1
|
Goel V, Ding J, Hatuwal B, Giri E, Deliberto TJ, Lowe J, Webby R, Emch M, Wan XF. Ecological drivers of evolution of swine influenza in the United States: a review. Emerg Microbes Infect 2025; 14:2455598. [PMID: 39817666 PMCID: PMC11780704 DOI: 10.1080/22221751.2025.2455598] [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] [Indexed: 01/18/2025]
Abstract
Influenza A viruses (IAVs) pose a major public health threat due to their wide host range and pandemic potential. Pigs have been proposed as "mixing vessels" for avian, swine, and human IAVs, significantly contributing to influenza ecology. In the United States, IAVs are enzootic in commercial swine farming operations, with numerous genetic and antigenic IAV variants having emerged in the past two decades. However, the dynamics of intensive swine farming systems and their interactions with ecological factors influencing IAV evolution have not been systematically analysed. This review examines the evolution of swine IAVs in commercial farms, highlighting the role of multilevel ecological factors. A total of 61 articles published after 2000 were reviewed, with most studies conducted after 2009 in Midwestern US, followed by Southeast and South-central US. The findings reveal that ecological factors at multiple spatial scales, such as regional transportation networks, interconnectedness of swine operations, farm environments, and presence of high-density, low-genetic diversity herds, can facilitate virus transmission and enhance virus evolution. Additionally, interactions at various interfaces, such as between commercial swine and feral swine, humans, or wild birds contribute to the increase in genetic diversity of swine IAVs. The review underscores the need for comprehensive studies and improved data collection to better understand the ecological dynamics influencing swine IAV evolution. This understanding is crucial for mitigating disease burden in swine production and reducing the risk of zoonotic influenza outbreaks.
Collapse
Affiliation(s)
- Varun Goel
- Department of Geography, University of South Carolina, Columbia, SC, USA
| | - Jessica Ding
- Department of Geography and Environment, University of North Carolina, Chapel Hill, NC, USA
| | - Bijaya Hatuwal
- NexGen Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA
- Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, MO, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Emily Giri
- NexGen Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Thomas J. Deliberto
- US Department of Agriculture Animal and Plant Health Inspection Service, Fort Collins, CO, USA
| | - James Lowe
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Richard Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael Emch
- Department of Geography and Environment, University of North Carolina, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Xiu-Feng Wan
- NexGen Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, USA
- Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, MO, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
2
|
Yao X, Lu WH, Qiao WT, Zhang YQ, Zhang BY, Li HX, Li JL. The highly pathogenic strain of porcine deltacoronavirus disrupts the intestinal barrier and causes diarrhea in newborn piglets. Virulence 2025; 16:2446742. [PMID: 39758030 DOI: 10.1080/21505594.2024.2446742] [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: 03/11/2024] [Revised: 08/30/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is increasingly prevalent in newborn piglets with diarrhea. With the development of research on the virus and the feasibility of PDCoV cross-species transmission, the biosafety and the development of pig industry have been greatly affected. In this study, a PDCoV strain CH/LNFX/2022 was isolated from diarrheal newborn piglets at a farm in China. A genome-wide based phylogenetic analysis suggests that 97.5% to 99.2% homology existed in the whole genomes of other strains. Five amino acid mutations are seen for the first time in the S protein. By constructing 3D models, it was found that the S1-NTD/CTD and S2-HR-C regions produced structural alterations. Protein functional analysis showed that the structural changes of the three regions changed the epitope of S protein, the O-GalNAc glycosylation site and the 3C-like protease cleavage site. In addition, oral administration of 107 TCID50 CH/LNFX/2022 to newborn piglets successfully reproduced obvious clinical signs of piglets, such as diarrhea and dehydration. Meanwhile, PDCoV antigen was detected by immunofluorescence in the small intestine, and microscopic lesions and intestinal mucosal barrier destruction were detected by histological observation and scanning electron microscopy. Our study confirmed that porcine coronavirus strains increased pathogenicity through evolution, damaged the intestinal barrier of newborn piglets, and caused diarrhea in pigs. This study provided the candidate strains and theoretical basis for establishing the prevention and control system of vaccine and diagnostic methods for piglet diarrhea.
Collapse
Affiliation(s)
- Xin Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Wei-Hong Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Wen-Ting Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Yu-Qian Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Bao-Ying Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
| | - Hui-Xin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, PR China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, PR China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, PR China
| |
Collapse
|
3
|
Yan M, Su A, Meyer D, Sosa GR, Fritsch H, Pitters M, Fischer N, Herrler G, Becher P. Precursor of H-type II histo-blood group antigen and subterminal sialic acids on gangliosides are significantly implicated in cell entry and infection by a porcine P[11] rotavirus. Emerg Microbes Infect 2025; 14:2447608. [PMID: 39726161 PMCID: PMC11727068 DOI: 10.1080/22221751.2024.2447608] [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: 08/01/2024] [Revised: 10/21/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Rotaviruses, non-enveloped viruses with a double-stranded RNA genome, are the leading etiological pathogen of acute gastroenteritis in young children and animals. The P[11] genotype of rotaviruses exhibits a tropism for neonates. In the present study, a binding assay using synthetic oligosaccharides demonstrated that the VP8* protein of P[11] porcine rotavirus (PRV) strain 4555 binds to lacto-N-neotetraose (LNnT) with the sequence Galβ1,4-GlcNAcβ1,3-Galβ1,4-Glc, one of the core parts of histo-blood group antigen (HBGA) and milk glycans. However, infections were significantly inhibited by blocking of endogenous monosialoganglioside (GM) GM1a with cholera toxin B subunit and preincubation of the virus with exogenous GM1a, suggesting that GM1a is involved in the infection of P[11] PRV 4555. In addition to GM1a, preincubation of the virus with exogenous disialogangliosides (GD) GD1a, GD1b, and trisialoganglioside (GT) GT1b also prevented infection. In contrast, exogenous ganglioside GM3 only inhibited infections at an early time point, and exogenous asyalosphingolipids GA1 and LacCer did not show any inhibitory effect on infections. This indicates that P[11] PRV 4555 preferentially utilizes gangliosides containing subterminal sialic acids. Further experiments revealed that P[11] PRV 4555 infections were prevented by preincubation of the virus with Neu5Ac and Neu5Gc. These results confirmed that sialic acids are essential for P[11] PRV 4555 cell entry, despite the classification as NA-resistant strain. Overall, our results proved that P[11] rotavirus not only binds to the Gal-GlcNAc motif but also utilizes gangliosides containing subterminal sialic acids.
Collapse
Affiliation(s)
- Miaomiao Yan
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ang Su
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Denise Meyer
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gleyder Roman Sosa
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Henrik Fritsch
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malte Pitters
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Paul Becher
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
4
|
Bardach A, Brizuela M, Berrueta M, Ciapponi A, Sambade JM, Ballivian J, Ortega V, Castellana N, Comandé D, Parker EPK, Kampmann B, Stegelmann K, Xiong X, Stergachis A, Munoz FM, Buekens P, Mazzoni A. Umbrella review of the safety of Chikungunya vaccine platforms used in other vaccines. Hum Vaccin Immunother 2025; 21:2463191. [PMID: 39932481 DOI: 10.1080/21645515.2025.2463191] [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: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/14/2025] Open
Abstract
Chikungunya virus (CHIKV), transmitted through Aedes mosquitoes, is a significant global health concern. Various vaccine platforms have been explored to combat CHIKV, including formalin inactivation, live-attenuated strains, virus-like particles (VLPs), viral vectors, and mRNA technologies. This umbrella review synthesizes evidence on the safety profiles of vaccine platforms used in Chikungunya vaccines that have been applied in other vaccines, focusing on adverse events of special interest (AESI) in pregnant persons, children, and adolescents. A comprehensive overview of systematic reviews (SRs) was conducted. Results: Seven systematic reviews were included and complemented with primary studies. Vaccines like influenza, human papillomavirus (HPV), and COVID-19, which share platforms with Chikungunya vaccines, showed no significant increase in AESI. Moderate-to high-quality SRs supported favorable safety profiles. Vaccines sharing platforms with Chikungunya vaccines generally exhibit acceptable safety profiles in pregnant persons, children, and adolescents.
Collapse
Affiliation(s)
- Ariel Bardach
- Center for Research in Epidemiology and Public Health (CIESP-IECS), CONICET, Buenos Aires, Argentina
| | - Martin Brizuela
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Mabel Berrueta
- Department of Mother and Child Health, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Agustín Ciapponi
- Argentine Cochrane Center, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Juan M Sambade
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Jamile Ballivian
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Vanesa Ortega
- Department of Mother and Child Health, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Noelia Castellana
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Daniel Comandé
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Edward P K Parker
- The Vaccine Centre, London School of Hygiene & Tropical Medicine, Keppel Street, London, UK
| | - Beate Kampmann
- Health Protection Research Unit in Immunisation, London School of Hygiene & Tropical Medicine, London, UK
- Charite Centre for Global Health, Charité, Universitätsmedizin, Vaccine Centre, Berlin, Germany
| | - Katharina Stegelmann
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| | - Xu Xiong
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Andy Stergachis
- Schools of Pharmacy and Public Health, University of Washington, Seattle, USA
| | - Flor M Munoz
- Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Pierre Buekens
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Agustina Mazzoni
- Department of Health Technology Assessment and Economic Evaluations, Institute for Clinical Effectiveness and Health Policy (IECS), Buenos Aires, Argentina
| |
Collapse
|
5
|
Arunsiripate TT, Groeltz-Thrush J, Saeng-Chuto K, Guo B, Michael A, Siepker C, Derscheid RJ, Rahe MC, Zhang J, Burrough E, Piñeyro PE. Diagnostic investigation of porcine hemagglutinating encephalomyelitis virus as potential pathogen associated with respiratory clinical signs and pulmonary lesions in pigs. Microb Pathog 2025; 203:107493. [PMID: 40120700 DOI: 10.1016/j.micpath.2025.107493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a member of the genus Betacoronavirus, known for its impact on the central and peripheral nervous systems in pigs. Traditionally associated with vomiting and wasting disease (VWD) and encephalomyelitis, PHEV was first reported in Canada in the late 1950s and has since been identified in numerous countries. Although serologic studies indicate global dissemination, the prevalence of PHEV remains unclear due to sporadic reporting and lack of active surveillance. Neonatal pigs are particularly vulnerable, with outbreaks resulting in high morbidity and mortality. Histopathological findings typically include non-suppurative encephalomyelitis and lymphoplasmacytic perivascular cuffs, gliosis, and neuronal degeneration. Recent observations have suggested a potential role for PHEV in respiratory disease, a hypothesis prompted by cases of influenza-like symptoms in pigs in Michigan in 2015 and corroborated by subsequent reports. This study aims to explore this possibility through a combination of clinical outbreak analysis and retrospective investigation. PHEV was confirmed via qPCR in 83.33 % of pigs examined for respiratory disease, with histological lesions such as necrotizing bronchitis and bronchiolitis. In-situ hybridization (ISH) confirmed the presence of PHEV mRNA in respiratory epithelium, and immunohistochemical analysis revealed significant macrophage infiltration in affected lung. Phylogenetic analysis indicated that PHEV strains from respiratory cases cluster closely with historical respiratory strains, though distinct from neurologic strains. This genetic differentiation suggests possible phenotypic variation contributing to respiratory tropism. The retrospective study identified PHEV in 7.62 % of cases with necrotizing bronchitis or bronchiolitis, reinforcing the virus's potential role in respiratory disease. Notably, PHEV co-infection with other respiratory pathogens such as PRRSV was observed, suggesting it may contribute to the porcine respiratory disease complex (PRDC). These findings suggest that PHEV is a significant respiratory pathogen in swine, warranting its inclusion in the differential diagnosis for respiratory disease in nursery pigs. Future research should focus on elucidating the pathogenesis of PHEV in respiratory disease, host-virus interactions, and the virus's impact on immune response and secondary infections. Understanding these factors will be crucial in developing effective preventive and therapeutic strategies against PHEV in swine.
Collapse
Affiliation(s)
- Trevor T Arunsiripate
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Jennifer Groeltz-Thrush
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Kepalee Saeng-Chuto
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Baoqing Guo
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Alyona Michael
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Christopher Siepker
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Rachel J Derscheid
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Michael C Rahe
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Population Health and Pathobiology, North Caroline State University, NC, USA
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Eric Burrough
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Pablo E Piñeyro
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA.
| |
Collapse
|
6
|
Tang J, Wang L, Fang W, Su CM, Kim J, Du Y, Yoo D. Coinfection with bacterial pathogens and genetic modification of PRRSV-2 for suppression of NF-κB and attenuation of proinflammatory responses. Virology 2025; 606:110484. [PMID: 40086205 DOI: 10.1016/j.virol.2025.110484] [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: 11/06/2024] [Revised: 02/17/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) infects pulmonary alveolar macrophages and induces inflammation in the respiratory system. In swine farms, coinfection with PRRSV and bacterial pathogens is common and can result in clinically complicated outcomes, including porcine respiratory disease complex. Coinfection can cause excessive expressions of proinflammatory mediators and may lead to cytokine-storm-like syndrome. An immunological hallmark of PRRSV-2 is the bidirectional regulation of NF-κB with the nucleocapsid (N) protein identified as the NF-κB activator. We generated an NF-κB-silencing mutant PRRSV-2 by mutating the N gene to block its binding to PIAS1 [protein inhibitor of activated STAT-1 (signal transducer and activator of transcription 1)]. PIAS1 functions as an NF-κB repressor, and thus, the PIAS1-binding modified N-mutant PRRSV-2 became NF-κB activation-resistant in its phenotype. During coinfection of pigs with PRRSV-2 and Streptococcus suis, the N-mutant PRRSV-2 decreased the expression of proinflammatory cytokines and showed clinical attenuation. This review describes the coinfection of pigs with various pathogens, the generation of mutant PRRSV for NF-κB suppression, inflammatory profiles during bacterial coinfection, and the potential application of these findings to designing a new vaccine candidate for PRRSV-2.
Collapse
Affiliation(s)
- Junyu Tang
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Leyi Wang
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Weihuan Fang
- Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chia-Ming Su
- Department of Biochemistry and Cell Biology, School of Medicine, Boston University, Boston, MA, USA
| | - Jineui Kim
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yijun Du
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, Shandong, China
| | - Dongwan Yoo
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
7
|
Sigal A, Neher RA, Lessells RJ. The consequences of SARS-CoV-2 within-host persistence. Nat Rev Microbiol 2025; 23:288-302. [PMID: 39587352 DOI: 10.1038/s41579-024-01125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
SARS-CoV-2 causes an acute respiratory tract infection that resolves in most people in less than a month. Yet some people with severely weakened immune systems fail to clear the virus, leading to persistent infections with high viral titres in the respiratory tract. In a subset of cases, persistent SARS-CoV-2 replication results in an accelerated accumulation of adaptive mutations that confer escape from neutralizing antibodies and enhance cellular infection. This may lead to the evolution of extensively mutated SARS-CoV-2 variants and introduce an element of chance into the timing of variant evolution, as variant formation may depend on evolution in a single person. Whether long COVID is also caused by persistence of replicating SARS-CoV-2 is controversial. One line of evidence is detection of SARS-CoV-2 RNA and proteins in different body compartments long after SARS-CoV-2 infection has cleared from the upper respiratory tract. However, thus far, no replication competent virus has been cultured from individuals with long COVID who are immunocompetent. In this Review, we consider mechanisms of viral persistence, intra-host evolution in persistent infections, the connection of persistent infections with SARS-CoV-2 variants and the possible role of SARS-CoV-2 persistence in long COVID. Understanding persistent infections may therefore resolve much of what is still unclear in COVID-19 pathophysiology, with possible implications for other emerging viruses.
