SARS-CoV-2 replication, serving as templates for the synthesis of structural and accessory proteins in infectious viral particles. This study aimed to use RT-qPCR to quantify sgRNA and negative RNA intermediates, assessing viral replication in virus samples inactivated by β-propiolactone (βPL). Inactivated viruses subjected to five blind serial passages (BSs) were amplified by RT-qPCR using primers to target the envelope (ENV) and nucleoproteins (N1 and N2) of genomic genes, subgenomic envelope RNA (sgENV), and intermediate envelope RNA (ENV-). All positive controls showed consistent viral titers across passages (10 log10 copies/mL in N1/N2 and 11 log10 copies/mL in ENV) during BSs. Inactivated viral samples for ENV and ENV- targets ranged from 11.34 log10 copies/mL in BS1 to 11.20 log10 copies/mL in BS5. The sgENV was no longer detected in the inactivated SARS-CoV-2 samples after the second passage, suggesting successful inactivation. Replication kinetics showed consistent profiles for N1/N2, ENV, and ENV- targets in the first three post-infection hours (pih) and maintained approximately 5 log10 copies/mL at 1 pih, 2 pih, and 3 pih. A sharp exponential increase in the viral titer was observed from 24 pih onwards, peaking at 11.64 log10 copies/mL at 48 pih. Transmission electron microscopy confirmed viral particles only in cells infected with active SARS-CoV-2. These results support the use of sgRNA as a reliable marker for SARS-CoV-2 replication, especially in distinguishing between active replication and non-viable particles and in the development of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Talita da Silva França
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | | | | | - Barbara Oliveira dos Santos
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | - Stephanie Almeida Silva
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | - José Henrique Resende Linhares
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | - Marcos Alexandre Nunes da Silva
- Viral Morphology and Morphogenesis Laboratory, Oswaldo Cruz Institute/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.A.N.d.S.); (D.F.B.-V.)
| | - Debora Ferreira Barreto-Vieira
- Viral Morphology and Morphogenesis Laboratory, Oswaldo Cruz Institute/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.A.N.d.S.); (D.F.B.-V.)
| | - Vanessa Salete de Paula
- Molecular Virology and Parasitology Laboratory, Oswaldo Cruz Institute/FIOCRUZ, Rio de Janeiro 21040-900, Brazil;
| | - Liliane Monteiro de Morais
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | - Renata Tourinho Santos
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| | - Gisela Freitas Trindade
- Virological Technology Laboratory, Bio-Manguinhos/FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.d.S.F.); (B.O.d.S.); (S.A.S.); (J.H.R.L.); (L.M.d.M.); (R.T.S.)
| |
Collapse
|
67
|
Pham NT, Ko J, Shah M, Rakkiyappan R, Woo HG, Manavalan B. Leveraging deep transfer learning and explainable AI for accurate COVID-19 diagnosis: Insights from a multi-national chest CT scan study. Comput Biol Med 2025; 185:109461. [PMID: 39631112 DOI: 10.1016/j.compbiomed.2024.109461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
The COVID-19 pandemic has emerged as a global health crisis, impacting millions worldwide. Although chest computed tomography (CT) scan images are pivotal in diagnosing COVID-19, their manual interpretation by radiologists is time-consuming and potentially subjective. Automated computer-aided diagnostic (CAD) frameworks offer efficient and objective solutions. However, machine or deep learning methods often face challenges in their reproducibility due to underlying biases and methodological flaws. To address these issues, we propose XCT-COVID, an explainable, transferable, and reproducible CAD framework based on deep transfer learning to predict COVID-19 infection from CT scan images accurately. This is the first study to develop three distinct models within a unified framework by leveraging a previously unexplored large dataset and two widely used smaller datasets. We employed five known convolutional neural network architectures, both with and without pretrained weights, on the larger dataset. We optimized hyperparameters through extensive grid search and 5-fold cross-validation (CV), significantly enhancing the model performance. Experimental results from the larger dataset showed that the VGG16 architecture (XCT-COVID-L) with pretrained weights consistently outperformed other architectures, achieving the best performance, on both 5-fold CV and independent test. When evaluated with the external datasets, XCT-COVID-L performed well with data with similar distributions, demonstrating its transferability. However, its performance significantly decreased on smaller datasets with lower-quality images. To address this, we developed other models, XCT-COVID-S1 and XCT-COVID-S2, specifically for the smaller datasets, outperforming existing methods. Moreover, eXplainable Artificial Intelligence (XAI) analyses were employed to interpret the models' functionalities. For prediction and reproducibility purposes, the implementation of XCT-COVID is publicly accessible at https://github.com/cbbl-skku-org/XCT-COVID/.
Collapse
Affiliation(s)
- Nhat Truong Pham
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea
| | - Jinsol Ko
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea
| | - Masaud Shah
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Rajan Rakkiyappan
- Department of Mathematics, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea; Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea; Ajou Translational Omics Center (ATOC), Ajou University Medical Center, Republic of Korea.
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
68
|
Haj Husin A, Ahrari H, Keep J. Predicting COVID-19 Patient Disposition Using the National Early Warning Score 2: A Retrospective Cohort Study. Cureus 2025; 17:e79610. [PMID: 40151731 PMCID: PMC11948155 DOI: 10.7759/cureus.79610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives To evaluate the effectiveness of the National Early Warning Score 2 (NEWS2) in predicting the need for hospital admission and close monitoring of suspected patients with COVID-19 presenting to the Emergency Department (ED). This study aims to determine whether NEWS2 can aid in identifying high-risk patients with COVID-19 requiring urgent care and admission. Methodology Retrospective data from electronic health records of 300 patients with COVID-19 who presented to Mediclinic Parkview Hospital ED between January 1, 2021, and June 30, 2021, were analyzed. Collected variables included age, gender, body mass index (BMI), vital signs, and patient disposition. Statistical analysis was conducted to assess the ability of NEWS2 to predict COVID-19 patient disposition. Results A total of 300 patients were included, and their NEWS2 scores were analyzed to predict clinical deterioration. NEWS2, with a cutoff value of 2, predicted hospital admission with 86% sensitivity and 75% specificity. It achieved an average area under the curve (AUC) of 0.86 for predicting outcomes at 24 to 72 hours from the time of initial presentation to the ED. Conclusions NEWS2 demonstrates high sensitivity in predicting the disposition of patients with COVID-19. Our findings support the use of NEWS2 as a useful tool for the initial assessment of patients with COVID-19 presenting to the ED, assisting in identifying patients at risk of deterioration.
Collapse
Affiliation(s)
- Asmaa Haj Husin
- Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, ARE
| | - Hind Ahrari
- Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, ARE
| | - Jeffrey Keep
- Emergency, Mediclinic Parkview Hospital, Dubai, ARE
| |
Collapse
|
69
|
Madhloom IH, Othman RM, Al-Bayati HAM. Molecular detection of coronavirus in camelids and bovines using real-time quantitative polymerase chain reaction in Wasit Province, Iraq. Open Vet J 2025; 15:765-773. [PMID: 40201811 PMCID: PMC11974302 DOI: 10.5455/ovj.2025.v15.i2.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/08/2025] [Indexed: 04/10/2025] Open
Abstract
Background Coronaviruses (CoVs) are a diverse group of RNA viruses that cause respiratory and gastrointestinal diseases in humans and animals. Over the past two decades, outbreaks of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) and bovine coronavirus (BCoV) have affected animal populations, especially in regions with close animal-human interactions, such as the Arabian Peninsula and Iraq. Given the potential for zoonotic transmission, understanding the prevalence and spread of CoVs among livestock is essential for managing potential risks to animal and human health. Aim This study aimed to investigate the prevalence of MERS-CoV in camels and BCoV in bovines within the Wasit Governorate of Iraq to assess the infection rates and potential interspecies transmission risks. Methods One hundred and fifty nasal swab samples (75 from camels and 75 from bovines) were collected between November 2022 and April 2023. The samples were analyzed for the presence of MERS-CoV and BCoV using real- time quantitative reverse transcription PCR (qRT-PCR) targeting the nucleocapsid (N) gene for each virus. Standard procedures for RNA extraction were followed, and qRT-PCR assays were conducted using specific primers to ensure high sensitivity and specificity. Results MERS-CoV was present in (42%) of the camel samples, whereas BCoV was detected in (34%) of the bovine samples. Statistical analysis indicated a significant difference (p < 0.05) in infection rates between camels and bovines, with a higher prevalence observed in camels. The clinical signs observed in infected camels included fever, nasal discharge, and appetite loss, whereas infected bovines exhibited symptoms such as diarrhea and respiratory distress. Conclusion The high prevalence of MERS-CoV and BCoV in camels and bovines in the Wasit region indicates a substantial risk for the continued spread of these viruses within animal populations. These findings underscore the importance of surveillance and biosecurity measures to control the spread of coronavirus among livestock, potentially reducing zoonotic transmission risks. Further research is required to understand the transmission dynamics of CoVs in mixed livestock farming systems.
Collapse
Affiliation(s)
- Ibrahim Hasan Madhloom
- Department of Microbiology, College of Veterinary Medicine, University of Basrah, Basrah, Iraq
| | - Rasha Munther Othman
- Department of Microbiology, College of Veterinary Medicine, University of Basrah, Basrah, Iraq
| | | |
Collapse
|
70
|
Ferreira FC, Pervin T, Tang WW, Hediger JA, Thomas LF, Cook WE, Cherry MJ, Neuman BW, Hamer GL, Hamer SA. Respiratory Shedding of Infectious SARS-CoV-2 Omicron XBB.1.41.1 Lineage among Captive White-Tailed Deer, Texas, USA. Emerg Infect Dis 2025; 31:267-274. [PMID: 39983708 PMCID: PMC11845158 DOI: 10.3201/eid3102.241458] [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: 02/23/2025] Open
Abstract
White-tailed deer (Odocoileus virginianus) have high value for research, conservation, agriculture, and recreation and might be key SARS-CoV-2 reservoirs. In November 2023, we sampled 15 female deer in a captive facility in Texas, USA. All deer had neutralizing antibodies to SARS-CoV-2; respiratory swab samples from 11 deer were SARS-CoV-2-positive by quantitative reverse transcription PCR, and 1 deer also had a positive rectal swab sample. Six of the 11 respiratory swab samples yielded infectious virus; replication kinetics of most samples displayed lower growth 24-48 hours postinfection in vitro than Omicron lineages isolated from humans in Texas in the same period. Virus growth was similar between groups by 72 hours, suggesting no strong attenuation of deer-derived virus. All deer viruses clustered in XBB Omicron clade and demonstrated more mutations than expected compared with contemporaneous viruses in humans, suggesting that crossing the species barrier was accompanied by a high substitution rate.