Collapse
Affiliation(s)
- Alex Sigal
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Richard A Neher
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Richard J Lessells
- KwaZulu-Natal Research Innovation & Sequencing Platform, School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
8
|
Monschein T, Zrzavy T, Rommer PS, Meuth SG, Chan A, Berger T, Hartung HP. SARS-CoV-2 Vaccines and Multiple Sclerosis: An Update. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200393. [PMID: 40279527 DOI: 10.1212/nxi.0000000000200393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/18/2025] [Indexed: 04/27/2025]
Abstract
The highly contagious zoonosis coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was declared a pandemic by the World Health Organization on March 11, 2020, and has led to a global health crisis with nearly 777 million confirmed infections and over 7 million deaths worldwide by November 10, 2024.1-3 Over time, various variants emerged, with Omicron and its sublines dominating the world over the past 3 years.4 In addition, there is increasing evidence regarding the immune response of SARS-CoV-2 vaccines, especially for people with multiple sclerosis (MS) receiving disease-modifying therapies. Hence, with this review, we aim to provide an updated overview and recommendations for clinical practice regarding MS and SARS-CoV-2 vaccines, including efficacy and safety, SARS-CoV-2 variants, vaccine hesitancy, and the immune response under treatment with respective disease-modifying therapies.
Collapse
Affiliation(s)
- Tobias Monschein
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Paulus S Rommer
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Universitätsklinikum Düsseldorf, Heinrich-Heine-University, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
- Department of Neurology, Medical Faculty, Universitätsklinikum Düsseldorf, Heinrich-Heine-University, Germany
- Brain and Mind Center, University of Sydney, Australia; and
- Department of Neurology, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
9
|
Zehr JD, Sun Q, Ceres K, Merrill A, Tyson GH, Ceric O, Guag J, Pauley S, McQueary HC, Sams K, Reboul G, Mitchell PK, Anderson R, Franklin-Guild R, Guarino C, Cronk BD, Burbick CR, Wolking R, Peak L, Zhang Y, McDowall R, Krishnamurthy A, Slavic D, Sekhon PK, Needle D, Gibson R, Cazer C, Rodriguez J, Harris B, Stanhope MJ, Goodman LB. Population and pan-genomic analyses of Staphylococcus pseudintermedius identify geographic distinctions in accessory gene content and novel loci associated with AMR. Appl Environ Microbiol 2025:e0001025. [PMID: 40272117 DOI: 10.1128/aem.00010-25] [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: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 04/25/2025] Open
Abstract
Staphylococcus pseudintermedius is a common representative of the normal skin microbiota of dogs and cats but is also a causative agent of a variety of infections. Although primarily a canine/feline bacterium, recent studies suggest an expanded host range including humans. This paper details population genomic analyses of the largest yet assembled and sequenced collection of S. pseudintermedius isolates from across the USA and Canada and assesses these isolates within a larger global population genetic context. We then employ a pan-genome-wide association study analysis of over 1,700 S. pseudintermedius isolates from sick dogs and cats, covering the period 2017-2020, correlating loci at a genome-wide level, with in vitro susceptibility data for 23 different antibiotics. We find no evidence from either core genome phylogenies or accessory genome content for separate lineages colonizing cats or dogs. Some core genome geographic clustering was evident on a global scale, and accessory gene content was noticeably different between various regions, some of which could be linked to known antimicrobial resistance (AMR) loci for certain classes of antibiotics (e.g., aminoglycosides). Analysis of genes correlated with AMR was divided into different categories, depending on whether they were known resistance mechanisms, on a plasmid, or a putatively novel resistance mechanism on the chromosome. We discuss several novel chromosomal candidates for follow-up laboratory experimentation, including, for example, a bacteriocin (subtilosin), for which the same protein from Bacillus subtilis has been shown to be active against Staphylococcus aureus infections, and for which the operon, present in closely related Staphylococcus species, is absent in S. aureus.IMPORTANCEStaphylococcus pseudintermedius is an important causative agent of a variety of canine and feline infections, with recent studies suggesting an expanded host range, including humans. This paper presents global population genomic data and analysis of the largest set yet sequenced for this organism, covering the USA and Canada as well as more globally. It also presents analysis of in vitro antibiotic susceptibility testing results for the North American (NA) isolates, as well as genetic analysis for the global set. We conduct a pan-genome-wide association study analysis of over 1,700 S. pseudintermedius isolates from sick dogs and cats from NA to correlate loci at a genome-wide level with the in vitro susceptibility data for 23 different antibiotics. We discuss several chromosomal loci arising from this analysis for follow-up laboratory experimentation. This study should provide insight regarding the development of novel molecular treatments for an organism of both veterinary and, increasingly, human medical concern.
Collapse
Affiliation(s)
| | - Qi Sun
- Cornell University, Ithaca, New York, USA
| | | | - Amy Merrill
- Office of Applied Science, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, USA
| | - Gregory H Tyson
- Office of Applied Science, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, USA
| | - Olgica Ceric
- Office of Applied Science, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, USA
| | - Jake Guag
- Office of Applied Science, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, USA
| | - Sarah Pauley
- Office of Applied Science, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, USA
| | | | - Kelly Sams
- Cornell University, Ithaca, New York, USA
| | | | | | | | | | | | | | - Claire R Burbick
- Washington Animal Disease Diagnostic Laboratory, Washington State University, Pullman, Washington, USA
| | - Rebecca Wolking
- Washington Animal Disease Diagnostic Laboratory, Washington State University, Pullman, Washington, USA
| | - Laura Peak
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Yan Zhang
- Ohio Department of Agriculture Animal Disease Diagnostic Laboratory, Reynoldsburg, Ohio, USA
| | - Rebeccah McDowall
- Animal Health Laboratory, University of Guelph, Guelph, Ontario, Canada
| | | | - Durda Slavic
- Animal Health Laboratory, University of Guelph, Guelph, Ontario, Canada
| | | | - David Needle
- Veterinary Diagnostic Laboratory, University of New Hampshire, Durham, New Hampshire, USA
| | - Robert Gibson
- Veterinary Diagnostic Laboratory, University of New Hampshire, Durham, New Hampshire, USA
| | | | | | - Beth Harris
- USDA APHIS National Animal Health Laboratory Network, Ames, Iowa, USA
| | | | | |
Collapse
|
10
|
Li Y, Yang S, Qian J, Liu S, Li Y, Song X, Cao Q, Guo R, Zhao Y, Sun M, Hu M, Li J, Zhang X, Fan B, Li B. Molecular characteristics of the immune escape of coronavirus PEDV under the pressure of vaccine immunity. J Virol 2025:e0219324. [PMID: 40237499 DOI: 10.1128/jvi.02193-24] [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: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Abstract
Coronaviruses have undergone evolutionary changes and mutations in response to the immune pressures exerted by vaccines and environmental factors, resulting in more severe consequences during breakthrough infections. Nevertheless, the specific correlation between the evolutionary mutations of coronaviruses and immune pressures remains ambiguous. Swine coronavirus-porcine epidemic diarrhea virus (PEDV)-has existed for decades. This study utilized in vivo preparation of polyclonal antibodies against the PEDV and identified critical neutralizing epitopes through serial in vitro passaging. Then, the recombinant mutated strains were successfully constructed. In vitro experiments confirmed the ability of the rA1273P strain to escape neutralization by polyclonal antibodies. Both in vitro cell studies and in vivo animal experiments revealed that the strain maintains virulence and pathogenicity while evading antibody pressure post-vaccination. The pathogenicity of the strain while evading immune pressure is comparable to wild-type strains. A comparison of the S protein gene between vaccine strains and clinical strains identified mutations in 1273 amino acid positions in clinical strains. In conclusion, this study identified a novel PEDV S protein neutralizing site under immune pressure through serial passaging, indicating that the 1,273th amino acid position is prone to mutation under prolonged antibody pressure, enhancing the virus's ability to escape hosts. This study offers new insights into the interpretation of coronavirus escape immune pressure and provides technical support for monitoring and predicting the variation and evolution of coronavirus.IMPORTANCECoronaviruses represent an ongoing public health threat because of high variability. Since 2010, the emergence of highly pathogenic porcine epidemic diarrhea virus (PEDV) strains has resulted in significant economic losses to the global pig industry. PEDV undergoes evolution and mutation under external immune pressure, rendering it an increasingly challenging target for prevention and control measures. Here, we prepared the polyclonal antibodies against PEDV and identified a novel neutralization epitope on the S protein (1,273th amino acids) through serial in vitro passaging. Furthermore, our findings indicate that the mutation of A1273P in the S protein did not alter the virulence of the PEDV but significantly enhanced its ability to escape and infect the host in vitro and in vivo. Finally, we found that the 1,273 amino acid position of the S gene has been mutated to varying degrees in clinical PEDV strains. This work provides a specific correlation between the evolutionary mutations of coronaviruses and immune pressures.
Collapse
Affiliation(s)
- Yunchuan Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Shanshan Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jiali Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Shiyu Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yupeng Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xu Song
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Qiuxia Cao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongxiang Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Min Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Mi Hu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xuehan Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| |
Collapse
|
11
|
Wu X, Lan S, Wang Y, Yang S, Shen Q, Wang X, Liu Y, Yang H, Ji L, Zhang W. LDHB suppresses the PDCoV proliferation by targeting viral nucleocapsid protein for autophagic degradation. Microbiol Spectr 2025:e0278724. [PMID: 40231829 DOI: 10.1128/spectrum.02787-24] [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: 11/04/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a newly identified enteric coronavirus that causes serious diarrhea and vomiting in pigs, leading to substantial economic losses globally. Studying the molecular interactions between virus and host proteins is crucial for developing new anti-PDCoV strategies. Here, the role and mechanism of lactate dehydrogenase B (LDHB) in PDCoV replication were investigated. LDHB suppresses PDCoV replication in a dose-dependent manner, whereas the knockdown of LDHB via RNA interference enhances virus proliferation in LLC-PK1 cells. Mechanistically, LDHB directly interacts with PDCoV N protein in the cytoplasm. LDHB mediated the autophagic degradation of PDCoV N protein, thereby inhibiting viral replication. To our interests, PDCoV infection or PDCoV N protein expression significantly reduces LDHB expression in cells. Further studies showed that PDCoV N protein, dependent on its LIR motif, binds to the LC3. It facilitates LDHB degradation, possibly as a strategy for viral evasion from host cell cytosolic defense mechanisms. Overall, the present study provided a novel regulatory mechanism of LDHB in PDCoV infection and suggested new avenues for the antiviral strategy. IMPORTANCE This study elucidates the intricate interaction between the PDCoV N protein and LDHB within the context of viral infection and immune evasion strategies. By demonstrating that LDHB can suppress PDCoV replication through a novel mechanism involving the autophagic degradation of the viral N protein, the research highlights the potential of targeting such interactions for antiviral strategies. The findings not only expand our understanding of how PDCoV manipulates host cell pathways to its advantage but also open up new avenues for therapeutic interventions that could mitigate the impact of this and similar viral pathogens.
Collapse
Affiliation(s)
- Xiaohan Wu
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shijin Lan
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shixing Yang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Quan Shen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaochun Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuwei Liu
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hongfeng Yang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Likai Ji
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wen Zhang
- Institute of Critical Care Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
12
|
Zhang S, Cao Y, Huang Y, Zhang X, Mou C, Qin T, Chen Z, Bao W. Abortive PDCoV infection triggers Wnt/β-catenin pathway activation, enhancing intestinal stem cell self-renewal and promoting chicken resistance. J Virol 2025; 99:e0013725. [PMID: 40135895 PMCID: PMC11998530 DOI: 10.1128/jvi.00137-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging coronavirus causing economic losses to swine industries worldwide. PDCoV can infect chickens under laboratory conditions, usually with no symptoms or mild symptoms, and may cause outbreaks in backyard poultry and wildfowl, posing a potential risk of significant economic loss to the commercial poultry industry. However, the reasons for such a subdued reaction after infection are not known. Here, using chicken intestinal organoid monolayers, we found that although PDCoV infects them nearly as well as porcine intestinal organoid monolayers, infection did not result in detectable amounts of progeny virus. In ex vivo and in vivo experiments using chickens, PDCoV infection failed to initiate interferon and inflammatory responses. Additionally, infection did not result in a disrupted intestinal barrier nor a reduced number of goblet cells and mucus secretion, as in pigs. In fact, the number of goblet cells increased as did the secreted mucus, thereby providing an enhanced protective barrier. Ex vivo PDCoV infection in chicken triggered activation of the Wnt/β-catenin pathway with the upregulation of Wnt/β-catenin pathway genes (Wnt3a, Lrp5, β-catenin, and TCF4) and Wnt target genes (Lgr5, cyclin D1, and C-myc). This activation stimulates the self-renewal of intestinal stem cells (ISCs), accelerating ISC-mediated epithelial regeneration by significant up-regulation of PCNA (transiently amplifying cells), BMI1 (ISCs), and Lyz (Paneth cells). Our data demonstrate that abortive infection of PDCoV in chicken cells activates the Wnt/β-catenin pathway, which facilitates the self-renewal and proliferation of ISCs, contributing to chickens' resistance to PDCoV infection.IMPORTANCEThe intestinal epithelium is the main target of PDCoV infection and serves as a physical barrier against pathogens. Additionally, ISCs are charged with tissue repair after injury, and promoting rapid self-renewal of intestinal epithelium will help to re-establish the physical barrier and maintain intestinal health. We found that PDCoV infection in chicken intestinal organoid monolayers resulted in abortive infection and failed to produce infectious virions, disrupt the intestinal barrier, reduce the number of goblet cells and mucus secretion, and induce innate immunity, but rather increased goblet cell numbers and mucus secretion. Abortive PDCoV infection activated the Wnt/β-catenin pathway, enhancing ISC renewal and accelerating the renewal and replenishment of shed PDCoV-infected intestinal epithelial cells, thereby enhancing chicken resistance to PDCoV infection. This study provides novel insights into the mechanisms underlying the mild or asymptomatic response to PDCoV infection in chickens, which is critical for understanding the virus's potential risks to the poultry industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
13
|
Kozanli E, Winkel AMAM, Han AX, van den Brink S, van den Brandt A, Haverkort ME, Euser SM, Russell CA, de Jong MD, van Houten MA, van Lelyveld SFL, Eggink D. Shortened SARS-CoV-2 Viral RNA Shedding in Saliva During Early Omicron Compared to Wild-Type Pandemic Phase. J Infect Dis 2025; 231:940-945. [PMID: 39823580 DOI: 10.1093/infdis/jiaf031] [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: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
This study compared the dynamics of SARS-CoV-2 viral shedding in saliva between wild-type virus-infected and Omicron-infected household cohorts. Preexisting immunity in participants likely shortens the viral RNA shedding duration and lowers viral load peaks. Frequent saliva sampling can be a convenient tool to study viral load dynamics.