Collapse
|
71
|
Figueiroa T, Galvão Bueno M, Bento Moura PE, de Oliveira MB, Passos Cordeiro JL, Santos-Cavalcante N, Camacho Antevere Mazzarotto GA, Wallau GL, Corrêa da Silva Junior L, Resende PC, Siqueira MMM, Ogrzewalska M. Alpha and Betacoronavirus Detection in Neotropical Bats from Northeast Brazil Suggests Wide Geographical Distribution and Persistence in Natural Populations. Animals (Basel) 2025; 15:332. [PMID: 39943102 PMCID: PMC11816360 DOI: 10.3390/ani15030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 02/16/2025] Open
Abstract
The emergence of zoonotic viral diseases, notably exemplified by the recent coronavirus disease pandemic in 2019 (COVID-19), underscores the critical need to understand the dynamics of viruses circulating in wildlife populations. This study aimed to investigate the diversity of coronaviruses in bat populations from northeastern Brazil, particularly in the state of Ceará, where little research on bat pathogens has been conducted previously. Bat sampling was performed between March 2021 and March 2022 across three municipalities, resulting in the collection of oral and rectal swabs from 298 captured individuals. Molecular analyses revealed alphacoronaviruses in multiple bat species. Additionally, a novel Betacoronavirus was identified in Artibeus planirostris, which did not fall within an established subgenus. Phylogenetic placement of these new coronavirus sequences suggests that closely related coronavirus lineages can infect a wide range of bat species sampled in distantly related Brazilian states and biomes. No SARS-CoV-2 and influenza A viruses were found in the sampled bats. These findings expand our understanding of coronavirus diversity in Brazilian bats. The detection of coronaviruses in various bat species underscores the importance of bats as reservoirs for these viruses. The absence of SARS-CoV-2 in the sampled bats indicates a lack of spillback events from human or environmental sources. However, the potential for future transmission events underscores the importance of ongoing surveillance and transmission mitigation protocols in wildlife management practices.
Collapse
Affiliation(s)
- Thays Figueiroa
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Marina Galvão Bueno
- Fundação Oswaldo Cruz, IOC, Laboratório de Virologia Comparada e Ambiental, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Patricia Emilia Bento Moura
- Fundação Oswaldo Cruz, IOC, Laboratório de Virologia Comparada e Ambiental, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Marcione Brito de Oliveira
- Museu Nacional, Departamento de Vertebrados, Setor de Mastozoologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 20940-040, RJ, Brazil;
| | - José Luís Passos Cordeiro
- Fundação Oswaldo Cruz, Unidade do Ceará, Área de Saúde e Ambiente, Eusébio, Ceará 61773-270, CE, Brazil;
- Plataforma Internacional para Ciência, Tecnologia e Inovação em Saúde (PICTIS), Via do Conhecimento, Edifício Central, 3830-352 Ílhavo, Portugal
| | - Nádia Santos-Cavalcante
- Museu de História Natural do Ceará Prof. Dias da Rocha, Universidade Estadual do Ceará, Pacoti, Ceará 62770-000, CE, Brazil;
- Fundação Oswaldo Cruz, Instituto Lêonidas and Maria Deane (ILDM), Unidade da Amazônia, Manaus 69057-070, AM, Brazil
| | - Giovanny A. Camacho Antevere Mazzarotto
- Fundação Oswaldo Cruz, Unidade do Ceará, Laboratório Analítico de Competências Moleculares e Epidemiológicas, Plataforma de Camelídeos e Produção de Nanocorpos, Eusébio, Ceará 61773-270, CE, Brazil;
| | - Gabriel Luz Wallau
- Fundação Oswaldo Cruz, Departamento de Entomologia e Núcleo de Bioinformática, Instituto Aggeu Magalhães (IAM), Cidade Universitária, Recife 50740-465, PE, Brazil;
- Department of Arbovirology and Entomology, Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, National Reference Center for Tropical Infectious Diseases, 20359 Hamburg, Germany
| | - Leonardo Corrêa da Silva Junior
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Paola Cristina Resende
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Marilda M. Mendonça Siqueira
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Maria Ogrzewalska
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| |
Collapse
|
72
|
Patel RS, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Kumar R, Tran A, Agrawal B. Mixed lipopeptide-based mucosal vaccine candidate induces cross-variant immunity and protects against SARS-CoV-2 infection in hamsters. Immunohorizons 2025; 9:vlae011. [PMID: 39849995 PMCID: PMC11841972 DOI: 10.1093/immhor/vlae011] [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: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
The global dissemination of SARS-CoV-2 led to a worldwide pandemic in March 2020. Even after the official downgrading of the COVID-19 pandemic, infection with SARS-CoV-2 variants continues. The rapid development and deployment of SARS-CoV-2 vaccines helped to mitigate the pandemic to a great extent. However, the current vaccines are suboptimal; they elicit incomplete and short-lived protection and are ineffective against evolving virus variants. Updating the spike antigen according to the prevailing variant and repeated boosters is not the long-term solution. We have designed a lipopeptide-based, mucosal, pan-coronavirus vaccine candidate, derived from highly conserved and/or functional regions of the SARS-CoV-2 spike, nucleocapsid, and membrane proteins. Our studies demonstrate that the designed lipopeptides (LPMix) induced both cellular and humoral (mucosal and systemic) immune responses upon intranasal immunization in mice. Furthermore, the antibodies bound to the wild-type and mutated S proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Delta and Omicron, and also led to efficient neutralization in a surrogate viral neutralization assay. Our sequence alignment and 3-dimensional molecular modeling studies demonstrated that spike-derived epitopes, P1 and P2, are sequentially and/or structurally conserved among the SARS-CoV-2 variants. The addition of a novel mucosal adjuvant, heat-killed Caulobacter crescentus, to the lipopeptide vaccine significantly bolstered mucosal antibody responses. Finally, the lipopeptide-based intranasal vaccine demonstrated significant improvement in lung pathologies in a hamster model of SARS-CoV-2 infection. These studies are fundamentally important and open new avenues in the investigation of an innovative, broadly protective intranasal vaccine platform for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Raj S Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, 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
| | - Melissa Hewitt
- Preclinical Imaging, 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
| | | | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
73
|
Zhang W, Gai X, Duan Z, Yan C, Huang C, Wu C, Zheng S, Lin Z, Zhou Q, Dai L, Yang P, Bao F, Jing H, Cai C, Ma Y, Sun Y. The immune landscape and viral shedding of Omicron SARS-CoV-2 variants implicate immune escape. Front Med (Lausanne) 2025; 11:1478466. [PMID: 39931433 PMCID: PMC11808361 DOI: 10.3389/fmed.2024.1478466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/26/2024] [Indexed: 02/13/2025] Open
Abstract
Background Three years into the SARS-CoV-2 pandemic, the virus continues to mutate despite widespread vaccination, posing ongoing challenges for epidemic prevention and control. The relationship between viral shedding and immune escape remains under investigation. This study aims to examine the association between viral shedding and immune escape in the BA.4/5 and BF.7 variants. Method We included 542 patients infected with the Omicron variant from Beijing Xiaotangshan shelter hospital. Based on the viral strain, patients were divided into BA.4/5 group and BF.7 group. Additionally, we categorized patients into rapid viral shedding and slow viral shedding groups according to their viral shedding rates. We explored the relationship between viral shedding and immune-related clinical indicators during this period. Result Of the 542 patients, 118 were infected with BA.4/5 variant, and 424 were infected with BF.7 variant. The viral shedding duration differed significantly between BA.4/5 and BF.7 groups (p < 0.0001). However, there was no statistically significant correlation between viral shedding duration and immune-related indicators, such as WBC, Hb, PLT, Neu, Lym, CRP, allergy, fever, and vaccination status (p > 0.05). Furthermore, viral shedding duration was not associated with vaccination status, intervals between vaccinations, or vaccine types (p > 0.05). Conclusion The duration of viral shedding in patients infected with Omicron variants BA.4/5 and BF.7 is not associated with WBC, Hb, Lym, CRP, fever, allergy, or vaccine-related indicators. This lack of association may be attributed to immune escape mechanisms.
Collapse
Affiliation(s)
- Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
| | - Zhonghui Duan
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Changjian Yan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Chunyuan Huang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Chaoling Wu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Siping Zheng
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Zixiang Lin
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Qingtao Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
| | - Lili Dai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Fang Bao
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Chao Cai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, and Center for Chronic Airway Diseases, Peking University Health Science Center, Peking University, Beijing, China
| |
Collapse
|
74
|
Liu L, Mu BR, Zhou Y, Wu QL, Li B, Wang DM, Lu MH. Research Trends and Development Dynamics of qPCR-based Biomarkers: A Comprehensive Bibliometric Analysis. Mol Biotechnol 2025:10.1007/s12033-024-01356-7. [PMID: 39843617 DOI: 10.1007/s12033-024-01356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025]
Abstract
Quantitative polymerase chain reaction (qPCR) is a vital molecular technique for biomarker detection; however, its clinical application is impeded by the scarcity of robust biomarkers and the inherent limitations of the technology. This study conducted a bibliometric analysis of 4063 qPCR-based biomarker studies sourced from the Web of Science (WOS) database, employing VOSviewer and CiteSpace to generate multi-dimensional structural insights into this field. The results reveal a growing trend in research within this domain, with gene expression analysis playing a central role in the identification of potential biomarkers. Among these, cancer-related biomarkers are the most prominent, while research on biomarkers for other diseases remains limited. Liquid biopsy biomarkers, including microRNA (miRNA), circulating free DNA (cfDNA), and circulating tumor DNA (ctDNA), are increasingly being explored. The integration of bioinformatics, omics analysis, and high-throughput technologies with qPCR is accelerating biomarker discovery. Furthermore, large-scale parallel sequencing is emerging as a potential alternative to relative quantification and microarray techniques. Nevertheless, qPCR remains essential for validating specific biomarkers, and further standardization of its protocols is necessary to enhance reliability. This study provides a systematic analysis of qPCR-based biomarker research and underscores the need for future technological integration and standardization to facilitate broader clinical applications.