Collapse
Affiliation(s)
- Eva Kozanli
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelique M A M Winkel
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Alvin X Han
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sharon van den Brink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Annemarie van den Brandt
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Mildred E Haverkort
- Department of Infectious Disease Control, Public Health Services Kennemerland, Haarlem, The Netherlands
| | - Sjoerd M Euser
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Epidemiology and Infection Prevention, Regional Public Health Laboratory Kennemerland, Haarlem, The Netherlands
| | - Colin A Russell
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Menno D de Jong
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marlies A van Houten
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Pediatrics, Spaarne Gasthuis, Haarlem and Hoofddorp, The Netherlands
| | - Steven F L van Lelyveld
- Spaarne Gasthuis Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Internal Medicine, Spaarne Gasthuis Hospital, Haarlem/Hoofddorp, The Netherlands
| | - Dirk Eggink
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Lv S, Ma R, Tang Q, Wang X, Wang C, Zhang K, Li H, Ye W, Zhou W. Discovery of 3,4-dihydropyrimidine derivatives as novel Anti-PEDV agents targeting viral internalization through a unique calcium homeostasis disruption mechanism. Eur J Med Chem 2025; 291:117637. [PMID: 40262295 DOI: 10.1016/j.ejmech.2025.117637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) poses critical challenges to global swine production, with current vaccines showing limited efficacy against emerging strains. To address this gap, we designed 41 novel 3,4-dihydropyrimidine derivatives via systematic structure-activity relationship (SAR) optimization. Compound D39, incorporating a C-4 2'-substituted biphenyl, C-2 thione, C-6 phenyl, and C-5 isopropanol substituents, emerged as the most potent anti-PEDV agent (EC50 = 0.09 μM, SI = 358.9), outperforming remdesivir (EC50 = 3.14 μM, SI > 40.8) by 35-fold. D39 exhibited broad-spectrum anti-coronavirus activity (FIPV, IDV) at micromolar levels and demonstrated acceptable metabolic stability (T1/2 = 78.75 min, Clint = 8.8 μL/min/mg) in porcine liver microsomes. Mechanistic studies revealed the antiviral actions was achieved by blocking PEDV early internalization via intracellular Ca2+ homeostasis modulation. These findings highlight D39 as a first-in-class anti-PEDV candidate with a unique dihydropyrimidine scaffold and a calcium-targeting mechanism, offering a promising therapeutic strategy against coronaviral infections.
Collapse
Affiliation(s)
- Sai Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Rumeng Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Qun Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenchong Ye
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| |
Collapse
|
15
|
Guillebaud J, Ou TP, Hul V, Hoem T, Meng C, Nuon S, Hoem S, Lim R, Khun L, Furey NM, Cappelle J, Duong V, Chevalier V. Study of coronavirus diversity in wildlife in Northern Cambodia suggests continuous circulation of SARS-CoV-2-related viruses in bats. Sci Rep 2025; 15:12628. [PMID: 40221475 PMCID: PMC11993651 DOI: 10.1038/s41598-025-92475-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/14/2025] Open
Abstract
Since SARS-CoV-2's emergence, studies in Southeast Asia, including Cambodia, have identified related coronaviruses (CoVs) in rhinolophid bats. This pilot study investigates the prevalence and diversity of CoVs in wildlife from two Cambodian provinces known for wildlife trade and environmental changes, factors favoring zoonotic spillover risk. Samples were collected from 2020 to 2022 using active (capture and swabbing of bats and rodents) and non-invasive (collection of feces from bat caves and wildlife habitats) methods. RNA was screened for CoVs using conventional pan-CoVs and real-time Sarbecovirus-specific PCR systems. Positive samples were sequenced and phylogenetic analysis was performed on the partial RdRp gene. A total of 2608 samples were collected: 867 rectal swabs from bats, 159 from rodents, 41 from other wild animals, and 1541 fecal samples. The overall prevalence of CoVs was 2.0%, with a 3.3% positive rate in bats, 2.5% in rodents, and no CoVs detected in other wildlife species. Alpha-CoVs were exclusive to bats, while Beta-CoVs were found in both bats and rodents. Seven SARS-CoV-2-related viruses were identified in Rhinolophus shameli bats sampled in August 2020, March 2021, and December 2021. Our results highlight diverse CoVs in Cambodian bats and rodents and emphasize bats as significant reservoirs. They also suggest continuous circulation of bat SARS-CoV-2-related viruses may occur in a region where ecological and human factors could favor virus emergence. Continuous surveillance and integrated approaches are crucial to managing and mitigating emerging zoonotic diseases.
Collapse
Affiliation(s)
- Julia Guillebaud
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France.
| | - Tey Putita Ou
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Vibol Hul
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Thavry Hoem
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Chana Meng
- Department of Wildlife and Biodiversity, Forestry Administration, Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Sithun Nuon
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sreyleak Hoem
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Reaksa Lim
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Limmey Khun
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Julien Cappelle
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Véronique Chevalier
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo, Madagascar
- CIRAD, UMR ASTRE, Antananarivo, Madagascar
| |
Collapse
|
16
|
Wang X, Wang L, Li D, Liu Y, Shang Q, Liu Y, Zhang L, Xu Z, Huang C, Song C. HDAC4 suppresses porcine epidemic diarrhea virus infection through negatively regulating MEF2A-GLUT1/3 axis- mediated glucose uptake. Vet Microbiol 2025; 305:110520. [PMID: 40250106 DOI: 10.1016/j.vetmic.2025.110520] [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: 12/16/2024] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV), a porcine enteropathogenic coronavirus, causes severe diarrhea and death in neonatal piglets. Histone deacetylase 4 (HDAC4), a member of class IIa deacetylases, controls a wide range of physiological processes, but, little is known about its role in PEDV infection. Here, we report a novel strategy by which PEDV manipulates HDAC4. First, HDAC4 expression was examined, and showed a significant down-regulation in PEDV-infected Vero and IPEC-J2 cells. Subsequently, knockdown of HDAC4 by specific small interfering RNA (siRNA) led to an increase in viral infection, whereas overexpression of HDAC4 remarkably suppressed PEDV infection. Mechanistically, we showed that HDAC4 significantly reduced glucose uptake, as glucose is required for PEDV infection. Through screening, we identified glucose transporters 1 and 3 (GLUT1 and GLUT3) as responsible for glucose uptake during PEDV infection. We further confirmed that HDAC4 regulated GLUT1 and GLUT3 expression through its converging hub, myocyte enhancer factor 2 A (MEF2A). Taken together, these findings contribute to a better understanding of a novel function of HDAC4 in regulating glucose uptake via MEF2A-GLUT1/3 to limit PEDV infection, and provide new strategies for the development of anti-PEDV drugs.
Collapse
Affiliation(s)
- Xiaomin Wang
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Lei Wang
- College of Life Sciences, Engineering Research Center for the Prevention and Control of Animal Original Zoonosis of Fujian Province University, Longyan University, Longyan, Fujian 364012, PR China
| | - Duan Li
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Yilong Liu
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Qi Shang
- Anhui Divinity Biological Products Co., LTD, Bozhou, Anhui 236800, PR China
| | - Yanling Liu
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Leyi Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 625014, PR China
| | - Zheng Xu
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China.
| | - Cuiqin Huang
- College of Life Sciences, Engineering Research Center for the Prevention and Control of Animal Original Zoonosis of Fujian Province University, Longyan University, Longyan, Fujian 364012, PR China.
| | - Changxu Song
- College of Animal Science, National Engineering Center for Swine Breeding Industry, State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, PR China.
| |
Collapse
|
17
|
Tian RF, Feng LL, Liang X, Shi Y, Wang H, Fan J, Fan XY, Zhang JJ, Ke Y, Yang T, Huo F, Fu X, Cui HY, Chen ZN, Li L. Carnitine palmitoyltransferase 2 as a novel prognostic biomarker and immunoregulator in colorectal cancer. Int J Biol Macromol 2025; 309:142945. [PMID: 40210071 DOI: 10.1016/j.ijbiomac.2025.142945] [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: 10/31/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Metabolic interventions are critical for enhancing immunotherapy efficacy, but reliable metabolic targets remain absent for colorectal cancer (CRC). This study aims to investigate the interplay between metabolic and immunological factors in CRC, identify metabolic immunoregulatory molecules, and propose targets for prognostic and therapeutic applications. METHODS Immune infiltration and metabolic pathways in CRC were analyzed using CIBERSORT and gene set variation analyses. Cox regression identified survival-related metabolic genes, forming a metabolic-related gene prognostic index (MRGPI), which was validated through survival analysis, timeROC, GSEA, CIBERSORT, and TIDE. Hub genes in the MRGPI were assessed using enrichment and co-expression network analyses. The expression of carnitine palmitoyltransferase 2 (CPT2) was validated through multiplex immunofluorescence of tissue microarrays. While its role was examined by western blot, CCK-8 assay, flow cytometry, qRT-PCR, Elisa, chemotaxis assays, etc. RESULTS: Fatty acid oxidation (FAO) pathways were significantly altered in CRC and correlated with immune cell infiltration and patient survival. The MRGPI, constructed from five survival-related metabolic genes, demonstrated strong prognostic and immunotherapeutic predictive value. Moreover, CPT2, a key hub gene in the MRGPI, whose lower expression in plasma cells predicts unfavorable patients' survival and could be an independent prognostic indicator, while its knockout in tumor cells significantly increases the infiltrating levels of CD8+ T cells via promoting the release of CCL25. CONCLUSION The FAO-dominated MRGPI is a promising biomarker for predicting patient outcomes and immunotherapy response. CPT2 holds potential as a prognostic marker and therapeutic target for CRC metabolic immunotherapy.
Collapse
Affiliation(s)
- Ruo-Fei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Le-Le Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang 110000, China
| | - Xin-Yu Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ting Yang
- Bayi Orthopedic Hospital, Chengdu 610031, China
| | - Fei Huo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Fu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hong-Yong Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
18
|
Zhou A, Subramanian PSG, El-Naggar S, Shisler JL, Verma V, Nguyen TH. Capsid and genome damage are the leading inactivation mechanisms of aerosolized porcine respiratory coronavirus at different relative humidities. Appl Environ Microbiol 2025:e0231924. [PMID: 40192313 DOI: 10.1128/aem.02319-24] [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: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Relative humidity (RH) varies widely in indoor environments based on temperature, outdoor humidity, heating systems, and other environmental conditions. This study explored how RH affects aerosolized porcine respiratory coronavirus (PRCV), a model for coronaviruses, over a time range from 0 min to a maximum of 1 h, and the molecular mechanism behind viral infectivity reduction. These questions were answered by quantifying: (i) viral-host receptor interactions, (ii) capsid integrity, (iii) viral genome integrity, and (iv) virus infectivity. We found RH did not alter PRCV-receptor interactions. RHs 45-55% and 65-75% damaged viral genomes (2 log10 reduction and 1 log10 reduction, respectively, in terms of median sample value), whereas RHs 55-65% decreased capsid integrity (2 log10 reduction). No apparent virion damage was observed in RH 75-85%. Two assays were used to quantify virus presence: qPCR for detecting the viral genomes and plaque-forming unit assay for detecting the virus replication. Our results indicated that the qPCR assay overestimated the concentrations of infectious viruses, and RNase treatment with long-range RT-qPCR performed better than one-step RT-qPCR. We propose that understanding the influence of RH on the stability of aerosolized viruses provides critical information for detecting and preventing the indoor transmission of coronaviruses. IMPORTANCE Indoor environments can impact the stability of respiratory viruses, which can then affect the transmission rates. The mechanisms of how relative humidity (RH) affects virus infectivity still remain unclear. This study found RH inactivates porcine respiratory coronavirus by damaging its capsid and genome. The finding highlights the potential role of controlling indoor RH levels as a strategy to reduce the risk of coronavirus transmission.
Collapse
Affiliation(s)
- Aijia Zhou
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
- The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - P S Ganesh Subramanian
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Salma El-Naggar
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Joanna L Shisler
- Department of Microbiology, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Vishal Verma
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Thanh H Nguyen
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
- The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
- Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
- Carle Illinois College of Medicine, Urbana, Illinois, USA
| |
Collapse
|
19
|
Meng Y, Zhang Z, Zhou C, Tang X, Hu X, Tian G, Yang J, Yao Y. Protein structure prediction via deep learning: an in-depth review. Front Pharmacol 2025; 16:1498662. [PMID: 40248099 PMCID: PMC12003282 DOI: 10.3389/fphar.2025.1498662] [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: 09/30/2024] [Accepted: 02/28/2025] [Indexed: 04/19/2025] Open
Abstract
The application of deep learning algorithms in protein structure prediction has greatly influenced drug discovery and development. Accurate protein structures are crucial for understanding biological processes and designing effective therapeutics. Traditionally, experimental methods like X-ray crystallography, nuclear magnetic resonance, and cryo-electron microscopy have been the gold standard for determining protein structures. However, these approaches are often costly, inefficient, and time-consuming. At the same time, the number of known protein sequences far exceeds the number of experimentally determined structures, creating a gap that necessitates the use of computational approaches. Deep learning has emerged as a promising solution to address this challenge over the past decade. This review provides a comprehensive guide to applying deep learning methodologies and tools in protein structure prediction. We initially outline the databases related to the protein structure prediction, then delve into the recently developed large language models as well as state-of-the-art deep learning-based methods. The review concludes with a perspective on the future of predicting protein structure, highlighting potential challenges and opportunities.
Collapse
Affiliation(s)
- Yajie Meng
- College of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, China
| | - Zhuang Zhang
- College of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, China
| | - Chang Zhou
- College of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, China
| | - Xianfang Tang
- College of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, China
| | - Xinrong Hu
- College of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, China
| | | | | | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou, China
- Key Laboratory of Data Science and Intelligence Education, Ministry of Education, Hainan Normal University, Haikou, China
- Key Laboratory of Computational Science and Application of Hainan Province, Hainan Normal University, Haikou, China
| |
Collapse
|
20
|
Feberwee A, Ferguson-Noel N, Catania S, Bottinelli M, Wawagema N, Gyuranecz M, Gautier-Bouchardon AV, Lysnyansky I, Wiegel J, Möller Palau-Ribes F, Ramirez AS. Mycoplasma gallisepticum and Mycoplasma synoviae in commercial poultry: current control strategies and future challenges. Avian Pathol 2025; 54:168-174. [PMID: 39471302 DOI: 10.1080/03079457.2024.2419037] [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: 04/01/2024] [Revised: 07/26/2024] [Accepted: 09/15/2024] [Indexed: 11/01/2024]
Abstract
Mycoplasma gallisepticum (Mg) and Mycoplasma synoviae (Ms) are regarded as the most important avian mycoplasma species for today's chicken and turkey farming industry from clinical and economical perspectives. Control strategies for Mg and Ms have become more efficient due to investments in mycoplasma research over the last 70 years. These investments have contributed to the further implementation of serological and molecular testing, the development of vaccines, and the improvement of antimicrobial treatment strategies. However, the increasing spotlight on welfare, the pressure on prudent use of antimicrobials, and the expected global increase in poultry production, are going to have an impact on the future control of avian mycoplasmas in commercial poultry. In this paper a group of avian mycoplasma experts discuss the future challenges in mycoplasma control considering the background of these expected changes and the relevance for future avian mycoplasma research.