Collapse
Affiliation(s)
- Li Liu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Ya Zhou
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Qing-Lin Wu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Bin Li
- Department of Respiratory Medicine, Guangyuan Hospital of Traditional Chinese Medicine, No.133 Jianshe Road, Lizhou District, Guangyuan, 628099, Sichuan, China
| | - Dong-Mei Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
75
|
Cardona-Ramírez C, Enríquez-Valencia CE, Méndez-Callejas G, Barreto GM, Tafur-Gómez GA, Sanjuanelo-Corredor DW. An integrated methodological framework for the validation and verification of clinical testing by qRT-PCR. Heliyon 2025; 11:e41088. [PMID: 39801958 PMCID: PMC11721250 DOI: 10.1016/j.heliyon.2024.e41088] [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: 05/20/2024] [Revised: 11/14/2024] [Accepted: 12/08/2024] [Indexed: 01/16/2025] Open
Abstract
This paper outlines a practical method for validating quantitative-qualitative techniques used to detect genetic material through qRT-PCR, specifically focusing on SARS-CoV-2 testing and adhering to ISO/IEC 17025:2018 accreditation standards. Despite the prevalence of quantitative-qualitative screening in genetic testing, comprehensive validation guidelines remain a notable gap in the field. Such guidelines could be applied to other molecular testing areas that rely on these techniques, particularly those involving sample handling, automated extraction, and amplification processes, which can significantly impact results. This work describes the statistical approaches behind qRT-PCR protocols, followed by a technical characterization profile of the validation process. Modifications to the gold standard method allowed us to establish a technical limit of detection (LOD) of 5,09 copies/reaction at a 95 % confidence interval.
Collapse
Affiliation(s)
- Carolina Cardona-Ramírez
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada GIBGA, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, Colombia
| | - Cruz Elena Enríquez-Valencia
- Grupo de Investigación Ciencia Animal, Facultad de Ciencias Agropecuarias, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, Colombia
| | - Gina Méndez-Callejas
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada GIBGA, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, Colombia
| | - Giovanna Meza Barreto
- Centro de Investigación y Diagnóstico Molecular BSL-3, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, Colombia
| | - Gabriel Andrés Tafur-Gómez
- Grupo de Investigación Ciencia Animal, Facultad de Ciencias Agropecuarias, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, Colombia
| | | |
Collapse
|
76
|
Alicia LB, María Ángeles OG, Desirée MG, Maximino R, Marilina GA. Utility of Protein Markers in COVID-19 Patients. Int J Mol Sci 2025; 26:653. [PMID: 39859366 PMCID: PMC11766239 DOI: 10.3390/ijms26020653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has been a challenge at the healthcare level not only in the early stages of the pandemic, but also in the subsequent appearance of long-term COVID-19. Several investigations have attempted to identify proteomic biomarkers in an attempt to improve clinical care, guide treatment and predict possible patient outcomes. Proteins such as C-reactive protein (CRP) or interleukin 6 (IL-6) are clear markers of severe disease, but many others have been proposed that could help in risk stratification and in the prediction of specific complications. This review aims to bring together the most relevant studies in this regard, providing information to identify the most notable biomarkers in relation to COVID-19 found to date.
Collapse
Affiliation(s)
- López-Biedma Alicia
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
| | - Onieva-García María Ángeles
- Preventive Medicine and Public Health Unit, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Preventive Medicine and Public Health Research Group, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Martín-García Desirée
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Redondo Maximino
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - García-Aranda Marilina
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
| |
Collapse
|
77
|
Futschik ME, Kulasegaran-Shylini R, Blandford E, Harper S, Chapman D, Turek E, Agrawal S, Phillips V, Fordham H, Chan L, Kidd M, Dodgson A, Klapper PE, Sudhanva M, Vipond R, Hopkins S, Peto T, Tunkel S, Fowler T. Effectiveness and user experience of nose and throat swabbing techniques for SARS-CoV-2 detection: results from the UK COVID-19 National Testing Programme. BMC GLOBAL AND PUBLIC HEALTH 2025; 3:5. [PMID: 39806484 PMCID: PMC11731392 DOI: 10.1186/s44263-024-00121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The UK's National Health Service Test and Trace (NHSTT) program aimed to provide the most effective and accessible SARS-CoV-2 testing approach possible. Early user feedback indicated that there were accessibility issues associated with throat swabbing. We report the results of service evaluations performed by NHSTT to assess the effectiveness and user acceptance of swabbing approaches, as well as qualitative findings of user experiences from research reports, surveys, and incident reports. Our intent is to present and summarize our findings about the application of alternative swabbing approaches during the COVID-19 pandemic in the UK. METHODS From May 2020 to December 2021, NHSTT conducted a series of service evaluations assessing self-swabbing and assisted swabbing of the nose and throat, and nose only (anterior nares/mid-turbinate) using polymerase chain reaction (PCR) and lateral flow devices (LFDs), for diagnostic suitability within the COVID-19 National Testing Programme. Outcomes included observational user feedback on swabbing approaches and quantitative testing performance (concordance, sensitivity, and specificity). A post-hoc indirect comparison of swabbing approaches was also performed. Additionally, an analysis of existing cross-service research was conducted in April 2021 to determine user feedback regarding swabbing approaches. RESULTS Observational data from cross-service research indicated a user preference for nose swabbing over throat swabbing. Significantly more users reported that nose swabbing was easier to perform than throat swabbing (50% vs. 12%) and there were significantly fewer reported incidents. In the service evaluations, while there was reduced sensitivity for nose-only swabbing for PCR (88%) compared with nose and throat swabbing, similar sensitivities were observed for nose-only and nose and throat swabbing for LFDs. The sensitivity of nose-only swabbing for LFDs was higher for individuals with higher viral concentrations. CONCLUSIONS User experience analyses supported a preference for nose-only swabbing. Nose-only swabbing for LFDs provided sufficient diagnostic accuracy, supporting its use as a viable option in the COVID-19 National Testing Programme. Less invasive swabbing approaches are important to maximize testing accessibility and alongside other behavioral interventions, increase user uptake.
Collapse
Affiliation(s)
- Matthias E Futschik
- UK Health Security Agency, London, UK
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, UK
| | | | | | | | - David Chapman
- UK Health Security Agency, London, UK
- Deloitte MCS Ltd, London, UK
| | - Elena Turek
- UK Health Security Agency, London, UK
- Deloitte MCS Ltd, London, UK
| | | | | | | | - Lee Chan
- UK Health Security Agency, London, UK
| | - Mike Kidd
- UK Health Security Agency, London, UK
| | | | - Paul E Klapper
- UK Health Security Agency, London, UK
- School of Biological Sciences, University of Manchester, Manchester, UK
| | - Malur Sudhanva
- UK Health Security Agency, London, UK
- Kings College Hospital NHS Foundation Trust, London, UK
| | | | - Susan Hopkins
- UK Health Security Agency, London, UK
- Health Protection Research Unit in Healthcare Associate Infections and Antimicrobial Resistance, National Institute for Health Research, Oxford, UK
| | - Tim Peto
- Health Protection Research Unit in Healthcare Associate Infections and Antimicrobial Resistance, National Institute for Health Research, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Tom Fowler
- UK Health Security Agency, London, UK.
- William Harvey Research Institute and the Barts Cancer Institute, Queen Mary University of London, London, UK.
- Public Health Wales, Cardiff, Wales, UK.
| |
Collapse
|
78
|
Wickenhagen A, Flagg M, Port JR, Yinda CK, Goldin K, Gallogly S, Schulz JE, Lutterman T, Williamson BN, Kaiser F, Mukesh RK, van Tol S, Smith B, van Doremalen N, Russell CA, de Wit E, Munster VJ. Evolution of Omicron lineage towards increased fitness in the upper respiratory tract in the absence of severe lung pathology. Nat Commun 2025; 16:594. [PMID: 39799119 PMCID: PMC11724920 DOI: 10.1038/s41467-025-55938-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
The emergence of the Omicron lineage represented a major genetic drift in SARS-CoV-2 evolution. This was associated with phenotypic changes including evasion of pre-existing immunity and decreased disease severity. Continuous evolution within the Omicron lineage raised concerns of potential increased transmissibility and/or disease severity. To address this, we evaluate the fitness and pathogenesis of contemporary Omicron variants XBB.1.5, XBB.1.16, EG.5.1, and JN.1 in the upper (URT) and lower respiratory tract (LRT). We compare in vivo infection in Syrian hamsters with infection in primary human nasal and lung epithelium cells and assess differences in transmissibility, antigenicity, and innate immune activation. Omicron variants replicate efficiently in the URT but display limited pathology in the lungs compared to previous variants and fail to replicate in human lung organoids. JN.1 is attenuated in both URT and LRT compared to other Omicron variants and fails to transmit in the male hamster model. Our data demonstrate that Omicron lineage evolution has favored increased fitness in the URT.