Collapse
Affiliation(s)
| | - Naola Ferguson-Noel
- Poultry Diagnostic and Research Center, The University of Georgia, Athens, GA, USA
| | - Salvatore Catania
- Istituto Zooprofilattico Sperimentale delle Venezie, Buttapietra, Italy
| | - Marco Bottinelli
- Istituto Zooprofilattico Sperimentale delle Venezie, Buttapietra, Italy
| | - Nadeeka Wawagema
- Melbourne Veterinary School, University of Melbourne, Melbourne, Australia
| | - Miklos Gyuranecz
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
| | | | | | | | | | - Ana S Ramirez
- Universidad de Las Palmas de Gran Canaria, Canary Islands, Spain
| |
Collapse
|
21
|
Shen J, Zhang X, Tang Y, Zhang P, Yan Y, Ye P, Zhang S, Huang Z. A comprehensive validation study on the influencing factors of cough-based COVID-19 detection through multi-center data with abundant metadata. J Biomed Inform 2025; 164:104798. [PMID: 39993588 DOI: 10.1016/j.jbi.2025.104798] [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: 10/15/2024] [Revised: 12/23/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE In recent years, COVID-19 has placed enormous burdens on healthcare systems. Currently, hundreds of thousands of new cases are reported monthly. World Health Organization is managing COVID-19 as a long-term disease, indicating that an efficient and low-cost detection method remains necessary. Previous studies have shown competitive results on cough-based COVID-19 detection combined with deep learning methods. However, most studies have focused only on improving classification performance on single-source data while neglecting the impact of various factors in real-world applications. METHODS To this end, we collected clinical and large-scale crowdsourced cough audios with abundant metadata to comprehensively validate the performance differences among different groups. Specifically, we leveraged self-supervised learning for pre-training and fine-tuned the model with data from different sources. Then based on the metadata, we compared the effects of factors such as cough types, symptoms, and infection stages on detection performance. Moreover, we recorded clinical indicators of viral load and antibody levels and observed the correlation between predicted probabilities and indicator values for the first time. Several open-source datasets were tested to verify the model generalizability. RESULTS The area under receiver operating characteristic curve is 0.79 for clinical data and 0.69 for crowdsourced data, indicating differences between clinical validation and real-world application. The performance in detecting symptomatic COVID-19 subjects is usually better than detecting asymptomatic COVID-19 subjects. The prediction results show weak correlation with clinical indicators on a small number of clinical data. Poor detection performance in recovery individuals and open-source datasets shows a limitation of existing cough-based detection models. CONCLUSION Our study validated the model performance and limitations using multi-source data with abundant metadata, which helped researchers evaluate the feasibility of cough-based COVID-19 detection model in practical applications.
Collapse
Affiliation(s)
- Jiakun Shen
- Key Laboratory of Speech Acoustics and Content Understanding, Institute of Acoustics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xueshuai Zhang
- Key Laboratory of Speech Acoustics and Content Understanding, Institute of Acoustics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yanfen Tang
- Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Pengyuan Zhang
- Key Laboratory of Speech Acoustics and Content Understanding, Institute of Acoustics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yonghong Yan
- Key Laboratory of Speech Acoustics and Content Understanding, Institute of Acoustics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Pengfei Ye
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Pediatrics, Beijing, China
| | | | - Zhihua Huang
- School of Information Science and Engineering, Xinjiang University, Urumchi, China
| |
Collapse
|
22
|
Lopez KP, Cool KR, Bold D, Gaudreault NN, Roberts BA, Maag E, Richt JA, Pogranichniy RM. Detection of SARS-CoV-2- specific antibodies in domestic cats using different ELISA tests. J Virol Methods 2025; 333:115099. [PMID: 39662743 DOI: 10.1016/j.jviromet.2024.115099] [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: 09/20/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
The emergence of SARS-CoV-2 raised concerns about the potential for interspecies transmission, particularly among domestic animals. We evaluated the seroprevalence of SARS-CoV-2 antibodies in domestic cats from various sites in North America. A total of 216 serum samples collected between December 2019 and February 2022, were analyzed using four different enzyme-linked immunosorbent assays (ELISAs), including a commercial surrogate virus neutralization test (sVNT), a commercial double antigen test (dN ELISA), and two in-house developed indirect ELISAS based on receptor-binding domain (RBD iELISA) and the nucleocapsid (N iELISA) proteins, respectively. Seropositive samples in the commercial ELISAs were subject to virus neutralization test (cVNT) employing the Wuhan-like USA-WA1/2020 SARS-CoV-2 isolate. Our findings revealed that, 6 % (12/216) of the cat serum samples tested positive by the sVNT, while 4 % (9/216) tested positive for the dN-ELISA. Interestingly, the N iELISA showed a higher seroprevalence, with 31 % of the samples testing positive, possibly due to cross-reactive antibodies against the N protein of other coronavirus commonly found in cats. There was a high concordance between sVNT, cVNT, and RBD iELISA. Among positive sVNT cat serum samples, 75 % (9/12) exhibited neutralizing titers with all samples also being positive by RBD iELISA. Notably, the RBD iELISA and sVNT demonstrated high sensitivity and specificity in detecting SARS-CoV-2 antibodies (100 and 79 %; 100 and 90 %, respectively). In conclusion, our study provides important insights into the seroprevalence of SARS-CoV-2 antibodies in domestic cats, highlighting the potential for interspecies transmission and the need for continued monitoring of SARS-CoV-2 in animal populations.
Collapse
Affiliation(s)
- Keyla P Lopez
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Konner R Cool
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Natasha N Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Bailey A Roberts
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Emma Maag
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Juergen A Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Roman M Pogranichniy
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA; Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
23
|
Lean FZ, Gallo G, Newman J, Ackroyd S, Spiro S, Cox R, Nymo IH, Bröjer C, Neimanis A, Suárez-Bonnet A, Priestnall SL, Everest H, Keep S, Bailey D, Delahay RJ, Seekings AH, McElhinney LM, Brookes SM, Núñez A. Distribution of aminopeptidase N coronavirus receptors in the respiratory and digestive tracts of domestic and wild artiodactyls and carnivores. J Gen Virol 2025; 106:002092. [PMID: 40184164 PMCID: PMC11971486 DOI: 10.1099/jgv.0.002092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Aminopeptidase N (APN) is a transmembrane protein that mediates the attachment of the spike protein of several clinically important coronaviruses (CoVs) responsible for respiratory and intestinal diseases in animals and humans. To assess the potential for APN-mediated viral tropism, we characterized APN receptor distribution in the respiratory and intestinal tissues of various artiodactyls (cervids, bovids, camelids and suids) and carnivores (canids, felids, mustelids and phocids) using immunohistochemistry. In the lungs, APN expression was limited to artiodactyls, with strong expression in the bronchiolar epithelium and weaker expression in pneumocytes. Nasal turbinate and tracheal samples, where available, showed stronger APN expression in artiodactyls over carnivores. APN was consistently detected on the microvilli of enterocytes in the small intestine across multiple taxa, while the presence in the colon was more variable. Of the animals examined, pig and alpaca consistently expressed the most abundant APN in the upper and lower respiratory tract. In silico evaluation of APN orthologue sequences from humans, artiodactyls and carnivores identified distinct evolutionary relationships. Further in silico binding predictions for alpaca alphacoronavirus and human coronavirus 229E with cognate and heterologous alpaca and human APN revealed substantial overlapping binding footprints with high conservation of amino acid residues, suggesting an evolutionary divergence and subsequent adaptation of a 229E-like or ancestral virus within a non-human animal host. This combined anatomical and in silico approach enhances understanding of host susceptibility, tissue tropism and viral transmission mechanisms in APN-dependent CoVs and has the potential to inform future strategies for disease modelling, surveillance and control.
Collapse
Affiliation(s)
- Fabian Z.X. Lean
- Pathology and Animal Sciences Department, Animal and Plant Health Agency, Addlestone, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, UK
| | | | | | - Stuart Ackroyd
- Pathology and Animal Sciences Department, Animal and Plant Health Agency, Addlestone, UK
| | | | - Ruth Cox
- National Wildlife Management Centre, Animal and Plant Health Agency, Sand Hutton, York, UK
| | | | - Caroline Bröjer
- Department of Pathology and Wildlife Diseases, Swedish Veterinary Agency, Uppsala, Sweden
| | - Aleksija Neimanis
- Department of Pathology and Wildlife Diseases, Swedish Veterinary Agency, Uppsala, Sweden
| | - Alejandro Suárez-Bonnet
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, UK
| | - Simon L. Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, UK
| | | | | | | | - Richard J. Delahay
- National Wildlife Management Centre, Animal and Plant Health Agency, Sand Hutton, York, UK
| | | | | | | | - Alejandro Núñez
- Pathology and Animal Sciences Department, Animal and Plant Health Agency, Addlestone, UK
| |
Collapse
|
24
|
Deming ME, Brown ER, McArthur MA, Schrag SJ, Arvay M, Humphrys M, Ravel J, Adelglass J, Essink B, Musante DB, Maguire R, Gorman R, Formentini E, Mason R, Robb ML, Neuzil KM, Rapaka RR, Wolff P, Kotloff KL. Vaccine efficacy of NVX-CoV2373 against SARS-CoV-2 infection in adolescents in the USA: an ancillary study to a phase 3, observer-blinded, randomised, placebo-controlled trial. THE LANCET. MICROBE 2025; 6:100984. [PMID: 39884302 DOI: 10.1016/j.lanmic.2024.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Although existing COVID-19 vaccines are known to be highly effective against severe disease and death, data are needed to assess their ability to reduce SARS-CoV-2 infection. We aimed to estimate the efficacy of the NVX-CoV2373 protein subunit vaccine against SARS-CoV-2 infection, regardless of symptoms, among adolescents. METHODS We performed an ancillary observational study (SNIFF) to the phase 3, observer-blinded, randomised, placebo-controlled PREVENT-19 trial that assessed vaccine efficacy against symptomatic COVID-19 in the USA. Participants in the PREVENT-19 trial included healthy adolescents aged 12-17 years and with no history of laboratory-confirmed SARS-CoV-2 infection. They were randomly assigned (2:1) to receive either the NVX-CoV2373 (Novavax, Gaithersburg, MD, USA) vaccine (immediate NVX-CoV2373 group) or placebo (delayed NVX-CoV2373 group) on days 0 and 21 (initial series). After 2 months, in a crossover series, participants received two doses, 21 days apart, of the intervention that they did not receive in their initial series. Participants at 47 of the PREVENT-19 sites were invited to participate in the SNIFF study and self-collect nasal swabs at home twice weekly for SARS-CoV-2 testing to assess vaccine efficacy against SARS-CoV-2 infection. This primary outcome was defined as the first identification of SARS-CoV-2 detected by RT-PCR, regardless of symptoms, with onset within 4 weeks after the second dose of the initial vaccination series until the second dose of the crossover series. Secondary outcomes were vaccine efficacy against asymptomatic and minimally symptomatic SARS-CoV-2 infection, durability of vaccine efficacy against SARS-CoV-2 infection, and durability of vaccine efficacy against asymptomatic and minimally symptomatic infections. Outcomes were analysed in the modified intention-to-treat population, which included all participants without previous SARS-CoV-2 infection and was restricted to participants enrolled within 4 weeks of the second dose of the primary (primary analysis population) or crossover (post-crossover analysis population) series. This study is registered with ClinicalTrials.gov (NCT04611802). FINDINGS Between June 1 and Dec 17, 2021, 1196 (53·2%) of the 2247 adolescent participants recruited in the PREVENT-19 trial enrolled in the SNIFF study. The primary analysis population included 471 participants in the immediate NVX-CoV2373 group and 220 in the delayed NVX-CoV2373 group. Incidence of SARS-CoV-2 infection was 14·9 cases per 100 person-years (95% CI 7·9-25·5) in the immediate group and 54·2 cases per 100 person-years (33·6-82·9) in the delayed group; vaccine efficacy was 73·5% (95% CI 47·1-86·7; p=0·0002). Incidence of minimally symptomatic or asymptomatic SARS-CoV-2 infection was 10·3 cases per 100 person-years (95% CI 4·7-19·6) in the immediate group and 36·1 cases per 100 person-years (19·8-60·7) in the delayed group; vaccine efficacy was 72·8% (95% CI 37·1-88·2; p=0·0023). After the second crossover dose, incidence of SARS-CoV-2 was 14·6 cases per 100 person-years (95% CI 8·6-23·0) in the immediate group (receiving placebo at crossover) and 9·1 cases per 100 person-years (3·0-21·3) in the delayed group, with a durability ratio of 160·3 (95% CI 59·5-431·6; p=0·35). Almost all infections after crossover were minimally symptomatic or asymptomatic, with a durability ratio of 151·4 (55·9-410·4; p=0·41). INTERPRETATION Among adolescents participating in the PREVENT-19 trial during the delta (B.1.617.2) variant wave of the COVID-19 pandemic, the NVX-CoV2373 vaccine was highly efficacious against SARS-CoV-2 infection regardless of symptoms, indicating its potential to reduce the reservoir of infections that contribute to community transmission. FUNDING US Department of Health and Human Services, Administration for Strategic Preparedness and Response, Biomedical Advanced Research and Development Authority, National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Meagan E Deming
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Monica A McArthur
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephanie J Schrag
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Melissa Arvay
- US COVID-19 Domestic Response and Division of Bacterial Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mike Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Rebecca Maguire
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard Gorman
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Robin Mason
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Merlin L Robb
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Wolff
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington, DC, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Yang X, Yin H, Liu M, Wang X, Song T, Song A, Xi Y, Zhang T, Sun Z, Li W, Niu S, Zainab F, Wang C, Zhang D, Wang H, Yang B. Isolation, phylogenetics, and characterization of a new PDCoV strain that affects cellular gene expression in human cells. Front Microbiol 2025; 16:1534907. [PMID: 40207165 PMCID: PMC11979167 DOI: 10.3389/fmicb.2025.1534907] [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: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that causes acute diarrhea, vomiting, dehydration, and even death in piglets, resulting in serious economic losses to the pork industry worldwide. PDCoV has received much attention owing to its broad host range, including humans, posing a potential threat to public health. However, the prevalence, characteristics, and host cellular gene expression of PDCoV remain poorly understood. Methods In this study, a new PDCoV strain (CHN/SX-Y/2023, GenBank number PQ373831) was successfully isolated, identified, and subjected to phylogenetic tree and transcriptome analysis in human hepatoma (Huh7) cells following PDCoV infection. Results The results showed that the CHN/SX-Y/2023 strain belongs to the Chinese lineage and causes cytopathic effects in canonical cell lines (LLC-PK1 and ST cells) and other cell lines (Huh7 and LMH cells). However, HEK-293T, EEC, MDBK, and Vero-CCL81 cells were not found to be susceptible in this study. Based on transcriptome analysis, 1,799 differentially expressed genes (DEGs) were upregulated and 771 were downregulated during PDCoV infection. Discussion Among the upregulated genes, FCGR1A, VSIG1, TNFRSF9, and PLCXD3 are associated with immunity, inflammation, and lipid catabolism. Moreover, Kyoto Encyclopedia of Genes and Genomes analysis revealed that the upregulated DEGs were significantly enriched in the MAPK, TNF, and NF-κB signaling pathways and viral protein interactions with cytokines and cytokine receptors. Protein-protein interaction networks showed that the upregulated genes CXCL8, DUSP1, PTGS2, and IL15 were associated with inflammation and immunity. In addition, the protein levels of p-IRF3, LC3-II, and ACSL4 increased, suggesting that PDCoV infection in Huh7 cells induces an intrinsic immune response, cellular autophagy, and ferroptosis. Collectively, our findings provide new insights into the characteristics and mechanisms of PDCoV infection.