Collapse
Affiliation(s)
- Arthur Wickenhagen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Meaghan Flagg
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julia R Port
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Laboratory of Transmission Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Claude Kwe Yinda
- 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
| | - Shane Gallogly
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan E Schulz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Tessa Lutterman
- 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
| | - Franziska Kaiser
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Reshma K Mukesh
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Sarah van Tol
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Brian Smith
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institutes of Health, Hamilton, MT, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Colin A Russell
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
79
|
Cavalcante FV, Martin CPS, Frio GS, Stabeli RG, Santos LMP. Laboratory readiness and genomic surveillance of Covid-19 in the capital of Brazil. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003289. [PMID: 39787152 PMCID: PMC11717220 DOI: 10.1371/journal.pgph.0003289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/14/2024] [Indexed: 01/12/2025]
Abstract
This study aims to analyze the diagnostic readiness to Covid-19 and the genomic surveillance of SARS-CoV-2 in Brasília, the capital of Brazil. This is a retrospective, cross-sectional study, with data from: cases/deaths-Ministry of Health; RT-PCR analyses Brasília Central Public Health Laboratory (LACEN); genomics-Global Initiative on Sharing All Influenza Data (GISAID). It was found that in March 2021, with the Gamma variant was predominant, RT-PCR diagnostic tests administered by LACEN reached their peak, followed by a reduction, possibly due to the start of vaccination. New peaks were observed in September 2021 and January 2022. The average time for releasing RT-PCR results was reduced from eight days (July 2020), to around eight hours in 2023. The participation of private laboratories was evident in sequencing the SARS-CoV-2 variants of concern in Brasília (n = 1,897). LACEN sequenced 50% of the samples received (571). A decrease in the incidence of cases and deaths due to Covid-19 was noted in the years 2022 to 2023, following the national trend. LACEN maintained RT-PCR diagnostic tests administered satisfactorily throughout the period. Regarding the genomic surveillance of SARS-CoV-2, the vast majority of samples were sequenced by private laboratories when compared to the public laboratory.
Collapse
Affiliation(s)
| | | | - Gustavo Saraiva Frio
- Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brasil
| | | | | |
Collapse
|
80
|
Molina Ramirez SR, Samiseresht N, Martínez-Roque MA, Catania F, Graef K, Rabe M, Offenhäusser A, Mayer D, Figueroa-Miranda G. A Truncated Multi-Thiol Aptamer-Based SARS-CoV-2 Electrochemical Biosensor: Towards Variant-Specific Point-of-Care Detection with Optimized Fabrication. BIOSENSORS 2025; 15:24. [PMID: 39852074 PMCID: PMC11763500 DOI: 10.3390/bios15010024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
With the goal of fast and accurate diagnosis of infectious diseases, this study presents a novel electrochemical biosensor that employs a refined aptamer (C9t) for the detection of spike (S) protein SARS-CoV-2 variants in a flexible multielectrode aptasensor array with PoC capabilities. Two aptamer modifications were employed: removing the primer binding sites and including two dithiol phosphoramidite anchor molecules. Thus, reducing fabrication time from 24 to 3 h and increasing the stability and sparseness for multi-thiol aptasensors compared to a standard aptasensor using single thiols, without a reduction in aptamer density. The biosensor fabrication, optimization, and detection were verified in detail by electrochemistry, QCM-D, SPR, and XPS. The analyte-receptor binding was further confirmed spectroscopically at the level of individual molecules by AFM-IR. The aptasensor possesses a low limit of detection (8.0 fg/mL), the highest sensitivity reported for S protein (209.5 signal per concentration decade), and a wide dynamic detection range (8.0 fg/mL-38 ng/mL) in nasopharyngeal samples, covering the clinically relevant range. Furthermore, the C9t aptasensor showed high selectivity for SARS-CoV-2 S proteins over biomarkers for MERS-CoV, RSV, and Influenza. Even more, it showed a three times higher sensitivity for the Omicron in comparison to the Wuhan strain (wild type), alpha, and beta variants of the SARS-CoV-2 virus. Those results demonstrate the creation of an affordable and variant-selective refined C9t aptasensor that outperformed current rapid diagnosis tests.
Collapse
Affiliation(s)
- Sergio Roberto Molina Ramirez
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Nafiseh Samiseresht
- Department of Interface Chemistry and Surface Engineering, Max Planck Institute for Sustainable Materials GmbH, 40237 Düsseldorf, Germany; (N.S.); (M.R.)
| | - Mateo Alejandro Martínez-Roque
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Ferdinando Catania
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Kevin Graef
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Martin Rabe
- Department of Interface Chemistry and Surface Engineering, Max Planck Institute for Sustainable Materials GmbH, 40237 Düsseldorf, Germany; (N.S.); (M.R.)
| | - Andreas Offenhäusser
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| | - Gabriela Figueroa-Miranda
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany; (S.R.M.R.); (M.A.M.-R.); (F.C.); (K.G.); (A.O.); (D.M.)
| |
Collapse
|
81
|
Franchitti E, Bottino P, Sidoti F, Carpino A, Pruccoli G, Ramenghi U, Costa C, Ala U, Parodi E, Traversi D. Investigating the Role of Gut Microbiota in Pediatric Patients with Severe COVID-19 or MIS-C. Microorganisms 2025; 13:83. [PMID: 39858851 PMCID: PMC11767266 DOI: 10.3390/microorganisms13010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Severe COVID-19 and MIS-C are rare but serious outcomes associated with SARS-CoV-2 infection. The onset of MIS-C often involves the gastrointestinal system, suggesting a potential connection with gut microbiota. This study aims to compare the gut microbiota of children with severe COVID-19 and those with MIS-C using various biomolecular approaches. Gut microbiota composition and specific microbial modulations were analyzed using fecal samples collected at hospital admission. The study included hospitalized patients (mean age 6 ± 5 years) diagnosed with severe COVID-19 (37 patients) or MIS-C (37 patients). Microbial differences were assessed using both NGS and qRT-PCR methodologies. In 75% of cases, pharmacological treatments included antibiotics and corticosteroids, which influenced the microbiota composition. Early age was found to have the most significant impact on microbiota diversity. Significant differences in alpha and beta diversity were observed between COVID-19 and MIS-C patients, particularly concerning low-abundance species. Levels of Bacteroides spp., Bifidobacterium spp., and Akkermansia muciniphila were comparable between groups, while an increased activity of Bifidobacterium spp. was noted in children with positive fecal samples (p = 0.019). An in-depth evaluation of lesser-known gut species may be key to reducing the risk of severe outcomes and developing microbiota-based biomarkers for the early diagnosis of MIS-C.
Collapse
Affiliation(s)
- Elena Franchitti
- Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.F.); (P.B.); (U.R.); (C.C.)
| | - Paolo Bottino
- Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.F.); (P.B.); (U.R.); (C.C.)
- Microbiology and Virology Laboratory, University Hospital “SS Antonio e Biagio e C. Arrigo”, Via Venezia 8 16, 15121 Alessandria, Italy
| | - Francesca Sidoti
- Division of Virology, Azienda Ospedaliero Universitaria—Città Della Salute e Della Scienza, Corso Bramante 88, 10126 Torino, Italy;
| | - Andrea Carpino
- Regina Margherita Paediartic Hospital, Azienda Ospedaliero Universitaria—Città Della Salute e Della Scienza, Piazza Polonia 94, 10126 Torino, Italy (G.P.)
| | - Giulia Pruccoli
- Regina Margherita Paediartic Hospital, Azienda Ospedaliero Universitaria—Città Della Salute e Della Scienza, Piazza Polonia 94, 10126 Torino, Italy (G.P.)
| | - Ugo Ramenghi
- Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.F.); (P.B.); (U.R.); (C.C.)
- Regina Margherita Paediartic Hospital, Azienda Ospedaliero Universitaria—Città Della Salute e Della Scienza, Piazza Polonia 94, 10126 Torino, Italy (G.P.)
| | - Cristina Costa
- Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.F.); (P.B.); (U.R.); (C.C.)
- Division of Virology, Azienda Ospedaliero Universitaria—Città Della Salute e Della Scienza, Corso Bramante 88, 10126 Torino, Italy;
| | - Ugo Ala
- Department of Veterinary Science, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, Italy;
| | - Emilia Parodi
- Struttura Complessa Pediatrica e Neonatologia, Azienda Ospedaliera Ordine Mauriziano Via Magellano 1, 10128 Torino, Italy;
| | - Deborah Traversi
- Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.F.); (P.B.); (U.R.); (C.C.)
| |
Collapse
|
82
|
Balistreri G. Nasal Mucociliary Epithelial Cell Culture Models for Studying Viral Infections. Methods Mol Biol 2025; 2890:237-252. [PMID: 39890731 DOI: 10.1007/978-1-0716-4326-6_13] [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: 02/03/2025]
Abstract
Respiratory nasal or lung epithelial cells serve as a valuable in vitro model for studying respiratory viral infections due to their physiological relevance and ability to recapitulate key aspects of the nasal or lung mucosa. In this chapter, we discuss the use of primary nasal epithelial cell cultures in studying viral infections, including their advantages, production methods, quality control, and identifiable disadvantages. Different methods for quantifying infection are presented with a special emphasis on how to adapt automated imaging methods and image analysis tools to the pseudostratified nasal epithelial cell models where cells are grown at the air-liquid interphase.
Collapse
Affiliation(s)
- Giuseppe Balistreri
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
83
|
Schrell L, Fuchs HL, Dickmanns A, Scheibner D, Olejnik J, Hume AJ, Reineking W, Störk T, Müller M, Graaf-Rau A, Diederich S, Finke S, Baumgärtner W, Mühlberger E, Balkema-Buschmann A, Dobbelstein M. Inhibitors of dihydroorotate dehydrogenase synergize with the broad antiviral activity of 4'-fluorouridine. Antiviral Res 2025; 233:106046. [PMID: 39638153 DOI: 10.1016/j.antiviral.2024.106046] [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: 10/05/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
RNA viruses present a constant threat to human health, often with limited options for vaccination or therapy. Notable examples include influenza viruses and coronaviruses, which have pandemic potential. Filo- and henipaviruses cause more limited outbreaks, but with high case fatality rates. All RNA viruses rely on the activity of a virus-encoded RNA-dependent RNA polymerase (RdRp). An antiviral nucleoside analogue, 4'-Fluorouridine (4'-FlU), targets RdRp and diminishes the replication of several RNA viruses, including influenza A virus and SARS-CoV-2, through incorporation into nascent viral RNA and delayed chain termination. However, the effective concentration of 4'-FlU varied among different viruses, raising the need to fortify its efficacy. Here we show that inhibitors of dihydroorotate dehydrogenase (DHODH), an enzyme essential for pyrimidine biosynthesis, can synergistically enhance the antiviral effect of 4'-FlU against influenza A viruses, SARS-CoV-2, henipaviruses, and Ebola virus. Even 4'-FlU-resistant mutant influenza A virus was re-sensitized towards 4'-FlU by DHODH inhibition. The addition of uridine rescued influenza A virus replication, strongly suggesting uridine depletion as a mechanism of this synergy. 4'-FlU was also highly effective against SARS-CoV-2 in a hamster model of COVID. We propose that the impairment of endogenous uridine synthesis by DHODH inhibition enhances the incorporation of 4'-FlU into viral RNAs. This strategy may be broadly applicable to enhance the efficacy of pyrimidine nucleoside analogues for antiviral therapy.
Collapse
Affiliation(s)
- Leon Schrell
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Hannah L Fuchs
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Antje Dickmanns
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - David Scheibner
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Adam J Hume
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Martin Müller
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Annika Graaf-Rau
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Sandra Diederich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02218, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02218, USA
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Matthias Dobbelstein
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany; Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
84
|
Cheng CY, Su YS, Chen CL, Chang M, Huang SW, Huang PN, Shih SR, Hsu YS, Chiu CH. Safety, tolerability, and efficacy of intranasally-administered detoxified LTh(αK) in mild-to-moderate COVID-19 patients: A randomized, double-blinded, placebo-controlled phase 2 study. Hum Vaccin Immunother 2024; 20:2432105. [PMID: 39611611 PMCID: PMC11610553 DOI: 10.1080/21645515.2024.2432105] [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: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
The objective of the study was to assess the safety, tolerability, and potential efficacy of intranasally administered AD17002, a detoxified form of Escherichia coli heat-labile enterotoxin, in treating individuals with mild-to-moderate coronavirus disease of 2019 (COVID-19). In this randomized, double-blinded, and placebo-controlled phase 2a study, a total of 30 adults aged 20-70 years with mild-to-moderate COVID-19 were recruited from three medical centers in Taiwan in 2022-2023. The trial comprised two cohorts, and participants were randomly assigned to receive intranasal administrations of either three doses of AD17002 immunomodulator or a placebo formulation buffer. Outcome analyses were conducted on the intention-to-treat set, and the safety set that included all randomized participants exposed to the AD17002. The proportion of cycle threshold (Ct) ≥30 and time to the recovery of key symptoms were assessed. An exploratory study was conducted to analyze the integrity of the viral genome after treatment. Administering 20 μg of AD17002 three times, either at 1-week or 1-day intervals, proved to be safe and well tolerated in subjects with mild-to-moderate COVID-19. AD17002 demonstrated a rapid and positive outcome in reducing the viral load in patients receiving the treatment. Impact of AD17002 treatment was further supported by the analysis of viral genome integrity following the treatment. The enhancement in clinical recovery by AD17002 within 5 days after symptom onset was observed but did not achieve statistical significance. According to the results, intranasal administration of AD17002 was safe, well-tolerated, and potentially effective for treating mild-to-moderate COVID-19.
Collapse
Affiliation(s)
- Chien-Yu Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- Institute of Public Health, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Shih Su
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan
| | - Chyi-Liang Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mingi Chang
- Advagene Biopharma Co., Ltd., Taipei, Taiwan
| | - Shu-Wei Huang
- Department of Orthopedics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Peng-Nien Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Shen Hsu
- Advagene Biopharma Co., Ltd., Taipei, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
85
|
Rusanganwa V, Nzabahimana I, Evander M. Quality and resilience of clinical laboratories in Rwanda: a need for sustainable strategies. Glob Health Action 2024; 17:2358633. [PMID: 38828509 PMCID: PMC11149573 DOI: 10.1080/16549716.2024.2358633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Quality healthcare is a global priority, reliant on robust health systems for evidence-based medicine. Clinical laboratories are the backbone of quality healthcare facilitating diagnostics, treatment, patient monitoring, and disease surveillance. Their effectiveness depends on sustainable delivery of accurate test results. Although the Strengthening Laboratory Management Towards Accreditation (SLMTA) programme has enhanced laboratory quality in low-income countries, the long-term sustainability of this improvement remains uncertain. OBJECTIVE To explore the sustainability of quality performance in clinical laboratories in Rwanda following the conclusion of SLMTA. METHODS A quasi-experimental design was adopted, involving 47 laboratories divided into three groups with distinct interventions. While one group received continuous mentorship and annual assessments (group two), interventions for the other groups (groups one and three) ceased following the conclusion of SLMTA. SLMTA experts collected data for 10 years through assessments using WHO's StepwiseLaboratory Quality Improvement Process Towards Accreditation (SLIPTA) checklist. Descriptive and t-test analyses were conducted for statistical evaluation. RESULTS Improvements in quality were noted between baseline and exit assessments across all laboratory groups (mean baseline: 35.3%, exit: 65.8%, p < 0.001). However, groups one and three experienced performance declines following SLMTA phase-out (mean group one: 64.6% in reference to 85.8%, p = 0.01; mean group three: 57.3% in reference to 64.7%, p < 0.001). In contrast, group two continued to enhance performance even years later (mean: 86.6%compared to 70.6%, p = 0.03). CONCLUSION A coordinated implementation of quality improvement plan that enables regular laboratory assessments to pinpoint and address the quality gaps is essential for sustaining quality services in clinical laboratories.
Collapse
Affiliation(s)
| | | | - Magnus Evander
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| |
Collapse
|
86
|
Hao S, Su J. Whispering gallery mode optical resonators for biological and chemical detection: current practices, future perspectives, and challenges. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2024; 88:016402. [PMID: 39626318 DOI: 10.1088/1361-6633/ad99e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
Sensors are important for a wide variety of applications include medical diagnostics and environmental monitoring. Due to their long photon confinement times, whispering gallery mode (WGM) sensors are among the most sensitive sensors currently in existence. We briefly discuss what are WGM sensors, the principles of WGM sensing, and the history of the field, beginning with Mie theory. We discuss recent work in the field on using these WGM resonators as sensors, focusing particularly on biological and chemical sensing applications. We discuss how sensorgrams are acquired and fundamental measurement limits. In addition, we discuss how to interpret binding curves and extract physical parameters such as binding affinity constants. We discuss the controversy surrounding single-molecule detection and discuss hybrid WGM nanoparticle sensors. In addition, we place these sensors in context with others sensing technologies both labeled and label-free. Finally, we discuss what we believe are the most promising applications for these devices, outline remaining challenges, and provide an outlook for the future.
Collapse
Affiliation(s)
- Shuang Hao
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, United States of America
| | - Judith Su
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, United States of America
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
87
|
Alsharksi AN, Sirekbasan S, Gürkök-Tan T, Mustapha A. From Tradition to Innovation: Diverse Molecular Techniques in the Fight Against Infectious Diseases. Diagnostics (Basel) 2024; 14:2876. [PMID: 39767237 PMCID: PMC11674978 DOI: 10.3390/diagnostics14242876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/15/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Infectious diseases impose a significant burden on global health systems due to high morbidity and mortality rates. According to the World Health Organization, millions die from infectious diseases annually, often due to delays in accurate diagnosis. Traditional diagnostic methods in clinical microbiology, primarily culture-based techniques, are time-consuming and may fail with hard-to-culture pathogens. Molecular biology advancements, notably the polymerase chain reaction (PCR), have revolutionized infectious disease diagnostics by allowing rapid and sensitive detection of pathogens' genetic material. PCR has become the gold standard for many infections, particularly highlighted during the COVID-19 pandemic. Following PCR, next-generation sequencing (NGS) has emerged, enabling comprehensive genomic analysis of pathogens, thus facilitating the detection of new strains and antibiotic resistance tracking. Innovative approaches like CRISPR technology are also enhancing diagnostic precision by identifying specific DNA/RNA sequences. However, the implementation of these methods faces challenges, particularly in low- and middle-income countries due to infrastructural and financial constraints. This review will explore the role of molecular diagnostic methods in infectious disease diagnosis, comparing their advantages and limitations, with a focus on PCR and NGS technologies and their future potential.
Collapse
Affiliation(s)
- Ahmed Nouri Alsharksi
- Department of Microbiology, Faculty of Medicine, Misurata University, Misrata 93FH+66F, Libya;
| | - Serhat Sirekbasan
- Department of Medical Laboratory Techniques, Şabanözü Vocational School, Çankırı Karatekin University, Çankırı 18650, Turkey
| | - Tuğba Gürkök-Tan
- Department of Field Crops, Food and Agriculture Vocational School, Çankırı Karatekin University, Çankırı 18100, Turkey;
| | - Adam Mustapha
- Department of Microbiology, Faculty of Life Sciences, University of Maiduguri, Maiduguri 600104, Nigeria;
| |
Collapse
|
88
|
Wang XS, Wang JY, Yu F, Shi D, Xie JJ, Li LJ, Wang BH. Microbiota-related metabolites correlated with the severity of COVID-19 patients. Hepatobiliary Pancreat Dis Int 2024:S1499-3872(24)00168-1. [PMID: 39734160 DOI: 10.1016/j.hbpd.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a global pandemic with high mortality, and the treatment options for the severe patients remain limited. Previous studies reported the altered gut microbiota in severe COVID-19. But there are no comprehensive data on the role of microbial metabolites in COVID-19 patients. METHODS We identified 153 serum microbial metabolites and assessed the changes in 72 COVID-19 patients upon admission and one-month after their discharge, comparing these changes to those in 133 healthy control individuals from the outpatient department during the same period. RESULTS Our study revealed that microbial metabolites varied across different stages and severity of COVID-19 patients. These altered microbial metabolites included tryptophan, bile acids, fatty acids, amino acids, vitamins and those containing benzene. A total of 13 distinct microbial metabolites were identified in COVID-19 patients compared to healthy controls. Notably, correlations were found among these disrupted metabolites and organ injury and inflammatory responses related to COVID-19. Furthermore, these metabolites did not restore to the normal levels one month after discharge. Importantly, two microbial metabolites were the core microbial metabolites related to the severity of COVID-19 patients. CONCLUSIONS The microbial metabolites were altered in the acute and recovery stage, correlating with disease severity of COVID-19. These results indicated the important role of gut microbiota in the progression of COVID-19, and facilitated the potential therapeutic microbial target for severe COVID-19 patients.