Collapse
Affiliation(s)
- Xiaozhu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Hanwei Yin
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Mengyuan Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xuemei Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Tao Song
- College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Aiai Song
- Xianyang Regional Wen's Animal Husbandry Co., Ltd., Xianyang, China
| | - Yibo Xi
- School of Management Shanxi Medical University, Taiyuan, China
| | - Ting Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Wei Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Sheng Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Farwa Zainab
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Chenyang Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
26
|
Lim J, Koprowski K, Wester M, Valera E, Bashir R. Review on biphasic blood drying method for rapid pathogen detection in bloodstream infections. SLAS Technol 2025; 32:100276. [PMID: 40127716 DOI: 10.1016/j.slast.2025.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/19/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Rapid and accurate detection of pathogenic microorganisms in blood is critical for diagnosing life-threatening conditions such as bloodstream infections (BSIs). Current methods for the detection and identification of bacteria from large volumes of blood (5 mL) involve culture steps followed by DNA extraction/purification/concentration and Polymerase Chain Reaction (PCR)-based nucleic acid amplification. DNA extraction and amplification directly from blood samples is hampered by the complexity of the blood matrix, resulting in time-consuming and labor-intensive processes. This review delves into recent advancements in molecular diagnostics based on blood drying, coined as 'biphasic reaction', and highlights this new technique that attempts to overcome the limitations of traditional sample preparation and amplification processes. The biphasic blood drying method, in combination with isothermal amplification methods such as loop-mediated isothermal amplification (LAMP) or recombinase polymerase amplification (RPA), has recently been shown to improve the sensitivity of detection of bacterial, viral, and fungal pathogens from ∼1 mL of whole blood, while minimizing DNA loss and avoiding the use of extraction/purification/concentration kits. Furthermore, the biphasic approach in combination with LAMP has been shown to be a culture-free method capable of detecting bacteria in clinical samples with a sensitivity of ∼1 CFU/mL in ∼2.5 h. This represents a significant reduction in detection and identification time compared to current clinical procedures based on bacterial culture prior to PCR amplification. This review paper aims to be a guide to identify new opportunities for future advancements and applications of the biphasic technology.
Collapse
Affiliation(s)
- Jongwon Lim
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Material Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Katherine Koprowski
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Matthew Wester
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Enrique Valera
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61801, USA; Chan Zuckerberg Biohub Chicago, Chicago, IL 60642, USA..
| |
Collapse
|
27
|
Yu Z, Shao Y, Zhang Y, Cheng F, Fang P, Tu J, Song X, Qi K, Wang Z. LAMP Assay Coupled with a Pyrococcus furiosus Argonaute System for the Rapid Detection of Porcine Epidemic Diarrhea Virus. ACS Synth Biol 2025; 14:689-698. [PMID: 39964196 DOI: 10.1021/acssynbio.4c00446] [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: 03/22/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) infection can lead to serious acute intestinal infectious disease, bringing huge economic losses to the pig industry. In addition to triggering an extremely high mortality rate for lactating piglets, there is currently a lack of effective treatments and vaccines. Therefore, rapid, accurate, sensitive, and specific detection of PEDV is critical for timely control. In this study, a nucleic acid detection method combining reverse transcription loop-mediated isothermal amplification (RT-LAMP) and Pyrococcus furiosus Argonaute (PfAgo) was established for the detection of PEDV and performed after optimizing the system (mainly for the design and screening of the LAMP primers and PfAgo gDNA). The optimized system had a detection limit as low as 2.4 copies/μL. To reach more timely on-site detection of PEDV and overcome the reliance on bulky and complex equipment, a lateral flow strip was introduced into the system, which could detect the target as low as 24 copies/μL. This RT-LAMP-PfAgo system took about 35 min to react, and the results could be observed and clarified with the naked eyes. Moreover, the method was highly specific and had no cross-reactivity with other swine pathogens. The detection results for the clinical samples were consistent with those obtained by the gold standard method, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), proving its applicability. In conclusion, the established RT-LAMP-PfAgo system can provide a new solution for the development of a portable, visual PEDV testing platform.
Collapse
Affiliation(s)
- Zhaorong Yu
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Ying Shao
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Yu Zhang
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Fanyu Cheng
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Peng Fang
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Jian Tu
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Xiangjun Song
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Kezong Qi
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Zhenyu Wang
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| |
Collapse
|
28
|
Phan T, Ribeiro RM, Edelstein GE, Boucau J, Uddin R, Marino C, Liew MY, Barry M, Choudhary MC, Tien D, Su K, Reynolds Z, Li Y, Sagar S, Vyas TD, Kawano Y, Sparks JA, Hammond SP, Wallace Z, Vyas JM, Li JZ, Siedner MJ, Barczak AK, Lemieux JE, Perelson AS. Modeling suggests SARS-CoV-2 rebound after nirmatrelvir-ritonavir treatment is driven by target cell preservation coupled with incomplete viral clearance. J Virol 2025; 99:e0162324. [PMID: 39902924 PMCID: PMC11915799 DOI: 10.1128/jvi.01623-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/22/2024] [Indexed: 02/06/2025] Open
Abstract
In a subset of SARS-CoV-2-infected individuals treated with the antiviral nirmatrelvir-ritonavir, the virus rebounds following treatment. The mechanisms driving this rebound are not well understood. We used a mathematical model to describe the longitudinal viral load dynamics of 51 individuals treated with nirmatrelvir-ritonavir, 20 of whom rebounded. Target cell preservation, either by a robust innate immune response or initiation of N-R near the time of symptom onset, coupled with incomplete viral clearance, appears to be the main factor leading to viral rebound. Moreover, the occurrence of viral rebound is likely influenced by the time of treatment initiation relative to the progression of the infection, with earlier treatments leading to a higher chance of rebound. A comparison with an untreated cohort suggests that early treatments with nirmatrelvir-ritonavir may be associated with a delay in the onset of an adaptive immune response. Nevertheless, our model demonstrates that extending the course of nirmatrelvir-ritonavir treatment to a 10-day regimen may greatly diminish the chance of rebound in people with mild-to-moderate COVID-19 and who are at high risk of progression to severe disease. Altogether, our results suggest that in some individuals, a standard 5-day course of nirmatrelvir-ritonavir starting around the time of symptom onset may not completely eliminate the virus. Thus, after treatment ends, the virus can rebound if an effective adaptive immune response has not fully developed. These findings on the role of target cell preservation and incomplete viral clearance also offer a possible explanation for viral rebounds following other antiviral treatments for SARS-CoV-2. IMPORTANCE Nirmatrelvir-ritonavir is an effective treatment for SARS-CoV-2. In a subset of individuals treated with nirmatrelvir-ritonavir, the initial reduction in viral load is followed by viral rebound once treatment is stopped. We show that the timing of treatment initiation with nirmatrelvir-ritonavir may influence the risk of viral rebound. Nirmatrelvir-ritonavir stops viral growth and preserves target cells but may not lead to full clearance of the virus. Thus, once treatment ends, if an effective adaptive immune response has not adequately developed, the remaining virus can lead to rebound. Our results provide insights into the mechanisms of rebound and can help develop better treatment strategies to minimize this possibility.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Gregory E. Edelstein
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Rockib Uddin
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Marino
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - May Y. Liew
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mamadou Barry
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manish C. Choudhary
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dessie Tien
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Karry Su
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zahra Reynolds
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yijia Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Shruti Sagar
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tammy D. Vyas
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yumeko Kawano
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey A. Sparks
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah P. Hammond
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary Wallace
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jatin M. Vyas
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark J. Siedner
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Amy K. Barczak
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob E. Lemieux
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
29
|
Lim J, Van AB, Koprowski K, Wester M, Valera E, Bashir R. Amplification-free, OR-gated CRISPR-Cascade reaction for pathogen detection in blood samples. Proc Natl Acad Sci U S A 2025; 122:e2420166122. [PMID: 40063799 PMCID: PMC11929484 DOI: 10.1073/pnas.2420166122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/03/2025] [Indexed: 03/25/2025] Open
Abstract
Rapid and accurate detection of DNA from disease-causing pathogens is essential for controlling the spread of infections and administering timely treatments. While traditional molecular diagnostics techniques like PCR are highly sensitive, they include nucleic acid amplification and many need to be performed in centralized laboratories, limiting their utility in point-of-care settings. Recent advances in CRISPR-based diagnostics (CRISPR-Dx) have demonstrated the potential for highly specific molecular detection, but the sensitivity is often constrained by the slow trans-cleavage activity of Cas enzymes, necessitating preamplification of target nucleic acids. In this study, we present a CRISPR-Cascade assay that overcomes these limitations by integrating a positive feedback loop that enables nucleic acid amplification-free detection of pathogenic DNA at atto-molar levels and achieves a signal-to-noise ratio greater than 1.3 within just 10 min. The versatility of the assay is demonstrated through the detection of bloodstream infection pathogens, including Methicillin-Sensitive Staphylococcus aureus (MSSA), Methicillin-Resistant Staphylococcus aureus (MRSA), Escherichia coli, and Hepatitis B Virus (HBV) spiked in whole blood samples. Additionally, we introduce a multiplexing OR-function logic gate, further enhancing the potential of the CRISPR-Cascade assay for rapid and accurate diagnostics in clinical settings. Our findings highlight the ability of the CRISPR-Cascade assay to provide highly sensitive and specific molecular detection, paving the way for advanced applications in point-of-care diagnostics and beyond.
Collapse
Affiliation(s)
- Jongwon Lim
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - An Bao Van
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- VinUni-Illinois Smart Health Center, Building G, VinUni Campus, Vinhomes Ocean Park, Gia Lam Dist, Hanoi 100000, Vietnam
| | - Katherine Koprowski
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Matthew Wester
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Enrique Valera
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61801
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60642
| |
Collapse
|
30
|
Li W, Hangalapura BN, van den Elzen P, van den Born E, van Kuppeveld FJM, Rottier PJM, Bosch BJ. Spike gene variability in porcine epidemic diarrhea virus as a determinant for virulence. J Virol 2025; 99:e0216524. [PMID: 40001283 PMCID: PMC11915861 DOI: 10.1128/jvi.02165-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a pathogenic coronavirus that targets the swine intestinal tract, leading to acute diarrhea and high mortality in neonatal piglets. PEDV is categorized into different genotypes based on genetic variations, especially in the spike (S) gene. The S protein is crucial for viral entry and a major immune target. Significant differences in virulence have been observed among PEDV genotypes, particularly between classical strains and newly emerging strains. In this study, we explored the impact of spike gene variability on PEDV pathogenicity. Using targeted RNA recombination, we generated recombinant PEDV (rPEDV) variants carrying spike genes from contemporary strains (moderately virulent strain UU and highly virulent strain GDU), all within the genetic background of the avirulent DR13 vaccine strain. Pathogenicity was assessed in 3-day-old piglets. The rPEDV carrying the DR13 spike gene was nonpathogenic, with no detectable viral RNA in feces. The rPEDV with the UU spike gene induced mild to severe diarrhea, with moderate viral shedding but no mortality. Conversely, the rPEDV with the GDU spike gene caused severe diarrhea, high viral titers, and high mortality. These findings highlight the critical role of the spike protein in PEDV virulence, informing future development of effective control strategies, including the design of live-attenuated vaccines.IMPORTANCEThis study significantly advances our understanding of how genetic variations in the spike (S) protein of porcine epidemic diarrhea virus (PEDV) influence its ability to cause disease. By engineering viruses with spike genes from different PEDV strains, variations in this protein could be directly linked to differences in disease severity. We found that the spike protein from highly virulent strains caused severe diarrhea and high mortality in piglets, while that from less virulent strains led to milder symptoms. These findings emphasize the central role of the spike protein in determining PEDV virulence, which may enable the design of more effective vaccines to combat PEDV and reduce its impact on the swine industry.
Collapse
Affiliation(s)
- Wentao Li
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | | | | | - Frank J M van Kuppeveld
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Peter J M Rottier
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Berend-Jan Bosch
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
31
|
Zhang Z, Wei Q, Wu C, Ye Z, Qin L, Chen T, Sun Z, Tian K, Li X. Isolation and pathogenicity of a novel recombinant pseudorabies virus from the attenuated vaccine and classical strains. Front Vet Sci 2025; 12:1579148. [PMID: 40166703 PMCID: PMC11955811 DOI: 10.3389/fvets.2025.1579148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Pseudorabies (PR) remains one of the most important swine diseases in China. Live attenuated vaccines have been widely deployed and have proven highly effective in controlling PR in the field. However, recent concerns regarding the evolution and recombination events involving pseudorabies virus (PRV) vaccine strains have raised substantial attention. In the present study, a novel recombinant PRV strain named HN2201 was isolated from one stillbirth case in Henan province in 2022. To assess the genetic and evolutionary features, the major immunogenic and virulence-associated genes, including gB, gC, gD, gG, gE and TK, were sequenced and analyzed. Phylogenetic and nucleotide homology analysis revealed that gB, gC, gD and gG genes of HN2201 displayed close relationship with Chinese classical strains. However, the TK gene of HN2201 contained a continuous deletion of 205 nucleotides, sharing the highest nucleotide homology (99.9%) with HB-98 vaccine strain. Additionally, a similar deletion was observed in the promoter region of the gE gene in both HN2201 and HB-98. Pathogenicity studies on 9-week-old piglets demonstrated that HN2201 exhibited attenuated virulence, characterized by transient clinical signs. The above results suggest that the naturally isolated HN2201 likely resulted from recombination events between the PRV classical strain and the HB-98 vaccine strain. Our findings provide valuable insights into the evolution of PRV in China and underscore the necessity of scientific and cautious use of PRV vaccines in the field.