Collapse
Affiliation(s)
- Xiao-Sen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing-Yu Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Fei Yu
- Department of Laboratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiao-Jiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lan-Juan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Jinan Microecological Biomedicine Shandong Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou 310000, China
| | - Bao-Hong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Jinan Microecological Biomedicine Shandong Laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou 310000, China.
| |
Collapse
|
89
|
van Wyk S, Moir M, Banerjee A, Bazykin GA, Biswas NK, Sitharam N, Das S, Ma W, Maitra A, Mazumder A, Karim WA, Lamarca AP, Li M, Nabieva E, Tegally H, San JE, Vasconcelos ATR, Xavier JS, Wilkinson E, de Oliveira T. "The COVID-19 pandemic in BRICS: Milestones, interventions, and molecular epidemiology". PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003023. [PMID: 39705269 DOI: 10.1371/journal.pgph.0003023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/02/2024] [Indexed: 12/22/2024]
Abstract
Brazil, Russia, India, China, and South Africa (BRICS) are a group of developing countries with shared economic, healthcare, and scientific interests. These countries navigate multiple syndemics, and the COVID-19 pandemic placed severe strain on already burdened BRICS' healthcare systems, hampering effective pandemic interventions. Genomic surveillance and molecular epidemiology remain indispensable tools for facilitating informed pandemic intervention. To evaluate the combined manner in which the pandemic unfolded in BRICS countries, we reviewed the BRICS pandemic epidemiological and genomic milestones, which included the first reported cases and deaths, and pharmaceutical and non-pharmaceutical interventions implemented in these countries. To assess the development of genomic surveillance capacity and efficiency over the pandemic, we analyzed the turnaround time from sample collection to data availability and the technologies used for genomic analysis. This data provided information on the laboratory capacities that enable the detection of emerging SARS-CoV-2 variants and highlight their potential for monitoring other pathogens in ongoing public health efforts. Our analyses indicated that BRICS suffered >105.6M COVID-19 infections, resulting in >1.7M deaths. BRICS countries detected intricate genetic combinations of SARS-CoV-2 variants that fueled country-specific pandemic waves. BRICS' genomic surveillance programs enabled the identification and characterization of the majority of globally circulating Variants of Concern (VOCs) and their descending lineages. Pandemic intervention strategies first implemented by BRICS countries included non-pharmaceutical interventions during the onset of the pandemic, such as nationwide lockdowns, quarantine procedures, the establishment of fever clinics, and mask mandates- which were emulated internationally. Vaccination rollout strategies complemented this, some representing the first of their kind. Improvements in BRICS sequencing and data generation turnaround time facilitated quicker detection of circulating and emerging variants, supported by investments in sequencing and bioinformatic infrastructure. Intra-BRICS cooperation contributed to the ongoing intervention in COVID-19 and other pandemics, enhancing collective capabilities in addressing these health challenges. The data generated continues to inform BRICS-centric pandemic intervention strategies and influences global health matters. The increased laboratory and bioinformatic capacity post-COVID-19 will support the detection of emerging pathogens.
Collapse
Affiliation(s)
- Stephanie van Wyk
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Monika Moir
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Anindita Banerjee
- BRICS-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Georgii A Bazykin
- A.A. Kharkevich Institute for Information Transmission Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Nidhan K Biswas
- BRICS-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Nikita Sitharam
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Saumitra Das
- BRICS-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
- Indian Institute of Science, Bengaluru, Karnataka, India
| | - Wentai Ma
- Beijing Institute of Genomics, CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences / China National Centre for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Arindam Maitra
- BRICS-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Anup Mazumder
- BRICS-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Wasim Abdool Karim
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Alessandra Pavan Lamarca
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Mingkun Li
- Beijing Institute of Genomics, CAS Key Laboratory of Genomic and Precision Medicine, Chinese Academy of Sciences / China National Centre for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Elena Nabieva
- A.A. Kharkevich Institute for Information Transmission Problems of the Russian Academy of Sciences, Moscow, Russia
- Princeton University, Princeton, New Jersey, United States of America
| | - Houriiyah Tegally
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - James Emmanuel San
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Ana Tereza R Vasconcelos
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Joicymara S Xavier
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- Institute of Agricultural Sciences, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Unaí, Brasil
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Eduan Wilkinson
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
90
|
Pinto PBA, Timis J, Chuensirikulchai K, Li QH, Lu HH, Maule E, Nguyen M, Alves RPDS, Verma SK, Ana-Sosa-Batiz F, Valentine K, Landeras-Bueno S, Kim K, Hastie K, Saphire EO, Alves A, Elong Ngono A, Shresta S. Co-immunization with spike and nucleocapsid based DNA vaccines for long-term protective immunity against SARS-CoV-2 Omicron. NPJ Vaccines 2024; 9:252. [PMID: 39702529 PMCID: PMC11659323 DOI: 10.1038/s41541-024-01043-3] [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: 03/06/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
The continuing evolution of SARS-CoV-2 variants challenges the durability of existing spike (S)-based COVID-19 vaccines. We hypothesized that vaccines composed of both S and nucleocapsid (N) antigens would increase the durability of protection by strengthening and broadening cellular immunity compared with S-based vaccines. To test this, we examined the immunogenicity and efficacy of wild-type SARS-CoV-2 S- and N-based DNA vaccines administered individually or together to K18-hACE2 mice. S, N, and S + N vaccines all elicited polyfunctional CD4+ and CD8+ T cell responses and provided short-term cross-protection against Beta and Omicron BA.2 variants, but only co-immunization with S + N vaccines provided long-term protection against Omicron BA.2. Depletion of CD4+ and CD8+ T cells reduced the long-term efficacy, demonstrating a crucial role for T cells in the durability of protection. These findings underscore the potential to enhance long-lived protection against SARS-CoV-2 variants by combining S and N antigens in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Paolla Beatriz Almeida Pinto
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kantinan Chuensirikulchai
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Hsueh Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Michael Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | | | | | | | - Kristen Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- University Cardenal Herrera-CEU, CEU Universities, Valencia, 46113, Spain
| | - Kenneth Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, 92093, USA
| | - Ada Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, 92093, USA.
| |
Collapse
|
91
|
Lu Z, Wang X, Chen J. AI-empowered visualization of nucleic acid testing. Life Sci 2024; 359:123209. [PMID: 39488264 DOI: 10.1016/j.lfs.2024.123209] [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: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
AIMS The visualization of nucleic acid testing (NAT) results plays a critical role in diagnosing and monitoring infectious and genetic diseases. The review aims to review the current status of AI-based NAT result visualization. It systematically introduces commonly used AI-based methods and techniques for NAT, emphasizing the importance of result visualization for accessible, clear, and rapid interpretation. This highlights the importance of developing a NAT visualization platform that is user-friendly and efficient, setting a clear direction for future advancements in making nucleic acid testing more accessible and effective for everyday applications. METHOD This review explores both the commonly used NAT methods and AI-based techniques for NAT result visualization. The focus then shifts to AI-based methodologies, such as color detection and result interpretation through AI algorithms. The article presents the advantages and disadvantages of these techniques, while also comparing the performance of various NAT platforms in different experimental contexts. Furthermore, it explores the role of AI in enhancing the accuracy, speed, and user accessibility of NAT results, highlighting visualization technologies adapted from other fields of experimentation. SIGNIFICANCE This review offers valuable insights for researchers and everyday users, aiming to develop effective visualization platforms for NAT, ultimately enhancing disease diagnosis and monitoring.
Collapse
Affiliation(s)
- Zehua Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China
| | - Xiaogang Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China.
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China.
| |
Collapse
|
92
|
Hurst JR, Naghibosadat M, Budowski P, Liu J, Samaan P, Budiman F, Kurtesi A, Qi F, Menon H, Krishnan R, Abioye J, Gingras AC, Ostrowski M, Orozco NM, Kozak RA. Comparison of a SARS-CoV-2 mRNA booster immunization containing additional antigens to a spike-based mRNA vaccine against Omicron BA.5 infection in hACE2 mice. PLoS One 2024; 19:e0314061. [PMID: 39625929 PMCID: PMC11614295 DOI: 10.1371/journal.pone.0314061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
The emergence of SARS-CoV-2 variants presents challenges to vaccine effectiveness, underlining the necessity for next-generation vaccines with multiple antigens beyond the spike protein. Here, we investigated a multiantigenic booster containing spike and a chimeric construct composed of nucleoprotein (N) and membrane (M) proteins, comparing its efficacy to a spike-only booster against Omicron BA.5 in K18-hACE2 mice. Initially, mice were primed and boosted with Beta (B.1.351) spike-only mRNA, showing strong spike-specific T cell responses and neutralizing antibodies, albeit with limited cross-neutralization to Omicron variants. Subsequently, a spike-NM multiantigenic vaccine was then examined as a second booster dose for protection in hACE2-transgenic mice. Mice receiving either homologous spike-only or heterologous spike-NM booster had nearly complete inhibition of infectious virus shedding in oral swabs and reduced viral burdens in both lung and nasal tissues following BA.5 challenge. Examination of lung pathology further revealed that both spike-only and spike-NM boosters provided comparable protection against inflammatory infiltrates and fibrosis. Moreover, the spike-NM booster demonstrated neutralization efficacy in a pseudovirus assay against Wuhan-Hu-1, Beta, and Omicron variants akin to the spike-only booster. These findings indicate that supplementing spike with additional SARS-CoV-2 targets in a booster immunization confers equivalent immunity and protection against Omicron BA.5. This work highlights a promising strategy for individuals previously vaccinated with spike-only vaccines, potentially offering enhanced protection against emerging coronaviruses.
Collapse
Affiliation(s)
- Jacklyn R. Hurst
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maedeh Naghibosadat
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Patrick Budowski
- Institute of Medical Sciences, University of Toronto, Ontario, Canada
| | - Jun Liu
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Frans Budiman
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Fredo Qi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Haritha Menon
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | | | - Jumai Abioye
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mario Ostrowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Robert A. Kozak
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
- Division of Microbiology, Sunnybrook Health Sciences Centre, Department of Laboratory Medicine and Molecular Diagnostics, Toronto, ON, Canada
| |
Collapse
|
93
|
Ling F, Xu Z, Sun J, Wang X, Feng Y, Liu Y, Chen Y, Wang J, Chen Z, Chen K. SARS-CoV-2 seroprevalence and antibody trajectories after easing of COVID-19 restrictions: a longitudinal study in China. Front Public Health 2024; 12:1420993. [PMID: 39691651 PMCID: PMC11650369 DOI: 10.3389/fpubh.2024.1420993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/29/2024] [Indexed: 12/19/2024] Open
Abstract
Background We aimed to evaluate the seroprevalence of SARS-CoV-2 and investigate the trajectories of protective immunity and associated risk factors in eastern China between March and November 2023 after the easing of COVID-19 restrictions. Materials and methods We conducted repeated population-based seroepidemiologic studies using a multistage, population-stratified, cluster random sampling method. We measured neutralizing antibodies (nAbs) using a fluorescence immunoassay. We calculated both overall and stratified seroprevalence. The latent class growth mixed model (LCGMM) was used to analyze the dynamic trajectories of antibodies, and a multinomial logistic regression model was used to identify factors associated with different antibody trajectory patterns. Results A total of 6,147 participants were included at baseline, with a median age of 53.61 years. Both observed and adjusted seroprevalence remained high and stable throughout the study period. The LCGMM identified four distinct antibody trajectories: 75.22% of participants had a high and stable antibody trajectory, while nearly 8% of them exhibited an increase, decline, or low-stable antibody trajectory. Younger participants, women, those fully vaccinated, and individuals with a history of previous infection were more likely to have high and stable antibody trajectories. Conclusion The majority of the population maintained sustained protective immunity after the outbreak, following the easing of COVID-19 restrictions across the country.
Collapse
Affiliation(s)
- Feng Ling
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zenghao Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jimin Sun
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Xiaoxiao Wang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yan Feng
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ying Liu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yijuan Chen
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jinna Wang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhiping Chen
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Kun Chen
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
94
|
Lucero Arteaga F, Nabaes Jodar M, Mondino M, Portu A, Boeris M, Jolly A, Jar A, Mundo S, Castro E, Alvarez D, Torres C, Viegas M, Bratanich A. An Outbreak of SARS-CoV-2 in Captive Armadillos Associated with Gamma Variant in Argentina. ECOHEALTH 2024; 21:183-194. [PMID: 38844740 DOI: 10.1007/s10393-024-01686-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 12/17/2024]
Abstract
The current pandemic produced by SARS-CoV-2 and its variants represent an example of the one health concept in which humans and animals are components of the same epidemiologic chain. Animal reservoirs of these viruses are thus the focus of surveillance programs, to monitor their circulation and evolution in potentially new hosts and reservoirs. In this work, we report the detection of the SARS-CoV-2 Gamma variant infection in four specimens of Chaetophractus villosus (big hairy armadillo/armadillo peludo) in Argentina. In addition to the finding of a new wildlife species susceptible to SARS-CoV-2 infection, the identification of the Gamma variant three months after its last detection in humans in Argentina is a noteworthy result, which can be due to alternative non-exclusive scenarios, such as unidentified viral reservoirs, unrecognized circulation in humans or species-specific variation in incubation periods.
Collapse
Affiliation(s)
- Franco Lucero Arteaga
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- CIDEF (Centro de Investigación y Desarrollo de Fármacos) y Bioterio, Facultad de Ciencias Veterinarias UNLPam, La Pampa, Argentina
| | - Mercedes Nabaes Jodar
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- Laboratorio de Virología Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD CABA, Buenos Aires, Argentina
| | - Mariela Mondino
- CIDEF (Centro de Investigación y Desarrollo de Fármacos) y Bioterio, Facultad de Ciencias Veterinarias UNLPam, La Pampa, Argentina
| | - Ana Portu
- CIDEF (Centro de Investigación y Desarrollo de Fármacos) y Bioterio, Facultad de Ciencias Veterinarias UNLPam, La Pampa, Argentina
| | - Mónica Boeris
- CIDEF (Centro de Investigación y Desarrollo de Fármacos) y Bioterio, Facultad de Ciencias Veterinarias UNLPam, La Pampa, Argentina
| | - Ana Jolly
- Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, Buenos Aires, Argentina
| | - Ana Jar
- Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, Buenos Aires, Argentina
| | - Silvia Mundo
- Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, INPA Conicet, Buenos Aires, Argentina
| | - Eliana Castro
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas, Universidad de San Martin (UNSAM), Buenos Aires, Argentina
| | - Diego Alvarez
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas, Universidad de San Martin (UNSAM), Buenos Aires, Argentina
| | - Carolina Torres
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
| | - Mariana Viegas
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina.
- Laboratorio de Virología Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD CABA, Buenos Aires, Argentina.
| | - Ana Bratanich
- Universidad de Buenos Aires, Facultad de Ciencias Veterinarias, INPA Conicet, Buenos Aires, Argentina.
| |
Collapse
|
95
|
Chen C, Wang Y, Kaur G, Adiga A, Espinoza B, Venkatramanan S, Warren A, Lewis B, Crow J, Singh R, Lorentz A, Toney D, Marathe M. Wastewater-based epidemiology for COVID-19 surveillance and beyond: A survey. Epidemics 2024; 49:100793. [PMID: 39357172 DOI: 10.1016/j.epidem.2024.100793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Abstract
The pandemic of COVID-19 has imposed tremendous pressure on public health systems and social economic ecosystems over the past years. To alleviate its social impact, it is important to proactively track the prevalence of COVID-19 within communities. The traditional way to estimate the disease prevalence is to estimate from reported clinical test data or surveys. However, the coverage of clinical tests is often limited and the tests can be labor-intensive, requires reliable and timely results, and consistent diagnostic and reporting criteria. Recent studies revealed that patients who are diagnosed with COVID-19 often undergo fecal shedding of SARS-CoV-2 virus into wastewater, which makes wastewater-based epidemiology for COVID-19 surveillance a promising approach to complement traditional clinical testing. In this paper, we survey the existing literature regarding wastewater-based epidemiology for COVID-19 surveillance and summarize the current advances in the area. Specifically, we have covered the key aspects of wastewater sampling, sample testing, and presented a comprehensive and organized summary of wastewater data analytical methods. Finally, we provide the open challenges on current wastewater-based COVID-19 surveillance studies, aiming to encourage new ideas to advance the development of effective wastewater-based surveillance systems for general infectious diseases.
Collapse
Affiliation(s)
- Chen Chen
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States.
| | - Yunfan Wang
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States.
| | - Gursharn Kaur
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Aniruddha Adiga
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Baltazar Espinoza
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Srinivasan Venkatramanan
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Andrew Warren
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Bryan Lewis
- Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| | - Justin Crow
- Virginia Department of Health, Richmond, 23219, United States.
| | - Rekha Singh
- Virginia Department of Health, Richmond, 23219, United States.
| | - Alexandra Lorentz
- Division of Consolidated Laboratory Services, Department of General Services, Richmond, 23219, United States.
| | - Denise Toney
- Division of Consolidated Laboratory Services, Department of General Services, Richmond, 23219, United States.
| | - Madhav Marathe
- Department of Computer Science, University of Virginia, Charlottesville, 22904, United States; Biocomplexity Institute and Initiative, University of Virginia, Charlottesville, 22904, United States.
| |
Collapse
|
96
|
Tebibi K, Ben Laamari R, Saied Z, Maghrebi O, Touzi H, Meddeb Z, Ben Sassi S, Triki H, Belghith M, Rezig D. Profile of Cytokines and T Cell Subsets Transcription Factors in Cerebrospinal Fluid of Patients with Viral Encephalitis. Viral Immunol 2024; 37:459-469. [PMID: 39527011 DOI: 10.1089/vim.2024.0058] [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: 11/16/2024] Open
Abstract
This study investigates the demographic, clinical characteristics, virological profiles, and immunological responses of patients with viral encephalitis (VE) compared with a control group. The VE group displayed a wide range of neurological symptoms. Virological analysis revealed the predominance of Herpesviridae family viruses. Immune responses in cerebrospinal fluid (CSF) from patients with VE were examined, highlighting an immunological shift toward T helper 1 (Th1) cells dominance, altered T helper 17 cells/regulatory T cells (Th17/Tregs) balance, and high interleukin-6 expression. These findings provide insights into the complex immunological landscape of VE, highlighting the role of specific cytokines and T cell subsets in its pathogenesis and potentially guiding targeted therapeutic strategies.