Collapse
Affiliation(s)
- Zhendong Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Qingteng Wei
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chengyue Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhengqin Ye
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Liting Qin
- Qingdao Jiazhi Biotechnology Co. Ltd., Qingdao, China
| | - Ting Chen
- Qingdao Jiazhi Biotechnology Co. Ltd., Qingdao, China
| | - Zhe Sun
- National Research Center for Veterinary Medicine, Luoyang, China
| | - Kegong Tian
- National Research Center for Veterinary Medicine, Luoyang, China
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
32
|
Pita-Juarez Y, Karagkouni D, Kalavros N, Melms JC, Niezen S, Delorey TM, Essene AL, Brook OR, Pant D, Skelton-Badlani D, Naderi P, Huang P, Pan L, Hether T, Andrews TS, Ziegler CGK, Reeves J, Myloserdnyy A, Chen R, Nam A, Phelan S, Liang Y, Gregory M, He S, Patrick M, Rane T, Wardhani A, Amin AD, Biermann J, Hibshoosh H, Veregge M, Kramer Z, Jacobs C, Yalcin Y, Phillips D, Slyper M, Subramanian A, Ashenberg O, Bloom-Ackermann Z, Tran VM, Gomez J, Sturm A, Zhang S, Fleming SJ, Warren S, Beechem J, Hung D, Babadi M, Padera RF, MacParland SA, Bader GD, Imad N, Solomon IH, Miller E, Riedel S, Porter CBM, Villani AC, Tsai LTY, Hide W, Szabo G, Hecht J, Rozenblatt-Rosen O, Shalek AK, Izar B, Regev A, Popov YV, Jiang ZG, Vlachos IS. A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients. Genome Biol 2025; 26:56. [PMID: 40087773 PMCID: PMC11907808 DOI: 10.1186/s13059-025-03499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/07/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The molecular underpinnings of organ dysfunction in severe COVID-19 and its potential long-term sequelae are under intense investigation. To shed light on these in the context of liver function, we perform single-nucleus RNA-seq and spatial transcriptomic profiling of livers from 17 COVID-19 decedents. RESULTS We identify hepatocytes positive for SARS-CoV-2 RNA with an expression phenotype resembling infected lung epithelial cells, and a central role in a pro-fibrotic TGFβ signaling cell-cell communications network. Integrated analysis and comparisons with healthy controls reveal extensive changes in the cellular composition and expression states in COVID-19 liver, providing the underpinning of hepatocellular injury, ductular reaction, pathologic vascular expansion, and fibrogenesis characteristic of COVID-19 cholangiopathy. We also observe Kupffer cell proliferation and erythrocyte progenitors for the first time in a human liver single-cell atlas. Despite the absence of a clinical acute liver injury phenotype, endothelial cell composition is dramatically impacted in COVID-19, concomitantly with extensive alterations and profibrogenic activation of reactive cholangiocytes and mesenchymal cells. CONCLUSIONS Our atlas provides novel insights into liver physiology and pathology in COVID-19 and forms a foundational resource for its investigation and understanding.
Collapse
Affiliation(s)
- Yered Pita-Juarez
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dimitra Karagkouni
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nikolaos Kalavros
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Spatial Technologies Unit, HMS Initiative for RNA Medicine / Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Johannes C Melms
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Sebastian Niezen
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Toni M Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam L Essene
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Deepti Pant
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Disha Skelton-Badlani
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Pourya Naderi
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Pinzhu Huang
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Liuliu Pan
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Tallulah S Andrews
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Carly G K Ziegler
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Program in Computational & Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Andriy Myloserdnyy
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rachel Chen
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Andy Nam
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Yan Liang
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Shanshan He
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Tushar Rane
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Amit Dipak Amin
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Jana Biermann
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Molly Veregge
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Zachary Kramer
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Christopher Jacobs
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Yusuf Yalcin
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Devan Phillips
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Michal Slyper
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | | | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zohar Bloom-Ackermann
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Victoria M Tran
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James Gomez
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexander Sturm
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuting Zhang
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephen J Fleming
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Deborah Hung
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Mehrtash Babadi
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sonya A MacParland
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, Toronto, ON, Canada
| | - Nasser Imad
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Isaac H Solomon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric Miller
- NanoString Technologies, Inc., Seattle, WA, USA
| | - Stefan Riedel
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Caroline B M Porter
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra-Chloé Villani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Linus T-Y Tsai
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Winston Hide
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Gyongyi Szabo
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jonathan Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Alex K Shalek
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA.
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Program in Computational & Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA.
- Columbia Center for Translational Immunology, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Program for Mathematical Genomics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - Yury V Popov
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Z Gordon Jiang
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Ioannis S Vlachos
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Spatial Technologies Unit, HMS Initiative for RNA Medicine / Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Harvard Medical School Initiative for RNA Medicine, Boston, MA, USA.
| |
Collapse
|
33
|
Chen J, Wang J, Zhao H, Tan X, Yan S, Zhang H, Wang T, Tang X. Molecular breeding of pigs in the genome editing era. Genet Sel Evol 2025; 57:12. [PMID: 40065264 PMCID: PMC11892312 DOI: 10.1186/s12711-025-00961-7] [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: 06/21/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To address the increasing demand for high-quality pork protein, it is essential to implement strategies that enhance diets and produce pigs with excellent production traits. Selective breeding and crossbreeding are the primary methods used for genetic improvement in modern agriculture. However, these methods face challenges due to long breeding cycles and the necessity for beneficial genetic variation associated with high-quality traits within the population. This limitation restricts the transfer of desirable alleles across different genera and species. This article systematically reviews past and current research advancements in porcine molecular breeding. It discusses the screening of clustered regularly interspaced short palindromic repeats (CRISPR) to identify resistance loci in swine and the challenges and future applications of genetically modified pigs. MAIN BODY The emergence of transgenic and gene editing technologies has prompted researchers to apply these methods to pig breeding. These advancements allow for alterations in the pig genome through various techniques, ranging from random integration into the genome to site-specific insertion and from target gene knockout (KO) to precise base and prime editing. As a result, numerous desirable traits, such as disease resistance, high meat yield, improved feed efficiency, reduced fat deposition, and lower environmental waste, can be achieved easily and effectively by genetic modification. These traits can serve as valuable resources to enhance swine breeding programmes. CONCLUSION In the era of genome editing, molecular breeding of pigs is critical to the future of agriculture. Long-term and multidomain analyses of genetically modified pigs by researchers, related policy development by regulatory agencies, and public awareness and acceptance of their safety are the keys to realizing the transition of genetically modified products from the laboratory to the market.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Haoran Zhao
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Xiao Tan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Shihan Yan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Tiefeng Wang
- College of Life Science, Baicheng Normal University, Baicheng, 137000, China.
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
34
|
Chen F, Zhao Z, Ren Z, Lu K, Yu Y, Wang W. Prediction of drug target interaction based on under sampling strategy and random forest algorithm. PLoS One 2025; 20:e0318420. [PMID: 40048461 PMCID: PMC11884685 DOI: 10.1371/journal.pone.0318420] [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: 05/27/2024] [Accepted: 01/16/2025] [Indexed: 03/09/2025] Open
Abstract
Drug target interactions (DTIs) play a crucial role in drug discovery and development. The prediction of DTIs based on computational method can effectively assist the experimental techniques for DTIs identification, which are time-consuming and expensive. However, the current computational models suffer from low accuracy and high false positive rate in the prediction of DTIs, especially for datasets with extremely unbalanced sample categories. To accurately identify the interaction between drugs and target proteins, a variety of descriptors that fully show the characteristic information of drugs and targets are extracted and applied to the integrated method random forest (RF) in this work. Here, the random projection method is adopted to reduce the feature dimension such that simplify the model calculation. In addition, to balance the number of samples in different categories, a down sampling method NearMiss (NM) which can control the number of samples is used. Based on the gold standard datasets (nuclear receptors, ion channel, GPCRs and enzymes), the proposed method achieves the auROC of 92.26%, 98.21%, 97.65%, 99.33%, respectively. The experimental results show that the proposed method yields significantly higher performance than that of state-of-the-art methods in predicting drug target interaction.
Collapse
Affiliation(s)
- Feng Chen
- School of Advanced Manufacturing Engineering, Hefei University, Hefei, China
| | - Zhigang Zhao
- School of Electrical and Information Engineering, Anhui University of Technology, Ma’anshan, Anhui, China
| | - Zheng Ren
- School of Electrical and Information Engineering, Anhui University of Technology, Ma’anshan, Anhui, China
| | - Kun Lu
- School of Electrical and Information Engineering, Anhui University of Technology, Ma’anshan, Anhui, China
| | - Yang Yu
- School of Advanced Manufacturing Engineering, Hefei University, Hefei, China
| | - Wenyan Wang
- School of Electrical and Information Engineering, Anhui University of Technology, Ma’anshan, Anhui, China
- Wuhu Technology and Innovation Research Institute, AHUT, Wuhu, China
| |
Collapse
|
35
|
Zhuang L, Zhao Y, Shen J, Sun L, Hao P, Yang J, Zhang Y, Shen Q. Advances in porcine epidemic diarrhea virus research: genome, epidemiology, vaccines, and detection methods. DISCOVER NANO 2025; 20:48. [PMID: 40029472 DOI: 10.1186/s11671-025-04220-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Porcine epidemic diarrhea (PED) is a highly contagious intestinal disease caused by the porcine epidemic diarrhea virus (PEDV). The economic impact of PEDV on the global pig industry has been significant, resulting in considerable losses. This paper presents a review of the latest research progress on PEDV genome, molecular epidemiology, vaccine development, and molecular detection methods. It was determined that the genetic diversity of the PEDV spike (S) gene was closely associated with the epidemiological trend of PEDV. The prevalence of S gene variants of different genotypes exhibited variability across regions and pig populations. Epidemiological analyses have demonstrated that PEDV can be transmitted via multiple routes, including direct contact, airborne aerosol, and water source contamination. With regard to vaccine research, the available vaccines can be classified into several categories, including live-attenuated vaccines, inactivated vaccines, subunit vaccines, bacterial vector vaccines, viral vector vaccines, mRNA vaccines, etc. Each of these has distinctive characteristics in terms of immunogenicity, protection efficiency, and safety. Molecular detection methods, including PCR-based methods, isothermal amplification techniques, immunological assays, and biosensors, play an important role in the diagnosis and monitoring of PEDV. Furthermore, this paper examines the current developments in PEDV research and identifies the key areas of future investigation. The objective of this paper is to establish a theoretical foundation for the prevention and control strategies of PED, and to provide a point of reference for further research on the genomics, epidemiology, vaccine development and detection methods of PEDV.
Collapse
Affiliation(s)
- Linlin Zhuang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China
| | - Jingyi Shen
- School of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 210038, People's Republic of China
| | - Li Sun
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Pan Hao
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Jianbo Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 211102, People's Republic of China.
| | - Qiuping Shen
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China.
| |
Collapse
|
36
|
Schuh AJ, Amman BR, Guito JC, Graziano JC, Sealy TK, Towner JS. Modeling natural coinfection in a bat reservoir shows modulation of Marburg virus shedding and spillover potential. PLoS Pathog 2025; 21:e1012901. [PMID: 40096181 PMCID: PMC11978059 DOI: 10.1371/journal.ppat.1012901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/08/2025] [Accepted: 01/12/2025] [Indexed: 03/19/2025] Open
Abstract
The Egyptian rousette bat (ERB) is a natural reservoir for Marburg virus (MARV; family Filoviridae), a putative reservoir for Sosuga virus (SOSV; family Paramyxoviridae), and a vertebrate reservoir for Kasokero virus (KASV; family Orthonairoviridae); however, the effect of naturally occurring coinfection by those viruses on MARV shedding and spillover potential is unknown. To answer this question, we experimentally infected one cohort of captive-bred ERBs with SOSV+MARV (n=12 bats) or MARV only (n=12 bats) and a second cohort with KASV+MARV (n=12 bats) or MARV only (n=12 bats), and then collected blood, oral swab, and rectal swab specimens throughout the course of infection to monitor viral shedding. Compared to the MARV-monoinfected bat group, the SOSV+MARV-coinfected bat group exhibited a significantly shortened duration of MARV oral shedding and a significantly decreased anti-MARV IgG response, which may increase the capacity for MARV reinfection. In contrast, relative to the MARV-monoinfected bat group, the KASV+MARV-coinfected bat group exhibited significantly increased peak magnitudes and durations of MARV viremia and oral shedding, as well as a significantly increased anti-MARV IgG response. Correspondingly, cumulative MARV shedding loads, a measure of infectiousness, were significantly higher in the KASV+MARV-coinfected bat group than the MARV-monoinfected bat group. Four of the KASV+MARV-coinfected bats were classified as MARV supershedders, together accounting for 72.5% of the KASV-MARV experimental cohort's total shedding. Our results demonstrate that SOSV+MARV and KASV+MARV coinfection of ERBs differentially modulates MARV shedding and anti-MARV IgG responses, thereby implicating MARV coinfection as playing a critical role in bat-to-bat MARV transmission dynamics and spillover potential.
Collapse
Affiliation(s)
- Amy J. Schuh
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- United States Public Health Service Commissioned Corps, Rockville, Maryland, United States of America
| | - Brian R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jonathan C. Guito
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James C. Graziano
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Tara K. Sealy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jonathan S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
37
|
Foglia EA, Mioulet V, Cavalera S, Baguisi J, Turgut Sİ, Sangula A, Khan S, Jamal SM, Bull H, Rosati S, Nogarol C, Pezzoni G, Bulut A, King DP, Anfossi L, Rosso F, Brocchi E, Grazioli S. Validation of two multiplex lateral flow devices for the rapid detection and typing of foot-and-mouth disease viruses. Res Vet Sci 2025; 185:105558. [PMID: 39904118 DOI: 10.1016/j.rvsc.2025.105558] [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: 10/11/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Lateral Flow Devices (LFDs) represent a simple tool for the rapid diagnosis of FMD, particularly in endemic regions, often lacking adequate and equipped laboratories. Other experimental prototypes offer a user-friendly FMD confirmation tool, but serotype identification is crucial for transmission patterns evaluation and for vaccine selection and their serotyping capacity was limited. This study describes the validation of two multiplex devices based on well-characterized monoclonal antibodies: LFD1 (O, A and Asia1) and LFD2 (SAT1 and SAT2). First, the ability of the LFDs to recognize the widest spectrum of strains was assessed. Eighty-three tissue-cultured FMDV strains were tested with LFD1 and 15 with LFD2, confirming the capability to recognize and serotype a broad range of lineages and sub-lineages belonging to all 7 endemic pools. The diagnostic sensitivity related to FMDV detection was evaluated using 224 real-time RT-PCR positive samples collected in endemic countries, resulting in 87.5 % for LFD1 (140/160 detected) and 70 % for LFD2 (45/64 detected). The diagnostic sensitivity of serotyping capabilities was assessed by testing field samples in parallel with an antigen-ELISA kit: applied on 178 samples, LFD1 correctly typed 96 %, 94 % and 83 % of samples positive for type O (n = 95), A (n = 54) and Asia1 (n = 29) respectively, while LFD2 correctly typed 77 % SAT1 (n = 52) and 82 % SAT2 (n = 40) in 92 positive field samples. A 100 % diagnostic specificity was found testing 60 epithelium homogenates collected from animals negative for FMD. Obtained data highlighted the devices' potential value for field use to support the enhanced surveillance of FMD.