Collapse
Affiliation(s)
- Khadija Tebibi
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rafika Ben Laamari
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Zakaria Saied
- Neurological Department of Mongi Ben Hmida Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Olfa Maghrebi
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Touzi
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Zina Meddeb
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Samia Ben Sassi
- Neurological Department of Mongi Ben Hmida Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Triki
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Meriam Belghith
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Dorra Rezig
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| |
Collapse
|
97
|
Goll A, Krupińska M, Nowicka J, Baranowicz K, Rabalski L, Lass A, Gorska A, Sironen T, Kant R, Grzybek M. Wild red foxes ( Vulpes vulpes) do not participate in SARS-CoV-2 circulation in Poland. One Health 2024; 19:100845. [PMID: 39071484 PMCID: PMC11278590 DOI: 10.1016/j.onehlt.2024.100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/10/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Biomonitoring is an essential activity for identifying possible vectors and reservoirs of pathogens and predicting potential outbreaks. Wild red foxes are present in both sylvatic and synanthropic environments, making them potential carriers of zoonotic pathogens. Experimental studies have shown that both coyotes and red foxes can transmit SARS-CoV-2. This study aimed to assess the prevalence and seroprevalence of SARS-CoV-2 in wild red foxes hunted in northern Poland. METHODS Oral swabs, blood clots or heat tissue samples were collected from 292 red foxes hunted in northern Poland. We used both molecular (RT-PCR) and serological (IFA) approaches to detect SARS-CoV-2 infections in the sampled animals. RESULTS We did not find any evidence of SARS-CoV-2 infection in the collected samples, using both molecular and serological methods. CONCLUSIONS Despite foxes having frequent contact with humans, human waste, and other animals, they do not appear to participate in the circulation of the SARS-CoV-2 virus in our geographical region. Nevertheless, we believe that continuous biomonitoring should be performed to assess the SARS-CoV-2 epidemiological situation in the wild.
Collapse
Affiliation(s)
- Aleksander Goll
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Martyna Krupińska
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Joanna Nowicka
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Karolina Baranowicz
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Lukasz Rabalski
- Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Gdansk, Poland
- Biological Threats Identification and Countermeasure Center, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Pulawy, Poland
| | - Anna Lass
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| | - Aleksandra Gorska
- Faculty of Medical Sciences, Medical University in Lublin, Lublin, Poland
| | - Tarja Sironen
- Department of Virology, Helsinki University, Helsinki, Finland
| | - Ravi Kant
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
- Department of Virology, Helsinki University, Helsinki, Finland
| | - Maciej Grzybek
- Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdynia, Poland
| |
Collapse
|
98
|
Spiess K, Petrillo M, Paracchini V, Leoni G, Lassaunière R, Polacek C, Marving EL, Larsen NB, Gunalan V, Ring A, Bull M, Buttinger G, Veneri C, Suffredini E, La Rosa G, Corbisier P, Querci M, Rasmussen M, Marchini A. Development of new RT-PCR assays for the specific detection of BA.2.86 SARS-CoV-2 and its descendent sublineages. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176365. [PMID: 39299334 DOI: 10.1016/j.scitotenv.2024.176365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
The SARS-CoV-2 BA.2.86 variant, also known as Pirola, has acquired over 30 amino acid changes in the Spike protein, evolving into >150 sublineages within ten months of its emergence. Among these, the JN.1, has been rapidly increasing globally becoming the most prevalent variant. To facilitate the identification of BA.2.86 sublineages, we designed the PiroMet-1 and PiroMet-2 assays in silico and validated them using BA.2.86 viral RNA and clinical samples to ascertain analytical specificity and sensitivity. Both assays resulted very specific with limit of detection of about 1-2 RNA copies/μL. The assays were then applied in a digital RT-PCR format to wastewater samples, combined with the OmMet assay (which identifies Omicron sublineages except BA.2.86 and its descendants) and the JRC-UCE.2 assay (which can universally recognize all SARS-CoV-2 variants). When used together with the OmMet and JRC-CoV-UCE.2 assays, the PiroMet assays accurately quantified BA.2.86 sublineages in wastewater samples. Our findings support the integration of these assays into routine SARS-CoV-2 wastewater surveillance as a timely and cost-effective complement to sequencing for monitoring the prevalence and spread of BA.2.86 sublineages within communities.
Collapse
Affiliation(s)
- Katja Spiess
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | | | | | - Gabriele Leoni
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Ria Lassaunière
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Charlotta Polacek
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Ellinor Lindberg Marving
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Nicolai Balle Larsen
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Vithiagaran Gunalan
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Aleksander Ring
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Maireid Bull
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | | | - Carolina Veneri
- National Center for Water Safety (CeNSiA), Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta Suffredini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppina La Rosa
- National Center for Water Safety (CeNSiA), Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Morten Rasmussen
- Virus Research & Development, Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut (SSI), Denmark
| | - Antonio Marchini
- European Commission, Joint Research Centre (JRC), Geel, Belgium.
| |
Collapse
|
99
|
Li J, Cheng R, Bian Z, Niu J, Xia J, Mao G, Liu H, Wu C, Hao C. Development of multiplex allele-specific RT-qPCR assays for differentiation of SARS-CoV-2 Omicron subvariants. Appl Microbiol Biotechnol 2024; 108:35. [PMID: 38183475 DOI: 10.1007/s00253-023-12941-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 01/08/2024]
Abstract
Quick differentiation of current circulating variants and the emerging recombinant variants of SARS-CoV-2 is essential to monitor their transmissions. However, the widely applied gene sequencing method is time-consuming and costly especially when facing recombinant variants, because a large part or whole genome sequencing is required. Allele-specific reverse transcriptase real time RT-PCR (RT-qPCR) represents a quick and cost-effective method for SNP (single nucleotide polymorphism) genotyping and has been successfully applied for SARS-CoV-2 variant screening. In the present study, we developed a panel of 5 multiplex allele-specific RT-qPCR assays targeting 20 key mutations for quick differentiation of the Omicron subvariants (BA.1 to BA.5 and their descendants) and the recombinant variants (XBB.1 and XBB.1.5). Two parallel multiplex RT-qPCR reactions were designed to separately target the prototype allele and the mutated allele of each mutation in the allele-specific RT-qPCR assay. Optimal annealing temperatures, primer and probe dosage, and time for annealing/extension for each reaction were determined by multi-factor and multi-level orthogonal test. The variation of Cp (crossing point) values (ΔCp) between the two multiplex RT-qPCR reactions was applied to determine if a mutation occurs or not. SARS-CoV-2 subvariants and related recombinant variants were differentiated by their unique mutation patterns. The developed multiplex allele-specific RT-qPCR assays exhibited excellent analytical sensitivities (with limits of detection (LoDs) of 1.47-18.52 copies per reaction), wide linear detection ranges (109-100 copies per reaction), good amplification efficiencies (88.25 to 110.68%), excellent reproducibility (coefficient of variations (CVs) < 5% in both intra-assay and inter-assay tests), and good clinical performances (99.5-100% consistencies with Sanger sequencing). The developed multiplex allele-specific RT-qPCR assays in the present study provide an alternative tool for quick differentiation of the SARS-CoV-2 Omicron subvariants and their recombinant variants. KEY POINTS: • A panel of five multiplex allele-specific RT-qPCR assays for quick differentiation of 11 SARS-CoV-2 Omicron subvariants (BA.1, BA.2, BA.4, BA.5, and their descendants) and 2 recombinant variants (XBB.1 and XBB.1.5). • The developed assays exhibited good analytical sensitivities and reproducibility, wide linear detection ranges, and good clinical performances, providing an alternative tool for quick differentiation of the SARS-CoV-2 Omicron subvariants and their recombinant variants.
Collapse
Affiliation(s)
- Jianguo Li
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China.
| | - Ruiling Cheng
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Zixin Bian
- College of Life Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Jiahui Niu
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Juan Xia
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Guoli Mao
- Shanxi Guoxin Caregeno Biotechnology Co., Ltd., Taiyuan, 030032, People's Republic of China
| | - Hulong Liu
- Shanxi Guoxin Caregeno Biotechnology Co., Ltd., Taiyuan, 030032, People's Republic of China
| | - Changxin Wu
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, People's Republic of China
| | - Chunyan Hao
- School of Environment and Resources, Taiyuan University of Science and Technology, Taiyuan, 030024, People's Republic of China.
| |
Collapse
|
100
|
Yaugel-Novoa M, Noailly B, Jospin F, Pizzorno A, Traversier A, Pozzetto B, Waeckel L, Longet S, Pillet S, Botelho-Nevers E, Rosa-Calatrava M, Bourlet T, Paul S. Impaired mucosal IgA response in patients with severe COVID-19. Emerg Microbes Infect 2024; 13:2401940. [PMID: 39358866 PMCID: PMC11451292 DOI: 10.1080/22221751.2024.2401940] [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: 06/12/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Several studies have investigated the antibody response to SARS-CoV-2, focusing particularly on the systemic humoral immune response and the production of immunoglobulin G (IgG) antibodies. IgA antibodies play a crucial role in protecting against respiratory viral infections but have also been associated with the pathophysiology of COVID-19. We performed a prospective study of 169 COVID-19 patients - 50 with critical/severe (ICU), 47 with moderate (Non-ICU), and 72 with asymptomatic COVID-19 - to explore the humoral immune response to SARS-CoV-2 infection. We found that the early systemic IgA response strongly induced in patients with severe disease did not block IgG neutralization functions and activated FcRs more effectively than IgG. However, even if SIgA levels were high, mucosal IgA antibodies could not control the infection effectively in patients with severe disease. Our findings highlight the complexity of the immune response to SARS-CoV-2 exhibiting high systemic levels of IgA with strong neutralizing capacity in severe cases, together with higher levels of IgA-FcR activation than in asymptomatic patients. They also suggest the need for further research to fully understand the role of IgA and its structural alterations in mucosal tissues in cases of severe disease and the impact of these antibodies on disease progression.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Blandine Noailly
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Fabienne Jospin
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Andrés Pizzorno
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aurélien Traversier
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Bruno Pozzetto
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Louis Waeckel
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphanie Longet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Sylvie Pillet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Diseases Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Manuel Rosa-Calatrava
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Thomas Bourlet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Lead contact
| |
Collapse
|