Collapse
Affiliation(s)
- Efrem Alessandro Foglia
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Brescia, Italy.
| | - Valerie Mioulet
- FAO World Reference Laboratory for Foot-and-Mouth Disease, The Pirbright Institute, Woking, United Kingdom
| | | | - Jozhel Baguisi
- FAO World Reference Laboratory for Foot-and-Mouth Disease, The Pirbright Institute, Woking, United Kingdom
| | | | - Abraham Sangula
- Foot-and-mouth Disease National Reference Laboratory, Embakasi, Nairobi, Kenya
| | - Salman Khan
- Department of Biotechnology, University of Malakand, Dir Lower, Pakistan
| | | | - Harry Bull
- FAO World Reference Laboratory for Foot-and-Mouth Disease, The Pirbright Institute, Woking, United Kingdom
| | - Sergio Rosati
- Department of Veterinary Science, University of Turin, Grugliasco, Torino, Italy
| | | | - Giulia Pezzoni
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Brescia, Italy
| | | | - Donald P King
- FAO World Reference Laboratory for Foot-and-Mouth Disease, The Pirbright Institute, Woking, United Kingdom
| | - Laura Anfossi
- Department of Chemistry, University of Turin, Torino, Italy
| | - Fabrizio Rosso
- European Commission for the control of Foot-and-Mouth Disease (EuFMD), FAO, Rome, Italy
| | - Emiliana Brocchi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Brescia, Italy
| | - Santina Grazioli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Brescia, Italy
| |
Collapse
|
38
|
Albert E, Biksi I, Laczkó L, Miló L, Cseri K, Bőkényné Tóth R, Papp D, Halmay D, Bódai E, Bakos Z. Streptococcus hillyeri isolated from septic pleuritis in a horse. J Vet Diagn Invest 2025; 37:393-397. [PMID: 39773117 PMCID: PMC11707762 DOI: 10.1177/10406387241306724] [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] [Indexed: 01/11/2025] Open
Abstract
Here we report the isolation of Streptococcus hillyeri from a thoracic sample from a horse. A 17-y-old Hungarian Sport Horse mare was referred to the equine clinic of the University of Veterinary Medicine Budapest, Hungary, with suspected pleuritis. Upon arrival, the horse was febrile and had tachycardia, severe inspiratory dyspnea, and tachypnea. Thoracic ultrasonography revealed severe bilateral pleural effusion, and a large area of lung consolidation. After sampling of both hemithoraces, 66 L of turbid exudate were drained. Based on these findings, a tentative diagnosis of septic pleuritis was made, and the horse was immediately started on a course of broad-spectrum antibiotics, a NSAID, an anticoagulant, and intravenous fluids. Despite intensive care, the clinical parameters deteriorated, and the horse was euthanized 6 d later. Cytology confirmed septic pleuritis, with short chains or groups of coccoid bacteria. Anaerobic culture yielded gram-positive cocci from both hemithoraces in almost pure culture, which we identified as S. hillyeri by 16S rDNA and whole-genome analysis. Additionally, we identified 4 previously unassigned Streptococcus sp. sequences as S. hillyeri. Of these, 3 were obtained from aborted equine fetuses and a fourth from a donkey mastitis case, supporting the pathogenic nature of S. hillyeri in these host species.
Collapse
Affiliation(s)
- Ervin Albert
- Departments of Pathology, University of Veterinary Medicine Budapest, Üllő, Hungary
- Institute of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - Imre Biksi
- Departments of Pathology, University of Veterinary Medicine Budapest, Üllő, Hungary
| | - Levente Laczkó
- One Health Institute, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Conservation Biology Research Group, University of Debrecen, Debrecen, Hungary
| | - László Miló
- One Health Institute, University of Debrecen, Debrecen, Hungary
| | - Karolina Cseri
- One Health Institute, University of Debrecen, Debrecen, Hungary
| | | | - Dalma Papp
- One Health Institute, University of Debrecen, Debrecen, Hungary
| | | | - Emese Bódai
- Clinic of Equine Medicine, University of Veterinary Medicine Budapest, Üllő, Hungary
| | - Zoltán Bakos
- Clinic of Equine Medicine, University of Veterinary Medicine Budapest, Üllő, Hungary
| |
Collapse
|
39
|
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
|
40
|
Gaffney A, Smyth EG, Moore Z, Patton D, Connor TO, Derwin R. Role of admission rapid antigen testing (RATs) for COVID-19 on patients transferred from acute hospitals to a postacute rehabilitation setting. Am J Infect Control 2025; 53:357-360. [PMID: 39489423 DOI: 10.1016/j.ajic.2024.10.031] [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: 08/07/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Rapid antigen tests (RATs) are suitable for point-of-care testing, require no laboratory time, and give immediate results. However, are RATs useful for detecting asymptomatic COVID-19 infection when compared with polymerase chain reaction (PCR) testing in health care settings? METHODS RAT testing was carried out on all new admissions without a history of confirmed COVID-19 infection within 3 months of admission. PCR testing was carried out on all patients with a positive RAT for confirmation purposes. The cycle threshold values of COVID-19-detected results on PCR testing were examined to determine the utility of the RATs. RESULTS A total of 1,403 patients were transferred to the postacute rehabilitation unit from January to December 2023. The results of the study revealed an 85% accuracy of RATs with a 15% rate of false-negative results at the time of admission. All patients who had a positive RAT at the time of admission also had a positive PCR test. CONCLUSIONS This testing algorithm resulted in early detection and prompt isolation of positive cases reducing the likely spread of COVID-19 infection, hospital outbreaks, and bed/ward closures.
Collapse
Affiliation(s)
- Ann Gaffney
- Clontarf Hospital, Post Acute Rehabilitation, Blackheath Park, Clontarf, Dublin 3, Ireland.
| | - Edmond G Smyth
- Clontarf Hospital, Post Acute Rehabilitation, Blackheath Park, Clontarf, Dublin 3, Ireland
| | - Zena Moore
- School of Nursing and Midwifery, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Nursing, Lida Institute, Shanghai, China; School of Nursing & Midwifery, Griffith University, Brisbane, Queensland, Australia; School of Health Sciences, Faculty of Life and Health Sciences Ulster University, Northern Ireland; School of Nursing, Cardiff University, Cardiff, Wales, UK; Department of Nursing, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia; Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Declan Patton
- School of Nursing and Midwifery, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Nursing, Fakeeh College of Health Sciences, Jeddah, Saudi Arabia; Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia
| | - Tom O' Connor
- School of Nursing and Midwifery, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Nursing, Fakeeh College of Health Sciences, Jeddah, Saudi Arabia
| | - Rosemarie Derwin
- School of Nursing, Cardiff University, Cardiff, Wales, UK; School of Nursing and Midwifery, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
41
|
Nawaz MS, Nawaz MZ, Gong Y, Fournier-Viger P, Diallo AB. In silico framework for genome analysis. FUTURE GENERATION COMPUTER SYSTEMS 2025; 164:107585. [DOI: 10.1016/j.future.2024.107585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Yim-Im W, Anderson TK, Böhmer J, Baliellas J, Stadejek T, Gauger PC, Krueger KM, Vermeulen CJ, Buter R, Kazlouski A, An T, Zhang J. Refining genetic classification of global porcine reproductive and respiratory syndrome virus type 1 (PRRSV-1) and investigating their geographic and temporal distributions. Vet Microbiol 2025; 302:110413. [PMID: 39904077 DOI: 10.1016/j.vetmic.2025.110413] [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: 11/23/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Porcine reproductive and respiratory syndrome virus type 1 (PRRSV-1) primarily circulates in Europe but is also detected in North America and Asia. Based on ORF5 sequences, previous studies classified PRRSV-1 into four subtypes. Subtype 1 was further classified into 12 clades (A-L) or into three lineages with lineage 1 including clades 1A-1G and lineage 3 including clades 3A-3G, but the systems are inconsistent and have not been adopted. In this study, we proposed a statistically supported PRRSV-1 genetic classification system based on 10,446 global PRRSV-1 ORF5 sequences spanning 1991-2023. We replaced the colloquial "subtype" designation with "lineage" to reflect evolutionary history and, subsequently, PRRSV-1 was classified into four lineages (L1-L4) with L1 including 18 sublineages (L1.1 to L1.18). The proposed classification system is flexible and may be amended if additional lineages, sublineages, or more granular classifications are needed to reflect contemporary PRRSV-1 detections and evolution. Geographic distributions of PRRSV-1 at lineage and sublineage levels were distinct, with L1 globally distributed and L2, L3 and L4 more restricted. Temporal dynamic changes in some countries were quantified. Classification and ORF5 nucleotide identity of six commercial PRRSV-1 vaccines to each lineage and sublineage and detection frequency of vaccine-like viruses were determined. The phylogenies based on whole-genome and ORF5 sequences demonstrated slightly different tree topologies. Recombination of PRRSV-1 was observed at within-sublineage and between-sublineage levels. A set of ORF5 reference sequences representing the refined classification is available for future diagnostic and epidemiological applications. This study provides a benchmark delineating the current genetic diversity of PRRSV-1 and introduces a refined classification system to support the global standardization and application of ORF5-based genetic classification for PRRSV-1.
Collapse
Affiliation(s)
- Wannarat Yim-Im
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, Ames, IA, United States
| | - Jan Böhmer
- IVD Gesellschaft für Innovative Veterinaerdiagnostik mbH, Seelzer-Letter, Germany
| | | | - Tomasz Stadejek
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Phillip C Gauger
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Karen M Krueger
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | | | - Rianne Buter
- Royal GD (GD Animal Health), P.O. Box 9, Deventer 7400 AA, the Netherlands
| | | | - Tongqing An
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Science, Harbin, China
| | - Jianqiang Zhang
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.
| |
Collapse
|
43
|
Woodbridge Y, Mandel M, Goldberg Y, Huppert A. Estimating Mean Viral Load Trajectory From Intermittent Longitudinal Data and Unknown Time Origins. Stat Med 2025; 44:e70033. [PMID: 39995297 PMCID: PMC11851093 DOI: 10.1002/sim.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
Viral load (VL) in the respiratory tract is the leading proxy for assessing infectiousness potential. Understanding the dynamics of disease-related VL within the host is of great importance, as it helps to determine different policies and health recommendations. However, normally the VL is measured on individuals only once, in order to confirm infection, and furthermore, the infection date is unknown. It is therefore necessary to develop statistical approaches to estimate the typical VL trajectory. We show here that, under plausible parametric assumptions, two measures of VL on infected individuals can be used to accurately estimate the VL mean function. Specifically, we consider a discrete-time likelihood-based approach to modeling and estimating partial observed longitudinal samples. We study a multivariate normal model for a function of the VL that accounts for possible correlation between measurements within individuals. We derive an expectation-maximization (EM) algorithm which treats the unknown time origins and the missing measurements as latent variables. Our main motivation is the reconstruction of the daily mean VL, given measurements on patients whose VLs were measured multiple times on different days. Such data should and can be obtained at the beginning of a pandemic with the specific goal of estimating the VL dynamics. For demonstration purposes, the method is applied to SARS-Cov-2 cycle-threshold-value data collected in Israel.
Collapse
Affiliation(s)
- Yonatan Woodbridge
- The Gertner Institute for Epidemiology & Health Policy Research, Sheba Medical CenterRamat GanIsrael
- Department of Computer ScienceHolon Institute of TechnologyHolonIsrael
| | - Micha Mandel
- Department of Statistics and Data ScienceThe Hebrew University of JerusalemJerusalemIsrael
| | - Yair Goldberg
- Faculty of Industrial Engineering and ManagementTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| | - Amit Huppert
- The Gertner Institute for Epidemiology & Health Policy Research, Sheba Medical CenterRamat GanIsrael
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
| |
Collapse
|
44
|
Didkowska A, Martín-Santander V, Wojciechowska M, Olech W, Anusz K, Fernández A, Davies JE, Gómez Á, Peña-Fresneda N, Arias M, Lacasta D, Ortín A, Pérez MD, Villanueva-Saz S, Marteles D. Presence of anti-SARS-CoV-2 antibodies in European bison (Bison bonasus) in Poland, 2019-2023. BMC Vet Res 2025; 21:120. [PMID: 40022124 PMCID: PMC11869555 DOI: 10.1186/s12917-025-04593-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND The origin of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains unknown. However, it is likely that the virus spillover occurred from an animal reservoir to humans. Identifying animal species susceptible to SARS-CoV-2 is crucial for understanding cross-species transmission to humans. This study distinguishes itself by focusing on the susceptibility of the European bison (Bison bonasus), an endangered species, to SARS-CoV-2. The objective of this study was to investigate the occurrence of SARS-CoV-2 antibodies in a substantial number (n = 238) of both free-living and captive Polish European bison using an in-house ELISA method and virus neutralization test (VNT). RESULTS The seroprevalence of SARS-CoV-2 infection was found to be 1.29% (3/232). None of the seropositive European bison tested positive in the virus neutralization test. All seropositive animals were part of captive herds. CONCLUSIONS This study represents the first report of SARS-CoV-2 seroprevalence in both free-ranging and captive European bison in Poland. Based on these findings, the European bison appears to be a less susceptible species to SARS-CoV-2. The most probable route of transmission was from humans to European bison, as all seropositive animals belonged to captive herds with contact with indirect human sources, such as tourists and keepers.
Collapse
Affiliation(s)
- Anna Didkowska
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 166, Warsaw, 02-787, Poland.
| | - Víctor Martín-Santander
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| | - Marlena Wojciechowska
- Department of Animal Genetics and Conservation, Warsaw University of Life Sciences (SGGW), Ciszewskiego 8, Warsaw, 02-786, Poland
| | - Wanda Olech
- Department of Animal Genetics and Conservation, Warsaw University of Life Sciences (SGGW), Ciszewskiego 8, Warsaw, 02-786, Poland
| | - Krzysztof Anusz
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 166, Warsaw, 02-787, Poland
| | - Antonio Fernández
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain
| | - Janine E Davies
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| | - Álex Gómez
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain
| | | | - Maykel Arias
- Aragon Health Research Institute (IIS Aragón), Zaragoza, 50009, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Delia Lacasta
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain
| | - Aurora Ortín
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain
| | - María Dolores Pérez
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain
- Department of Animal Production and Sciences of the Food, Veterinary Faculty, University of Zaragoza, Zaragoza, Spain
| | - Sergio Villanueva-Saz
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain.
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain.
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, 50013, Spain.
| | - Diana Marteles
- Department of Animal Pathology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
- Clinical Immunology Laboratory, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| |
Collapse
|
45
|
Barzin Tond S, Abolghasemi S, Khatami SH, Ehtiati S, Zarei T, Shateri S, Mahmoodi Baram S, Yarahmadi S, Fallah S, Salmani F, Shahmohammadi MR, Khajavirad N, Tafakhori A, Riazi A, Karima S. Boswellic Acids Reduce Systemic Inflammation in Patients with Moderate COVID-19 Through Modulation of NF-κB Pathway. J Diet Suppl 2025; 22:365-381. [PMID: 40012185 DOI: 10.1080/19390211.2025.2468484] [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: 02/28/2025]
Abstract
Prevention and/or management of the dysregulated immune response in patients with COVID-19 is expected to help in the treatment of COVID-19. Boswellic acids (BAs) have great therapeutic potential because they have anti-inflammatory and immunomodulatory effects. Here, we aimed to investigate the mechanism of action of a BA formulation, Inflawell syrup, which was previously shown to be effective in reducing disease symptoms in patients who suffer from mild to moderate COVID-19. Patients with mild to moderate COVID-19 were treated with either Inflawell containing boswellic acids or a placebo for 14 days. The serum levels of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-8, IL-1α, IL-17, IL-1Ra, and Monocyte Chemoattractant Protein-1 (MCP-1), were measured both at study onset and on day 14 after treatment started. In addition, to further investigate the signaling pathway(s) underlying the changes in cytokine levels, we evaluated the expression of tumor necrosis factor receptor 1 (TNFR1), tumor necrosis factor receptor 2 (TNFR2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 mRNAs and phospho-inhibitor of nuclear factor kappa B (IκB) and IκB proteins. In our study, a significant decrease in the serum levels of IL-1α (p < .009), IL-8 (p < .04), TNF-α (p < .0001), and MCP-1 (p < .007) was detected in patients treated with Inflawell. Additionally, our data revealed a decrease in phospho-IκB protein levels (p < .02) and NF-κB p65 mRNA levels (p < .002), whereas the amount of IκB protein (p < .01) in the Inflawell group was significantly greater than that in the placebo group. Furthermore, despite the decreasing trend in the expression of TNFR1 and TNFR2 in the Inflawell group, there was no statistically significant difference compared with that in the placebo group. In general, treatment with Inflawell syrup led to a lower level of proinflammatory cytokines and a decrease in the activity of the TNF-α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Sepideh Barzin Tond
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Sara Abolghasemi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medial Sciences (SBMU), Tehran, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayebe Zarei
- Clinical Trial Department, Behbalin Co., Ltd, Tehran, Iran
| | - Somayyeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | | | - Sahar Yarahmadi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Fallah
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Nasim Khajavirad
- Internal Medicine Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| |
Collapse
|
46
|
Wang Y, Xia B, Gao Z. A comprehensive review of current insights into the virulence factors of SARS-CoV-2. J Virol 2025; 99:e0204924. [PMID: 39878471 PMCID: PMC11852741 DOI: 10.1128/jvi.02049-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
The evolution of SARS-CoV-2 pathogenicity has been a major focus of attention. However, the determinants of pathogenicity are still unclear. Various hypotheses have attempted to elucidate the mechanisms underlying the evolution of viral pathogenicity, but a definitive conclusion has yet to be reached. Here, we review the potential impact of all proteins in SARS-CoV-2 on the viral pathogenic process and analyze the effects of their mutations on pathogenicity evolution. We aim to summarize which virus-encoded proteins are crucial in influencing viral pathogenicity, defined as disease severity following infection. Mutations in these key proteins, which are the virulence factors in SARS-CoV-2, may be the driving forces behind the evolution of viral pathogenicity. Mutations in the S protein can impact viral entry and fusogenicity. Mutations in proteins such as NSP2, NSP5, NSP14, and ORF7a can alter the virus's ability to suppress host protein synthesis and innate immunity. Mutations in NSP3, NSP4, NSP6, N protein, NSP5, and NSP12 may alter viral replication efficiency. The combined effects of mutations in the S protein and NSP6 can significantly reduce viral replication. In addition, various viral proteins, including ORF3a, ORF8, NSP4, Spike protein, N protein, and E protein, directly participate in the inflammatory process. Mutations in these proteins can modulate the levels of inflammation following infection. Collectively, these viral protein mutations can influence SARS-CoV-2 pathogenicity by impacting viral immune evasion, replication capacity, and the level of inflammation mediated by infection. In conclusion, the evolution of SARS-CoV-2 pathogenicity is likely determined by multiple virulence factors.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bingqing Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
47
|
Chen R, Zhou G, Yang J, Yuan R, Sun Y, Liang Y, Wu R, Wen Y, Wang Y, Zhao Q, Du S, Yan Q, Cao S, Huang X. A novel neutralizing antibody recognizing a conserved conformational epitope in PDCoV S1 protein and its therapeutic efficacy in piglets. J Virol 2025; 99:e0202524. [PMID: 39840987 PMCID: PMC11853068 DOI: 10.1128/jvi.02025-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an enteric pathogen that burdens the global pig industry and is a public health concern. The development of effective antiviral therapies is necessary for the prevention and control of PDCoV, yet to date, there are few studies on the therapeutic potential of PDCoV-neutralizing antibodies. Here, we investigate the therapeutic potential of a novel monoclonal antibody (mAb 4A6) which targets the PDCoV S1 protein and effectively neutralizes PDCoV, both pre- and post-attachment on cells, with IC50 values of 0.537 and 8.487 µg/mL, respectively. A phage-display peptide library was used to determine the epitope recognized by mAb 4A6, and two mimotopes, QYPVSYA (P1) and FPHWPTI (P2), were identified. KLH-P1 reacted with PDCoV-positive sera but failed to induce PDCoV-specific IgG and neutralizing antibodies in mice, suggesting P1 does not fully mimic the conformational epitope. Molecular docking and alanine scanning mutagenesis revealed that S461, P462, T463, E465, and Y467 on the S protein are essential for mAb 4A6 binding. Antibody therapy experiments in PDCoV-infected piglets showed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Taken together, our findings demonstrate that mAb 4A6 holds promise as a treatment against PDCoV, and the amino acids recognized by mAb 4A6 will be valuable for developing novel epitope-based vaccines or antiviral drugs. IMPORTANCE Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that poses a potential threat to public health. Developing effective antiviral therapies is crucial for its prevention and control. Here, we demonstrated that mAb 4A6 shows promise as a treatment against PDCoV. Antibody therapy experiments conducted on PDCoV-infected piglets revealed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Furthermore, we characterized the conformational epitope (S461, P462, T463, E465, and Y467) recognized by mAb 4A6 through an integrated approach involving phage display peptide library, molecular docking, and alanine scanning mutagenesis. More importantly, mAb 4A6 exhibits a broad-spectrum neutralizing activity against different PDCoV strains. These findings indicate that mAb 4A6 has promising therapeutic value for PDCoV-infected piglets, and the identification of mAb 4A6 recognized epitope may provide a new idea for the identification of conformational epitopes.
Collapse
Affiliation(s)
- Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guiping Zhou
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junpeng Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rong Yuan
- Chengdu Livestock and Poultry Genetic Resources Protection Center, Chengdu, China
| | - Ying Sun
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yixiao Liang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| |
Collapse
|
48
|
Gallichotte EN, Bashor L, Erbeck K, Croft L, Stache K, Long J, VandeWoude S, Johnson JC, Pabilonia KL, Ebel GD. SARS-CoV-2 outbreak in lions, tigers, and hyenas at Denver Zoo. mSphere 2025; 10:e0098924. [PMID: 39912638 PMCID: PMC11853051 DOI: 10.1128/msphere.00989-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
In late 2019, SARS-CoV-2 spilled over from an animal host into humans, where it efficiently spread, resulting in the COVID-19 pandemic. Through both natural and experimental infections, we learned that many animal species are susceptible to SARS-CoV-2. Importantly, animals in close proximity to humans, including companion, farmed, and those at zoos and aquariums, became infected, and many studies demonstrated transmission to/from humans in these settings. In this study, we first review the literature of SARS-CoV-2 infections in tigers and lions and compare species, sex, age, virus and antibody detection assay, and types, frequency, and length of clinical signs, demonstrating broad heterogeneity among infections. We then describe a SARS-CoV-2 outbreak in lions, tigers, and hyenas at Denver Zoo in late 2021. Animals were tested for viral RNA (vRNA) for 4 months. Lions had significantly more vRNA in nasal swabs than both tigers and hyenas, and many individual lions experienced viral recrudescence after weeks of undetectable vRNA. Infectious virus was correlated with high levels of vRNA and was more likely to be detected earlier during infection. Four months post-infection, all tested animals generated robust neutralizing antibody titers. Animals were infected with Delta lineage AY.20 identical to a variant circulating at less than 1% in Colorado humans at that time, suggesting a single spillover event from an infected human spread within and between species housed at the zoo. Better understanding of epidemiology and susceptibility of SARS-CoV-2 infections in animals is critical to limit the current and future spread and protect animal and human health.IMPORTANCESurveillance and experimental testing have shown many animal species, including companion, wildlife, and conservatory, are susceptible to SARS-CoV-2. Early in the COVID-19 pandemic, big cats at zoological institutions were among the first documented cases of naturally infected animals; however, challenges in the ability to collect longitudinal samples in zoo animals have limited our understanding of SARS-CoV-2 kinetics and clearance in these settings. We measured SARS-CoV-2 infections over 4 months in lions, tigers, and hyenas at Denver Zoo and detected viral RNA, infectious virus, neutralizing antibodies, and recrudescence after initial clearance. We found lions had longer and higher levels of virus compared to the other species. All animals were infected by a rare viral lineage circulating in the human population, suggesting a single spillover followed by interspecies transmission. These data are important in better understanding natural SARS-CoV-2 spillover, spread, and infection kinetics within multiple species of zoo animals.
Collapse
Affiliation(s)
- Emily N. Gallichotte
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Laura Bashor
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Katelyn Erbeck
- Veterinary Diagnostic Laboratories, Colorado State University, Colorado, Fort Collins, USA
| | | | | | | | - Sue VandeWoude
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | | | - Kristy L. Pabilonia
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
- Veterinary Diagnostic Laboratories, Colorado State University, Colorado, Fort Collins, USA
| | - Gregory D. Ebel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
49
|
Fernandes-Santos RC, Warren K, Vaughan-Higgins R, Micheletti T, Bruce M. Disease dynamics and mortality risk in tapirs (Perissodactyla: Tapiridae) through a systematic literature review: Implications for preventive medicine and conservation. Prev Vet Med 2025; 239:106470. [PMID: 40010001 DOI: 10.1016/j.prevetmed.2025.106470] [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: 10/24/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
The impact of diseases on tapir mortality and potential implications for preventive medicine and conservation remain unclear. A systematic literature review was conducted using seven databases and grey literature to address these gaps. The PRISMA statement was adopted to report results, and boosted regression tree models were employed for data analysis. After screening 5323 records and removing duplicates, the title and abstract of 2484 records were assessed. Out of 502 eligible studies, only 206 met all inclusion criteria. These were published between 1924 and 2023 in ten languages, comprising mainly case reports (45.1 %) and cross-sectional studies (41.3 %). Infectious diseases were found in 72.9 % of the reports, and 27.2 % presented clinical signs. The most affected systems were gastroenteric (22.7 %), integumentary (22.1 %), and respiratory (17.5 %). Respiratory diseases were associated with increased mortality. Factors affecting tapir mortality included species (relative influence 41.5 %), followed by geographic location (23.5 %) and captivity (16.8 %). Clinical signs were the least important variable (4 %). While infectious diseases were associated with higher mortality risk, tapirs were more likely to become ill from non-infectious than infectious diseases. Captive individuals were also more likely to present with illness than their wild counterparts. When considering external causes, vehicle collisions represented the most significant cause of death (52.2 %), followed by hunting (38.2 %). Diseases (8.7 %) were the third most important, with bacterial infections the leading cause of death. This review represents the most comprehensive overview on tapir health to date and provides novel ways to collate epidemiological data from disparate study designs.
Collapse
Affiliation(s)
- Renata Carolina Fernandes-Santos
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia; Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia.
| | - Kristin Warren
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia; Centre for Terrestrial Ecosystem Science and Sustainability, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia.
| | - Rebecca Vaughan-Higgins
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia; Centre for Terrestrial Ecosystem Science and Sustainability, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia; Perth Zoo, South Perth, WA 6151, Australia.
| | - Tatiane Micheletti
- Faculty of Forestry, University of British Columbia, Vancouver, BC V6T 1Z2, Canada; Institute of Forest Growth and Forest Computer Sciences, TU Dresden, Dresden 01062, Germany.
| | - Mieghan Bruce
- School of Veterinary Medicine, College of Environmental and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia; Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Murdoch, WA 6150, Australia.
| |
Collapse
|
50
|
Li J, Zhang Y, Liu J, Xu S, Gao X, Li X, DanBaZhaXi, Zhao Q, Zhou EM, Chen Y, Liu B. Identification and pathogenicity of avian hepatitis E virus from quail. BMC Vet Res 2025; 21:79. [PMID: 39972467 PMCID: PMC11837710 DOI: 10.1186/s12917-025-04531-3] [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: 03/16/2023] [Accepted: 01/29/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Avian hepatitis E virus (HEV) has caused economic losses in the poultry industry and has shown a broad spectrum of infections. In 2022, a quail farm (YangLing, China) exhibited a decrease in egg production, an increase in mortality and hepatosplenomegaly. These characteristics were similar to those of avian HEV infection. To determine whether avian HEV existed on this farm and further clarify the pathogenicity caused by avian HEV under experimental conditions, the livers and spleens were collected from the diseased quails in the field for gross lesion observation and avian HEV detection; then, the pathogenicity was characterized. RESULTS In the field, the results showed enlargement of the liver and spleen and hemorrhage spots on the liver, and the amplified fragment (330-bp length) of HEV shared 100% identity with the Chinese avian HEV strain. The pathogenicity of this virus in quail was characterized by decreased egg production, seroconversion, viremia, fecal virus shedding, liver lesions and HEV antigen in the liver under experimental conditions. These differences indicated that there may be other pathogens or factors causing this disease together on the quail farm in addition to avian HEV, and further detection should be performed. CONCLUSIONS Overall, this is the first study to detect HEV RNA in quails, and an avian HEV strain can successfully infect quails under experimental conditions.
Collapse
Affiliation(s)
- Jinyao Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jingyu Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shixuan Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xueyan Gao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinru Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - DanBaZhaXi
- General Station of Animal Husbandry and Veterinary Technology Promotion, Naqu, Tibet, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yiyang Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Baoyuan Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|